WO2022155212A1 - Cell-fusion based immune agents - Google Patents

Cell-fusion based immune agents Download PDF

Info

Publication number
WO2022155212A1
WO2022155212A1 PCT/US2022/012136 US2022012136W WO2022155212A1 WO 2022155212 A1 WO2022155212 A1 WO 2022155212A1 US 2022012136 W US2022012136 W US 2022012136W WO 2022155212 A1 WO2022155212 A1 WO 2022155212A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cancer
protein
biological cell
cells
Prior art date
Application number
PCT/US2022/012136
Other languages
French (fr)
Inventor
Eric Paul Dixon
Jeff T. Hutchins
Original Assignee
Heat Biologics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Heat Biologics, Inc. filed Critical Heat Biologics, Inc.
Publication of WO2022155212A1 publication Critical patent/WO2022155212A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector

Definitions

  • the present disclosure relates to, in part, cell-fusion based immune agents, and methods of making thereof, that are useful for treatment of cancer and infectious diseases.
  • Immunotherapy is defined as a therapeutic approach that targets or manipulates the immune system. See Papaioannou et al., Ann. Transl. Med. 36 199-201. 10.21037/atm.2016.04.01. The goal of immunotherapy is to harness the host's adaptive and innate immune response to cause sustained elimination of diseased cells. See Naran et al., Front Microbiol. 2018;9:3158. Published 2018 Dec 21. doi:10.3389/fmicb.2018.03158.
  • infectious pathogens create a hospitable environment within the host and modulate host metabolic functions to support their nutritional requirements, while suppressing host defenses by altering host's regulatory mechanisms. See Naran et al. (2016).
  • targeted immunotherapy-based therapeutic approaches to infectious diseases are being developed, including those that enhance antigen-specific B cell antibody and T cell responses.
  • therapies are still in infancy, and much work remains to be done to expand immunotherapy approaches to treatment of infectious diseases.
  • the present disclosure provides, in part, methods for making and using in a treatment a biological cell, which is made using cell fusion.
  • the biological cell can be created using one or more fusion events, such that it can express a secretable vaccine protein and a T cell costimulatory fusion protein, and, In embodiments, one or more diseases antigens.
  • the disease antigens can be infectious disease antigens.
  • a method of making a biological cell comprises obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein; obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein; and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
  • the secretable vaccine protein is a secretable gp96-lg fusion protein, which optionally lacks the gp96 KDEL (SEQ ID NO: 3) sequence.
  • a method of making a biological cell comprises obtaining a first biological cell comprising a nucleotide sequence encoding a vaccine protein; obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein; and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
  • the vaccine protein is a native gp96 protein. In embodiments, the vaccine protein is secretable vaccine protein, such as a gp96-lg fusion protein.
  • the Ig tag in the gp96-lg fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or lgE.
  • the T cell costimulatory fusion protein is selected from OX40L-lg, ICOSL-lg, 4-1 BBL-lg, LAG3-lg, CD40L-lg, TL1 A-lg, CD70-lg, GITRL-lg, and CD28-lg.
  • the Ig tag in the T cell costimulatory fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE.
  • the T cell costimulatory fusion protein is selected from OX40L-lg or a portion thereof that binds specifically to 0X40, ICOSL-lg or a portion thereof that binds specifically to IGOS, 4-1 BBL-lg, or a portion thereof that binds specifically to 4-1 BBR, CD40L-lg, or a portion thereof that binds specifically to CD40, CD70-lg, or a portion thereof that binds specifically to CD27, TL1 A-lg or a portion thereof that binds specifically to TNFRSF25, or GITRL-lg or a portion thereof that binds specifically to GITR.
  • the T cell costimulatory fusion protein is OX40L-lg or a portion thereof that binds specifically to 0X40.
  • the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to the patient.
  • the disclosure provides sequential fusion combination (e.g., a single cell is transduced with gp96-lg, and a single cell is transduced with T cell costimulatory fusion protein, e.g. OX40L-lg, then the cells are fused together).
  • sequential fusion combination e.g., a single cell is transduced with gp96-lg, and a single cell is transduced with T cell costimulatory fusion protein, e.g. OX40L-lg, then the cells are fused together.
  • the provided methods result in considerably greater expression of secretable vaccine protein (e.g., without limitation, gp96-lg) and/or T cell costimulatory fusion protein (e.g, without limitation OX40L-lg), as compared to an unfused cell that has been caused to express (e.g. transduced with nucleic acids encoding) the secretable vaccine protein (e.g, without limitation, gp96-lg) and/or T cell costimulatory fusion protein (e.g, without limitation OX40L-lg).
  • secretable vaccine protein e.g., without limitation, gp96-lg
  • T cell costimulatory fusion protein e.g, without limitation OX40L-lg
  • the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of from about 1 : 1 to about 1 :5. In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1 : 1. In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1 :3.
  • the fused biological cell expresses the secretable vaccine protein and OX40L-lg fusion protein in a ratio of from about 1 : 1 to about 1 :3.
  • the fused biological cell further comprises a nucleotide sequence encoding one or more disease antigens.
  • the fused biological cell is made by fusing together at least two biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, and the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein.
  • the fused biological cell is made by fusing together at least three biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens.
  • the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens; wherein contacting the first biological cell and the second biological cell with the fusion agent further comprises contacting the third biological cell with the fusion agent, to result in the fused biological cell being created such that the fused biological cell comprises a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • the fused biological cell (created by fusing the first and second biological cells) is further fused with a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens, to thereby result in the fused biological cell (which can also be referred to as "a second fused biological cell”) that can express a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • a second fused biological cell which can express a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding one or more disease antigens; and contacting the third biological cell with the fusion agent; to result in a second fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • the second fused biological cell is immortalized.
  • one or more of the first, second, and third biological cells is a human tumor cell. In embodiments, all of the first, second, and third biological cells are human tumor cells. In embodiments, the human tumor cells are human primary humor cells.
  • the human tumor cell is a cell from an established non-small cell lung cancer (NSCLC), bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
  • NSCLC non-small cell lung cancer
  • bladder cancer bladder cancer
  • melanoma ovarian cancer
  • renal cell carcinoma prostate carcinoma
  • sarcoma breast carcinoma
  • squamous cell carcinoma head and neck carcinoma
  • pancreatic carcinoma pancreatic carcinoma
  • the third biological cell is a trophoblast or the third biological cell is fused with a trophoblast. In embodiments, one or both the first and second biological cells are fused with a trophoblast.
  • one or more of the first, second, and third biological cells are fused with a trophoblast or are trophoblasts.
  • one or more of the first, second, and third biological cells is a cell or is derived from a cell other than a cancer cell line.
  • all of the first, second, and third biological cells are cells or are derived from cells other than a cancer cell line.
  • the fused biological cell can be produced by fusing two or more biological cells other than cells from a cancer cell line. In this way, the fused biological cell does not include cancer cells.
  • one or more of the first, second, and third biological cells is a fibroblast or a fibroblast-like cell, or is derived from a fibroblast or a fibroblast-like cell.
  • one or more of the first, second, and third biological cells are derived from a cell line such as, without limitation, MRC-5 or WI-38.
  • the fused biological cell is immortalized. In embodiments, at least one of the first, second, and third biological cells is immortalized.
  • the nucleotide sequence is a mammalian expression vector.
  • the expression vector comprises DNA.
  • the expression vector comprises RNA.
  • the expression vector is incorporated into a virus or virus-like particle.
  • the expression vector comprises a pCEP4-EGFP plasmid.
  • the fusion agent comprises a protein from Paramyxoviridae Genus paramyxovirus.
  • the protein can be, for example, inactivated hemagglutinating virus of Japan envelope (HVJ-E).
  • the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof.
  • PEG poly(ethyleneglycol)
  • the PEG moiety comprises PEG-1500.
  • one or more of the first biological cell, the second biological cell, and the third biological cell comprise at least one tumor antigen.
  • the tumor antigen is a cancer testis (CT) antigen.
  • the fused biological cell expresses one or more cancer testis (CT) antigens.
  • CT cancer testis
  • one or more of the first, second, and third biological cells expresses one or more cancer testis (CT) antigens.
  • CT cancer testis
  • the fused biological cell is enriched for genes encoding one or more cancer testis (CT) antigens.
  • CT cancer testis
  • one or more of the first, second, and third biological cells expresses one or more cancer testis (CT) antigens.
  • CT cancer testis
  • the one or more disease antigens comprise one of more infectious disease antigens.
  • the one or more infectious disease antigens comprise an antigenic fragment of a betacoronavirus protein or an alphacoronavirus protein, optionally wherein the betacoronavirus protein is selected from a SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1 , and HCoV-OC43 protein, or the alphacoronavirus protein is selected from a HCoV-NL63 and HCoV-229E protein.
  • the betacoronavirus protein is a SARS-CoV-2 protein (a 2019-nCoV protein).
  • the SARS-CoV-2 protein is selected from a spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N.
  • the SARS-CoV-2 fragment comprises amino acid residues F486, N487, Q493, Q498, T500, N501 of the SARS-CoV-2 surface glycoprotein.
  • the SARS-CoV-2 peptide is a fragment of the SARS-CoV-2 RBD or the spike protein, including the wild type or a variant (also referred to as lineages). In embodiments, the SARS-CoV-2 peptide is a fragment of SEQ ID NO: 19 or a variant thereof. In embodiments, the wild type SARS-CoV-2 coronavirus is the "Wuhan strain.”
  • the present vaccine is pan-antigenic, thus providing immune response to the wild type (e.g., "Wuhan strain”) and numerous variants of the coronavirus.
  • the present vaccine comprises one or more peptides of the wild type and/or a variants of the spike proteins, or RBD thereof.
  • the vaccine includes two or more peptides of a respective variant, lineage, or strain of a coronavirus protein.
  • the variants can include a coronavirus protein having a mutation (e.g., without limitation, a substitution, deletion, or insertion) in any part of the spike, or the RBD thereof, protein, such as in the S1 subunit ( e.g, in the RBD of the Spike protein), or in the S2 subunit.
  • a mutation is in a glycosylation site of the Spike protein.
  • the variant (also referred to as lineages) is one or more of B.1.1.7, B.1.351 , B.1.617.2, B.1.427, B.1.429, B.1.525, B.1.526, B.1.617.1 , B.1.617.3, B.1 , B.1.1.28, B.1.2, CAL.20C, B.6, P.1 , and P.2 variants and/or any other variants, or antigenic fragments thereof.
  • the lineages include A.1 , A.2, A.3, A.4, A.5, A.6, A.7,
  • the variant is one or more of alpha (including B.1.1.7 and Q lineages), beta (including B.1.351 and descendent lineages), gamma (P.1 and descendent lineages), epsilon (including B.1.427 and B.1.429), eta (including B.1.525), iota (including B.1.526), kappa (including B.1.617.1), 1.617.3, mu (including B.1.621 , B.1.621.1), zeta (including P.2), delta (including B.1.617.2 and AY lineages), and omicron (including B.1.1.529).
  • alpha including B.1.1.7 and Q lineages
  • beta including B.1.351 and descendent lineages
  • gamma P.1 and descendent lineages
  • epsilon including B.1.427 and B.1.429
  • eta including B.1.525
  • iota including B.1.526
  • kappa including B.1.617.1
  • the SARS-CoV-2 variant is B.1.1.7, also known as the Alpha variant.
  • the B.1.1.7 (“Alpha”) variant comprises one or more mutations selected from 69del, 70del, 144del, (E484K*), (S494P*), N501Y, A570D, D614G, P681 H, T716I, S982A, and D1118H (K1191 N*), or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.351 , also known as the Beta variant.
  • the B.1.351 (“Beta”) variant comprises one or more mutations selected from D80A, D215G, 241 del, 242del, 243del, K417N, E484K, N501Y, D614G, and A701V, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.617.2, also known as the Delta variant.
  • the B.1.617.2 (“Delta”) variant comprises one or more mutations selected from T19R, (G142D*), 156del, 157del, R158G, L452R, T478K, D614G, P681 R, and D950N, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is P.1, also known as the Gamma variant.
  • the P.1 ("Gamma”) variant comprises one or more mutations selected from L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, D614G, H655Y, and T1027I, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.427, also known as the Epsilon variant.
  • the B.1.427 (“Epsilon”) variant comprises one or more mutations selected from L452R and D614G, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.429, also known as the Epsilon variant.
  • the B.1.429 (“Epsilon”) variant comprises one or more mutations selected from S13I, W152C, L452R, and D614G, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.525, also known as the Eta variant.
  • the B.1.525 (“Eta”) variant comprises one or more mutations selected from A67V, 69del, 70del, 144del, E484K, D614G, Q677H, and F888L, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.526, also known as the lota variant.
  • the B.1.526 (“Iota”) variant comprises one or more mutations selected from L5F, (D80G*), T95I, (Y144-*), (F157S*), D253G, (L452R*), (S477N*), E484K, D614G, A701V, (T859N*), (D950H*), and (Q957R*), or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.617.1 , also known as the Kappa variant.
  • the B.1.617.1 (“Kappa”) variant comprises one or more mutations selected from (T95I), G142D, E154K, L452R, E484Q, D614G, P681 R, and Q1071 H, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is B.1.617.3.
  • the B.1.617.3 variant comprises one or more mutations selected from T19R, G142D, L452R, E484Q, D614G, P681R, D950N, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is P.2, also known as the Zeta variant.
  • the P.2 (“Zeta”) variant comprises one or more mutations selected from E484K, (F565L*), D614G, and V1176F, or an antigenic fragment thereof.
  • the SARS-CoV-2 variant is Delta (B.1.617.2). This variant was first identified in India in late 2020. The Delta variant is shown to be more infectious and spreads more rapidly than other variants.
  • the Delta variant contains mutations in the spike protein: T19R, del157/158, L452R, T478K, D614G, P681 R, and D950N. See Dhar, Mahesh S et al. "Genomic characterization and epidemiology of an emerging SARS-CoV-2 variant in Delhi, India.” Science (New York, N.Y.) vol. 374,6570 (2021): 995-999. doi: 10.1126/science.abj9932.
  • the Delta variant has more than a dozen mutations and more likely to result in hospitalization in the unvaccinated human population. See Twohig eta/., "Hospital admission and emergency care attendance risk for SARS-CoV-2 delta (B.1.617.2) compared with alpha (B.1.1.7) variants of concern: a cohort study.” published online August 27, 2021 , The Lancet. 22(1): 25-42. doi:10.1016/S1473- 3099(21)00475-8.
  • the SARS-CoV-2 variant is the Omicron variant (B.1.1.529). This variant was discovered in South Africa in November 2021 . The World Health Organization has proposed the Omicron variant to be a variant of concern.
  • Omicron carries about 50 mutations, 26 unique to the variant and more than 30 mutations on the spike protein.
  • the Omicron variant contains substitutions in the spike protein: A67V, del69-70, T95I, del142-144, Y145D, del211, L212I, ins214EPE, G339D, S371 L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681 H, N764K, D796Y, N856K, Q954H, N969K, and L981 F.
  • the Omicron variant contains receptor binding domain substitutions in the spike protein: G339D, S371 L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, and Y505H.
  • G339D S371 L, S373P, S375F
  • K417N N440K
  • G446S S477N
  • T478K T478K
  • E484A Q493R
  • G496S Q498R
  • N501Y and Y505H.
  • a variant is a SARS-CoV-2 protein having a variation in a glycosylation site of a Spike protein.
  • a variant is a Spike protein having one or more of D614G, E484K, N501Y, K417N, S477G, and S477N mutations or an antigenic fragment thereof.
  • a variant is a Spike protein having a mutation in the RBD of the Spike protein.
  • the mutation in the RBD of the Spike protein is a mutation in a glycosylation site in the RBD.
  • a variant is a Spike protein having a mutation outside the RBD of the Spike protein.
  • a method of treating cancer comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • the cancer comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia.
  • a method of treating an infectious disease provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • the infectious disease comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection.
  • the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae.
  • the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E.
  • the infectious disease comprises a coronavirus infection 2019 (COVID-19).
  • a method of stimulating a patient's antibody response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • a method of stimulating a patient T cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • a method of stimulating one or both a patient's antibody response and T cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • a method of stimulating a patient B cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with any of the embodiments of the present disclosure.
  • a method of stimulating one or both a patient's antibody response and B cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • the T cell-driven cellular immune response induces a CD8+ T cell response in the patient. In embodiments, the T cell-driven cellular immune response induces a CD4+ T cell response in the patient.
  • a method induces an immune response, selected from a CD8+ T cell, a CD4+ T cell, a cytotoxic T lymphocyte (CTL), a TH1 response, a TH2 response, or a combination thereof.
  • an immune response selected from a CD8+ T cell, a CD4+ T cell, a cytotoxic T lymphocyte (CTL), a TH1 response, a TH2 response, or a combination thereof.
  • CTL cytotoxic T lymphocyte
  • a method results in reduction of a number of regulatory T cells (Tregs).
  • a method results in the decrease in the functionality of Tregs. In embodiments, the method does not result in the decrease in the number of Tregs but results in the decrease in the functionality of Tregs.
  • a method that makes use of a fused cell generated in accordance with embodiments of the present disclosure comprises administering to the patient an inhibitor of an immune checkpoint molecule.
  • the immune checkpoint molecule is selected from PD-1 , PD-L1 , PD-L2, CTLA-4, ICOS, LAG3, 0X40, OX40L, and TIM3.
  • the immune checkpoint molecule is PD-1.
  • a lentivirus vector encodes the nucleotide sequence encoding the secretable vaccine protein, and a lentivirus vector encodes the nucleotide sequence encoding the T cell costimulatory fusion protein.
  • the method further comprises merging together the first biological cell and the second biological cell.
  • a method for generating a cellular therapy comprising: (a) obtaining a lentivirus, the lentivirus comprising: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, and an immunoglobulin, or a fragment thereof; and (b) introducing into a biological cell the lentivirus of step (a).
  • a method for generating a cellular therapy comprising: (a) obtaining a lentivirus, the lentivirus comprising: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject; and (b) introducing into a biological cell the lentivirus of step (a).
  • a method for generating a cellular therapy comprising: (a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a first biological cell; (b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject, and (i) introducing the nucleic acid into a second biological cell, and (c) merging together the first biological cell and the second biological cell.
  • a method for generating a cellular therapy comprising: (a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a first biological cell; (b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a second biological cell, and (c) merging together the first biological cell and the second biological cell.
  • FIG. 1 shows both an image and a bar graph of clones developed from lentivirus transduction generated from DNA encoding for either gp96-lg or OX40L-lg, or both gp96-lg and OX40L-lg.
  • FIG. 2 is a graph illustrating expression of cancer testis antigens (CTAs) in HEK293 and HS112 (AD-100 expressing gp96) cells, and in a fusion of the HEK293 and HS122 cells.
  • CTAs cancer testis antigens
  • FIG. 3 is a graph illustrating ELISA analysis of GP96-lgG and OX40L-lgG levels in individual lentiviral clones before and after cellular fusion.
  • the present disclosure is based, in part, on the discovery that cell fusion can be used to produce immune or immunotherapy agents for treatment of cancer and infectious diseases.
  • One, two, or more than two biological cells can be fused to result in a fused biological cell comprising a nucleotide sequence encoding a vaccine protein (e.g., a secretable vaccine protein) and a nucleotide sequence encoding a T cell costimulatory fusion protein.
  • Cell fusion is a mechanism that enables genetic material to move from one cell to another and produce viable hybrid progeny.
  • APCs antigen presenting cells
  • DC-tumor cell hybrids induced higher antigen-specific T-cell expansion as compared to simple mixture of tumor cells and DCs.
  • the inventors have recognized and appreciated that, among other factors, the similarities between cancer cells and trophoblasts can be exploited in making immune agents, which can be made using cell fusion.
  • the present disclosure provides methods for making a fused biological cell that can be generated as a result of one, two, or more than two fusion events.
  • the fused biological cell can be made by fusing a first biological cell and a second biological cell, which can be then fused with a third biological cell.
  • the fused biological cell can be made by fusing a first biological cell, a second biological cell, and a third biological cell.
  • more than three biological cells can be fused via one, two, or three cell fusion events.
  • the fused cell is created by fusing at least a first biological cell comprising a nucleotide sequence that encodes a secretable vaccine protein (such as exogenous gp96-lg fusion protein, which can lack KDEL (SEQ ID NO: 3) sequence) and a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein.
  • a first biological cell comprises a nucleotide sequence that encodes a vaccine protein such as a native (non secreted) gp96 protein, or a secreted gp96-lg fusion protein.
  • One or both the first biological cell and the second biological cell can be derived from a cell line producing at least one tumor antigen, such as, without limitation, a cancer testis (CT) antigen.
  • a cell line is a polyploidy cell line that expresses multiple CT antigens.
  • the cell line can be created by fusing cancer tissue specific ⁇ e.g., breast, colon, pancreatic, lung, liver, etc.) cells with an established non-small cell lung cancer (NSCLC) cell line.
  • NSCLC non-small cell lung cancer
  • the cell line is created by fusing an established NSCLC cell line with a trophoblast.
  • one or more of the first, second, and third biological cells is a cell or is derived from a cell other than a cancer cell line.
  • all of the first, second, and third biological cells are cells or are derived from cells other than a cancer cell line.
  • the fused biological cell produced in accordance with embodiments of the present disclosure is used for treatment of an infectious disease
  • the fused biological cell is produced by fusing two or more biological cells other than cells from a cancer cell line.
  • the fused biological cell does not include cancer cells.
  • the fused biological cell can similarly be produced by fusing two or more biological cells other than cells from a cancer cell line.
  • one or more of the first, second, and third biological cells is a fibroblast or a fibroblast-like cell, or is derived from a fibroblast or a fibroblast-like cell. In embodiments, one or more of the first, second, and third biological cells are derived from a cell line such as, without limitation, MRC-5 or WI-38.
  • a method of making a biological cell comprises obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
  • a method of making a fused biological cell comprises obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and causing the first biological cell and the second biological cell to fuse by contacting the first biological cell and the second biological cell with a fusion agent, thereby generating a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
  • the secretable vaccine protein is a heat shock protein (hsp) gp96 that is localized in the endoplasmic reticulum (ER) and serves as a chaperone for peptides on their way to MHC class I and II molecules.
  • Gp96 obtained from tumor cells and used as a vaccine can induce specific tumor immunity, presumably through the transport of tumorspecific peptides to antigen-presenting cells (APCs) (Yamazaki et al., J Immunol 1999, 163(10):5178-5182).
  • APCs antigen-presenting cells
  • gp96-associated peptides are cross-presented to CD8 cells by dendritic cells (DCs).
  • DCs dendritic cells
  • Gp96-based vaccination modality has also been shown to provide protection against mucosal infection caused by simian immunodeficiency virus. Strbo et al., J Immunol. 2013; 190 (6): 2495-2499.
  • the present compositions and methods use gp96 to trigger mucosal immunity by activating both B and T cell responses at the point of pathogen entry.
  • the gp96-based composition activates the immune system thereby.
  • the secretable vaccine protein is a secretable gp96-lg fusion protein, which optionally lacks the gp96 KDEL (SEQ ID NO: 3) sequence.
  • the Ig tag in the gp96-lg fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE.
  • a nucleotide sequence that encodes a gp96-lg fusion protein.
  • the coding region of human gp96 is 2,412 bases in length (SEQ ID NO: 1), and encodes an 803 amino acid protein (SEQ ID NO: 2) that includes a 21 amino acid signal peptide at the amino terminus, a potential transmembrane region rich in hydrophobic residues, and an ER retention peptide sequence at the carboxyl terminus (GENBANK® Accession No. X15187; see Maki et al., Proc Natl Acad Sc/ USA 1990, 87:5658-5562).
  • a nucleic acid encoding a gp96-lg fusion sequence can be produced using the methods described in U.S. Patent No. 8,685,384, which is incorporated herein by reference in its entirety.
  • the gp96 portion of a gp96-lg fusion protein can contain all or a portion of a wild type gp96 sequence (e.g., the human sequence set forth in SEQ ID NO: 2).
  • a secretable gp96-lg fusion protein can include the first 799 amino acids of SEQ ID NO: 2, such that it lacks the C-terminal KDEL (SEQ ID NO: 3) sequence.
  • the gp96 portion of the fusion protein can have an amino acid sequence that includes one or more substitutions, deletions, or additions, as compared to the first 799 amino acids of the wild type gp96 sequence, such that it has at least 90% (e.g., at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) sequence identity to the wild type polypeptide.
  • 90% e.g., at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%
  • the gp96 portion of a nucleotide sequence encoding a gp96-lg fusion polypeptide can encode an amino acid sequence that differs from the wild type gp96 polypeptide at one or more amino acid positions, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms.
  • the Ig portion ("tag”) of a gp96-lg fusion protein can contain, for example, a non-variable portion of an immunoglobulin molecule (e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule).
  • an immunoglobulin molecule e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule.
  • such portions contain at least functional CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. Fusions also can be made using the carboxyl terminus of the Fc portion of a constant domain, or a region immediately amino-terminal to the CH1 of the heavy or light chain.
  • the Ig tag can be from a mammalian (e.g., human, mouse, monkey, or rat) immunoglobulin, but human immunoglobulin can be particularly useful when the gp96-lg fusion is intended for in vivo use for humans.
  • mammalian e.g., human, mouse, monkey, or rat
  • human immunoglobulin can be particularly useful when the gp96-lg fusion is intended for in vivo use for humans.
  • gp96 genetically fused to an immunoglobulin domain (e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule), activates TLR2 and TLR4 on professional antigen-presenting cells (APCs).
  • an immunoglobulin domain e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule
  • APCs professional antigen-presenting cells
  • a gp96 peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen et al., J Immunol 1996, 156:442-449).
  • This region of the lgG1 molecule contains three cysteine residues that normally are involved in disulfide bonding with other cysteines in the Ig molecule. Since none of the cysteines are required for the peptide to function as a tag, one or more of these cysteine residues can be substituted by another amino acid residue, such as, for example, serine.
  • leader sequences known in the art also can be used for efficient secretion of gp96-lg fusion proteins from bacterial and mammalian cells (see, von Heijne, J Mol Biol 1985, 184:99-105).
  • Leader peptides can be selected based on the intended host cell, and may include bacterial, yeast, viral, animal, and mammalian sequences.
  • the herpes virus glycoprotein D leader peptide is suitable for use in a variety of mammalian cells.
  • leader peptide for use in mammalian cells can be obtained from the V-J2-C region of the mouse immunoglobulin kappa chain (Bernard et al., Proc Natl Acad Sci USA 1981 , 78:5812-5816). DNA sequences encoding peptide tags or leader peptides are known or readily available from libraries or commercial suppliers, and are suitable in the fusion proteins described herein.
  • a nucleic acid sequence encodes a native gp96.
  • one may substitute the gp96 of the present disclosure with one or more vaccine proteins.
  • various heat shock proteins are among the vaccine proteins.
  • the heat shock protein is one or more of a small hsp, hsp40, hsp60, hsp70, hsp90, and hsp110 family member, inclusive of fragments, variants, mutants, derivatives or combinations thereof (Hickey, et al., 1989, Mol. Cell. Biol. 9:2615-2626; Jindal, 1989, Mol. Cell. Biol. 9:2279-2283)
  • nucleotide sequences can be introduced into host cells for producing secreted vaccine proteins (e.g., gp96-lg), T cell costimulatory fusion proteins, and one or more disease antigens.
  • secreted vaccine proteins e.g., gp96-lg
  • T cell costimulatory fusion proteins e.g., T cell costimulatory fusion proteins
  • disease antigens e.g., gp96-lg
  • Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, polymer-based systems, DEAE-dextran, viral transduction, the calcium phosphate precipitation method, etc.
  • liposomes For in vivo gene transfer, a number of techniques and reagents may also be used, including liposomes; natural polymer-based delivery vehicles, such as chitosan and gelatin; viral vectors are also suitable for in vivo transduction.
  • a targeting agent such as an antibody or a ligand specific for a cell surface membrane protein.
  • proteins which bind to a cell surface membrane protein associated with endocytosis may be used for targeting and/or to facilitate uptake, e.g., capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, proteins that target intracellular localization and enhance intracellular half-life.
  • gene delivery agents such as, e.g., integration sequences can also be employed.
  • Numerous integration sequences are known in the art (see, e.g, Nunes-Duby et al., Nucleic Acids Res. 26:391-406, 1998; Sadwoski, J. Bacteriol., 165:341-357, 1986; Bestor, Cell, 122 (3): 322-325, 2005; Plasterk et al., TIG 15:326-332, 1999; Kootstra et al., Ann. Rev. Pharm. Toxicol., 43:413-439, 2003). These include recombinases and transposases. Examples include Cre (Sternberg & Hamilton, J. Mol.
  • Cells may be cultured in vitro or genetically engineered, for example.
  • Host cells can be obtained from normal or affected subjects, including healthy humans, cancer patients, and patients with an infectious disease, private laboratory deposits, public culture collections such as the American Type Culture Collection, or from commercial suppliers.
  • biological cells that can be used for production and secretion of gp96-lg fusion proteins or T cell costimulatory fusion proteins are human tumor cells.
  • the human tumor cells are human primary tumor cells.
  • biological cells that can be used for production and secretion of gp96-lg fusion proteins and T cell costimulatory fusion proteins in vivo include, without limitation, epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes, blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, or granulocytes, various stem or progenitor cells, such as hematopoietic stem or progenitor cells (e.g., as obtained from bone marrow), umbilical cord blood, peripheral blood, fetal liver, etc., and tumor cells (e.g., human tumor cells).
  • epithelial cells include, without limitation, epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes, blood cells such as T lymphocytes, B lymphocytes, mon
  • Different biological host cells have characteristic and specific mechanisms for post-translational processing and modification of proteins.
  • a biological cell may be chosen which modifies and processes the expressed gene products in a specific fashion similar to the way the recipient processes its heat shock proteins (hsps).
  • hsps heat shock proteins
  • the type of host cell has been used for expression of heterologous genes, and is reasonably well characterized and developed for large-scale production processes.
  • the host cells are autologous to the patient to whom the present fusion or recombinant cells secreting the present fusion proteins are subsequently administered.
  • a fused biological cell in addition to a secretable vaccine protein, such as a gp96-lg fusion protein, a fused biological cell is made such that it also includes a nucleotide sequence encoding one or more biological response modifiers.
  • the fused biological cell comprises a nucleotide sequence encoding one or more T cell costimulatory molecules.
  • the T cell costimulatory fusion protein is selected from OX40L-lg, ICOSL-lg, 4-1 BBL-lg, LAG3-lg, CD40L-lg, TL1 A-lg, CD70-lg, GITRL-lg, and CD28-lg.
  • the T cell costimulatory fusion protein is selected from OX40L-lg or a portion thereof that binds to 0X40, ICOSL-lg or a portion thereof that binds to IGOS, 4-1 BBL-lg or a portion thereof that binds to 4-1 BBR, TL1 A-lg or a portion thereof that binds to TNFRSF25, GITRL-lg or a portion thereof that binds to GITR, CD40-lg or a portion thereof that binds to CD40, or CD70-I g or a portion thereof that binds to CD27, among others.
  • the CD28-lg fusion protein binds to the costimulatory ligands CD80 and CD86 to provide a costimulatory signal to T cells.
  • the Ig tag in the T cell costimulatory fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE.
  • the T cell costimulatory fusion protein is OX40L-lg. In embodiments, the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to the patient.
  • the lentivirus and/or transfer plasmid can comprise one or more biological response modifiers.
  • the lentivirus and/or transfer plasmid can encode one or more T cell costimulatory molecules or fusion proteins.
  • ICOS is an inducible T cell costimulatory receptor molecule that displays some homology to CD28 and CTLA-4, and interacts with B7-H2 expressed on the surface of antigen-presenting cells. ICOS has been implicated in the regulation of cell-mediated and humoral immune responses.
  • 4-1 BB is a type 2 transmembrane glycoprotein belonging to the TNF superfamily, and is expressed on activated T Lymphocytes.
  • 0X40 (also referred to as CD134 or TNFRSF4) is a T cell costimulatory molecule that is engaged by OX40L, and frequently is induced in antigen presenting cells and other cell types. 0X40 is known to enhance cytokine expression and survival of effector T cells.
  • GITR TNFRSF18
  • GITRL GITRL
  • FoxP3+ regulatory T cells GITR plays a significant role in the maintenance and function of Treg within the tumor microenvironment.
  • TNFRSF25 is a T cell costimulatory molecule that is preferentially expressed in CD4+ and CD8+ T cells following antigen stimulation. Signaling through TNFRSF25 is provided by TL1A, and functions to enhance T cell sensitivity to IL-2 receptor mediated proliferation in a cognate antigen dependent manner.
  • CD40 is a costimulatory protein found on various antigen presenting cells which plays a role in their activation.
  • the binding of CD40L (CD154) on TH cells to CD40 activates antigen presenting cells and induces a variety of downstream effects.
  • CD27 a T cell costimulatory molecule belonging to the TNF superfamily which plays a role in the generation and longterm maintenance of T cell immunity. It binds to a ligand CD70 in various immunological processes. See van de Ven & Borst (2015) Immunotherapy 7(6): 655-67.
  • LAG3 (CD223) is mainly expressed in activated T and natural killer (NK) cells and was identified to as a marker for the activation of CD4+ and CD8+T cells. Puhr & Ilhan-Mutlu, ESMO Open 2019;4:e000482. doi: 10.1136/esmoopen-2018- 000482. LAG3-lg fusion protein binds to cell surface MHC class II. Andrews et al. (2017) Immunological reviews vol. 276,1 ; 80-96.
  • Additional costimulatory molecules that may be used in embodiments of the present disclosure include, but are not limited to, HVEM, CD30, CD30L, CD40, CD70, LIGHT (CD258), B7-1 , and B7-2.
  • the present lentivirus and/or transfer plasmid comprises an agonist of 0X40 (e.g, an 0X40 ligand-lg (OX40L-lg) fusion, or a fragment thereof that binds 0X40), an agonist of inducible T-cell costimulator (IGOS) (e.g., an IGOS ligand-lg (ICOSL-lg) fusion, or a fragment thereof that binds IGOS), an agonist of CD40 (e.g., a CD40L-lg fusion protein, or fragment thereof), an agonist of CD27 (e.g.
  • IGOS inducible T-cell costimulator
  • CD40 e.g., a CD40L-lg fusion protein, or fragment thereof
  • CD27 e.g.
  • CD70-lg fusion protein or fragment thereof a CD70-lg fusion protein or fragment thereof), or an agonist of 4-1 BB (e.g, a 4-1 BB ligand-lg (4-1 BBL-lg) fusion, or a fragment thereof that binds 4-1 BB).
  • 4-1 BB e.g, a 4-1 BB ligand-lg (4-1 BBL-lg) fusion, or a fragment thereof that binds 4-1 BB.
  • the lentivirus and/or transfer plasmid comprises an agonist of TNFRSF25 (e.g, a TL1 A-lg fusion, or a fragment thereof that binds TNFRSF25), or an agonist of glucocorticoid-induced tumor necrosis factor receptor (GITR) (e.g, a GITR ligand-lg (GITRL-lg) fusion, or a fragment thereof that binds GITR), or an agonist of CD40 (e.g, a CD40 ligand-lg (CD40L-lg) fusion, or a fragment thereof that binds CD40); or an agonist of CD27 (e.g, a CD27 ligand-lg (e.g CD70L- Ig) fusion, or a fragment thereof that binds CD40).
  • GITR glucocorticoid-induced tumor necrosis factor receptor
  • CD40 e.g, a CD40 ligand-lg (
  • the Ig portion ("tag”) of the T cell costimulatory fusion protein can include a non-variable portion of an immunoglobulin molecule (e.g, an lgG1, lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule). Such portions typically contain at least functional CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain.
  • a T cell costimulatory peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen etal., J Immunol 1996, 156:442-449).
  • the Ig tag can be from a mammalian (e.g, human, mouse, monkey, or rat) immunoglobulin, but human immunoglobulin can be particularly useful when the fusion protein is intended for in vivo use for humans.
  • DNAs encoding immunoglobulin light or heavy chain constant regions are known or readily available from cDNA libraries.
  • Various leader sequences as described above also can be used for secretion of T cell costimulatory fusion proteins from bacterial and mammalian cells.
  • a representative nucleotide optimized sequence (SEQ ID NO:4) encoding the extracellular domain of human ICOSL fused to Ig, and the amino acid sequence of the encoded fusion (SEQ ID NO:5) are provided.
  • the sequence of the entire fusion, or just the ICOSL component fused to another Fc domain is used:
  • a representative nucleotide optimized sequence (SEQ ID NO:6) encoding the extracellular domain of human 4-1 BBL fused to Ig, and the encoded amino acid sequence (SEQ ID NO:7) are provided.
  • the sequence of the entire fusion, or just the 4-1 BBL component fused to another Fc domain is used:
  • a representative nucleotide optimized sequence (SEQ ID NO:8) encoding the extracellular domain of human TL1A fused to Ig, and the encoded amino acid sequence (SEQ ID NO:9) are provided.
  • the sequence of the entire fusion, or just the TL1A component fused to another Fc domain is used:
  • a representative nucleotide optimized sequence (SEQ ID NO:10) encoding human QX40L-lg, and the encoded amino acid sequence (SEQ ID NO:11) are provided.
  • the sequence of the entire fusion, or just the OX40L component fused to another Fc domain is used:
  • TL1A Representative nucleotide and amino acid sequences for human TL1A are set forth in SEQ ID NO:12 and SEQ ID NO: 13, respectively. In embodiments, the sequence of the entire fusion, or just the TL1A component fused to another Fc domain is used:
  • VMDAVPARRWKEFVRTLGLREAEIEAVEVEIGRFRDQQYEMLKRWRQQQPAGLGAVYA ALERMGLDGCVEDLRSRLQRGP (SEQ ID NO: 13).
  • HVEM human HVEM
  • SEQ ID NO:26 accession no. CR456909
  • SEQ ID NO:27 accession no. CR456909
  • sequence of the entire fusion, or just the HVEM component fused to another Fc domain is used:
  • nucleotide and amino acid sequences for human CD28 are set forth in SEQ ID NO:28 (accession no. NM_006139) and SEQ ID NO:29, respectively.
  • sequence of the entire fusion, or just the CD28 component fused to another Fc domain is used:
  • nucleotide and amino acid sequences for human CD30L are set forth in SEQ ID NQ:30 (accession no. L09753) and SEQ ID NO:31, respectively.
  • sequence of the entire fusion, or just the CD30L component fused to another Fc domain is used: CCAAGTCACATGATTCAGGATTCAGGGGGAGAATCCTTCTTGGAACAGAGATGGGCC
  • nucleotide and amino acid sequences for human CD40 are set forth in SEQ ID NO:32 (accession no. NM_001250) and SEQ ID NO:33, respectively.
  • sequence of the entire fusion, or just the CD40 component fused to another Fc domain is used:
  • nucleotide and amino acid sequences for human CD70 are set forth in SEQ ID NO:34 (accession no. NM_001252) and SEQ ID NO:35, respectively.
  • sequence of the entire fusion, or just the CD70 component fused to another Fc domain is used:
  • a representative amino acid sequences for human GITRL are set forth in SEQ ID NO:52 (accession no. Q9UNG2), respectively.
  • sequence of the entire fusion, or just the GITRL component fused to another Fc domain is used:
  • variants comprising any of the sequences described herein, for instance, a sequence having at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71 %, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81 %, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or
  • an amino acid sequence is provided that has one or more amino acid mutations relative to any of the protein sequences described herein.
  • the one or more amino acid mutations may be independently selected from conservative or non-conservative substitutions, insertions, deletions, and truncations as described herein.
  • a "conservative substitution” denotes the replacement of an amino acid residue by another, biologically similar, residue.
  • biological similarity reflects substitutions on the wild type sequence with conserved amino acids. For example, conservative amino acid substitutions would be expected to have little or no effect on biological activity, particularly if they represent less than 10% of the total number of residues in the polypeptide or protein. Conservative substitutions may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved.
  • the 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1) hydrophobic: Met, Ala, Vai, Leu, lie; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gin; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe. Accordingly, conservative substitutions may be affected by exchanging an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide.
  • glycine and proline may be substituted for one another based on their ability to disrupt o-helices.
  • Additional examples of conserved amino acid substitutions include, without limitation, the substitution of one hydrophobic residue for another, such as isoleucine, valine, leucine, or methionine, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like.
  • conserved amino acid substitution also includes the use of a substituted amino acid residue in place of an un-substituted parent amino acid residue, provided that antibodies raised to the substituted polypeptide also immunoreact with the un-substituted polypeptide.
  • non-conservative substitutions are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1) to (6) shown above.
  • the substitutions may also include non-classical amino acids (e.g. selenocysteine, pyrrolysine, N- formylmethionine p-alanine, GABA and 6-Aminolevulinic acid, 4-aminobenzoic acid (PABA), D-isomers of the common amino acids, 2,4-diaminobutyric acid, o-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, y-Abu, s-Ahx, 6-amino hexanoic acid, Alb, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosme, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexy
  • mutations may also be made to the nucleotide sequences of the present fusion proteins by reference to the genetic code, including taking into account codon degeneracy.
  • the gp96-lg fusion protein and/or a T cell costimulatory fusion protein comprises a linker.
  • the linker may be derived from naturally-occurring multi-domain proteins or are empirical linkers as described, for example, in Chichili et al. (2013) Protein Sci. 22(2): 153-167; Chen et al. (2013), Adv Drug Deliv Rev. 65(10): 1357-1369, the entire contents of which are hereby incorporated by reference.
  • the linker may be designed using linker designing databases and computer programs such as those described in Chen et al. (2013) Adv Drug Deliv Rev. 65(10): 1357-1369 and Crasto et. al. (2000) Protein Eng. 13(5):309-312, the entire contents of which are hereby incorporated by reference.
  • the linker is a synthetic linker such as PEG.
  • the linker is a polypeptide. In embodiments, the linker is less than about 100 amino acids long. For example, the linker may be less than about 100, about 95, about 90, about 85, about 80, about 75, about 70, about 65, about 60, about 55, about 50, about 45, about 40, about 35, about 30, about 25, about 20, about 19, about 18, about 17, about 16, about 15, about 14, about 13, about 12, about 11 , about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, or about 2 amino acids long. In embodiments, the linker is flexible. In another embodiment, the linker is rigid. In embodiments, the linker is substantially comprised of glycine and serine residues (e.g. about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 95%, or about 97% glycines and serines).
  • glycine and serine residues e.g. about 30%, or about 40%, or about 50%, or about 60%, or about 70%,
  • the linker is a hinge region of an antibody (e.g., of IgG, IgA, IgD, and IgE, inclusive of subclasses (e.g. lgG1, lgG2, lgG3, and lgG4, and lgA1 and lgA2)).
  • the hinge region found in IgG, IgA, IgD, and IgE class antibodies, acts as a flexible spacer, allowing the Fab portion to move freely in space.
  • the hinge domains are structurally diverse, varying in both sequence and length among immunoglobulin classes and subclasses. For example, the length and flexibility of the hinge region varies among the IgG subclasses.
  • the hinge region of lgG1 encompasses amino acids 216-231 and, because it is freely flexible, the Fab fragments can rotate about their axes of symmetry and move within a sphere centered at the first of two inter-heavy chain disulfide bridges.
  • I gG2 has a shorter hinge than lgG1 , with 12 amino acid residues and four disulfide bridges.
  • the hinge region of lgG2 lacks a glycine residue, is relatively short, and contains a rigid poly-proline double helix, stabilized by extra inter-heavy chain disulfide bridges. These properties restrict the flexibility of the lgG2 molecule.
  • lgG3 differs from the other subclasses by its unique extended hinge region (about four times as long as the lgG1 hinge), containing 62 amino acids (including 21 prolines and 11 cysteines), forming an inflexible poly-proline double helix.
  • the Fab fragments are relatively far away from the Fc fragment, giving the molecule a greater flexibility.
  • the elongated hinge in lgG3 is also responsible for its higher molecular weight compared to the other subclasses.
  • the hinge region of lgG4 is shorter than that of lgG1 and its flexibility is intermediate between that of IgG 1 and I gG2. The flexibility of the hinge regions reportedly decreases in the order lgG3>lgG 1 >lgG4>lgG2.
  • the linker may be functional.
  • the linker may function to improve the folding and/or stability, improve the expression, improve the pharmacokinetics, and/or improve the bioactivity of the present compositions.
  • the linker may function to target the compositions to a particular cell type or location.
  • An expression vector was engineered to include both the gp96-lg fusion protein and a T cell costimulatory fusion protein, such that a combination immunotherapy could be achieved by vector re-engineering to obviate the need for vaccine/antibody/fusion protein regimens, which may reduce a cost of therapy and the risk of systemic toxicity.
  • a therapy additionally includes a checkpoint inhibitor. Further, the need for using separate different drug products ⁇ e.g., a vaccine, a T cell costimulatory protein, and a checkpoint inhibitor) is obviated.
  • the present disclosure further improves the combination therapy by fusing first and second biological cells including nucleotide sequences encoding a vaccine protein (such as a secretable vaccine protein) and a T cell costimulatory fusion protein, respectively, such that a ratio of a secretable vaccine protein and a T cell costimulatory fusion protein is controlled.
  • the first and second biological cells can be generated to express a known quantity of a vaccine protein and a T cell costimulatory fusion protein, respectively, such that the fused biological cell expresses known, desired quantities of the vaccine protein and the T cell costimulatory fusion protein.
  • the vaccine protein and the T cell costimulatory fusion protein can be expressed in the fused cell at a desired ratio. This allows creating compositions for treating cancer or an infectious disease with an appropriate dosage and ratio of components.
  • the vaccine protein can be a secretable vaccine protein, such as a gp96-lg fusion protein, or a native gp96 protein.
  • the fused biological cell expresses the vaccine protein (e.g., the secretable gp96-lg fusion protein or gp96 in its native form) and the T cell costimulatory fusion protein in a ratio of from about 1:1 to about 1:5.
  • the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1:1, or about 1:1.5, or about 1:2, or about 1:2.5, or about 1:3, or about 1:3.5, or about 1:4, or about 1:4.5, or about 1:5.
  • the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1:3.
  • the fused biological cell expresses the vaccine protein, such as gp96-lg fusion protein or a native gp96 protein, and OX40L- 1 g fusion protein in a ratio of about 1 :3.
  • the vaccine protein such as gp96-lg fusion protein or a native gp96 protein, and OX40L- 1 g fusion protein in a ratio of about 1 :3.
  • a fused biological cell is created such that an amount of a vaccine protein (e.g., gp96-lg or native gp96) is lower than the expression of a T cell costimulatory fusion protein, (e.g., OX40-lg or any of the other fusion proteins including those described herein).
  • a vaccine protein e.g., gp96-lg or native gp96
  • a T cell costimulatory fusion protein e.g., OX40-lg or any of the other fusion proteins including those described herein.
  • a ratio of an amount of a vaccine protein (e.g., gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is about 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1:25, 1:30, 1:40, 1:50, 1:60, 1:70, 1:80. 1:90, or 1:100.
  • a ratio of an amount of a vaccine protein (e.g., gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is from about 1:6 to about 1:100, inclusive of all endpoints.
  • secretable vaccine protein (e.g., gp96-lg) secretion is lower than the expression of a T cell costimulatory fusion protein (e.g, OX40-lg).
  • a ratio of secretable vaccine protein (e.g, gp96-lg) secretion to the expression of aT cell costimulatory fusion protein (e.g, OX40-lg) is about 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1:25, 1:30, 1:40, 1:50, 1:60, 1:70, 1:80. 1:90, or 1:100.
  • a ratio of secretable vaccine protein (e.g, gp96-lg) secretion to the expression of a T cell costimulatory fusion protein (e.g, OX40-lg) is from about 1:6 to about 1:100, inclusive of all endpoints.
  • an amount of a vaccine protein (e.g, gp96-lg or native gp96) is higher than the expression of a T cell costimulatory fusion protein (e.g, OX40-lg).
  • the ratio of an amount of vaccine protein (e.g, gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g, OX40-lg) is about 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 25:1, 30:1, 40:1, or 50:1.
  • the ratio of an amount of vaccine protein (e.g, gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g, OX40-lg) is from about 2:1 to about 50:1, inclusive of all endpoints.
  • secretable vaccine protein e.g, gp96-lg
  • secretable vaccine protein secretion is higher than the expression of a T cell costimulatory fusion protein (e.g., OX40-lg).
  • costimulatory fusion protein e.g., OX40-lg.
  • the ratio of the vaccine protein (e.g., gp96-lg) secretion to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is about 2:1 , 3:1 , 4: 1 , 5: 1 , 6: 1 , 7:1 , 8: 1, 9: 1 , 10:1 , 11 :1 , 12: 1 , 13:1 , 14: 1 , 15:1 , 16: 1, 17: 1 , 18:1 , 19: 1 , 20:1 , 25:1 , 30: 1 , 40:1 , or 50: 1.
  • the ratio of the vaccine protein (e.g., gp96-lg) secretion to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is from about 2: 1 to about 50: 1 , inclusive of all endpoints.
  • a combination therapy in accordance with embodiments of the present disclosure involves the use of a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein, as well as a checkpoint inhibitor.
  • the fused biological cell can express a gp96-lg fusion protein and a T cell costimulatory fusion protein such as, without limitation, OX40L, and a checkpoint inhibitor such as, without limitation, checkpoint inhibitors that block immune checkpoints such as PD-1 , PD-L1 , PD-L2, IGOS, LAG3, TIM3, or CTLA-4.
  • checkpoint inhibitors are, without limitation, antibodies such as an anti-PD1 antibody, anti-PDL1 antibody, anti-PDL2 antibody, anti-ICOS antibody, anti-CTLA-4 antibody, anti-TIM-3 antibody, and/or anti-LAG-3 antibody.
  • checkpoint inhibitors include, without limitation, ipilimumab, pembrolizumab, nivolumab, pidilizumab, and others.
  • a tri-functional immunotherapy includes the use of a fused biological cell encoding a gp96-lg fusion protein and a T cell costimulatory fusion protein (e.g., without limitation, OX40L), and a checkpoint inhibitor such as, without limitation, a PD1 inhibitor.
  • a fused biological cell encoding a gp96-lg fusion protein and a T cell costimulatory fusion protein (e.g., without limitation, OX40L), and a checkpoint inhibitor such as, without limitation, a PD1 inhibitor.
  • the fused biological cell is made by fusing together at least two biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, and the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein.
  • a fused biological cell includes both a nucleotide sequence encoding a secretable vaccine protein and a nucleotide sequence encoding a T cell costimulatory fusion protein.
  • the fused biological cell can further include a nucleotide sequence encoding the one or more disease antigens, as a result of a fusion with a third biological cell.
  • the fused biological cell is made by fusing more than three biological cells together.
  • the fused biological cell is made by fusing four, five, six, or more than six biological cells.
  • the fused biological cell is made by fusing together at least three biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens.
  • the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens; wherein contacting the first biological cell and the second biological cell with the fusion agent further comprises contacting the third biological cell with the fusion agent, to result in the fused biological cell being created such that the fused biological cell comprises a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • the fused biological cell (created by fusing the first and second biological cells) is further fused with a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens, to thereby result in the fused biological cell (which can also be referred to as "a second fused biological cell”) that can express a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • a second fused biological cell which can express a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding one or more disease antigens, and contacting the third biological cell with the fusion agent, to result in a second fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
  • the fused cell increases tumor-specific immune response by simulating innate immunoactivation mechanisms as more types of tumor antigens are introduced into the fused cell.
  • the fused cell can display antigen presenting capacity to activate T cells due to the presence of antigen peptide MHC class I and class II molecules and costimulatory molecules.
  • the fused cell overexpress cancer tumor antigens in sync with T cell costimulatory molecules.
  • tumor antigens expressed by the fused cell be recognized by dendritic cells (DCs), triggering DC maturation followed by the T cell activation.
  • DCs dendritic cells
  • one or more of the first and second biological cells, or a fused biological cell secrete a variety of antigens.
  • antigens that can be secreted are: MART-1/Melan-A, gp100, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC)-0017- 1A/GA733, Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, aml1 , Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1 , PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), T-cell receptor/CD3-zeta chain, MAGE-family of tumor antigens (e.g., MAGE-A1 , MAGE-A2, MAGE-A
  • DPPIV Dipeptidyl peptid
  • the antigens are human endogenous retroviral antigens.
  • Illustrative antigens can also include antigens from human endogenous retroviruses which include, but are not limited to, epitopes derived from at least a portion of Gag, at least a portion of Tat, at least a portion of Rev, a least a portion of Nef, and at least a portion of gp160.
  • Various fused cells can be made in accordance with embodiments of the present disclosure.
  • one, two, or three biological cells are provided that include the nucleotide sequences in accordance with the present disclosure in various ways.
  • a nucleotide sequence encoding a secretable vaccine protein and a nucleotide sequence encoding a T cell costimulatory fusion protein are present in one biological cell, whereas a nucleotide sequence encoding one or more disease antigens is present on another biological cell.
  • a nucleotide sequence encoding a secretable vaccine protein and a nucleotide sequence encoding one or more disease antigens are present in one biological cell, whereas a nucleotide sequence encoding a T cell costimulatory fusion is present on another biological cell.
  • a nucleotide sequence encoding a T cell costimulatory fusion and a nucleotide sequence encoding one or more disease antigens are present in one biological cell, whereas a nucleotide sequence encoding a secretable vaccine protein is present on another biological cell.
  • the biological cells are contacted with a fusion agent to result in a fused cell in accordance with embodiments of the present disclosure.
  • a fused biological cell can be fused with another biological cell. Also, in embodiments, a fused biological cell can be fused with another fused biological cell.
  • one or more of the biological cells used to make a fused cell are immortalized. In embodiments, the fused biological cell is immortalized.
  • one or more of biological cells used to make a fused cell are derived from a human tumor cell line.
  • a human tumor cell line is a primary tumor cell line.
  • one or more of biological cells used to make a fused cell are Expi293F human cells that are derived from the 293 cell line.
  • a first biological cell comprising a nucleotide sequence encoding a vaccine protein comprises human kidney cells, e.g., Expi293 cells derived from the 293 cell line.
  • human tumor cells provide antigenic peptides that become non-covalently associated with the expressed gp96-lg fusion proteins.
  • Cell lines derived from a preneoplastic lesion, cancer tissue, or cancer cells also can be used, provided that the cells of the cell line have at least one or more antigenic determinant in common with the antigens on the target cancer cells.
  • Cancer tissues, cancer cells, cells infected with a cancer-causing agent, other preneoplastic cells, and cell lines of human origin can be used.
  • Cancer cells excised from the patient to whom the compositions in accordance with the present disclosure are ultimately to be administered can be particularly useful, although allogeneic cells can also be used.
  • one or more of the first, second, and third biological cells is fused with a human tumor cell.
  • one or more of the first, second, and third biological cells is a human tumor cell.
  • the human tumor cell is a cell from an established non-small cell lung cancer (NSCLC), bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
  • NSCLC non-small cell lung cancer
  • the human tumor cell is a cell from an established NSCLC cell line.
  • the NSCLC cell line is a human lung adenocarcinoma cell line (e.g., AD100).
  • the human tumor cell is a cell from a squamous cell carcinoma or large cell lung cancer cell line.
  • the third biological cell is a trophoblast. In embodiments, the third biological cell is fused with a trophoblast. In embodiments, one or both of the first and second biological cells is a trophoblast or is fused with a trophoblast.
  • the fused biological cell is immortalized. In embodiments, at least one of the first, second, and third biological cells is immortalized.
  • two or more biological cells that are fused are wild type, transfected, stable, transduced, or any combinations thereof.
  • the nucleotide sequence is a mammalian expression vector.
  • the nucleotide sequence from any of the first, second, and third biological cells can be a mammalian expression vector.
  • the expression vector comprises DNA. In embodiments, the expression vector comprises RNA. In embodiments, the expression vector is incorporated into a virus or virus-like particle.
  • the expression vector comprises a pCEP4-EGFP plasmid DNA.
  • prokaryotic and eukaryotic vectors can be used for expression of a secretable vaccine protein (e.g., gp96-lg) and T cell costimulatory fusion proteins in biological cells provided herein.
  • Prokaryotic vectors include constructs based on E. coli sequences (see, e.g., Makrides, Microbiol Rev 1996, 60:512-538).
  • Non-limiting examples of regulatory regions that can be used for expression in E. coli include lac, trp, Ipp, phoA, recA, tac, T3, T7 and APL.
  • Non-limiting examples of prokaryotic expression vectors may include the Agt vector series such as Agt11 (Huynh et al., in "DNA Cloning Techniques, Vol.
  • Prokaryotic host-vector systems cannot perform much of the post-translational processing of mammalian cells, however. Thus, eukaryotic host-vector systems may be particularly useful.
  • various fusion (or fusogenic) agents can be used to allow two or more biological cells to fuse.
  • the fusion agent comprises a protein from Paramyxoviridae Genus paramyxovirus.
  • the protein can be, for example, hemagglutinating virus of Japan (HVJ) envelope (HVJ-E).
  • HVJ is also known as Sendai virus. See, e.g., Okada (1993) Methods Enzymol. 221 : 18—41 .
  • the HVJ-E is a nonproliferative and noninfectious vesicle purified after inactivation of Sendai virus genomic RNA. See, e.g., Kaneda et al. (2002) Molecular Therapy 6, 219-226; see also Lund et al. (2010) Pharm Res. 27(3):400-20, the entire contents of which are hereby incorporated by reference.
  • the fusion agent comprises a membrane fusion protein from an envelope of a virus selected from Orthomyxoviridae, Herpesviridae, Hepadnaviridae, and Flaviviridae.
  • the fusion agent comprises a virosome derived from the influenza virus (Orthomyxovirus) or the Sendai virus.
  • a virosome is defined as a vector derived from an inactive enveloped virus or a reconstituted envelope that contains viral envelope proteins or viral envelope particles. Saga & Kaneda (2013). BioMed research international, 2013, 764706.
  • the fusion agent comprises a Sendai virosome containing pCEP4-EGFP plasmid.
  • the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof.
  • PEG poly(ethyleneglycol)
  • the PEG moiety comprises PEG-1500.
  • the fusion agent comprises PEG-300, PEG- 400, PEG-550, PEG-550, or PEG-1000, and other PEG molecules having an average molecular weight of less than 2000. In embodiments, PEG molecules have an average molecular weight of greater than 2000.
  • cell fusion is performed using electrofusion. See, e.g., Skelley et al. (2009) Nat. Methods 6: 147-152; Kjaergaard et al. (2003) Cell. Immunol. 225:65-74; Trevor et al. (2004) Cancer Immunol. Immunother. 53:705-714; Scott-Taylor et al. (2000) Biochim. Biophys. Acta. 2000; 1500:265-279.
  • one or both the first biological cell and the second biological cell comprise at least one tumor antigen.
  • the tumor antigen is a cancer testis (CT) antigen.
  • CT cancer testis
  • the fused biological cell expresses one or more CT antigens.
  • one or more of the first, second, and third biological cells expresses one or more CT antigens.
  • the fused biological cell is enriched for genes encoding one or more CT antigens.
  • one or more of the first, second, and third biological cells expresses one or more CT antigens.
  • the fused biological cell further comprises a nucleotide sequence encoding one or more disease antigens.
  • the one or more disease antigens comprise one of more infectious disease antigens.
  • This approach can be used in developing compositions (e.g., vaccines) for treatment of disease vaccines, by fusing cancer cells with other biological cells expressing either mini-genes or bacterial or viral proteins capable of driving B and T cell responses.
  • a "mini-gene” is a part of a disease antigen, such as an immunogenic portion of the disease antigen which is sufficient to induce immune response specific to the disease antigen.
  • the fused cell in accordance with the present disclosure leverages gp96 to effectively present one or more disease antigens and activate the immune system.
  • the gp96-based system utilizes natural immune process to induce long-lasting memory responses and can effectively present multiple antigens and activate the immune system.
  • the described methods and compositions aim to trigger mucosal immunity by activating both B and T cell responses at the point of pathogen entry.
  • the one or more infectious disease antigens comprise an antigenic fragment of a betacoronavirus protein or an alphacoronavirus protein, optionally wherein the betacoronavirus protein is selected from a SARS-CoV- 2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43 protein, or the alphacoronavirus protein is selected from a HCoV-NL63 and HCoV-229E protein.
  • the betacoronavirus protein is a SARS-CoV-2 protein, also called 2019-nCoV protein.
  • the SARS-CoV-2 protein comprises an amino acid encoded by the nucleic acid of SEQ ID NO: 38, or an antigenic fragment thereof.
  • the SARS-CoV-2 protein comprises the amino acid that encompasses an amino acid of sequence of SEQ ID NO: 39, an amino acid of sequence of SEQ ID NO: 40, an amino acid of sequence of SEQ ID NO: 41 , an amino acid of sequence of SEQ ID NO: 42, an amino acid of sequence of SEQ ID NO: 43, an amino acid of sequence of SEQ ID NO: 44, an amino acid of sequence of SEQ ID NO: 45, and an amino acid of sequence of SEQ ID NO: 46, or an antigenic fragment thereof.
  • the SARS-CoV-2 is selected from a spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N.
  • the coronavirus protein is a SARS-CoV-2 protein, or an antigenic fragment thereof, selected from the spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N.
  • the spike surface glycoprotein comprises the amino acid sequence of SEQ ID NO: 39
  • the envelope protein E comprises the amino acid sequence of SEQ ID NO: 41
  • the membrane glycoprotein precursor M comprises the amino acid sequence of SEQ ID NO: 42
  • the nucleocapsid phosphoprotein N comprises the amino acid sequence of SEQ ID NO: 46, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity with any of the foregoing, or an antigenic fragment of any of the foregoing.
  • the spike protein comprises the following amino acid sequence:
  • the envelope protein comprises the following amino acid sequence:
  • the membrane protein comprises the following amino acid sequence: MADSNGTITVEELKKLLEQWNLVIGFLFLTWICLLQFAYANRNRFLYIIKLIFLWLLWPVTLACFVLAAVYRINWITGGIAI AMACLVGLMWLSYFIASFRLFARTRSMWSFNPETNILLNVPLHGTILTRPLLESELVIGAVILRGHLRIAGHHLGRCDIK DLPKEITVATSRTLSYYKLGASQRVAGDSGFAAYSRYRIGNYKLNTDHSSSSDNIALLVQ (SEQ ID NO: 42).
  • the nucleocapsid phosphoprotein comprises the following amino acid sequence:
  • the 2019-nCoV protein may comprise an amino acid sequence that has at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71%, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81 %, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 92%, or at least
  • the SARS-CoV-2 protein may comprise an amino acid sequence that has at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71 %, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81%, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 92%, or at
  • a SARS-CoV-2 protein portion can encode an amino acid sequence that differs from any known wild type amino acid sequence of the SARS-CoV-2 protein or a SARS-CoV-2 amino acid sequence disclosed herein having a mutation, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms.
  • a fused biological cell can be made in various ways.
  • cell fusion between two biological cells can be performed using a method that is similar to conventional method for fusing immune cells and myeloma cells, for example, as described in Milstein et al. Methods Enzymol., 73, 3-46 (1981)).
  • two or more biological cells that are fused are wild type, transfected, stable, transduced cells, or any combinations thereof.
  • a fused biological cell is prepared in a conventional nutrient medium in the presence of a fusogenic or fusion agent.
  • the fusion promoter can be polyethylene glycol (PEG), Hemagglutinating virus (HVJ), or another type of a fusion promoter.
  • an auxiliary agent such as, e.g, dimethylsulfoxide, can be used to enhance fusion efficiency.
  • a fusion agent comprises a protein suitable to cause a protein-mediated membrane fusion.
  • a protein can be a membrane fusion protein of an inactivated virus, such as an envelope protein of an inactivated virus.
  • Viruses having an envelope comprising a membrane fusion protein include Paramyxoviridae, Orthomyxoviridae, Herpesviridae, Hepadnaviridae and Flaviviridae viruses. These viruses utilize their membrane fusion activity to infect host cells through fusion with the plasma membranes of the host cells.
  • An "inactivated” virus can be defined as a virus with inactivated genes such that the virus can no longer replicate its genes and has lost its proliferation potency and infectivity.
  • a virus can be inactivated using UV irradiation, irradiation of radioactive rays, or treatment with an alkylating agent, or a combination thereof.
  • a virus envelope generally is formed of small spike-like structures made of spike proteins encoded by a virus gene, and a lipid bilayer derived from the host and that does not have proliferation ability.
  • Spike proteins constituting the small spikes comprise a membrane fusion protein which functions to confer a membrane fusion activity.
  • an inactivated virus envelope having membrane fusion activity can be used for making a fused biological cell.
  • the fusion agent comprises a membrane fusion protein from an envelope of an inactivated virus selected from Orthomyxoviridae, Herpesviridae, Hepadnaviridae, and Flaviviridae.
  • a fusion agent comprises a protein from Paramyxoviridae Genus paramyxovirus.
  • the protein can be, for example, hemagglutinating virus of Japan (HVJ) envelope (HVJ-E).
  • HVJ also known as Sendai virus, an enveloped virus with a nonsegmented negative-strand RNA genome. See, e.g., Okada (1993) Methods Enzymol. 221 :18-41.
  • the HVJ-E is a nonproliferative and noninfectious vesicle purified after inactivation of Sendai virus genomic RNA. See, e.g., Kaneda ef a/. (2002) Molecular Therapy 6, 219-226; see a/so Lund etal. (2010) Pharm Res. 27(3):400- 20, the entire contents of which are hereby incorporated by reference.
  • the fusion agent comprises inactivated Sendai virus envelope protein.
  • the fusion agent is inactivated Sendai virus HVJ-E.
  • the fusion agent comprises a Sendai virosome comprising pCEP4-EGFP plasmid.
  • Sendai virus can be cultivated, for example, in chicken fertilized eggs or by persistent infection in mammalian cell or tissue cultures, which can include a hydrolase such as trypsin.
  • An inactivated virus envelope having membrane fusion activity can be prepared using ultracentrifugation, isolation by column chromatography, reconstitution and the like. Isolation can be carried out by ion chromatography, as described, for example, in Example 1 (1) and Examples 7 (1) to (5) of U.S. Patent Application Publication No. 20040253272, which is incorporated herein by reference in its entirety.
  • a freeze-dried composition of an inactivated virus envelope having membrane fusion activity can be prepared, e.g., as described in U.S. Patent No. 8,043,610, which is incorporated herein by reference in its entirety.
  • the freeze-dried composition can be prepared, for example, by adding a protein hydrolysate, an amino acid or a polysaccharide at a predetermined concentration, and optionally adding a known pH adjusting agent, a known isotonicity agent, a known stabilizer or a known preservative, to the virus envelope having membrane fusion activity above.
  • the protein hydrolysate, the amino acid or the polysaccharide may be dissolved in aqueous solvent(s) ⁇ e.g., distilled water, a buffer solution, etc.) respectively before added at predetermined concentrations to the virus envelope having membrane fusion activity.
  • aqueous solvent(s) e.g., distilled water, a buffer solution, etc.
  • the resulting mixture can be freeze-dried by ordinary methods.
  • a known method such as tray freeze-drying, spray freeze-drying or vial freeze- drying under ordinarily used conditions may be used.
  • an antibiotic-containing media is used for cell fusion of biological cells.
  • the media comprises one or more of G418 (Geneticin), Puromycin, & Hygromycin.
  • the antibiotic-containing media comprises from about 100 ug/mL (micrograms per milliliter) to about 800 ug/mL of one or more of an antibiotic. In embodiments, the media comprises from about 100 ug/mL to about 1000 ug/mL of one or more antibiotic. In embodiments, the media comprises about 100 ug/mL, or about 200 ug/mL, or about 300 ug/mL, or about 400 ug/mL, or about 500 ug/mL, or about 600 ug/mL, or about 700 ug/mL, or about 800 ug/mL, or about 900 ug/mL, or about 1000 ug/mL of an antibiotic.
  • the antibiotic-containing media comprises about 500 ug/mL G418 and about 200 ug/mL Hygromycin.
  • a fused biological cell is resuspended in a buffering agent such as, without limitation, PBS (phosphate- buffered saline) or tris-buffered saline (TBS).
  • a buffering agent comprises, without limitation, one or more of PBS, triethanolamine, Tris, bicine, TAPS, tricine, HEPES, TES, MOPS, PIPES, cacodylate, MES, acetate, citrate, succinate, histidine or other pharmaceutically acceptable buffers.
  • the buffering agent in embodiments includes Bovine serum albumin (BSA).
  • BSA Bovine serum albumin
  • the buffering agent comprises PBS including from about 1 % to about 10% BSA.
  • the buffering agent comprises PBS including about 1 % BSA.
  • the buffering agent comprises TBS including from about 1 % to about 10% BSA.
  • the buffering agent comprises TBS including about 1 % BSA.
  • the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof.
  • PEG poly(ethyleneglycol)
  • the PEG moiety comprises PEG-1500.
  • the fusion agent comprises PEG-300, PEG- 400, PEG-550, PEG-550, or PEG-1000, and other PEG molecules having an average molecular weight of less than 2000. In embodiments, PEG molecules have an average molecular weight of greater than 2000.
  • cell fusion is performed using electrofusion. See, e.g., Skelley et al. (2009) Nat. Methods 6: 147-152; Kjaergaard et al. (2003) Cell. Immunol. 225:65-74; Trevor et al. (2004) Cancer Immunol. Immunother. 53:705-714; Scott-Taylor et al. (2000) Biochim. Biophys. Acta. 2000; 1500:265-279.
  • a non-limiting example of cell fusion is as follows.
  • AD-100 (the 293 Cell Line) transduced (OX40L, 10E5 cells) cells and transfected human kidney cells (Expi293 cells expressing gp96, 10E5 cells) are seeded in 12- well plates and grown for 24 hours at 37°C.
  • AD-100 transduced (OX40L, 10E5 cells) cells or transfected human kidney cells (Expi293 cells expressing gp96, 10E5 cells) are seeded in 12-well plates and grown for 24 hours at 37°C.
  • the gp96 can be native gp96 or a gp96-lg fusion protein.
  • the cells are washed and replaced with antibiotic- free media and then incubated at 37°C in the presence of Sendai virosomes containing pCEP4-EGFP plasmid DNA. After de novo protein expression and a 24-hour time period of transfection, serum-free wells are replaced by antibioticcontaining media (500 ug/mL G418 and 200 ug/mL Hygromycin), and the cells are then incubated further for 48 hours at 37°C and 5% CO 2 . After 48 hours of incubation, the fused cells are harvested and then resuspended in 500 uL of PBS containing 1 % BSA and evaluated by immunofluorescence.
  • antibioticcontaining media 500 ug/mL G418 and 200 ug/mL Hygromycin
  • the disclosure provides sequential fusion combination ⁇ e.g., a single cell is transduced with gp96-lg, and a single cell is transduced with T cell costimulatory fusion protein, e.g. OX40L-lg, then the cells are fused together).
  • sequential fusion combination ⁇ e.g., a single cell is transduced with gp96-lg, and a single cell is transduced with T cell costimulatory fusion protein, e.g. OX40L-lg, then the cells are fused together).
  • the provided methods result in considerably greater expression and/or secretion (e.g. about or at least about 2-fold, or about or at least about 3-fold, or about or at least about 4-fold, or about or at least about 5-fold, or about or at least about 6-fold, or about or at least about 7-fold, or about or at least about 8-fold, or about or at least about 9-fold, or about or at least about 10-fold, or about or at least about 15-fold, or about or at least about 20-fold, or about or at least about 50% greater, or about or at least about 75% greater, or about or at least about 100% greater, or about or at least about 200% greater, or about or at least about 300% greater, or about or at least about 400% greater, or about or at least about 500% greater) of secretable vaccine protein (e.g., without limitation, gp96-l g) and/or T cell costimulatory fusion protein (e.g., without limitation OX40L-lg), as compared to an unfused cell that
  • the disclosure relates to a method for generating a cellular therapy, comprising: (a) obtaining a lentivirus, the lentivirus comprising: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject; and (b) introducing into a biological cell the lentivirus of step (a).
  • the disclosure relates to a method for generating a cellular therapy, comprising: (a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a first biological cell; (b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject, and (i) introducing the nucleic acid into a second biological cell, and (c) merging together the first biological cell and the second biological cell.
  • Lentiviruses or lentiviral vectors are powerful tools to ensure stable and long-term gene expression by transfer of genes into dividing and non-dividing cells.
  • lentiviral expression vectors are based on human immunodeficiency virus- 1 (HIV-1).
  • Therapeutic lentiviruses have been modified from the original HIV- 1 genome to avoid replication in host cells.
  • the components necessary for virus production are split across multiple plasmids.
  • the lentivirus is produced using either a second generation lentiviral packaging system, in which the lentivirus components are split across three plasmids, or a third generation lentiviral packaging system, in which the lentivirus components are split across four plasmids.
  • the lentivirus is produced using a third-generation lentiviral packaging system.
  • the lentiviral packaging system comprises a transfer plasmid, a packaging plasmid, and an envelope plasmid.
  • the packaging plasmid comprises two plasmids.
  • the packaging plasmid encodes a Gag and Pol gene.
  • the packaging plasmid encodes a Rev gene.
  • the envelope plasmid comprises a VSV-G envelope protein.
  • the transfer plasmid comprises a transgene, or a nucleic acid of interest.
  • the transfer plasmid comprises: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject.
  • the sequence for the transgene or the nucleic acid of interest is flanked by long terminal repeat (LTR) sequences, which facilitate integration of the transfer plasmid sequences into the host genome. Typically, it is the sequences between and including the LTRs that are integrated into the host genome upon viral transduction.
  • LTR long terminal repeat
  • the lentivirus is replication incompetent.
  • the lentivirus comprises a deletion in a long terminal repeat (LTR) sequence, rendering it self-inactivating (SIN) after integration.
  • the transfer plasmid is optimized.
  • the transfer plasmid utilizes a hybrid LTR promoter.
  • Tat is eliminated from the third-generation lentiviral packaging system through the addition of a chimeric 5' LTR fused to a heterologous promoter on the transfer plasmid.
  • the expression of the transgene from this promoter is no longer dependent on Tat transactivation.
  • the transfer plasmid comprises additional or specialized promoters, such as a U6 promoter.
  • the transfer plasmid comprises Tet- or Cre-based regulation element.
  • the transfer plasmid comprises fluorescent fusions or reporters.
  • Lentiviruses are produced using established processes that are well known in the art.
  • the lentivirus is produced by contacting a cell with the third-generation lentiviral packaging system and isolating the lentivirus from the cell.
  • the cell contacted is a host cell, such as an A293T cell.
  • the one transfer plasmid, one or two packaging plasmids, and one envelope plasmid are transfected into a host cell.
  • supernatant containing the lentivirus is removed and stored or centrifuged to concentrate the lentivirus.
  • crude or concentrated lentivirus can then be introduced into the cells of interest.
  • Lentiviruses can be used to make stable cell lines in the same manner as standard retroviruses. For instance, many lentiviral genomes have selectable markers, such as the puromycin resistance gene, conferring antibiotic resistance to infected host cells. When selection antibiotics are added to the growth medium of the host cells, they kill off any cells that have not incorporated the lentiviral genome and those clonal cells that survive can be expanded to create stable cell lines, which have incorporated the lentiviral genome and harbor the genetic information encoded by that genome.
  • selectable markers such as the puromycin resistance gene
  • the lentivirus and/or transfer plasmid comprises one or more selection markers, optionally selected from puromycin, neomycin, zeocin, and hygromycin.
  • the selection marker is a gene product which confers resistance to an antibiotic, including but not limited to ampicillin, kanamycin, neomycin/G418, tetracycline, geneticin, triclosan, puromycin, zeocin, and hygromycin.
  • the selection marker is one or more of kanamycin resistance genes, puromycin resistance genes, zeocin resistance genes, neomycin/G418 resistance genes, hygromycin resistance genes, histidinol resistance genes, tetracycline resistance genes, geneticin resistance genes, triclosan resistance genes, R-fluroorotic acid resistance genes, 5-fluorouracil resistance genes and ampicillin resistance genes. Combination of any of the selection markers described herein is contemplated.
  • each plasmid comprises a selection marker.
  • each plasmid has the same selection marker.
  • each plasmid within the system comprises a different selection marker.
  • lentiviral transfer plasmids do not have selectable markers conferring resistance to an antibiotic, but do encode another marker, such as GFP.
  • FACS is used to sort cells expressing GFP, wherein these cells are later expanded into a cell line.
  • high titer lentiviruses can be introduced into cancer cell lines, such as AD-100 or HEK293, to express dominant B and T cell epitopes.
  • techniques suitable for the transfer of nucleic acids into mammalian cells in vitro are used, including the use of liposomes, electroporation, microinjection, cell fusion, polymer-based systems, DEAE-dextran, viral transduction, the calcium phosphate precipitation method, etc.
  • a number of techniques and reagents are used, including electroporation, liposomes; natural polymer-based delivery vehicles, such as chitosan and gelatin.
  • viral vectors are used for in vivo transduction.
  • multiplicity of infection is optimized to for use in: (I) manufacturing, when many billions of cells are needed for infectious disease vaccination, or (II) generating stable cell lines containing three or more proteins.
  • the high titer of the lentiviruses is about 0.5x10 8 transducing units (TU)/ ml. In embodiments, the high titer of the lentiviruses is about 1x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 1.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 2x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 2.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 3x10 8 TU/ml.
  • the high titer of the lentiviruses is about 3.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 4x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 4.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is about 5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is between about 0.5x10 8 TU/ml to about 1x10 8 TU/ml.
  • the high titer of the lentiviruses is between about 1x10 8 TU/ml to about 1.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is between about 1.5x10 8 TU/ml to about 2x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is between about 2x10 8 TU/ml to about 2.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is between about 2.5x10 8 TU/ml to about 3x10 8 TU/ml.
  • the high titer of the lentiviruses is between about 3x10 8 TU/ml to about 3.5x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is between about 3.5x10 8 TU/ml to about 4x10 8 TU/ml. In embodiments, the high titer of the lentiviruses is between about 4x10 8 TU/ml to about 4.5x10 8 TU/ml.
  • the lentivirus and/or transfer plasmid of the present disclosure comprises a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof.
  • chaperone proteins chaperone antigens to the immune system for MHC-l-mediated antigen crosspresentation, leading to a robust cytotoxic CD8+ T cell response.
  • the chaperone protein is selected from the group consisting of: gp96, Hsp70, BiP, and Grp78.
  • the chaperone protein comprises an amino acid sequence of any one of SEQ ID NOs: 1 , 2, 3, and 4, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the amino acid sequence (SEQ ID NO: 2) and nucleic acid sequence (SEQ ID NO: 1) of chaperone protein, gp96, are provided below:
  • the chaperone protein is gp96.
  • the coding region of human gp96 is 2,412 bases in length, and encodes an 803 amino acid protein that includes a 21 amino acid signal peptide at the amino terminus, a potential transmembrane region rich in hydrophobic residues, and an ER retention peptide sequence at the carboxyl terminus (GEN BANK® Accession No. X15187; see, Maki ef a/., Proc Natl Acad Sc/ USA 1990, 87:5658-5562).
  • the chaperone protein is gp96 comprising the amino acid sequence of SEQ ID NO: 2.
  • the chaperone protein is gp96 comprising the nucleic acid sequence of SEQ ID NO: 1.
  • any of the nucleic acid sequences described herein may be codon optimized.
  • the chaperone protein of the secretable fusion protein is a secretable gp96-lg fusion protein which optionally lacks the gp96 KDEL (SEQ ID NO: 3) sequence.
  • gp96 genetically fused to an immunoglobulin domain (e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule), activates TLR2 and TLR4 on professional antigen-presenting cells (APCs).
  • an immunoglobulin domain e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule
  • the gp96 portion of the secretable fusion protein comprises an amino acid sequence that has at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71%, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81 %, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 9
  • the gp96 portion of nucleic acid encoding a gp96-lg fusion polypeptide can encode an amino acid sequence that differs from the wild type gp96 polypeptide at one or more amino acid positions, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms as described previously.
  • the chaperone protein may be a heat shock protein.
  • the heat shock protein is one or more of hsp40, hsp60, hsp70, hsp90, and hsp110 family members, inclusive of fragments, variants, mutants, derivatives or combinations thereof (Hickey, et al., 1989, Mol. Cell. Biol. 9:2615-2626; Jindal, 1989, Mol. Cell. Biol. 9:2279-2283).
  • the secretable fusion protein comprises an immunoglobulin or antibody.
  • the antibody may be any type of antibody, i.e., immunoglobulin, known in the art.
  • the antibody is an antibody of class or isotype IgA, IgD, IgE, IgG, or IgM.
  • the antibody described herein comprises one or more alpha, delta, epsilon, gamma, and/or mu heavy chains.
  • the antibody described herein comprises one or more kappa or light chains.
  • the antibody is an IgG antibody and optionally is one of the four human subclasses: lgG1, lgG2, lgG3 and lgG4.
  • the antibody In embodiments is a monoclonal antibody. In other embodiments, the antibody is a polyclonal antibody. In embodiments, the antibody is structurally similar to or derived from a naturally-occurring antibody, e.g, an antibody isolated and/or purified from a mammal, e.g., mouse, rabbit, goat, horse, chicken, hamster, human, and the like.
  • the antibody may be considered as a mammalian antibody, e.g., a mouse antibody, rabbit antibody, goat antibody, horse antibody, chicken antibody, hamster antibody, human antibody, and the like.
  • the antibody comprises sequence of only mammalian antibodies.
  • the antibody is a humanized antibody, such as a humanized mouse, rabbit, goat, horse, chicken, hamster, or human antibody.
  • the antibody is a humanized monoclonal antibody.
  • the antibody is an antibody or antibody-like molecule such as a single-domain antibody, a recombinant heavy-chain-only antibody (VHH), a single-chain antibody (scFv), a shark heavy-chain-only antibody (VNAR), a microprotein (cysteine knot protein, knottin), a DARPin; a Tetranectin; an Affibody; a Transbody; an Anticalin; an AdNectin; an Affilin; a Microbody; a peptide aptamer; an alterase; a plastic antibody; a phylomer; a stradobody; a maxibody; an evibody; a fynomer, an armadillo repeat protein, a Kunitz domain, an avimer, an atrimer, a probody, an immunobody, a triomab, a troybody; a pepbody; a vaccibody, a UniBody; Af
  • the secretable fusion protein comprises a scaffold protein.
  • the scaffold protein is Affilin, Min-23, and/or Kunitz domain.
  • the fusion protein comprises a fragment of an immunoglobulin or antibody.
  • Antibody fragments include, but are not limited to, the F(ab')2 fragment which may be produced by pepsin digestion of the antibody molecule; the Fab' fragments which may be generated by reducing the disulfide bridges of the F(ab')2 fragment, and the two Fab' fragments which may be generated by treating the antibody molecule with papain and a reducing agent.
  • a gp96 peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen et al., J Immunol 1996, 156:442-449).
  • This region of the lgG1 molecule contains three cysteine residues that normally are involved in disulfide bonding with other cysteines in the Ig molecule. Since none of the cysteines are required for the peptide to function as a tag, one or more of these cysteine residues can be substituted by another amino acid residue, such as, for example, serine.
  • the secretable fusion protein comprises an Fc fragment of an immunoglobulin.
  • the Fc fragment comprises the amino acid sequence of SEQ ID NO: 51 , or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the immunoglobulin is an lgG1 immunoglobulin.
  • the immunoglobulin comprises an Ig tag of the gp96-lg fusion protein comprising the Fc region of human I gG 1 , lgG2, 1 gG3, lgG4, IgM, IgA, or Ig E.
  • Such Ig tags typically include at least functional CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain.
  • a T cell costimulatory peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen etal., J Immunol 1996, 156:442-449).
  • the Ig tag can be from a mammalian (e.g, human, mouse, monkey, or rat) immunoglobulin, but human immunoglobulin can be particularly useful when the fusion protein is intended for in vivo use for humans.
  • the secretable fusion protein comprises the amino acid sequence of SEQ ID NO: 51 , or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
  • the amino acid sequences of an Fc fragment of an lgG1 antibody (SEQ ID NO: 50) and of gp96 fused to an Fc fragment of an lgG1 antibody (SEQ ID NO: 51) are provided below:
  • the Fc fragment can have an amino acid sequence that differs from the Fc fragment polypeptide at one or more amino acid positions, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms as described herein.
  • the gp96-lg fusion protein further comprises a linker.
  • the linker may be derived from naturally-occurring multi-domain proteins or are empirical linkers as described, for example, in Chichili et al., (2013), Protein Sci. 22(2): 153-167, Chen et al., (2013), Adv Drug Deliv Rev. 65(10):1357-1369, the entire contents of which are hereby incorporated by reference.
  • the linker may be designed using linker designing databases and computer programs such as those described in Chen et al., (2013), Adv Drug Deliv Rev. 65(10): 1357-1369 and Crasto et. al., (2000), Protein Eng.
  • the linker is a synthetic linker such as PEG. In other embodiments, the linker is a polypeptide. In embodiments, the linker is less than about 100 amino acids long. For example, the linker may be less than about 100, about 95, about 90, about 85, about 80, about 75, about 70, about 65, about 60, about 55, about 50, about 45, about 40, about 35, about 30, about 25, about 20, about 19, about 18, about 17, about 16, about 15, about 14, about 13, about 12, about 11 , about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, or about 2 amino acids long. In embodiments, the linker is flexible. In another embodiment, the linker is rigid.
  • the linker is substantially comprised of glycine and serine residues (e.g. about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 95%, or about 97% glycines and serines).
  • the linker is a hinge region of an antibody (e.g., of IgG, IgA, IgD, and IgE, inclusive of subclasses (e.g. lgG1, lgG2, lgG3, and lgG4, and lgA1 and lgA2)).
  • the hinge region found in IgG, IgA, IgD, and IgE class antibodies, acts as a flexible spacer, allowing the Fab portion to move freely in space.
  • the hinge domains are structurally diverse, varying in both sequence and length among immunoglobulin classes and subclasses. For example, the length and flexibility of the hinge region varies among the IgG subclasses.
  • the hinge region of lgG1 encompasses amino acids 216-231 and, because it is freely flexible, the Fab fragments can rotate about their axes of symmetry and move within a sphere centered at the first of two inter-heavy chain disulfide bridges.
  • I gG2 has a shorter hinge than lgG1 , with 12 amino acid residues and four disulfide bridges.
  • the hinge region of lgG2 lacks a glycine residue, is relatively short, and contains a rigid poly-proline double helix, stabilized by extra inter-heavy chain disulfide bridges. These properties restrict the flexibility of the lgG2 molecule.
  • lgG3 differs from the other subclasses by its unique extended hinge region (about four times as long as the lgG1 hinge), containing 62 amino acids (including 21 prolines and 11 cysteines), forming an inflexible poly-proline double helix.
  • the Fab fragments are relatively far away from the Fc fragment, giving the molecule a greater flexibility.
  • the elongated hinge in lgG3 is also responsible for its higher molecular weight compared to the other subclasses.
  • the hinge region of lgG4 is shorter than that of lgG1 and its flexibility is intermediate between that of IgG 1 and I gG2. The flexibility of the hinge regions reportedly decreases in the order lgG3>lgG 1 >lgG4>lgG2.
  • the linker may be functional.
  • the linker may function to improve the folding and/or stability, improve the expression, improve the pharmacokinetics, and/or improve the bioactivity of the present compositions.
  • the linker may function to target the compositions to a particular cell type or location.
  • Fused biological cells generated in accordance with embodiments of the present disclosure can be used in various methods of treatment of cancer and infections.
  • the infection can be, for example, an acute infection or a chronic infection.
  • the infection can be an infection by coronavirus, hepatitis C virus, hepatitis B virus, human immunodeficiency virus, or malaria.
  • the methods can include administering to a subject a fused biological cell, under conditions wherein the progression or a symptom of a clinical condition in the subject is reduced in a therapeutic manner.
  • the present disclosure pertains to treatment of cancers and/or tumors.
  • Cancers or tumors refer to an uncontrolled growth of cells and/or abnormal increased cell survival and/or inhibition of apoptosis which interferes with the normal functioning of the bodily organs and systems. Included are benign and malignant cancers, polyps, hyperplasia, as well as dormant tumors or micrometastases. Also, included are cells having abnormal proliferation that is not impeded by the immune system ⁇ e.g. virus infected cells).
  • the cancer may be a primary cancer or a metastatic cancer.
  • the primary cancer may be an area of cancer cells at an originating site that becomes clinically detectable, and may be a primary tumor.
  • the metastatic cancer may be the spread of a disease from one organ or part to another non-adjacent organ or part.
  • the metastatic cancer may be caused by a cancer cell that acquires the ability to penetrate and infiltrate surrounding normal tissues in a local area, forming a new tumor, which may be a local metastasis.
  • the cancer may also be caused by a cancer cell that acquires the ability to penetrate the walls of lymphatic and/or blood vessels, after which the cancer cell is able to circulate through the bloodstream (thereby being a circulating tumor cell) to other sites and tissues in the body.
  • the cancer may be due to a process such as lymphatic or hematogeneous spread.
  • the cancer may also be caused by a tumor cell that comes to rest at another site, repenetrates through the vessel or walls, continues to multiply, and eventually forms another clinically detectable tumor.
  • the cancer may be this new tumor, which may be a metastatic (or secondary) tumor.
  • the cancer may be caused by tumor cells that have metastasized, which may be a secondary or metastatic tumor.
  • the cells of the tumor may be like those in the original tumor.
  • the secondary tumor while present in the liver, is made up of abnormal breast or colon cells, not of abnormal liver cells.
  • the tumor in the liver may thus be a metastatic breast cancer or a metastatic colon cancer, not liver cancer.
  • the cancer may have an origin from any tissue.
  • the cancer may originate from melanoma, colon, breast, or prostate, and thus may be made up of cells that were originally skin, colon, breast, or prostate, respectively.
  • the cancer may also be a hematological malignancy, which may be lymphoma.
  • the cancer may invade a tissue such as liver, lung, bladder, or intestinal.
  • Illustrative cancers that may be treated include, but are not limited to, carcinomas, e.g. various subtypes, including, for example, adenocarcinoma, basal cell carcinoma, squamous cell carcinoma, and transitional cell carcinoma), sarcomas (including, for example, bone and soft tissue), leukemias (including, for example, acute myeloid, acute lymphoblastic, chronic myeloid, chronic lymphocytic, and hairy cell), lymphomas and myelomas (including, for example, Hodgkin and non-Hodgkin lymphomas, light chain, non-secretory, MGUS, and plasmacytomas), and central nervous system cancers (including, for example, brain ⁇ e.g.
  • gliomas ⁇ e.g. astrocytoma, oligodendroglioma, and ependymoma
  • meningioma ⁇ e.g. astrocytoma, oligodendroglioma, and ependymoma
  • meningioma ⁇ e.g. astrocytoma, oligodendroglioma, and ependymoma
  • meningioma ⁇ e.g. astrocytoma, oligodendroglioma, and ependymoma
  • meningioma ⁇ e.g. astrocytoma, oligodendroglioma, and ependymoma
  • meningioma ⁇ e.g. astrocytoma, oligodendroglioma, and ependymoma
  • meningioma ⁇ e.
  • compositions and methods in accordance with embodiments of the present disclosure activate an innate, humoral (i.e. antibody response), and/or cellular (i.e. T cell) response in a subject receiving the present compositions.
  • a method is suitable for increasing the subject's T-cell response as compared to the T-cell response of a subject that was not administered the compositions in accordance with embodiments of the present disclosure.
  • the method is suitable for increasing the subject's antibody response as compared to the antibody response of a subject that was not administered the present compositions.
  • the method is suitable for increasing the subject's innate immune response as compared to the innate immune response of a subject that was not administered the present compositions.
  • the method is suitable for increasing the subject's T-cell response, antibody response, and innate immune response as compared to the T-cell response, antibody response, and innate immune responses of a subject that was not administered the present compositions.
  • the method is suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell population(s) of a subject that was not administered the present compositions.
  • the subject's T cells include T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells.
  • the method is suitable for increasing and/or restoring the subject's CD4+ helper cells population(s) as compared to the CD4+ helper cells population(s) of a subject that was not administered the present compositions.
  • the method is suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell population(s) of a subject that was administered another composition.
  • the subject's T cells include T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells.
  • the method is suitable for increasing and/or restoring the subject's CD4+ helper cells population(s) as compared to the CD4+ helper cells population(s) of a subject that was administered another composition.
  • a method of treating cancer comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • the cancer comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia.
  • the cancer is selected from one or more of the basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer; glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer; melanoma; myeloma; neuroblastoma; oral cavity cancer; ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular
  • the cancer is selected from one or more of basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer;
  • a method of treating or preventing an infectious disease comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • the infectious disease comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection.
  • the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae.
  • the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E.
  • the infectious disease comprises a coronavirus infection 2019 (COVID-19).
  • a fused biological cell in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell is used to eliminate intracellular pathogens.
  • the fused biological cells are used to treat one or more infections.
  • methods of treating viral infections are provided, including, for example, HIV and HCV; parasitic infections (including, for example, malaria); and bacterial infections.
  • the infections induce immunosuppression.
  • HIV infections often result in immunosuppression in the infected subjects.
  • the treatment of such infections may involve, in embodiments, modulating the immune system with the present fused biological cell to favor immune stimulation over immune inhibition.
  • embodiments of the present disclosure provide methods for treating infections that induce immunoactivation.
  • intestinal helminth infections have been associated with chronic immune activation.
  • the treatment of such infections may involve modulating the immune system with the present fused biological cell to favor immune inhibition over immune stimulation.
  • the viral infections include, without limitation, acute or chronic viral infections, for example, of the respiratory tract, of papilloma virus infections, of herpes simplex virus (HSV) infection, of human immunodeficiency virus (HIV) infection, and of viral infection of internal organs such as infection with hepatitis viruses.
  • the viral infection is caused by a virus of family Flaviviridae.
  • the virus of family Flaviviridae is selected from Yellow Fever Virus, West Nile virus, Dengue virus, Japanese Encephalitis Virus, St. Louis Encephalitis Virus, and Hepatitis C Virus.
  • the viral infection is caused by a virus of family Picornaviridae, e.g, poliovirus, rhinovirus, coxsackievirus.
  • the viral infection is caused by a member of Orthomyxoviridae, e.g., an influenza virus.
  • the viral infection is caused by a member of Retroviridae, e.g, a lentivirus.
  • the viral infection is caused by a member of Paramyxoviridae, e.g., respiratory syncytial virus, a human parainfluenza virus, rubulavirus ⁇ e.g., mumps virus), measles virus, and human metapneumovirus.
  • the viral infection is caused by a member of Bunyaviridae, e.g., hantavirus. In other embodiments, the viral infection is caused by a member of Reoviridae, e.g., a rotavirus.
  • methods of treating parasitic infections such as, e.g., protozoan or helminths infections, are provided.
  • the parasitic infection is by a protozoan parasite.
  • the oritiziab parasite is selected from intestinal protozoa, tissue protozoa, or blood protozoa.
  • Illustrative protozoan parasites include, but are not limited to, Entamoeba hystolytica, Giardia lamblia, Cryptosporidium muris, Trypanosomatida gambiense, Trypanosomatida rhodesiense, Trypanosomatida crusi, Leishmania mexicana, Leishmania braziliensis, Leishmania tropica, Leishmania donovani, Toxoplasma gondii, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Plasmodium falciparum, Trichomonas vaginalis, and Histomonas meleagridis.
  • the parasitic infection is by a helminthic parasite such as nematodes (e.g., Adenophorea).
  • the parasite is selected from Secementea (e.g., Trichuris trichiura, Ascaris lumbricoides, Enterobius vermicularis, Ancylostoma duodenale, Necator americanus, Strongyloides stercoralis, Wuchereria bancrofti, Dracunculus medinensis).
  • the parasite is selected from trematodes (e.g. blood flukes, liver flukes, intestinal flukes, and lung flukes).
  • the parasite is selected from: Schistosoma mansoni, Schistosoma haematobium, Schistosoma japonicum, Fasciola hepatica, Fasciola gigantica, Heterophyes, Paragonimus westermani.
  • the parasite is selected from cestodes (e.g, Taenia solium, Taenia saginata, Hymenolepis nana, and Echinococcus granulosus).
  • the bacteria infection is by a gram-positive bacteria, gram-negative bacteria, aerobic and/or anaerobic bacteria.
  • the bacteria is selected from, but not limited to, Staphylococcus, Lactobacillus, Streptococcus, Sarcina, Escherichia, Enterobacter, Klebsiella, Pseudomonas, Acinetobacter, Mycobacterium, Proteus, Campylobacter, Citrobacter, Nisseria, Baccillus, Bacteroides, Peptococcus, Clostridium, Salmonella, Shigella, Serratia, Haemophilus, Brucella and other organisms.
  • the bacteria is selected from, but not limited to, Pseudomonas aeruginosa, Pseudomonas fluorescens, Pseudomonas acidovorans, Pseudomonas alcaligenes, Pseudomonas putida, Stenotrophomonas maltophilia, Burkholderia cepacia, Aeromonas hydrophilia, Escherichia coli, Citrobacter freundii, Salmonella typhimurium, Salmonella typhi, Salmonella paratyphi, Salmonella enteritidis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Enterobacter cloacae, Enterobacter aerogenes, Klebsiella pneumoniae, Klebsiella oxytoca, Serratia marcescens, Francisella tularensis, Morganella morganii, Proteus mirabilis
  • the fused biological cell made in accordance with embodiments of the present disclosure which is to be administered to a patient, can be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecular structures, or mixtures of compounds such as, for example, liposomes, receptor or cell targeted molecules, or oral, topical or other formulations for assisting in uptake, distribution and/or absorption.
  • an expression vector can be contained within a cell that is administered to a subject, or contained within a virus or virus-like particle.
  • the fused biological cell can be included in a pharmaceutical composition or can be in the form of a pharmaceutical composition.
  • the pharmaceutical composition comprises a checkpoint inhibitor.
  • the checkpoint inhibitor is an anti-PD1 antibody.
  • the fused biological cell to be administered can be in combination with a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier is a physiologically and pharmaceutically acceptable carrier.
  • the physiologically and pharmaceutically acceptable carrier can include any of the well-known components useful for immunization.
  • the carrier can facilitate or enhance an immune response to antigen(s) expressed by the fused biological cell.
  • the cell formulations can contain buffers to maintain a preferred pH range, salts or other components that present antigen(s) to a subject in a composition that stimulates an immune response to the antigen(s).
  • the physiologically acceptable carrier also can include one or more adjuvants that enhance the immune response to the antigens.
  • Pharmaceutically acceptable carriers include, for example, pharmaceutically acceptable solvents, suspending agents, or any other pharmacologically inert vehicles for delivering compounds to a subject.
  • Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties, when combined with one or more therapeutic compounds and any other components of a given pharmaceutical composition.
  • Typical pharmaceutically acceptable carriers include, without limitation: water, saline solution, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose or dextrose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g, starch or sodium starch glycolate), and wetting agents (e.g, sodium lauryl sulfate).
  • Compositions can be formulated for subcutaneous, intramuscular, or intradermal administration, or in any manner acceptable for delivery of a fused cell to a subject.
  • An adjuvant refers to a substance which, when added to an immunogenic agent such as a fused cell expressing secreted vaccine protein (e.g, gp96-lg), a T cell costimulatory fusion polypeptide, and optionally one or more disease antigens, nonspecifical ly enhances or potentiates an immune response to the agent in the recipient host upon exposure to the mixture.
  • Adjuvants can include, for example, oil-in-water emulsions, water-in oil emulsions, alum (aluminum salts), liposomes and microparticles, such as, polysytrene, starch, polyphosphazene and poly lactide/polyglycosides.
  • Adjuvants can also include, for example, squalene mixtures (SAF-I), muramyl peptide, saponin derivatives, mycobacterium cell wall preparations, monophosphoryl lipid A, mycolic acid derivatives, nonionic block copolymer surfactants, Quil A, cholera toxin B subunit, polyphosphazene and derivatives, and immunostimulating complexes (ISCOMs) such as those described by Takahashi et al., Nature 1990, 344:873-875.
  • SAF-I squalene mixtures
  • muramyl peptide saponin derivatives
  • mycobacterium cell wall preparations monophosphoryl lipid A
  • mycolic acid derivatives nonionic block copolymer surfactants
  • Quil A cholera toxin B subunit
  • polyphosphazene and derivatives and immunostimulating complexes
  • IFA Incomplete Freund's Adjuvant
  • adjuvants include, for example, bacille Calmett-Guerin (BCG), DETOX (containing cell wall skeleton of Mycobacterium phlei (CWS) and monophosphoryl lipid A from Salmonella minnesota (MPL)), and the like (see, for example, Hoover etal., J Clin Oncol 1993, 11 :390; and Woodlock etal., J Immunother 1999, 22:251-259).
  • a fused biological cell can be administered to a subject one or more times (e.g, once, twice, two to four times, three to five times, five to eight times, six to ten times, eight to 12 times, or more than 12 times).
  • a fused biological cell as provided herein can be administered one or more times per day, one or more times per week, every other week, one or more times per month, once every two to three months, once every three to six months, or once every six to 12 months.
  • a fused biological cell can be administered over any suitable period of time, such as a period from about 1 day to about 12 months.
  • the period of administration can be from about 1 day to 90 days; from about 1 day to 60 days; from about 1 day to 30 days; from about 1 day to 20 days; from about 1 day to 10 days; from about 1 day to 7 days.
  • the period of administration can be from about 1 week to 50 weeks; from about 1 week to 50 weeks; from about 1 week to 40 weeks; from about 1 week to 30 weeks; from about 1 week to 24 weeks; from about 1 week to 20 weeks; from about 1 week to 16 weeks; from about 1 week to 12 weeks; from about 1 week to 8 weeks; from about 1 week to 4 weeks; from about 1 week to 3 weeks; from about 1 week to 2 weeks; from about 2 weeks to 3 weeks; from about 2 weeks to 4 weeks; from about 2 weeks to 6 weeks; from about 2 weeks to 8 weeks; from about 3 weeks to 8 weeks; from about 3 weeks to 12 weeks; or from about 4 weeks to 20 weeks.
  • a boosting dose can be administered about 10 to 30 days, about 15 to 35 days, about 20 to 40 days, about 25 to 45 days, or about 30 to 50 days after a priming dose.
  • the methods provided herein can be used for controlling solid tumor growth (e.g., breast, prostate, melanoma, renal, colon, or cervical tumor growth) and/or metastasis.
  • the methods can include administering an effective amount of a fused biological cell as described herein to a subject in need thereof.
  • the subject is a mammal (e.g., a human).
  • the methods provided herein can be useful for stimulating an immune response against a tumor. Such immune response is useful in treating or alleviating a sign or symptom associated with the tumor.
  • "treating” is defined as reducing, preventing, and/or reversing the symptoms in the individual to which a fused biological cell in accordance with embodiments of the present disclosure has been administered, as compared to the symptoms of an individual not being treated.
  • a practitioner will appreciate that the methods described herein are to be used in concomitance with continuous clinical evaluations by a skilled practitioner (physician or veterinarian) to determine subsequent therapy. Such evaluations will aid and inform in evaluating whether to increase, reduce, or continue a particular treatment dose, mode of administration, etc.
  • the methods provided herein can thus be used to treat a tumor, including, for example, a cancer.
  • the methods can be used, for example, to inhibit the growth of a tumor by preventing further tumor growth, by slowing tumor growth, or by causing tumor regression.
  • the methods can be used, for example, to treat a cancer such as a lung cancer.
  • a cancer such as a lung cancer.
  • the subject to which a compound is administered need not suffer from a specific traumatic state.
  • the fused biological cells described herein may be administered prophylactically, prior to development of symptoms (e.g., a patient in remission from cancer).
  • the terms "therapeutic” and “therapeutically,” and permutations of these terms, are used to encompass therapeutic, palliative, and prophylactic uses.
  • treating or alleviating the symptoms is meant reducing, preventing, and/or reversing the symptoms of the individual to which a therapeutically effective amount of a composition has been administered, as compared to the symptoms of an individual receiving no such administration.
  • the terms “effective amount” and “therapeutically effective amount” refer to an amount sufficient to provide the desired therapeutic (e.g., anti-cancer, anti-tumor, or anti-infection) effect in a subject (e.g., a human diagnosed as having cancer or an infection).
  • Anti-tumor and anti-cancer effects include, without limitation, modulation of tumor growth (e.g, tumor growth delay), tumor size, or metastasis, the reduction of toxicity and side effects associated with a particular anti-cancer agent, the amelioration or minimization of the clinical impairment or symptoms of cancer, extending the survival of the subject beyond that which would otherwise be expected in the absence of such treatment, and the prevention of tumor growth in an animal lacking tumor formation prior to administration, i.e., prophylactic administration.
  • administration of an effective amount of a fused biological cell or a composition including the fused biological cell can increase the activation or proliferation of tumor antigen specific T cells in a subject.
  • the activation or proliferation of tumor antigen specific T cells in the subject can be is increased by at least 10 percent (e.g, at least 25 percent, at least 50 percent, or at least 75 percent) as compared to the level of activation or proliferation of tumor antigen specific T cells in the subject prior to the administration.
  • Anti-infection effects include, for example, a reduction in the number of infective agents (e.g, viruses or bacteria).
  • administration of a vector as provided herein can stimulate the activation or proliferation of pathogenic antigen specific T cells in the subject.
  • administration of the vector can lead to activation of antigen-specific T cells in the subject to a level greater than that achieved by gp96-lg vaccination alone.
  • an effective amount of a fused biological cell may be lowered or increased by fine tuning and/or by administering more than one dose. This document therefore provides a method for tailoring the administration/treatment to the particular exigencies specific to a given mammal.
  • Therapeutically effective amounts can be determined by, for example, starting at relatively low amounts and using step-wise increments with concurrent evaluation of beneficial effects.
  • the methods provided herein thus can be used alone or in combination with other well- known tumor therapies, to treat a patient having a tumor.
  • One skilled in the art will readily understand advantageous uses of the fused biological cells and methods provided herein, for example, in prolonging the life expectancy of a cancer patient and/or improving the quality of life of a cancer patient (e.g., a lung cancer patient).
  • a method of stimulating a patient's antibody response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • a method of stimulating a patient T cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • a method of stimulating one or both a patient's antibody response and T cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • the T cell-driven cellular immune response induces a CD8+ T cell response in the patient. In embodiments, the T cell-driven cellular immune response induces a CD4+ T cell response in the patient.
  • a method induces an immune response, selected from a CD8+ T cell, a CD4+ T cell, a cytotoxic T lymphocyte (CTL), a TH1 response, a TH2 response, or a combination thereof.
  • an immune response selected from a CD8+ T cell, a CD4+ T cell, a cytotoxic T lymphocyte (CTL), a TH1 response, a TH2 response, or a combination thereof.
  • CTL cytotoxic T lymphocyte
  • a method results in reduction of a number of regulatory T cells (Tregs).
  • Tregs regulatory T cells
  • the depletion of Tregs during an acute phase of infection was shown to result in enhanced effector T cell function and decreased viral loads. See Dietze et al. (2013) PLoS pathogens vol. 9: 12, e1003798. doi:10.1371/journal.ppat.1003798; see also Zelinskyy et al. (2009). Blood 114: 3199-3207.
  • the role of Treg depletion in antitumor immunity has also been observed. See, e.g., Dannull et al. (2005) J Clin Invest 115: 3623-3633.
  • a method results in the decrease in the functionality of Tregs. In embodiments, the method does not result in the decrease in the number of Tregs but results in the decrease in the functionality of Tregs. In embodiments, the method results in a decrease in the frequency of Tregs. In embodiments, the method results in the decrease in the number of functional Tregs.
  • a method of stimulating one or both a patient's antibody response and B cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
  • a method of stimulating a patient B cell-driven cellular immune response comprises administering to a patient in need thereof the fused biological cell generated in accordance with any of the embodiments of the present disclosure.
  • the method of stimulating or enhancing one or both a patient's antibody response and B cell-driven cellular immune response is performed without depleting the T cell population in the patient.
  • Embodiments of the present disclosure also provide a composition comprising a fused biological cell as described herein, and an excipient, carrier, or diluent.
  • the composition is a pharmaceutical composition.
  • the composition may comprise virus particles containing the fused biological cell.
  • Embodiments of the present disclosure also provide a composition comprising a lentivirus and/or transfer plasmid or a biological or host cell or a population of cells, as described herein, and an excipient, carrier, or diluent.
  • the composition is a pharmaceutical composition.
  • a cellular therapy in accordance with embodiments of the present disclosure may be used as a standalone cellular therapy or as a cellular therapy in combination with other cellular therapies that drive humoral immunity, to provide an added layer of cellular immunity.
  • the cellular therapy comprises a therapeutic biological cell (e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease).
  • the present cellular therapy can be used in combination with one or cellular therapies or any other type.
  • the composition is a sterile composition.
  • the composition is suitable for administration to a human.
  • the composition comprises a unit dose of fused biological cells.
  • the unit dose is about 10 5 , about 10 6 , about 10 7 , about 10 8 , about 10 9 , about 10 10 , about 10 11 , about 10 12 , about 10 13 , about 10 14 , about 10 15 , or more fused biological cells.
  • the composition comprises at least or about 10 6 fused cells.
  • the composition may comprise lentivirus particles containing a transfer plasmid.
  • the composition is a sterile composition.
  • the composition is suitable for administration to a human.
  • the composition comprises a unit dose of biological or host cells.
  • the unit dose is about 10 5 , about 10 6 , about 10 7 , about 10 8 , about 10 9 , about 10 10 , about 10 11 , about 10 12 , about 10 13 , about 10 14 , about 10 15 , or more biological or host cells into which the lentivirus and/or transfer plasmid is introduced.
  • the composition comprises at least or about 10 6 cells into with the lentivirus and/or transfer plasmid is introduced.
  • the pharmaceutical composition can comprise any pharmaceutically acceptable ingredient, including, for example, acidifying agents, additives, adsorbents, aerosol propellants, air displacement agents, alkalizing agents, anticaking agents, anticoagulants, antimicrobial preservatives, antioxidants, antiseptics, bases, binders, buffering agents, chelating agents, coating agents, coloring agents, desiccants, detergents, diluents, disinfectants, disintegrants, dispersing agents, dissolution enhancing agents, dyes, emollients, emulsifying agents, emulsion stabilizers, fillers, film forming agents, flavor enhancers, flavoring agents, flow enhancers, gelling agents, granulating agents, humectants, lubricants, mucoadhesives, ointment bases, ointments, oleaginous vehicles, organic bases, pastille bases, pigments, plasticizers, polishing agents, preservatives, sequestering agents, skin penet
  • the pharmaceutical compositions may be formulated to achieve a physiologically compatible pH.
  • the pH of the pharmaceutical composition may be at least 5, at least 5.5, at least 6, at least 6.5, at least 7, at least 7.5, at least 8, at least 8.5, at least 9, at least 9.5, at least 10, or at least 10.5 up to and including pH 11, depending on the formulation and route of administration, for example between 4 and 7, or 4.5 and 5.5.
  • the pharmaceutical compositions may comprise buffering agents to achieve a physiological compatible pH.
  • the buffering agents may include any compounds capable of buffering at the desired pH such as, for example, phosphate buffers (e.g., PBS), triethanolamine, Tris, bicine, TAPS, tricine, HEPES, TES, MOPS, PIPES, cacodylate, MES, acetate, citrate, succinate, histidine or other pharmaceutically acceptable buffers.
  • phosphate buffers e.g., PBS
  • triethanolamine Tris
  • Tris bicine
  • TAPS tricine
  • HEPES TES
  • MOPS MOPS
  • PIPES PIPES
  • cacodylate MES
  • acetate citrate
  • succinate histidine or other pharmaceutically acceptable buffers.
  • a buffering agent comprises PBS (phosphate-buffered saline) including a blocking buffer, such as, e.g., Bovine serum albumin (BSA).
  • PBS phosphate-buffered saline
  • BSA Bovine serum albumin
  • the buffering agent comprises PBS including from about 1 % to about 10% BSA. In embodiments, the buffering agent comprises PBS including about 1 % BSA.
  • a buffering agent comprises tris-buffered saline (TBS) including a blocking buffer, such as, e.g, BSA.
  • TBS tris-buffered saline
  • the buffering agent comprises TBS including from about 1 % to about 10% BSA.
  • the buffering agent comprises TBS including about 1 % BSA.
  • the present disclosure therefore provides compositions including pharmaceutical compositions including a fused biological cell as described herein, in combination with a physiologically and pharmaceutically acceptable carrier.
  • the present disclosure therefore provides compositions including pharmaceutical compositions containing packaged lentivirus or a cell containing the lentivirus and/or transfer plasmid as described herein, in combination with a physiologically and pharmaceutically acceptable carrier.
  • the physiologically and pharmaceutically acceptable carrier can include any of the well-known components useful for immunization.
  • the carrier can facilitate or enhance an immune response to an antigen administered in a vaccine.
  • the cell formulations can contain buffers to maintain a preferred pH range, salts or other components that present an antigen to an individual in a composition that stimulates an immune response to the antigen.
  • the physiologically acceptable carrier also can contain one or more adjuvants that enhance the immune response to an antigen.
  • Pharmaceutically acceptable carriers include, for example, pharmaceutically acceptable solvents, suspending agents, or any other pharmacologically inert vehicles for delivering compounds to a subject.
  • Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties, when combined with one or more therapeutic compounds and any other components of a given pharmaceutical composition.
  • Typical pharmaceutically acceptable carriers include, without limitation: water, saline solution, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose or dextrose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g, starch or sodium starch glycolate), and wetting agents (e.g, sodium lauryl sulfate).
  • binding agents e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose
  • fillers e.g., lactose or dextrose and other sugars, gelatin, or calcium sulfate
  • lubricants e.g., starch, polyethylene glycol, or sodium acetate
  • disintegrates e.g, starch or sodium starch glycolate
  • wetting agents e.g, sodium la
  • An adjuvant refers to a substance which, when added to an immunogenic agent such as a cell containing an expression vector system, nonspecifically enhances or potentiates an immune response to the agent in the recipient host upon exposure to the mixture.
  • Adjuvants can include, for example, oil-in-water emulsions, water-in oil emulsions, alum (aluminum salts), liposomes and microparticles, such as, polysytrene, starch, polyphosphazene and polylactide/polyglycosides.
  • Adjuvants can also include, for example, squalene mixtures (SAF-I), muramyl peptide, saponin derivatives, mycobacterium cell wall preparations, monophosphoryl lipid A, mycolic acid derivatives, nonionic block copolymer surfactants, Quil A, cholera toxin B subunit, polyphosphazene and derivatives, and immunostimulating complexes (ISCOMs) such as those described by Takahashi et al., Nature 1990, 344:873-875.
  • SAF-I squalene mixtures
  • muramyl peptide saponin derivatives
  • mycobacterium cell wall preparations monophosphoryl lipid A
  • mycolic acid derivatives nonionic block copolymer surfactants
  • Quil A cholera toxin B subunit
  • polyphosphazene and derivatives and immunostimulating complexes
  • IFA Incomplete Freund's Adjuvant
  • Additional adjuvants include, for example, bacille Calmett-Guerin (BCG), DETOX (containing cell wall skeleton of Mycobacterium phlei (CWS) and monophosphoryl lipid A from Salmonella minnesota (MPL)), and the like (see, for example, Hoover et al., J Clin Oncol 1993, 11 :390; and Woodlock et al., J Immunother 1999, 22:251-259).
  • BCG Bacille Calmett-Guerin
  • DETOX containing cell wall skeleton of Mycobacterium phlei
  • MPL monophosphoryl lipid A from Salmonella minnesota
  • Methods of administering cells to a subject include, but not limited to perfusions, infusions, and injections. See, e.g, Burch et al., Clin Cancer Res 6(6): 2175-2182 (2000), Dudley et al., J Clin Oncol 26(32): 5233- 5239 (2008); Khan et al., Cell Transplant 19:409-418 (2010); Gridelli et al., Liver Transpl 18:226-237 (2012).
  • a fused biological cell prepared in accordance with embodiments of the present disclosure is administered by injection.
  • the fused biological cell (In embodiments in combination with a checkpoint inhibitor and/or another agent) is administered in a pharmaceutically acceptable formulation, including a formulation suitable for one or more of injection, e.g., subcutaneous injection, intradermal injection (including to the dermis or epidermis), subdermal injection, intratumoral injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, and intraportal injection.
  • a pharmaceutically acceptable formulation including a formulation suitable for one or more of injection, e.g., subcutaneous injection, intradermal injection (including to the dermis or epidermis), subdermal injection, intratumoral injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, and
  • methods in accordance with the present disclosure comprise administering an additional agent to a subject.
  • chemotherapeutic agents include, but are not limited to, alkylating agents such as thiotepa and CYTOXAN cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins ⁇ e.g, bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; cally statin; CC-1065 (including its adozelesin
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo- 5-oxo-L-norleucine, ADRIAMYCIN doxorubicin (including morpholino- doxorubicin, cyanomorpholino-doxorubicin, 2- pyrrolino-doxorubicin and deoxy doxorubicin), epi
  • the present disclosure pertains to anti- infectives as additional agents.
  • the anti-infective is an anti-viral agent including, but not limited to, Abacavir, Acyclovir, Adefovir, Amprenavir, Atazanavir, Cidofovir, Darunavir, Delavirdine, Didanosine, Docosanol, Efavirenz, Elvitegravir, Emtricitabine, Enfuvirtide, Etravirine, Famciclovir, and Foscarnet.
  • the anti- infective is an anti-bacterial agent including, but not limited to, cephalosporin antibiotics (cephalexin, cefuroxime, cefadroxil, cefazolin, cephalothin, cefaclor, cefamandole, cefoxitin, cefprozil, and ceftobiprole); fluoroquinolone antibiotics (cipro, Levaquin, floxin, tequin, avelox, and norflox); tetracycline antibiotics (tetracycline, minocycline, oxytetracycline, and doxycycline); penicillin antibiotics (amoxicillin, ampicillin, penicillin V, dicloxacillin, carbenicillin, vancomycin, and methicillin); monobactam antibiotics (aztreonam); and carbapenem antibiotics (ertapenem, doripenem, imipenem/cilastatin, and meropenem).
  • cephalosporin antibiotics ce
  • the anti-infectives include anti-malarial agents (e.g, chloroquine, quinine, mefloquine, primaquine, doxycycline, artemether/lumefantrine, atovaquone/proguanil and sulfadoxine/pyrimethamine), metronidazole, tinidazole, ivermectin, pyrantel pamoate, and albendazole.
  • anti-malarial agents e.g, chloroquine, quinine, mefloquine, primaquine, doxycycline, artemether/lumefantrine, atovaquone/proguanil and sulfadoxine/pyrimethamine
  • metronidazole e.g, chloroquine, quinine, mefloquine, primaquine, doxycycline, artemether/lumefantrine, atovaquone/proguanil and sulfadoxine/
  • additional agents include blocking antibodies targeted to an immune checkpoint molecules.
  • checkpoint molecules include CTLA4 (cytotoxic T lymphocyte antigen-4), PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), LAG-3 (lymphocyte activation gene-3), TIM-3 (T cell immunoglobulin and mucin protein-3), LAG-3 (lymphocyte activation gene-3), and others. See Pardoll, 2012, Nature Reviews Cancer 12:252-64; Nirschl & Drake, 2013, Clin Cancer Res 19:4917-24; He et al., 2018, OncoTargets and therapy, vol. 11 7005-7009; Qin et al., 2019. Mol Cancer 18, 155.
  • a biological cell e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease
  • host cell comprising any of the lentiviruses as described herein.
  • the term "host cell” refers to any type of cell that can contain the lentivirus and/or transfer plasmid, e.g., without limitation, a cell that produces the lentivirus.
  • the host cell can be a eukaryotic cell, e.g, plant, animal, fungi, or algae, or can be a prokaryotic cell, e.g., bacteria or protozoa.
  • the host cell can be a cultured cell or a primary cell, i.e., isolated directly from an organism, e.g., a human.
  • the host cell can be an adherent cell or a suspended cell, i.e., a cell that grows in suspension.
  • the host cell is a mammalian host cell.
  • the host cell is a human host cell.
  • the human host cell is an NIH 3T3 cell or an HEK293 cell. The presently disclosed host cells are not limited to just these two types of cells, however, and may be any cell type described herein.
  • the cells that can be used include, without limitation, epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, or granulocytes, various stem or progenitor cells, such as hematopoietic stem or progenitor cells (e.g., as obtained from bone marrow), umbilical cord blood, peripheral blood, fetal liver, etc., and tumor cells (e.g., human tumor cells).
  • the cells are irradiated.
  • a population of cells comprising at least one biological cell (e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) or host cell described herein.
  • the population of cells can be a heterogeneous population comprising the host cell comprising any of the lentiviruses described, in addition to at least one other cell.
  • the population of cells can be a substantially homogeneous population, in which the population comprises mainly host cells (e.g., consisting essentially of) comprising the lentivirus.
  • the population also can be a clonal population of cells, in which all cells of the population are clones of a single host cell comprising the lentivirus, such that all cells of the population comprise the lentivirus.
  • the population of cells is a clonal population comprising host cells comprising the lentivirus as described herein.
  • the cell population of the present disclosure is one wherein at least 50% of the cells are host cells as described herein.
  • the cell population of the present disclosure is one wherein at least 60%, at least 70%, at least 80% or at least 90% or more of the cells are host cells as described herein.
  • the biological cell is selected from AD-100, HEK293, and NIH 3T3.
  • the biological cell is a human tumor cell.
  • the biological cell is an irradiated or live and attenuated human tumor cell.
  • the human tumor cell is a cell from an established NSCLC, bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
  • a biological cell comprising a first recombinant protein having an amino acid sequence of at least 95% sequence identity with SEQ ID NO: 2 and a second recombinant protein having an amino acid sequence of at least 95% sequence identity with the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 42, the amino acid sequence of SEQ ID NO: 41 , the amino acid sequence of SEQ ID NO: 46, the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 41 , the amino acid sequence of SEQ ID NO: 43, the amino acid sequence of SEQ ID NO: 45, the amino acid sequence of SEQ ID NO: 42, or an antigenic fragment of any of the foregoing.
  • the first recombinant protein has at least 97% sequence identity with SEQ ID NO: 2 and the second recombinant protein having an amino acid sequence of at least 97% sequence identity with the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 42, the amino acid sequence of SEQ ID NO: 43, or the amino acid sequence of SEQ ID NO: 46, or an antigenic fragment of any of the foregoing.
  • the first recombinant protein has at least 98% sequence identity with SEQ ID NO: 2 and the second recombinant protein having an amino acid sequence of at least 98% sequence identity with the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 42, the amino acid sequence of SEQ ID NO: 41 , the amino acid sequence of SEQ ID NO: 46, the amino acid sequence of SEQ ID NO: 5, the amino acid sequence of SEQ ID NO: 7, the amino acid sequence of SEQ ID NO: 9, the amino acid sequence of SEQ ID NO: 11 , the amino acid sequence of SEQ ID NO: 13, the amino acid sequence of SEQ ID NO: 27, the amino acid sequence of SEQ ID NO: 13, the amino acid sequence of SEQ ID NO: 15, the amino acid sequence of SEQ ID NO: 17, the amino acid sequence of SEQ ID NO: 19, or the amino acid sequence of SEQ ID NO: 21 , or an antigenic fragment of any of the foregoing.
  • SEQ ID NO: 39 the amino acid sequence of SEQ ID NO: 42, the amino
  • the first biological cell comprising a lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof
  • the second biological cell comprising a lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject
  • the third biological cell comprising a lentivirus comprising a nucleic acid encoding a protein, or an antigenic portion thereof.
  • the disclosure relates to a cellular therapy (e.g., a therapeutic biological cell, and/or a lentivirus as disclosed herein) comprising one or more secretable fusion proteins, T cell costimulatory fusion proteins, and/or fragments thereof, which can stimulate immune responses in a subject.
  • a cellular therapy e.g., a therapeutic biological cell, and/or a lentivirus as disclosed herein
  • secretable fusion proteins e.g., T cell costimulatory fusion proteins, and/or fragments thereof, which can stimulate immune responses in a subject.
  • the methods of the present disclosure advantageously rely on the chaperone function of the secreted fusion protein.
  • the fusion protein chaperones the one or more proteins or antigen portions thereof, which are efficiently taken up by activated antigen presenting cells (APCs).
  • APCs act to cross-present the proteins or antigen portions thereof via MHC I to CD8+ CTLs, whereupon an avid, antigen specific, cytotoxic CD8+ T cell response is stimulated.
  • the proteins of the present disclosure are advantageously capable of initiating both an innate, humoral immune response (including, e.g., activation of APCs, pro-inflammatory cytokine release, activation of NK cells), and an adaptive immune response (including, e.g., priming, activation and proliferation of antigen specific CTLs).
  • an innate, humoral immune response including, e.g., activation of APCs, pro-inflammatory cytokine release, activation of NK cells
  • an adaptive immune response including, e.g., priming, activation and proliferation of antigen specific CTLs.
  • the present disclosure provides a method of administering to the subject the lentivirus as disclosed herein, or a population of cells transfected with the lentivirus.
  • the present disclosure provides a method of generating a cellular therapy that is suitable for treating or preventing a disease.
  • the cellular therapy generated by the methods of the disclosure prevent, alleviate, and/or treat one or more symptoms associated with a disease.
  • symptoms that may be treated include, but are not limited to fever, cough ⁇ e.g., dry cough), shortness of breath and other breathing difficulties, fatigue, diarrhea, upper respiratory symptoms ⁇ e.g. sneezing, runny nose, sore throat), and/or pneumonia.
  • the cellular therapy is suitable for eliciting innate immunity when administered to a subject.
  • the cellular therapy is suitable for eliciting humoral immunity (i.e. antibody response) when administered to a subject.
  • the cellular therapy is suitable for eliciting cell-mediated immunity (i.e. T-cell response) when administered to a subject.
  • the subject is immunocompromised due to one or more factors, including, without limitation, a genetic predisposition, age, immune status, and disease.
  • the cellular therapy stimulates, promotes, or increases one or more of a T-cell response, antibody response, and activation of innate immunity.
  • the cellular therapy stimulates, promotes, or increases all three of the T-cell response, antibody response, and activation of innate immunity, thereby activating all three arms of the subject's immune system.
  • the chaperone protein is gp96.
  • an expression vector system or a population of cells transfected with the expression vector system is designed to use gp96 so as to trigger mucosal immunity by activating both B and T cell responses at the point of pathogen entry.
  • the gp96-based cellular therapy effectively presents multiple antigens and activates the immune system thereby.
  • the gp96-based cellular therapy utilizes natural and adaptive immune process to induce long- lasting memory responses against a disease.
  • the gp96-based cellular therapy, compositions, and biological cells in accordance with the present disclosure activate all three of innate immune response, cellular immune response (i.e. T cell response), and humoral immune response (i.e. antibody response).
  • the present vaccine activates innate immunity via Toll-Like Receptor (TLRs), as, without wishing to be bound by the theory, gp96 activates Toll-Like Receptor 4/2 (TLR4 and TLR2) on macrophages and dendritic cells.
  • TLRs Toll-Like Receptor
  • gp96 activates Toll-Like Receptor 4/2 (TLR4 and TLR2) on macrophages and dendritic cells.
  • present cellular therapy adapted to present multiple antigens, in accordance with embodiments of the present disclosure, stimulates, promotes, or increases a cellular immune response via CD4 and CD8 T cells, in addition to the humoral immune response, via neutralizing IgG antibody.
  • the present cellular therapy is suitable for increasing the subject's T-cell response as compared to the T-cell response of a subject that was not administered the cellular therapy.
  • the cellular therapy is suitable for increasing the subject's antibody response as compared to the antibody response of a subject that was not administered the cellular therapy.
  • the cellular therapy is suitable for increasing the subject's innate immune response as compared to the innate immune response of a subject that was not administered the cellular therapy.
  • the cellular therapy is suitable for increasing the subject's T-cell response, antibody response, and innate immune response as compared to the T-cell response, antibody response, and innate immune responses of a subject that was not administered the cellular therapy.
  • the cellular therapy is suitable for increasing the subject's innate immune response as compared to the innate immune response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's adaptive immune response as compared to the adaptive immune response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's innate immune response and adaptive immune response as compared to the innate and adaptive immune responses of a subject that was not administered the cellular therapy. In embodiments, methods and compositions of the present disclosure are for improving and/or increasing vaccine efficacy in a patient and include maintaining and/or increasing the patient's T cell populations (e.g., CD4+ and/or CD8+ T cell populations).
  • T cell populations e.g., CD4+ and/or CD8+ T cell populations
  • methods and compositions of the present disclosure are for improving and/or increasing vaccine efficacy in a patient and include maintaining and/or increasing the patient's antigen-specific antibody titers (e.g., IgG, IgM and IgA).
  • methods of the present disclosure provide for mitigation of age-related immunosenescence as measured by an increase or restoration of a patient's antigen-specific antibody titers (e.g., IgG, IgM and IgA).
  • the present cellular therapy, and methods for use thereof are suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell populations of a subject that was not administered the present cellular therapy.
  • the subject's T cells including T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells, are increased and/or restored as compared to the T cell populations of a subject that was not administered the cellular therapy.
  • compositions are provided that includes a cellular therapy in accordance with embodiments of the present disclosure, and at least one other cellular therapy.
  • the present cellular therapy can be administered alone or in combination with at least one other cellular therapy.
  • the method is suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell population(s) of a subject that was administered another cellular therapy.
  • the subject's T cells including T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells, are increased and/or restored as compared to the T cell populations of a subject that was administered another cellular therapy.
  • the subject's CD4+ helper cells population(s) are increased and/or restored as compared to the CD4+ helper cells population(s) of a subject that was not administered the present cellular therapy.
  • OX40L co-stimulation expands CD4 helper T cells that promote B-cell differentiation and IgG/lgA antibody class switching.
  • the present disclosure provides compositions and methods for improving and/or increasing cellular therapy efficacy in a patient, as measured by an increase and/or restoration of the patient's T cell subsets.
  • the T cells are T helper cells (e.g, Th cells).
  • T helper cells secrete cytokines that attract one or more of macrophages, neutrophils, other lymphocytes, and other cytokines to further direct these cells.
  • CD4+ T helper cells are one of several subsets, including, Th1 , Th2, Th17, Th9, and Tfh, with each subset having a different function.
  • T cells are cytotoxic cells that optionally produce IL-2 and IFNy cytokines. In embodiments, these T cells are cytotoxic CD8+ T cells (also known as Tc cells or T-killer cells).
  • memory T cells elicited by the compositions and methods of the present disclosure are long-lived and can expand to large numbers of effector T cells when re-exposed to their cognate antigen.
  • the memory T cells elicited by methods of the present disclosure can persist in a subject for at least about 1 year, or at least about 2 years, or at least about 3 years, or at least about 4 years, or at least about 5 years, or at least about 6 years, or at least about 7 years, or at least about 8 years, or at least about 9 years, or at least about 10 years, or at least about 15 years, or at least about 20 years, or at least about 30 years, or at least about 40 years, or at least about 50 years, or at least about 60 years, or at least about 70 years, or at least about 80 years.
  • memory T cells elicited by the compositions and methods of the present disclosure can last for the entire lifespan of a subject.
  • memory T cells provide a patient's immune system with memory against previously encountered pathogens.
  • memory T cell populations include, but are not limited to, tissue-resident memory T (T rm ) cells, stem memory TSCM cells, and virtual memory T cells.
  • T rm tissue-resident memory T
  • memory T cells are classified as CD4+ or CD8+ and express CD45RO.
  • memory T cells are further differentiated into various subsets.
  • memory T cell subsets include: Central memory T cells (TCM cells), which can express CD45RO, C-C chemokine receptor type 7 (CCR7), L-selectin (CD62L), and CD44; Effector memory T cells (T E M cells and T EMRA cells), which express CD45RO and CD44 but lack expression of CCR7 and CD62L; Tissue resident memory T cells (TRM), which is associated with the integrin oep7; and Virtual memory T cells.
  • TCM cells Central memory T cells
  • CD45RO CD45RO
  • CCR7 C-C chemokine receptor type 7
  • CD62L L-selectin
  • CD44 Effector memory T cells
  • T E M cells and T EMRA cells Effector memory T cells
  • TRM Tissue resident memory T cells
  • the term "treat,” as well as words related thereto, do not necessarily imply 100% or complete treatment. Rather, there are varying degrees of treatment of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect.
  • the methods of treating an infection of the present disclosure can provide any amount or any level of treatment.
  • the treatment provided by the method of the present disclosure may include treatment of one or more conditions or symptoms or signs of the infection, being treated.
  • the treatment provided by the methods of the present disclosure may encompass slowing the progression of the infection.
  • the methods can treat the infection by virtue of eliciting an immune response against the infection, stimulating or activating CD8+ T cells specific for the infection, to proliferate, and the like.
  • the term “prevent” and words stemming therefrom encompasses inhibiting or otherwise blocking an infection.
  • the term “inhibit” and words stemming therefrom may not be a 100% or complete inhibition or abrogation. Rather, there are varying degrees of inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the presently disclosed expression vector systems or host cells may inhibit infection to any amount or level.
  • the inhibition provided by the methods of the present disclosure is at least or about a 10% inhibition (e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition).
  • a 10% inhibition e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition.
  • the present cellular therapy and biological cells may be administered to a subject by any route considered appropriate by a medical practitioner.
  • routes of administration include, for example: oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, by electroporation, or topically. Administration can be local or systemic.
  • the cellular therapy or the biological cell can be administered to a subject one or more times (e.g., once, twice, two to four times, three to five times, five to eight times, six to ten times, eight to 12 times, or more than 12 times).
  • a cellular therapy or biological cell as provided herein can be administered one or more times per day, one or more times per week, every other week, one or more times per month, once every two to three months, once every three to six months, or once every six to 12 months.
  • a cellular therapy or biological cell can be administered over any suitable period of time, such as a period from about 1 day to about 12 months.
  • the period of administration can be from about 1 day to 90 days; from about 1 day to 60 days; from about 1 day to 30 days; from about 1 day to 20 days; from about 1 day to 10 days; from about 1 day to 7 days.
  • the period of administration can be from about 1 week to 50 weeks; from about 1 week to 50 weeks; from about 1 week to 40 weeks; from about 1 week to 30 weeks; from about 1 week to 24 weeks; from about 1 week to 20 weeks; from about 1 week to 16 weeks; from about 1 week to 12 weeks; from about 1 week to 8 weeks; from about 1 week to 4 weeks; from about 1 week to 3 weeks; from about 1 week to 2 weeks; from about 2 weeks to 3 weeks; from about 2 weeks to 4 weeks; from about 2 weeks to 6 weeks; from about 2 weeks to 8 weeks; from about 3 weeks to 8 weeks; from about 3 weeks to 12 weeks; or from about 4 weeks to 20 weeks.
  • RT reverse transcription PCR RT reverse transcription PCR
  • the cellular therapy of the present disclosure is co-administered in conjunction with additional therapeutic agent(s), including vaccines.
  • Co-administration can be simultaneous or sequential.
  • the additional therapeutic agent is an agent that is used to provide relief to symptoms of infections.
  • agents include remdesivir; favipiravir; galidesivir; prezcobix; lopinavir and/or ritonavir and/or arbidol; mRNA-1273; MSCs-derived exosomes; lopinavir/ ritonavir and/or ribavirin and/or I FN-beta; xiyanping; anti-VEGF-A (e.g.
  • the additional therapeutic agent is chloroquine, including chloroquine phosphate.
  • the additional therapeutic agent is a composition comprising one or more HIV drugs.
  • the composition comprises a combination of one or more of lopinavir and/or ritonavir and/or arbidol.
  • the additional therapeutic agent comprises one or more vaccines. In embodiments, the additional therapeutic agent comprises one or more vaccines.
  • the cellular therapy in accordance with embodiments of the present disclosure which employs gp- 96- (e.g., without limitation, a gp96-based cellular therapy), may be delivered alone (e.g., as a standalone cellular therapy) or in combination with other cellular therapies that drive humoral immunity, to provide an added layer of cellular immunity.
  • the present cellular therapy can be administered in combination with one or more other cellular therapies, e.g., without limitation, flu vaccines, and other vaccines.
  • the vaccine in accordance with embodiments of the present disclosure in combination with other vaccines (including conventional vaccines), induces effective and durable immune responses.
  • a combination of the present cellular therapy and other cellular therapies may boost immunity in certain types of patients, including elderly patients, patents with comorbidities, and patients with compromised immune system.
  • the present cellular therapy enhances effect of other vaccines by providing an added layer of T-cell immunity boost to generate an effective and long-term immune response.
  • Various v cellular therapies can be co-administered with the present cellular therapy.
  • the present cellular therapy is administered in combination with a cellular therapy either simultaneously or sequentially.
  • the cellular therapy is in the exploratory, preclinical, clinical, post-clinical, or approved stage.
  • the cellular therapy comprises one or more of: a live attenuated virus, an inactivated virus, a nonreplicating viral vector, a replicating viral vector, a recombinant protein, a peptide, a virus-like particle, DNA, RNA, mRNA, another macromolecule, and a fragment thereof.
  • the subject and/or animal is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, rabbit, sheep, or non-human primate, such as a monkey, chimpanzee, or baboon.
  • the subject and/or animal is a non-mammal, such, for example, a zebrafish.
  • the subject and/or animal may comprise fluorescently-tagged cells (with e.g. GFP).
  • the subject and/or animal is a human.
  • the human is a pediatric human.
  • the human is an adult human.
  • the human is a geriatric human.
  • the human may be referred to as a patient.
  • the human has an age in a range of from about 0 months to about 6 months old, from about 6 to about 12 months old, from about 6 to about 18 months old, from about 18 to about 36 months old, from about 1 to about 5 years old, from about 5 to about 10 years old, from about 10 to about 15 years old, from about 15 to about 20 years old, from about 20 to about 25 years old, from about 25 to about 30 years old, from about 30 to about 35 years old, from about 35 to about 40 years old, from about 40 to about 45 years old, from about 45 to about 50 years old, from about 50 to about 55 years old, from about 55 to about 60 years old, from about 60 to about 65 years old, from about 65 to about 70 years old, from about 70 to about 75 years old, from about 75 to about 80 years old, from about 80 to about 85 years old, from about 85 to about 90 years old, from about 90 to about 95 years old or from about 95 to about 100 years old.
  • the subject is a non-human animal, and therefore the disclosure pertains to veterinary use.
  • the non-human animal is a household pet.
  • the non-human animal is a livestock animal.
  • the cellular therapy, compositions, cells, and methods employ a cellular therapy (e.g, a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) which can be, without limitation, a gp96/OX40L-lg cellular therapy, and which can activate robust T-cell immunity along with humoral immunity.
  • a gp96-based cellular therapy in accordance with embodiments of the present disclosure can have any other T cell costimulatory fusion protein, as the cellular therapy is not limited to the OX40L-I g T cell costimulatory fusion protein.
  • the present cellular therapy is useful for harnessing natural antigen presentation and T-cell activation pathways in, without limitations, elderly patients (e.g., patients over the age of 65), patients with comorbidities, and/or in patients with a compromised immune system.
  • the patient can be selected for treatment in accordance with embodiments of the present disclosure based on one or more of that patient's age, the status of the patient's immune system, and based on whether or not the patient has a comorbidity.
  • the comorbidity can be defined as the simultaneous presence of two or more chronic diseases or conditions in the patient.
  • kits comprising a fused biological cell or a composition comprising any one of the foregoing of the present disclosure are also provided.
  • an exemplary kit of the disclosure comprises any composition described herein in a unit dosage form.
  • the unit dosage form is a container, such as a pre-filled syringe, which can be sterile, containing any agent described herein and a pharmaceutically acceptable carrier, diluent, excipient, or vehicle.
  • the kit comprises a sterile, GMP-grade unit dose of the cells.
  • a unit dose of cells comprises 10 5 , 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 10 13 , or more than 10 15 cells made in accordance with embodiments of the present disclosure.
  • the unit dose of cells are packaged in an intravenous bag.
  • the unit dose of cells are provided in a cryogenic form.
  • the unit dose of cells are ready to use.
  • the unit dose of cells are provided in a tube, a flask, a dish, or like container.
  • Kits can simplify the administration of any agent described herein.
  • the kit can further comprise a label or printed instructions instructing the use of any agent described herein.
  • the kit comprises a container containing an effective amount of a composition of the disclosure and an effective amount of another composition, such those described herein.
  • the cells are cryopreserved. In illustrative aspects, the cells are not frozen.
  • an “effective amount,” when used in connection with medical uses is an amount that is effective for providing a measurable treatment, prevention, or reduction in the rate of pathogenesis of a disease of interest.
  • something is "decreased” if a read-out of activity and/or effect is reduced by a significant amount, such as by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, or more, up to and including at least about 100%, in the presence of an agent or stimulus relative to the absence of such modulation.
  • activity is decreased and some downstream read-outs will decrease but others can increase.
  • activity is "increased” if a read-out of activity and/or effect is increased by a significant amount, for example by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, or more, up to and including at least about 100% or more, at least about 2-fold, at least about 3-fold, at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, at least about 10-fold, at least about 50-fold, at least about 100-fold, in the presence of an agent or stimulus, relative to the absence of such agent or stimulus.
  • compositional percentages are by weight of the total composition, unless otherwise specified.
  • the word "include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the compositions and methods of this technology.
  • the terms “can” and “may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.
  • the words "preferred” and “preferably” refer to embodiments of the technology that afford certain benefits, under certain circumstances. However, other embodiments may also be preferred, under the same or other circumstances. Furthermore, the recitation of one or more preferred embodiments does not imply that other embodiments are not useful, and is not intended to exclude other embodiments from the scope of the technology.
  • compositions described herein needed for achieving a therapeutic effect may be determined empirically in accordance with conventional procedures for the particular purpose.
  • the therapeutic agents are given at a pharmacologically effective dose.
  • a “pharmacologically effective amount,” “pharmacologically effective dose,” “therapeutically effective amount,” or “effective amount” refers to an amount sufficient to produce the desired physiological effect or amount capable of achieving the desired result, particularly for treating the disorder or disease.
  • An effective amount as used herein would include an amount sufficient to, for example, delay the development of a symptom of the disorder or disease, alter the course of a symptom of the disorder or disease (e.g, slow the progression of a symptom of the disease), reduce or eliminate one or more symptoms or manifestations of the disorder or disease, and reverse a symptom of a disorder or disease.
  • administration of therapeutic agents to a patient suffering from cancer provides a therapeutic benefit not only when the underlying condition is eradicated or ameliorated, but also when the patient reports a decrease in the severity or duration of the symptoms associated with the disease, e.g., a decrease in tumor burden, a decrease in circulating tumor cells, an increase in progression free survival.
  • Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized.
  • Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to about 50% of the population) and the ED50 (the dose therapeutically effective in about 50% of the population).
  • the dosage can vary depending upon the dosage form employed and the route of administration utilized.
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50.
  • compositions and methods that exhibit large therapeutic indices are preferred.
  • a therapeutically effective dose can be estimated initially from in vitro assays, including, for example, cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 as determined in cell culture, or in an appropriate animal model.
  • Levels of the described compositions in plasma can be measured, for example, by high performance liquid chromatography.
  • the effects of any particular dosage can be monitored by a suitable bioassay. The dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
  • the effect will result in a quantifiable change of at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 70%, or at least about 90%. In embodiments, the effect will result in a quantifiable change of about 10%, about 20%, about 30%, about 50%, about 70%, or even about 90% or more.
  • Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized.
  • a pharmacologically effective amount that will treat cancer will modulate the symptoms typically by at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%. In exemplary embodiments, such modulations will result in, for example, statistically significant and quantifiable changes in the numbers of cancerous cells.
  • Example 1 The experiments of this example demonstrate methods for generating a cellular therapy using a lentivirus transduced with DNA encoding for gp96-lg or CX40L-lg, or both gp96-lg and OX40L-lg.
  • stable expressing cells were transduced with a replication incompetent lentivirus vectors having DNA encoding for either gp96-lg or CX40L-lg, or both gp96-lg and OX40L-lg. As shown in FIG.
  • the expression levels of individual clones varied from low to high across cells transduced with gp96-lg or CX40L-lg, or both gp96-lg and OX40L-lg.
  • individual stable expressing cells were transduced with either gp96-lg or CX40L-lg, and the individual clones were then subsequently fused together to engineer multiple target expressor clones.
  • the expression levels can be varied depending on the clone type (e.g., a single cell transduced with either gp96-lg or CX40L-lg, or both gp96-lg and CX40L-lg), and/or the sequential fusion combination (e.g., a single cell transduced with gp96-lg, and a single cell transduced with OX40L-I g, then fused together), and how these engineered clones can be used for a cellular therapy to treat oncology, infectious diseases, and/or autoimmune related diseases.
  • the sequential fusion combination e.g., a single cell transduced with gp96-lg, and a single cell transduced with OX40L-I g, then fused together
  • FIG. 2 illustrates cell fusion expression profile of a fusion of a HEK293 cell and an HS112 cell (which is an AD-100 cell expressing gp96).
  • the HEK293 cell expresses such CTAs as LIN28B, SRSF12, TEX15, TEX19, SYNGR4 and GAPDHS; and the HS112 cell expresses COX7B2, MAGEA6, and MAGEC1 CTAs, and gp96-lg.
  • RPS18 and GAPDH which are housekeeping genes (translation and metabolism, respectively), were used as endogenous control, to normalize the quantitative real-time PCR (qPCR) data.
  • CTAs RPS18 and GAPDH are expressed by both HEK293 cell and the HS112 cell.
  • the HEK293 to HS112 cells ratio was 1 :10 (1 repetiion) and 9:10 (2 repetitions).
  • the expression of genes in the HEK293 cell was tested via one repetition, and the expression of genes in the HS112 cell was tested via two repetitions.
  • FIG. 2 illustrates dCt for various genes - COX7B2, MAGEA6, gp-96lg, LIN28B, SRSF12, TEX15, TEX19, SYNGR4, MAGEC1 , and GAPDHS.
  • the expression is shown, in this order, in six bars - HEK293 (11/13), HS112 (5/19), HS112 (6/16/), Fusion 9: 10 (12/28), Fusion 9: 10 (12/30), and Fusion 1 :10 (1/14).
  • FIG. 2 illustrates dCt for various genes - COX7B2, MAGEA6, gp-96lg, LIN28B, SRSF12, TEX15, TEX19, SYNGR4, MAGEC1 , and GAPDHS.
  • the expression is shown, in this order, in six bars - HEK293 (11/13), HS112 (5/19), HS112 (6/16/), Fusion 9: 10 (12/28), Fusion 9: 10
  • FIG. 2 shows that CTAs of interest in the HEK293 cell line show strong activation after fusion with the HS112 cell line, without sacrificing the pre-existing HS112 CTA profile.
  • Example 3 Two successive rounds of transduction were performed in AD100 cells 24 hours apart. One million cells were seeded into 6-well dishes and allowed to adhere. 1 ml of fresh media was added for 24 hours, collected, and used to assess levels of the two factors produced by the clones (FIG. 3). Cells were transduced with lentivirus encoding human GP96- IgG (vGP96) or human OX40L-lgG (vOX40L) at an MOI of 100. 72 hours after the second round, the cells were passaged into a larger culture vessel and allowed to recover and expand for another 72 hours. After sufficient cell expansion, selection began using the respective antibiotic encoded by the lentivirus construct.
  • vGP96 human GP96- IgG
  • vOX40L human OX40L-lgG
  • Single-cell clones were further expanded up and screened for levels of either hGP96-lgG or hOX40L-lgG. Protein levels were measured by ELISA using media conditioned for 24 hours by 1 million cells. The highest expressing clone for hGP96-lgG and hOX40L was selected for cell fusion to generate a hybrid cell that expresses both factors.
  • Cellular fusion was achieved using the GenomeONETM -CF EX kit and utilized the viral HV J-envelope.
  • the cell fusion was stochastic, but different antibiotic resistance markers allowed for selection ef fusion events containing DNA of both cells.
  • the fusions were allowed to recover and expand for 72 hours followed by dual drug treatment to eliminate cells that do not contain both antibiotic resistance genes. After 7 days of selection, the remaining cells underwent another round of single cell isolation by limiting dilution.
  • These clones were expanded up and screened for high level expression of both hGP96-lgG and hOX40L-lgG. These data highlight unprecedented levels of GP96-lg and OX40L-lg expression in the AD-100 cell line.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Immunogenic agents are provided that are generated by fusing two or more biological cells into a fused biological cell, for use in treatment of cancer and infectious diseases in a manner that allows control of at least a ratio of a vaccine protein and a T cell costimulatory fusion protein expressed by the fused cell.

Description

CELL-FUSION BASED IMMUNE AGENTS
FIELD OF THE DISCLOSURE
The present disclosure relates to, in part, cell-fusion based immune agents, and methods of making thereof, that are useful for treatment of cancer and infectious diseases.
RELATED APPLICATIONS
The present application claims priority to U.S. Provisional Patent Application Nos. 63/136,842, filed January 13, 2021, and 63/222,503, filed July 26, 2021 , the entire contents of all of which are hereby incorporated by reference.
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY
The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (filename: HTB_038PC_ST25.txt; date recorded: January 12, 2022; file size: 405,694 bytes).
BACKGROUND
Cancer and various infectious diseases are significant health problems worldwide, taking millions of lives each year and thus taking an enormous toll on human resources and the economy. Despite recent advances that have been made in detection and therapy of cancer, no vaccine or other universally successful method for prevention or treatment is currently available. Treatment of infectious diseases, although generally more advanced and managed using preventative vaccines in many cases, faces issues such as strain diversity and appearance of new strains, including those carrying (multi)antibiotics resistance. Also, new diseases, such as, for example, novel coronavirus disease 2019 (COVID-19), appear, for which no accepted treatments have been developed.
Immunotherapy is defined as a therapeutic approach that targets or manipulates the immune system. See Papaioannou et al., Ann. Transl. Med. 36 199-201. 10.21037/atm.2016.04.01. The goal of immunotherapy is to harness the host's adaptive and innate immune response to cause sustained elimination of diseased cells. See Naran et al., Front Microbiol. 2018;9:3158. Published 2018 Dec 21. doi:10.3389/fmicb.2018.03158.
Various immunotherapy approaches have been undertaken to modulate a patient's immune system to direct it towards an anti-cancer effect. For instance, cell-based cancer vaccines haven proven effective in clinical trials against lung cancer (e.g. using an immortalized lung cancer cell line that is altered to release a secretable from of the chaperone protein gp96). However, even these promising therapies suffer from difficulties in manufacturing and a lack of personalization that may render them ineffective for certain patients (e.g. due to a lack of tumor neo-antigens).
Similar to cancer pathogenesis, infectious pathogens create a hospitable environment within the host and modulate host metabolic functions to support their nutritional requirements, while suppressing host defenses by altering host's regulatory mechanisms. See Naran et al. (2018). Thus, targeted immunotherapy-based therapeutic approaches to infectious diseases are being developed, including those that enhance antigen-specific B cell antibody and T cell responses. However, such therapies are still in infancy, and much work remains to be done to expand immunotherapy approaches to treatment of infectious diseases.
Therefore, there is a need for effective and targeted approaches to treatment of cancer and infectious diseases.
SUMMARY
Accordingly, the present disclosure provides, in part, methods for making and using in a treatment a biological cell, which is made using cell fusion. The biological cell can be created using one or more fusion events, such that it can express a secretable vaccine protein and a T cell costimulatory fusion protein, and, In embodiments, one or more diseases antigens. The disease antigens can be infectious disease antigens.
In aspects, a method of making a biological cell is provided that comprises obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein; obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein; and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein. In embodiments, the secretable vaccine protein is a secretable gp96-lg fusion protein, which optionally lacks the gp96 KDEL (SEQ ID NO: 3) sequence.
In aspects, a method of making a biological cell is provided that comprises obtaining a first biological cell comprising a nucleotide sequence encoding a vaccine protein; obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein; and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
In embodiments, the vaccine protein is a native gp96 protein. In embodiments, the vaccine protein is secretable vaccine protein, such as a gp96-lg fusion protein.
In embodiments, the Ig tag in the gp96-lg fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or lgE.
In embodiments, the T cell costimulatory fusion protein is selected from OX40L-lg, ICOSL-lg, 4-1 BBL-lg, LAG3-lg, CD40L-lg, TL1 A-lg, CD70-lg, GITRL-lg, and CD28-lg. In embodiments, the Ig tag in the T cell costimulatory fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE.
In embodiments, the T cell costimulatory fusion protein is selected from OX40L-lg or a portion thereof that binds specifically to 0X40, ICOSL-lg or a portion thereof that binds specifically to IGOS, 4-1 BBL-lg, or a portion thereof that binds specifically to 4-1 BBR, CD40L-lg, or a portion thereof that binds specifically to CD40, CD70-lg, or a portion thereof that binds specifically to CD27, TL1 A-lg or a portion thereof that binds specifically to TNFRSF25, or GITRL-lg or a portion thereof that binds specifically to GITR.
In embodiments, the T cell costimulatory fusion protein is OX40L-lg or a portion thereof that binds specifically to 0X40.
In embodiments, the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to the patient.
In embodiments, the disclosure provides sequential fusion combination (e.g., a single cell is transduced with gp96-lg, and a single cell is transduced with T cell costimulatory fusion protein, e.g. OX40L-lg, then the cells are fused together).
In embodiments, the provided methods result in considerably greater expression of secretable vaccine protein (e.g., without limitation, gp96-lg) and/or T cell costimulatory fusion protein (e.g, without limitation OX40L-lg), as compared to an unfused cell that has been caused to express (e.g. transduced with nucleic acids encoding) the secretable vaccine protein (e.g, without limitation, gp96-lg) and/or T cell costimulatory fusion protein (e.g, without limitation OX40L-lg).
In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of from about 1 : 1 to about 1 :5. In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1 : 1. In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1 :3.
In embodiments, the fused biological cell expresses the secretable vaccine protein and OX40L-lg fusion protein in a ratio of from about 1 : 1 to about 1 :3.
In embodiments, the fused biological cell further comprises a nucleotide sequence encoding one or more disease antigens.
In embodiments, the fused biological cell is made by fusing together at least two biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, and the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein.
In embodiments, the fused biological cell is made by fusing together at least three biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens. Accordingly, In embodiments, the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens; wherein contacting the first biological cell and the second biological cell with the fusion agent further comprises contacting the third biological cell with the fusion agent, to result in the fused biological cell being created such that the fused biological cell comprises a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
In embodiments, the fused biological cell (created by fusing the first and second biological cells) is further fused with a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens, to thereby result in the fused biological cell (which can also be referred to as "a second fused biological cell”) that can express a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens. Accordingly, In embodiments, the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding one or more disease antigens; and contacting the third biological cell with the fusion agent; to result in a second fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
In embodiments, the second fused biological cell is immortalized.
In embodiments, one or more of the first, second, and third biological cells is a human tumor cell. In embodiments, all of the first, second, and third biological cells are human tumor cells. In embodiments, the human tumor cells are human primary humor cells.
In embodiments, the human tumor cell is a cell from an established non-small cell lung cancer (NSCLC), bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line. In embodiments, the human tumor cell is a cell from an established NSCLC cell line.
In embodiments, the third biological cell is a trophoblast or the third biological cell is fused with a trophoblast. In embodiments, one or both the first and second biological cells are fused with a trophoblast.
In embodiments, one or more of the first, second, and third biological cells are fused with a trophoblast or are trophoblasts.
In embodiments, one or more of the first, second, and third biological cells is a cell or is derived from a cell other than a cancer cell line.
In embodiments, all of the first, second, and third biological cells are cells or are derived from cells other than a cancer cell line. For example, in embodiments in which the fused biological cell is produced in accordance with embodiments of the present disclosure for treatment of an infectious disease, the fused biological cell can be produced by fusing two or more biological cells other than cells from a cancer cell line. In this way, the fused biological cell does not include cancer cells. In embodiments, one or more of the first, second, and third biological cells is a fibroblast or a fibroblast-like cell, or is derived from a fibroblast or a fibroblast-like cell. In embodiments, one or more of the first, second, and third biological cells are derived from a cell line such as, without limitation, MRC-5 or WI-38.
In embodiments, the fused biological cell is immortalized. In embodiments, at least one of the first, second, and third biological cells is immortalized.
In embodiments, the nucleotide sequence is a mammalian expression vector. In embodiments, the expression vector comprises DNA. In embodiments, the expression vector comprises RNA.
In embodiments, the expression vector is incorporated into a virus or virus-like particle.
In embodiments, the expression vector comprises a pCEP4-EGFP plasmid.
In embodiments, the fusion agent comprises a protein from Paramyxoviridae Genus paramyxovirus. The protein can be, for example, inactivated hemagglutinating virus of Japan envelope (HVJ-E).
In embodiments, the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof. In embodiments, the PEG moiety comprises PEG-1500.
In embodiments, one or more of the first biological cell, the second biological cell, and the third biological cell comprise at least one tumor antigen. In embodiments, the tumor antigen is a cancer testis (CT) antigen.
In embodiments, the fused biological cell expresses one or more cancer testis (CT) antigens.
In embodiments, one or more of the first, second, and third biological cells expresses one or more cancer testis (CT) antigens.
In embodiments, the fused biological cell is enriched for genes encoding one or more cancer testis (CT) antigens.
In embodiments, one or more of the first, second, and third biological cells expresses one or more cancer testis (CT) antigens.
In embodiments, the one or more disease antigens comprise one of more infectious disease antigens. In embodiments, the one or more infectious disease antigens comprise an antigenic fragment of a betacoronavirus protein or an alphacoronavirus protein, optionally wherein the betacoronavirus protein is selected from a SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1 , and HCoV-OC43 protein, or the alphacoronavirus protein is selected from a HCoV-NL63 and HCoV-229E protein.
In embodiments, the betacoronavirus protein is a SARS-CoV-2 protein (a 2019-nCoV protein).
In embodiments, the SARS-CoV-2 protein is selected from a spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N. In embodiments, the SARS-CoV-2 fragment comprises amino acid residues F486, N487, Q493, Q498, T500, N501 of the SARS-CoV-2 surface glycoprotein.
In embodiments, the SARS-CoV-2 peptide is a fragment of the SARS-CoV-2 RBD or the spike protein, including the wild type or a variant (also referred to as lineages). In embodiments, the SARS-CoV-2 peptide is a fragment of SEQ ID NO: 19 or a variant thereof. In embodiments, the wild type SARS-CoV-2 coronavirus is the "Wuhan strain.”
In embodiments, the present vaccine is pan-antigenic, thus providing immune response to the wild type (e.g., "Wuhan strain”) and numerous variants of the coronavirus. In embodiments, the present vaccine comprises one or more peptides of the wild type and/or a variants of the spike proteins, or RBD thereof. Accordingly, in embodiments, the vaccine includes two or more peptides of a respective variant, lineage, or strain of a coronavirus protein. For example, the variants can include a coronavirus protein having a mutation (e.g., without limitation, a substitution, deletion, or insertion) in any part of the spike, or the RBD thereof, protein, such as in the S1 subunit ( e.g, in the RBD of the Spike protein), or in the S2 subunit. In embodiments, a mutation is in a glycosylation site of the Spike protein.
In embodiments, the variant (also referred to as lineages) is one or more of B.1.1.7, B.1.351 , B.1.617.2, B.1.427, B.1.429, B.1.525, B.1.526, B.1.617.1 , B.1.617.3, B.1 , B.1.1.28, B.1.2, CAL.20C, B.6, P.1 , and P.2 variants and/or any other variants, or antigenic fragments thereof. In embodiments, the lineages include A.1 , A.2, A.3, A.4, A.5, A.6, A.7,
A.8, A.9, B, B.1 , B.1.1 , B.1.1.1 , B.2, B.3, B.4, B.5, B.6, B.7, B.9, B.10, B.11, B.12, B.13, B.14, B.15, B.16, B.17, B.18,
B.19, B.20, B.21 , B.22, B.23, B.24, B.25, B.26, B.27, C.1 , C.2, C.3, D.1 , and D2.
In embodiments, the variant is one or more of alpha (including B.1.1.7 and Q lineages), beta (including B.1.351 and descendent lineages), gamma (P.1 and descendent lineages), epsilon (including B.1.427 and B.1.429), eta (including B.1.525), iota (including B.1.526), kappa (including B.1.617.1), 1.617.3, mu (including B.1.621 , B.1.621.1), zeta (including P.2), delta (including B.1.617.2 and AY lineages), and omicron (including B.1.1.529).
In embodiments, the SARS-CoV-2 variant is B.1.1.7, also known as the Alpha variant. In embodiments, the B.1.1.7 ("Alpha”) variant comprises one or more mutations selected from 69del, 70del, 144del, (E484K*), (S494P*), N501Y, A570D, D614G, P681 H, T716I, S982A, and D1118H (K1191 N*), or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.351 , also known as the Beta variant. In embodiments, the B.1.351 ("Beta”) variant comprises one or more mutations selected from D80A, D215G, 241 del, 242del, 243del, K417N, E484K, N501Y, D614G, and A701V, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.617.2, also known as the Delta variant. In embodiments, the B.1.617.2 ("Delta”) variant comprises one or more mutations selected from T19R, (G142D*), 156del, 157del, R158G, L452R, T478K, D614G, P681 R, and D950N, or an antigenic fragment thereof. In embodiments, the SARS-CoV-2 variant is P.1, also known as the Gamma variant. In embodiments, the P.1 ("Gamma”) variant comprises one or more mutations selected from L18F, T20N, P26S, D138Y, R190S, K417T, E484K, N501Y, D614G, H655Y, and T1027I, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.427, also known as the Epsilon variant. In embodiments, the B.1.427 ("Epsilon”) variant comprises one or more mutations selected from L452R and D614G, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.429, also known as the Epsilon variant. In embodiments, the B.1.429 ("Epsilon”) variant comprises one or more mutations selected from S13I, W152C, L452R, and D614G, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.525, also known as the Eta variant. In embodiments, the B.1.525 ("Eta”) variant comprises one or more mutations selected from A67V, 69del, 70del, 144del, E484K, D614G, Q677H, and F888L, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.526, also known as the lota variant. In embodiments, the B.1.526 ("Iota”) variant comprises one or more mutations selected from L5F, (D80G*), T95I, (Y144-*), (F157S*), D253G, (L452R*), (S477N*), E484K, D614G, A701V, (T859N*), (D950H*), and (Q957R*), or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.617.1 , also known as the Kappa variant. In embodiments, the B.1.617.1 ("Kappa”) variant comprises one or more mutations selected from (T95I), G142D, E154K, L452R, E484Q, D614G, P681 R, and Q1071 H, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is B.1.617.3. In embodiments, the B.1.617.3 variant comprises one or more mutations selected from T19R, G142D, L452R, E484Q, D614G, P681R, D950N, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is P.2, also known as the Zeta variant. In embodiments, the P.2 ("Zeta”) variant comprises one or more mutations selected from E484K, (F565L*), D614G, and V1176F, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 variant is Delta (B.1.617.2). This variant was first identified in India in late 2020. The Delta variant is shown to be more infectious and spreads more rapidly than other variants. The Delta variant contains mutations in the spike protein: T19R, del157/158, L452R, T478K, D614G, P681 R, and D950N. See Dhar, Mahesh S et al. "Genomic characterization and epidemiology of an emerging SARS-CoV-2 variant in Delhi, India.” Science (New York, N.Y.) vol. 374,6570 (2021): 995-999. doi: 10.1126/science.abj9932. The Delta variant has more than a dozen mutations and more likely to result in hospitalization in the unvaccinated human population. See Twohig eta/., "Hospital admission and emergency care attendance risk for SARS-CoV-2 delta (B.1.617.2) compared with alpha (B.1.1.7) variants of concern: a cohort study.” published online August 27, 2021 , The Lancet. 22(1): 25-42. doi:10.1016/S1473- 3099(21)00475-8. In embodiments, the SARS-CoV-2 variant is the Omicron variant (B.1.1.529). This variant was discovered in South Africa in November 2021 . The World Health Organization has proposed the Omicron variant to be a variant of concern. Omicron carries about 50 mutations, 26 unique to the variant and more than 30 mutations on the spike protein. The Omicron variant contains substitutions in the spike protein: A67V, del69-70, T95I, del142-144, Y145D, del211, L212I, ins214EPE, G339D, S371 L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, Y505H, T547K, D614G, H655Y, N679K, P681 H, N764K, D796Y, N856K, Q954H, N969K, and L981 F. The Omicron variant contains receptor binding domain substitutions in the spike protein: G339D, S371 L, S373P, S375F, K417N, N440K, G446S, S477N, T478K, E484A, Q493R, G496S, Q498R, N501Y, and Y505H. See Qin etal., "Genome Characterization and Potential Risk Assessment of the Novel SARS-CoV-2 Variant Omicron (B.1.1.529).” Zoonoses. Vol. 1 (1). DOI: 10.15212/ZOONQSES-2021-0024.
In embodiments, a variant is a SARS-CoV-2 protein having a variation in a glycosylation site of a Spike protein.
In embodiments, a variant is a Spike protein having one or more of D614G, E484K, N501Y, K417N, S477G, and S477N mutations or an antigenic fragment thereof.
In embodiments, a variant is a Spike protein having a mutation in the RBD of the Spike protein. In embodiments, the mutation in the RBD of the Spike protein is a mutation in a glycosylation site in the RBD.
In embodiments, a variant is a Spike protein having a mutation outside the RBD of the Spike protein.
In aspects, a method of treating cancer is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In embodiments, the cancer comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia.
In aspects, a method of treating an infectious disease provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In embodiments, the infectious disease comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection. In embodiments, the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae. In embodiments, the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E. In embodiments, the infectious disease comprises a coronavirus infection 2019 (COVID-19).
In aspects, a method of stimulating a patient's antibody response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In aspects, a method of stimulating a patient T cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In aspects, a method of stimulating one or both a patient's antibody response and T cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In aspects, a method of stimulating a patient B cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with any of the embodiments of the present disclosure.
In aspects, a method of stimulating one or both a patient's antibody response and B cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In embodiments, the T cell-driven cellular immune response induces a CD8+ T cell response in the patient. In embodiments, the T cell-driven cellular immune response induces a CD4+ T cell response in the patient.
In embodiments, a method induces an immune response, selected from a CD8+ T cell, a CD4+ T cell, a cytotoxic T lymphocyte (CTL), a TH1 response, a TH2 response, or a combination thereof.
In embodiments, a method results in reduction of a number of regulatory T cells (Tregs).
In embodiments, a method results in the decrease in the functionality of Tregs. In embodiments, the method does not result in the decrease in the number of Tregs but results in the decrease in the functionality of Tregs.
In embodiments, the method results in a decrease in the frequency of Tregs. In embodiments, the method results in the decrease in the number of functional Tregs. In aspects, a method that makes use of a fused cell generated in accordance with embodiments of the present disclosure comprises administering to the patient an inhibitor of an immune checkpoint molecule. In embodiments, the immune checkpoint molecule is selected from PD-1 , PD-L1 , PD-L2, CTLA-4, ICOS, LAG3, 0X40, OX40L, and TIM3. In embodiments, the immune checkpoint molecule is PD-1.
In embodiments, a lentivirus vector encodes the nucleotide sequence encoding the secretable vaccine protein, and a lentivirus vector encodes the nucleotide sequence encoding the T cell costimulatory fusion protein.
In embodiments, the method further comprises merging together the first biological cell and the second biological cell.
In aspects, disclosed herein is a method for generating a cellular therapy, comprising: (a) obtaining a lentivirus, the lentivirus comprising: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, and an immunoglobulin, or a fragment thereof; and (b) introducing into a biological cell the lentivirus of step (a).
In aspects, disclosed herein is a method for generating a cellular therapy, comprising: (a) obtaining a lentivirus, the lentivirus comprising: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject; and (b) introducing into a biological cell the lentivirus of step (a).
In aspects, disclosed herein is a method for generating a cellular therapy, comprising: (a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a first biological cell; (b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject, and (i) introducing the nucleic acid into a second biological cell, and (c) merging together the first biological cell and the second biological cell.
In aspects, disclosed herein is a method for generating a cellular therapy, comprising: (a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a first biological cell; (b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a second biological cell, and (c) merging together the first biological cell and the second biological cell. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows both an image and a bar graph of clones developed from lentivirus transduction generated from DNA encoding for either gp96-lg or OX40L-lg, or both gp96-lg and OX40L-lg.
FIG. 2 is a graph illustrating expression of cancer testis antigens (CTAs) in HEK293 and HS112 (AD-100 expressing gp96) cells, and in a fusion of the HEK293 and HS122 cells.
FIG. 3 is a graph illustrating ELISA analysis of GP96-lgG and OX40L-lgG levels in individual lentiviral clones before and after cellular fusion.
DETAILED DESCRIPTION
The present disclosure is based, in part, on the discovery that cell fusion can be used to produce immune or immunotherapy agents for treatment of cancer and infectious diseases. One, two, or more than two biological cells can be fused to result in a fused biological cell comprising a nucleotide sequence encoding a vaccine protein (e.g., a secretable vaccine protein) and a nucleotide sequence encoding a T cell costimulatory fusion protein.
Cell fusion is a mechanism that enables genetic material to move from one cell to another and produce viable hybrid progeny. Bastida-Ruiz et al. (2016) Int. J. Mol. Sci.Vf ), 638. Cancer cells express selective antigens that are recognized frequently by the immune system and in some cases can promote natural remission of the tumor. See, e.g., Wirth et al., Frontiers in immunology vol. 8 1848. 19 Dec. 2017, doi:10.3389/fimmu.2017.01848. Studies have shown that formation of hybrid-cell vaccines that are produced through fusion of antigen presenting cells (APCs) with tumor cells, have been used to successfully exploit this natural defense using the mechanisms required for antigen presentation to express tumor antigens. See Gong et al. (1997), Nat Med 3, 558-561; Liu et al. (2019) Nat Commun 10, 3199. Cancer cell fusions can resemble other types of natural cell fusions. See Bastida-Ruiz (2016). One of the most prominent cell fusion events occurs during trophoblastic development. See, e.g., Omata et al. (2013) PLoS One 8(11):e81003. Genes and proteins in trophoblasts and cancer cells have many similarities. See, e.g., Bastida-Ruiz et al. (2016). Also, it has been shown that dendritic cell (DC)-tumor cell hybrids induced higher antigen-specific T-cell expansion as compared to simple mixture of tumor cells and DCs. Pereira Pinho et al. (2016) Cytotherapy Apr; 18(4):570-80. doi: 10.1016/j.jcyt.2016.01.005. The inventors have recognized and appreciated that, among other factors, the similarities between cancer cells and trophoblasts can be exploited in making immune agents, which can be made using cell fusion.
Accordingly, In aspects, the present disclosure provides methods for making a fused biological cell that can be generated as a result of one, two, or more than two fusion events. The fused biological cell can be made by fusing a first biological cell and a second biological cell, which can be then fused with a third biological cell. In embodiments, the fused biological cell can be made by fusing a first biological cell, a second biological cell, and a third biological cell. Also, more than three biological cells can be fused via one, two, or three cell fusion events.
In embodiments, the fused cell is created by fusing at least a first biological cell comprising a nucleotide sequence that encodes a secretable vaccine protein (such as exogenous gp96-lg fusion protein, which can lack KDEL (SEQ ID NO: 3) sequence) and a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein. In embodiments, a first biological cell comprises a nucleotide sequence that encodes a vaccine protein such as a native (non secreted) gp96 protein, or a secreted gp96-lg fusion protein. One or both the first biological cell and the second biological cell can be derived from a cell line producing at least one tumor antigen, such as, without limitation, a cancer testis (CT) antigen. In embodiments, a cell line is a polyploidy cell line that expresses multiple CT antigens.
In embodiments, the cell line can be created by fusing cancer tissue specific {e.g., breast, colon, pancreatic, lung, liver, etc.) cells with an established non-small cell lung cancer (NSCLC) cell line. In embodiments, the cell line is created by fusing an established NSCLC cell line with a trophoblast.
In embodiments, one or more of the first, second, and third biological cells is a cell or is derived from a cell other than a cancer cell line.
In embodiments, all of the first, second, and third biological cells are cells or are derived from cells other than a cancer cell line. For example, in embodiments in which the fused biological cell produced in accordance with embodiments of the present disclosure is used for treatment of an infectious disease, the fused biological cell is produced by fusing two or more biological cells other than cells from a cancer cell line. In this way, the fused biological cell does not include cancer cells. In embodiments in which the fused biological cell is used in another non-oncology application, the fused biological cell can similarly be produced by fusing two or more biological cells other than cells from a cancer cell line. In embodiments, one or more of the first, second, and third biological cells is a fibroblast or a fibroblast-like cell, or is derived from a fibroblast or a fibroblast-like cell. In embodiments, one or more of the first, second, and third biological cells are derived from a cell line such as, without limitation, MRC-5 or WI-38.
In aspects, a method of making a biological cell is provided that comprises obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
In embodiments, a method of making a fused biological cell is provided that comprises obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and causing the first biological cell and the second biological cell to fuse by contacting the first biological cell and the second biological cell with a fusion agent, thereby generating a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
In embodiments, the secretable vaccine protein is a heat shock protein (hsp) gp96 that is localized in the endoplasmic reticulum (ER) and serves as a chaperone for peptides on their way to MHC class I and II molecules. Gp96 obtained from tumor cells and used as a vaccine can induce specific tumor immunity, presumably through the transport of tumorspecific peptides to antigen-presenting cells (APCs) (Yamazaki et al., J Immunol 1999, 163(10):5178-5182). For example, gp96-associated peptides are cross-presented to CD8 cells by dendritic cells (DCs). Gp96-based vaccination modality has also been shown to provide protection against mucosal infection caused by simian immunodeficiency virus. Strbo et al., J Immunol. 2013; 190 (6): 2495-2499.
In embodiments in accordance with the present disclosure, the present compositions and methods use gp96 to trigger mucosal immunity by activating both B and T cell responses at the point of pathogen entry. The gp96-based composition activates the immune system thereby.
In embodiments, the secretable vaccine protein is a secretable gp96-lg fusion protein, which optionally lacks the gp96 KDEL (SEQ ID NO: 3) sequence. In embodiments, the Ig tag in the gp96-lg fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE.
In embodiments, a nucleotide sequence is provided that encodes a gp96-lg fusion protein. The coding region of human gp96 is 2,412 bases in length (SEQ ID NO: 1), and encodes an 803 amino acid protein (SEQ ID NO: 2) that includes a 21 amino acid signal peptide at the amino terminus, a potential transmembrane region rich in hydrophobic residues, and an ER retention peptide sequence at the carboxyl terminus (GENBANK® Accession No. X15187; see Maki et al., Proc Natl Acad Sc/ USA 1990, 87:5658-5562). The DNA and protein sequences of human gp96 follow: atgagggccctgtgggtgctgggcctctgctgcgtcctgctgaccttcgggtcggtcagagctgacgatgaagttgatgtggat ggtacagtagaagaggatctgggtaaaagtagagaaggatcaaggacggatgatgaagtagtacagagagaggaaga agctattcagttggatggattaaatgcatcacaaataagagaacttagagagaagtcggaaaagtttgccttccaagccgaa g ttaacag aatg atgaaacttatcatcaattcattgtataaaaataaag ag attttcctg ag ag aactg atttcaaatgcttctg at gctttagataagataaggctaatatcactgactgatgaaaatgctctttctggaaatgaggaactaacagtcaaaattaagtgt gataaggagaagaacctgctgcatgtcacagacaccggtgtaggaatgaccagagaagagttggttaaaaaccttggtac catagccaaatctgggacaagcgagtttttaaacaaaatgactgaagcacaggaagatggccagtcaacttctgaattgatt ggccagtttggtgtcggtttctattccgccttccttgtagcagataaggttattgtcacttcaaaacacaacaacgatacccagca catctgggagtctgactccaatgaattttctgtaattgctgacccaagaggaaacactctaggacggggaacgacaattaccc ttgtcttaaaagaagaagcatctgattaccttgaattggatacaattaaaaatctcgtcaaaaaatattcacagttcataaactttc ctatttatgtatggagcagcaagactgaaactgttgaggagcccatggaggaagaagaagcagccaaagaagagaaag aagaatctgatgatgaagctgcagtagaggaagaagaagaagaaaagaaaccaaagactaaaaaagttgaaaaaact gtctgggactgggaacttatgaatgatatcaaaccaatatggcagagaccatcaaaagaagtagaagaagatgaatacaa agctttctacaaatcattttcaaaggaaagtgatgaccccatggcttatattcactttactgctgaaggggaagttaccttcaaatc aattttatttgtacccacatctgctccacgtggtctgtttgacgaatatggatctaaaaagagcgattacattaagctctatgtgcgc cgtgtattcatcacagacgacttccatgatatgatgcctaaatacctcaattttgtcaagggtgtggtggactcagatgatctccc cttgaatgtttcccgcgagactcttcagcaacataaactgcttaaggtgattaggaagaagcttgttcgtaaaacgctggacat gatcaagaagattgctgatgataaatacaatgatactttttggaaagaatttggtaccaacatcaagcttggtgtgattgaagac cactcgaatcgaacacgtcttgctaaacttcttaggttccagtcttctcatcatccaactgacattactagcctagaccagtatgtg gaaagaatgaaggaaaaacaagacaaaatctacttcatggctgggtccagcagaaaagaggctgaatcttctccatttgttg agcgacttctgaaaaagggctatgaagttatttacctcacagaacctgtggatgaatactgtattcaggcccttcccgaatttgat gggaagaggttccagaatgttgccaaggaaggagtgaagttcgatgaaagtgagaaaactaaggagagtcgtgaagcag ttgagaaagaatttgagcctctgctgaattggatgaaagataaagcccttaaggacaagattgaaaaggctgtggtgtctcag cgcctgacagaatctccgtgtgctttggtggccagccagtacggatggtctggcaacatggagagaatcatgaaagcacaa gcgtaccaaacgggcaaggacatctctacaaattactatgcgagtcagaagaaaacatttgaaattaatcccagacacccg ctgatcagagacatgcttcgacgaattaaggaagatgaagatgataaaacagttttggatcttgctgtggttttgtttgaaacag caacgcttcggtcagggtatcttttaccagacactaaagcatatggagatagaatagaaagaatgcttcgcctcagtttgaac attgaccctgatgcaaaggtggaagaagagcccgaagaagaacctgaagagacagcagaagacacaacagaagaca cagagcaagacgaagatgaagaaatggatgtgggaacagatgaagaagaagaaacagcaaaggaatctacagctga aaaagatgaattgtaa (SEQ ID N0:1)
MRALWVLGLCCVLLTFGSVRADDEVDVDGTVEEDLGKSREGSRTDDEWQREEEAIQL DGLNASQIRELREKSEKFAFQAEVNRMMKLIINSLYKNKEIFLRELISNASDALDKIRLISLT DENALSGNEELTVKIKCDKEKNLLHVTDTGVGMTREELVKNLGTIAKSGTSEFLNKMTEA QEDGQSTSELIGQFGVGFYSAFLVADKVIVTSKHNNDTQHIWESDSNEFSVIADPRGNTL GRGTTITLVLKEEASDYLELDTIKNLVKKYSQFINFPIYVWSSKTETVEEPMEEEEAAKEEK EESDDEAAVEEEEEEKKPKTKKVEKTVWDWELMNDIKPIWQRPSKEVEEDEYKAFYKSF SKESDDPMAYIHFTAEGEVTFKSILFVPTSAPRGLFDEYGSKKSDYIKLYVRRVFITDDFH DMMPKYLNFVKGWDSDDLPLNVSRETLQQHKLLKVIRKKLVRKTLDMIKKIADDKYNDT FWKEFGTNIKLGVIEDHSNRTRLAKLLRFQSSHHPTDITSLDQYVERMKEKQDKIYFMAG SSRKEAESSPFVERLLKKGYEVIYLTEPVDEYCIQALPEFDGKRFQNVAKEGVKFDESEK TKESREAVEKEFEPLLNWMKDKALKDKIEKAWSQRLTESPCALVASQYGWSGNMERIM KAQAYQTGKDISTNYYASQKKTFEINPRHPLIRDMLRRIKEDEDDKTVLDLAWLFETATL RSGYLLPDTKAYGDRIERMLRLSLNIDPDAKVEEEPEEEPEETAEDTTEDTEQDEDEEMD VGTDEEEETAKESTAEKDEL (SEQ ID NO: 2).
A nucleic acid encoding a gp96-lg fusion sequence can be produced using the methods described in U.S. Patent No. 8,685,384, which is incorporated herein by reference in its entirety. In embodiments, the gp96 portion of a gp96-lg fusion protein can contain all or a portion of a wild type gp96 sequence (e.g., the human sequence set forth in SEQ ID NO: 2). For example, a secretable gp96-lg fusion protein can include the first 799 amino acids of SEQ ID NO: 2, such that it lacks the C-terminal KDEL (SEQ ID NO: 3) sequence. Alternatively, the gp96 portion of the fusion protein can have an amino acid sequence that includes one or more substitutions, deletions, or additions, as compared to the first 799 amino acids of the wild type gp96 sequence, such that it has at least 90% (e.g., at least 90%, at least 91 %, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%) sequence identity to the wild type polypeptide.
In embodiments, the gp96 portion of a nucleotide sequence encoding a gp96-lg fusion polypeptide can encode an amino acid sequence that differs from the wild type gp96 polypeptide at one or more amino acid positions, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms.
The Ig portion ("tag”) of a gp96-lg fusion protein can contain, for example, a non-variable portion of an immunoglobulin molecule (e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule). Typically, such portions contain at least functional CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. Fusions also can be made using the carboxyl terminus of the Fc portion of a constant domain, or a region immediately amino-terminal to the CH1 of the heavy or light chain. The Ig tag can be from a mammalian (e.g., human, mouse, monkey, or rat) immunoglobulin, but human immunoglobulin can be particularly useful when the gp96-lg fusion is intended for in vivo use for humans.
In embodiments, gp96, genetically fused to an immunoglobulin domain (e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule), activates TLR2 and TLR4 on professional antigen-presenting cells (APCs).
In embodiments, a gp96 peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen et al., J Immunol 1996, 156:442-449). This region of the lgG1 molecule contains three cysteine residues that normally are involved in disulfide bonding with other cysteines in the Ig molecule. Since none of the cysteines are required for the peptide to function as a tag, one or more of these cysteine residues can be substituted by another amino acid residue, such as, for example, serine.
Various leader sequences known in the art also can be used for efficient secretion of gp96-lg fusion proteins from bacterial and mammalian cells (see, von Heijne, J Mol Biol 1985, 184:99-105). Leader peptides can be selected based on the intended host cell, and may include bacterial, yeast, viral, animal, and mammalian sequences. For example, the herpes virus glycoprotein D leader peptide is suitable for use in a variety of mammalian cells. Another leader peptide for use in mammalian cells can be obtained from the V-J2-C region of the mouse immunoglobulin kappa chain (Bernard et al., Proc Natl Acad Sci USA 1981 , 78:5812-5816). DNA sequences encoding peptide tags or leader peptides are known or readily available from libraries or commercial suppliers, and are suitable in the fusion proteins described herein.
In embodiments, a nucleic acid sequence encodes a native gp96. Furthermore, in embodiments, one may substitute the gp96 of the present disclosure with one or more vaccine proteins. For instance, various heat shock proteins are among the vaccine proteins. In embodiments, the heat shock protein is one or more of a small hsp, hsp40, hsp60, hsp70, hsp90, and hsp110 family member, inclusive of fragments, variants, mutants, derivatives or combinations thereof (Hickey, et al., 1989, Mol. Cell. Biol. 9:2615-2626; Jindal, 1989, Mol. Cell. Biol. 9:2279-2283)
In embodiments, nucleotide sequences can be introduced into host cells for producing secreted vaccine proteins (e.g., gp96-lg), T cell costimulatory fusion proteins, and one or more disease antigens. There are a variety of techniques available for introducing nucleic acids into viable cells. Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro include the use of liposomes, electroporation, microinjection, cell fusion, polymer-based systems, DEAE-dextran, viral transduction, the calcium phosphate precipitation method, etc. For in vivo gene transfer, a number of techniques and reagents may also be used, including liposomes; natural polymer-based delivery vehicles, such as chitosan and gelatin; viral vectors are also suitable for in vivo transduction. In some situations, it is desirable to provide a targeting agent, such as an antibody or a ligand specific for a cell surface membrane protein. Where liposomes are employed, proteins which bind to a cell surface membrane protein associated with endocytosis may be used for targeting and/or to facilitate uptake, e.g., capsid proteins or fragments thereof tropic for a particular cell type, antibodies for proteins which undergo internalization in cycling, proteins that target intracellular localization and enhance intracellular half-life. The technique of receptor-mediated endocytosis is described, for example, by Wu et al., J. Biol. Chem. 262, 4429-4432 (1987); and Wagner et al., Proc. Natl. Acad. Sci. USA 87, 3410-3414 (1990).
Where appropriate, gene delivery agents such as, e.g., integration sequences can also be employed. Numerous integration sequences are known in the art (see, e.g, Nunes-Duby et al., Nucleic Acids Res. 26:391-406, 1998; Sadwoski, J. Bacteriol., 165:341-357, 1986; Bestor, Cell, 122 (3): 322-325, 2005; Plasterk et al., TIG 15:326-332, 1999; Kootstra et al., Ann. Rev. Pharm. Toxicol., 43:413-439, 2003). These include recombinases and transposases. Examples include Cre (Sternberg & Hamilton, J. Mol. Biol., 150:467-486, 1981), lambda (Nash, Nature, 247, 543-545, 1974), Flp (Broach et al., Cell, 29:227-234, 1982), R (Matsuzaki et al., J. Bacteriology, 172:610-618, 1990), cpC31 (see, e.g., Groth et al., J. Mol. Biol. 335:667-678, 2004), sleeping beauty, transposases of the mariner family (Plasterk et al., supra), and components for integrating viruses such as AAV, retroviruses, and antiviruses having components that provide for virus integration such as the LTR sequences of retroviruses or lentivirus and the ITR sequences of AAV (Kootstra et al., Ann. Rev. Pharm. Toxicol., 43:413-439, 2003).
Cells may be cultured in vitro or genetically engineered, for example. Host cells can be obtained from normal or affected subjects, including healthy humans, cancer patients, and patients with an infectious disease, private laboratory deposits, public culture collections such as the American Type Culture Collection, or from commercial suppliers. In embodiments, biological cells that can be used for production and secretion of gp96-lg fusion proteins or T cell costimulatory fusion proteins are human tumor cells. In embodiments, the human tumor cells are human primary tumor cells.
In embodiments, biological cells that can be used for production and secretion of gp96-lg fusion proteins and T cell costimulatory fusion proteins in vivo include, without limitation, epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes, blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, or granulocytes, various stem or progenitor cells, such as hematopoietic stem or progenitor cells (e.g., as obtained from bone marrow), umbilical cord blood, peripheral blood, fetal liver, etc., and tumor cells (e.g., human tumor cells). The choice of cell type depends on the type of tumor or infectious disease being treated, and can be determined by one of skill in the art.
Different biological host cells have characteristic and specific mechanisms for post-translational processing and modification of proteins. A biological cell may be chosen which modifies and processes the expressed gene products in a specific fashion similar to the way the recipient processes its heat shock proteins (hsps). For the purpose of producing large amounts of gp96-lg, it can be preferable that the type of host cell has been used for expression of heterologous genes, and is reasonably well characterized and developed for large-scale production processes. In embodiments, the host cells are autologous to the patient to whom the present fusion or recombinant cells secreting the present fusion proteins are subsequently administered.
In embodiments, in addition to a secretable vaccine protein, such as a gp96-lg fusion protein, a fused biological cell is made such that it also includes a nucleotide sequence encoding one or more biological response modifiers. In embodiments, the fused biological cell comprises a nucleotide sequence encoding one or more T cell costimulatory molecules.
In embodiments, the T cell costimulatory fusion protein is selected from OX40L-lg, ICOSL-lg, 4-1 BBL-lg, LAG3-lg, CD40L-lg, TL1 A-lg, CD70-lg, GITRL-lg, and CD28-lg.
In embodiments, the T cell costimulatory fusion protein is selected from OX40L-lg or a portion thereof that binds to 0X40, ICOSL-lg or a portion thereof that binds to IGOS, 4-1 BBL-lg or a portion thereof that binds to 4-1 BBR, TL1 A-lg or a portion thereof that binds to TNFRSF25, GITRL-lg or a portion thereof that binds to GITR, CD40-lg or a portion thereof that binds to CD40, or CD70-I g or a portion thereof that binds to CD27, among others.
The CD28-lg fusion protein binds to the costimulatory ligands CD80 and CD86 to provide a costimulatory signal to T cells.
In embodiments, the Ig tag in the T cell costimulatory fusion protein comprises the Fc region of human lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE.
In embodiments, the T cell costimulatory fusion protein is OX40L-lg. In embodiments, the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to the patient.
In addition to a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, the lentivirus and/or transfer plasmid provided herein can comprise one or more biological response modifiers. In embodiments, the lentivirus and/or transfer plasmid can encode one or more T cell costimulatory molecules or fusion proteins.
ICOS is an inducible T cell costimulatory receptor molecule that displays some homology to CD28 and CTLA-4, and interacts with B7-H2 expressed on the surface of antigen-presenting cells. ICOS has been implicated in the regulation of cell-mediated and humoral immune responses.
4-1 BB is a type 2 transmembrane glycoprotein belonging to the TNF superfamily, and is expressed on activated T Lymphocytes.
0X40 (also referred to as CD134 or TNFRSF4) is a T cell costimulatory molecule that is engaged by OX40L, and frequently is induced in antigen presenting cells and other cell types. 0X40 is known to enhance cytokine expression and survival of effector T cells.
GITR (TNFRSF18) is a T cell costimulatory molecule that is engaged by GITRL and is preferentially expressed in FoxP3+ regulatory T cells. GITR plays a significant role in the maintenance and function of Treg within the tumor microenvironment.
TNFRSF25 is a T cell costimulatory molecule that is preferentially expressed in CD4+ and CD8+ T cells following antigen stimulation. Signaling through TNFRSF25 is provided by TL1A, and functions to enhance T cell sensitivity to IL-2 receptor mediated proliferation in a cognate antigen dependent manner.
CD40 is a costimulatory protein found on various antigen presenting cells which plays a role in their activation. The binding of CD40L (CD154) on TH cells to CD40 activates antigen presenting cells and induces a variety of downstream effects.
CD27 a T cell costimulatory molecule belonging to the TNF superfamily which plays a role in the generation and longterm maintenance of T cell immunity. It binds to a ligand CD70 in various immunological processes. See van de Ven & Borst (2015) Immunotherapy 7(6): 655-67.
LAG3 (CD223) is mainly expressed in activated T and natural killer (NK) cells and was identified to as a marker for the activation of CD4+ and CD8+T cells. Puhr & Ilhan-Mutlu, ESMO Open 2019;4:e000482. doi: 10.1136/esmoopen-2018- 000482. LAG3-lg fusion protein binds to cell surface MHC class II. Andrews et al. (2017) Immunological reviews vol. 276,1 ; 80-96. doi:10.1111/imr.12519 Additional costimulatory molecules that may be used in embodiments of the present disclosure include, but are not limited to, HVEM, CD30, CD30L, CD40, CD70, LIGHT (CD258), B7-1 , and B7-2.
In embodiments, the present lentivirus and/or transfer plasmid comprises an agonist of 0X40 (e.g, an 0X40 ligand-lg (OX40L-lg) fusion, or a fragment thereof that binds 0X40), an agonist of inducible T-cell costimulator (IGOS) (e.g., an IGOS ligand-lg (ICOSL-lg) fusion, or a fragment thereof that binds IGOS), an agonist of CD40 (e.g., a CD40L-lg fusion protein, or fragment thereof), an agonist of CD27 (e.g. a CD70-lg fusion protein or fragment thereof), or an agonist of 4-1 BB (e.g, a 4-1 BB ligand-lg (4-1 BBL-lg) fusion, or a fragment thereof that binds 4-1 BB). In embodiments, the lentivirus and/or transfer plasmid comprises an agonist of TNFRSF25 (e.g, a TL1 A-lg fusion, or a fragment thereof that binds TNFRSF25), or an agonist of glucocorticoid-induced tumor necrosis factor receptor (GITR) (e.g, a GITR ligand-lg (GITRL-lg) fusion, or a fragment thereof that binds GITR), or an agonist of CD40 (e.g, a CD40 ligand-lg (CD40L-lg) fusion, or a fragment thereof that binds CD40); or an agonist of CD27 (e.g, a CD27 ligand-lg (e.g CD70L- Ig) fusion, or a fragment thereof that binds CD40).
The Ig portion ("tag”) of the T cell costimulatory fusion protein can include a non-variable portion of an immunoglobulin molecule (e.g, an lgG1, lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule). Such portions typically contain at least functional CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. In embodiments, a T cell costimulatory peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen etal., J Immunol 1996, 156:442-449). The Ig tag can be from a mammalian (e.g, human, mouse, monkey, or rat) immunoglobulin, but human immunoglobulin can be particularly useful when the fusion protein is intended for in vivo use for humans. DNAs encoding immunoglobulin light or heavy chain constant regions are known or readily available from cDNA libraries. Various leader sequences as described above also can be used for secretion of T cell costimulatory fusion proteins from bacterial and mammalian cells.
A representative nucleotide optimized sequence (SEQ ID NO:4) encoding the extracellular domain of human ICOSL fused to Ig, and the amino acid sequence of the encoded fusion (SEQ ID NO:5) are provided. In embodiments, the sequence of the entire fusion, or just the ICOSL component fused to another Fc domain is used:
ATGAGACTGGGAAGCCCTGGCCTGCTGTTTCTGCTGTTCAGCAGCCTGAGAGCCGA CACCCAGGAAAAAGAAGTGCGGGCCATGGTGGGAAGCGACGTGGAACTGAGCTGC GCCTGTCCTGAGGGCAGCAGATTCGACCTGAACGACGTGTACGTGTACTGGCAGAC CAGCGAGAGCAAGACCGTCGTGACCTACCACATCCCCCAGAACAGCTCCCTGGAAA ACGTGGACAGCCGGTACAGAAACCGGGCCCTGATGTCTCCTGCCGGCATGCTGAGA GGCGACTTCAGCCTGCGGCTGTTCAACGTGACCCCCCAGGACGAGCAGAAATTCCA CTGCCTGGTGCTGAGCCAGAGCCTGGGCTTCCAGGAAGTGCTGAGCGTGGAAGTGA CCCTGCACGTGGCCGCCAATTTCAGCGTGCCAGTGGTGTCTGCCCCCCACAGCCCT TCTCAGGATGAGCTGACCTTCACCTGTACCAGCATCAACGGCTACCCCAGACCCAAT GTGTACTGGATCAACAAGACCGACAACAGCCTGCTGGACCAGGCCCTGCAGAACGA TACCGTGTTCCTGAACATGCGGGGCCTGTACGACGTGGTGTCCGTGCTGAGAATCG CCAGAACCCCCAGCGTGAACATCGGCTGCTGCATCGAGAACGTGCTGCTGCAGCAG AACCTGACCGTGGGCAGCCAGACCGGCAACGACATCGGCGAGAGAGACAAGATCA CCGAGAACCCCGTGTCCACCGGCGAGAAGAATGCCGCCACCTCTAAGTACGGCCCT CCCTGCCCTTCTTGCCCAGCCCCTGAATTTCTGGGCGGACCCTCCGTGTTTCTGTTC CCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGACCCCCGAAGTGACCTGCGT GGTGGTGGATGTGTCCCAGGAAGATCCCGAGGTGCAGTTCAATTGGTACGTGGACG GGGTGGAAGTGCACAACGCCAAGACCAAGCCCAGAGAGGAACAGTTCAACAGCACC TACCGGGTGGTGTCTGTGCTGACCGTGCTGCACCAGGATTGGCTGAGCGGCAAAGA GTACAAGTGCAAGGTGTCCAGCAAGGGCCTGCCCAGCAGCATCGAAAAGACCATCA GCAACGCCACCGGCCAGCCCAGGGAACCCCAGGTGTACACACTGCCCCCTAGCCA GGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTGTCTCGTGAAGGGCTTCTACC CCTCCGATATCGCCGTGGAATGGGAGAGCAACGGCCAGCCAGAGAACAACTACAAG ACCACCCCCCCAGTGCTGGACAGCGACGGCTCATTCTTCCTGTACTCCCGGCTGAC AGTGGACAAGAGCAGCTGGCAGGAAGGCAACGTGTTCAGCTGCAGCGTGATGCACG AAGCCCTGCACAACCACTACACCCAGAAGTCCCTGTCTCTGTCCCTGGGCAAATGA (SEQ ID NO:4)
MRLGSPGLLFLLFSSLRADTQEKEVRAMVGSDVELSCACPEGSRFDLNDVYVYWQTSE SKTWTYHIPQNSSLENVDSRYRNRALMSPAGMLRGDFSLRLFNVTPQDEQKFHCLVLS QSLGFQEVLSVEVTLHVAANFSVPWSAPHSPSQDELTFTCTSINGYPRPNVYWINKTDN SLLDQALQNDTVFLNMRGLYDWSVLRIARTPSVNIGCCIENVLLQQNLTVGSQTGNDIG ERDKITENPVSTGEKNAATSKYGPPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVT CVWDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLSGK EYKCKVSSKGLPSSIEKTISNATGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDI AVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSSWQEGNVFSCSVMHEALHNH
YTQKSLSLSLGK (SEQ ID NO:5).
A representative nucleotide optimized sequence (SEQ ID NO:6) encoding the extracellular domain of human 4-1 BBL fused to Ig, and the encoded amino acid sequence (SEQ ID NO:7) are provided. In embodiments, the sequence of the entire fusion, or just the 4-1 BBL component fused to another Fc domain is used:
ATGTCTAAGTACGGCCCTCCCTGCCCTAGCTGCCCTGCCCCTGAATTTCTGGGCGG ACCCAGCGTGTTCCTGTTCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGA CCCCCGAAGTGACCTGCGTGGTGGTGGATGTGTCCCAGGAAGATCCCGAGGTGCA GTTCAATTGGTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCCAGAG AGGAACAGTTCAACAGCACCTACCGGGTGGTGTCCGTGCTGACCGTGCTGCACCAG GATTGGCTGAGCGGCAAAGAGTACAAGTGCAAGGTGTCCAGCAAGGGCCTGCCCAG CAGCATCGAGAAAACCATCAGCAACGCCACCGGCCAGCCCAGGGAACCCCAGGTGT ACACACTGCCCCCTAGCCAGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTGT CTCGTGAAGGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGAGCAACGGCCA GCCTGAGAACAACTACAAGACCACCCCCCCAGTGCTGGACAGCGACGGCTCATTCT TCCTGTACAGCAGACTGACCGTGGACAAGAGCAGCTGGCAGGAAGGCAACGTGTTC AGCTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTC TCTGAGCCTGGGCAAGGCCTGTCCATGGGCTGTGTCTGGCGCTAGAGCCTCTCCTG GATCTGCCGCCAGCCCCAGACTGAGAGAGGGACCTGAGCTGAGCCCCGATGATCCT GCCGGACTGCTGGATCTGAGACAGGGCATGTTCGCCCAGCTGGTGGCCCAGAACGT GCTGCTGATCGATGGCCCCCTGAGCTGGTACAGCGATCCTGGACTGGCTGGCGTGT CACTGACAGGCGGCCTGAGCTACAAAGAGGACACCAAAGAACTGGTGGTGGCCAAG GCCGGCGTGTACTACGTGTTCTTTCAGCTGGAACTGCGGAGAGTGGTGGCCGGCGA AGGATCCGGCTCTGTGTCTCTGGCTCTGCATCTGCAGCCCCTGAGATCTGCTGCTG GCGCTGCTGCTCTGGCCCTGACAGTGGACCTGCCTCCTGCCTCTAGCGAGGCCAGA AACAGCGCATTCGGGTTTCAAGGCAGACTGCTGCACCTGTCTGCCGGCCAGAGACT GGGAGTGCATCTGCACACAGAGGCCAGAGCCAGGCACGCCTGGCAGCTGACTCAG GGCGCTACAGTGCTGGGCCTGTTCAGAGTGACCCCCGAGATTCCAGCCGGCCTGCC TAGCCCCAGATCCGAATGA (SEQ ID NO:6)
MSKYGPPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNW YVDGVEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLSGKEYKCKVSSKGLPSSIEKTIS NATGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSRLTVDKSSWQEGNVFSCSVMHEALHNHYTQKSLSLSLGKACPWAV SGARASPGSAASPRLREGPELSPDDPAGLLDLRQGMFAQLVAQNVLLIDGPLSWYSDP GLAGVSLTGGLSYKEDTKELWAKAGVYYVFFQLELRRVVAGEGSGSVSLALHLQPLRS AAGAAALALTVDLPPASSEARNSAFGFQGRLLHLSAGQRLGVHLHTEARARHAWQLTQ GATVLGLFRVTPEIPAGLPSPRSE (SEQ ID N0:7).
A representative nucleotide optimized sequence (SEQ ID NO:8) encoding the extracellular domain of human TL1A fused to Ig, and the encoded amino acid sequence (SEQ ID NO:9) are provided. In embodiments, the sequence of the entire fusion, or just the TL1A component fused to another Fc domain is used:
ATGTCTAAGTACGGCCCTCCCTGCCCTAGCTGCCCTGCCCCTGAATTTCTGGGCGG ACCCAGCGTGTTCCTGTTCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGA CCCCCGAAGTGACCTGCGTGGTGGTGGATGTGTCCCAGGAAGATCCCGAGGTGCA GTTCAATTGGTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCCAGAG AGGAACAGTTCAACAGCACCTACCGGGTGGTGTCCGTGCTGACCGTGCTGCACCAG GATTGGCTGAGCGGCAAAGAGTACAAGTGCAAGGTGTCCAGCAAGGGCCTGCCCAG CAGCATCGAGAAAACCATCAGCAACGCCACCGGCCAGCCCAGGGAACCCCAGGTGT ACACACTGCCCCCTAGCCAGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTGT CTCGTGAAGGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGAGCAACGGCCA GCCTGAGAACAACTACAAGACCACCCCCCCAGTGCTGGACAGCGACGGCTCATTCT TCCTGTACAGCAGACTGACCGTGGACAAGAGCAGCTGGCAGGAAGGCAACGTGTTC AGCTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTC TCTGAGCCTGGGCAAGATCGAGGGCCGGATGGATAGAGCCCAGGGCGAAGCCTGC GTGCAGTTCCAGGCTCTGAAGGGCCAGGAATTCGCCCCCAGCCACCAGCAGGTGTA CGCCCCTCTGAGAGCCGACGGCGATAAGCCTAGAGCCCACCTGACAGTCGTGCGG CAGACCCCTACCCAGCACTTCAAGAATCAGTTCCCCGCCCTGCACTGGGAGCACGA ACTGGGCCTGGCCTTCACCAAGAACAGAATGAACTACACCAACAAGTTTCTGCTGAT CCCCGAGAGCGGCGACTACTTCATCTACAGCCAAGTGACCTTCCGGGGCATGACCA GCGAGTGCAGCGAGATCAGACAGGCCGGCAGACCTAACAAGCCCGACAGCATCAC CGTCGTGATCACCAAAGTGACCGACAGCTACCCCGAGCCCACCCAGCTGCTGATGG GCACCAAGAGCGTGTGCGAAGTGGGCAGCAACTGGTTCCAGCCCATCTACCTGGGC GCCATGTTTAGTCTGCAAGAGGGCGACAAGCTGATGGTCAACGTGTCCGACATCAG CCTGGTGGATTACACCAAAGAGGACAAGACCTTCTTCGGCGCCTTTCTGCTCTGA (SEQ ID NO:8)
MSKYGPPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNW YVDGVEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLSGKEYKCKVSSKGLPSSIEKTIS NATGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSRLTVDKSSWQEGNVFSCSVMHEALHNHYTQKSLSLSLGKIEGRMDR AQGEACVQFQALKGQEFAPSHQQVYAPLRADGDKPRAHLTVVRQTPTQHFKNQFPALH WEHELGLAFTKNRMNYTNKFLLIPESGDYFIYSQVTFRGMTSECSEIRQAGRPNKPDSIT WITKVTDSYPEPTQLLMGTKSVCEVGSNWFQPIYLGAMFSLQEGDKLMVNVSDISLVDY TKEDKTFFGAFLL (SEQ ID NO:9). A representative nucleotide optimized sequence (SEQ ID NO:10) encoding human QX40L-lg, and the encoded amino acid sequence (SEQ ID NO:11) are provided. In embodiments, the sequence of the entire fusion, or just the OX40L component fused to another Fc domain is used:
ATGTCTAAGTACGGCCCTCCCTGCCCTAGCTGCCCTGCCCCTGAATTTCTGGGCGG ACCCAGCGTGTTCCTGTTCCCCCCAAAGCCCAAGGACACCCTGATGATCAGCCGGA CCCCCGAAGTGACCTGCGTGGTGGTGGATGTGTCCCAGGAAGATCCCGAGGTGCA GTTCAATTGGTACGTGGACGGCGTGGAAGTGCACAACGCCAAGACCAAGCCCAGAG AGGAACAGTTCAACAGCACCTACCGGGTGGTGTCCGTGCTGACCGTGCTGCACCAG GATTGGCTGAGCGGCAAAGAGTACAAGTGCAAGGTGTCCAGCAAGGGCCTGCCCAG CAGCATCGAGAAAACCATCAGCAACGCCACCGGCCAGCCCAGGGAACCCCAGGTGT ACACACTGCCCCCTAGCCAGGAAGAGATGACCAAGAACCAGGTGTCCCTGACCTGT CTCGTGAAGGGCTTCTACCCCTCCGATATCGCCGTGGAATGGGAGAGCAACGGCCA GCCTGAGAACAACTACAAGACCACCCCCCCAGTGCTGGACAGCGACGGCTCATTCT TCCTGTACAGCAGACTGACCGTGGACAAGAGCAGCTGGCAGGAAGGCAACGTGTTC AGCTGCAGCGTGATGCACGAGGCCCTGCACAACCACTACACCCAGAAGTCCCTGTC TCTGAGCCTGGGCAAGATCGAGGGCCGGATGGATCAGGTGTCACACAGATACCCCC GGATCCAGAGCATCAAAGTGCAGTTTACCGAGTACAAGAAAGAGAAGGGCTTTATCC TGACCAGCCAGAAAGAGGACGAGATCATGAAGGTGCAGAACAACAGCGTGATCATC AACTGCGACGGGTTCTACCTGATCAGCCTGAAGGGCTACTTCAGTCAGGAAGTGAAC ATCAGCCTGCACTACCAGAAGGACGAGGAACCCCTGTTCCAGCTGAAGAAAGTGCG GAGCGTGAACAGCCTGATGGTGGCCTCTCTGACCTACAAGGACAAGGTGTACCTGA ACGTGACCACCGACAACACCAGCCTGGACGACTTCCACGTGAACGGCGGCGAGCTG
ATCCTGATTCACCAGAACCCCGGCGAGTTCTGCGTGCTCTGA (SEQ ID NO: 10)
MSKYGPPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNW YVDGVEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLSGKEYKCKVSSKGLPSSIEKTIS NATGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSRLTVDKSSWQEGNVFSCSVMHEALHNHYTQKSLSLSLGKIEGRMD QVSHRYPRIQSIKVQFTEYKKEKGFILTSQKEDEIMKVQNNSVIINCDGFYLISLKGYFSQE VNISLHYQKDEEPLFQLKKVRSVNSLMVASLTYKDKVYLNVTTDNTSLDDFHVNGGELILI HQNPGEFCVL (SEQ ID NO:11).
Representative nucleotide and amino acid sequences for human TL1A are set forth in SEQ ID NO:12 and SEQ ID NO: 13, respectively. In embodiments, the sequence of the entire fusion, or just the TL1A component fused to another Fc domain is used:
TCCCAAGTAGCTGGGACTACAGGAGCCCACCACCACCCCCGGCTAATTTTTTGTATT TTTAGTAGAGACGGGGTTTCACCGTGTTAGCCAAGATGGTCTTGATCACCTGACCTC GTGATCCACCCGCCTTGGCCTCCCAAAGTGCTGGGATTACAGGCATGAGCCACCGC GCCCGGCCTCCATTCAAGTCTTTATTGAATATCTGCTATGTTCTACACACTGTTCTAG GTGCTGGGGATGCAACAGGGGACAAAATAGGCAAAATCCCTGTCCTTTTGGGGTTG ACATTCTAGTGACTCTTCATGTAGTCTAGAAGAAGCTCAGTGAATAGTGTCTGTGGTT GTTACCAGGGACACAATGACAGGAACATTCTTGGGTAGAGTGAGAGGCCTGGGGAG GGAAGGGTCTCTAGGATGGAGCAGATGCTGGGCAGTCTTAGGGAGCCCCTCCTGGC ATGCACCCCCTCATCCCTCAGGCCACCCCCGTCCCTTGCAGGAGCACCCTGGGGAG CTGTCCAGAGCGCTGTGCCGCTGTCTGTGGCTGGAGGCAGAGTAGGTGGTGTGCTG GGAATGCGAGTGGGAGAACTGGGATGGACCGAGGGGAGGCGGGTGAGGAGGGGG GCAACCACCCAACACCCACCAGCTGCTTTCAGTGTTCTGGGTCCAGGTGCTCCTGG
CTGGCCTTGTGGTCCCCCTCCTGCTTGGGGCCACCCTGACCTACACATACCGCCAC TGCTGGCCTCACAAGCCCCTGGTTACTGCAGATGAAGCTGGGATGGAGGCTCTGAC CCCACCACCGGCCACCCATCTGTCACCCTTGGACAGCGCCCACACCCTTCTAGCAC CTCCTGACAGCAGTGAGAAGATCTGCACCGTCCAGTTGGTGGGTAACAGCTGGACC CCTGGCTACCCCGAGACCCAGGAGGCGCTCTGCCCGCAGGTGACATGGTCCTGGG ACCAGTTGCCCAGCAGAGCTCTTGGCCCCGCTGCTGCGCCCACACTCTCGCCAGAG TCCCCAGCCGGCTCGCCAGCCATGATGCTGCAGCCGGGCCCGCAGCTCTACGACG TGATGGACGCGGTCCCAGCGCGGCGCTGGAAGGAGTTCGTGCGCACGCTGGGGCT GCGCGAGGCAGAGATCGAAGCCGTGGAGGTGGAGATCGGCCGCTTCCGAGACCAG CAGTACGAGATGCTCAAGCGCTGGCGCCAGCAGCAGCCCGCGGGCCTCGGAGCCG TTTACGCGGCCCTGGAGCGCATGGGGCTGGACGGCTGCGTGGAAGACTTGCGCAG CCGCCTGCAGCGCGGCCCGTGACACGGCGCCCACTTGCCACCTAGGCGCTCTGGT GGCCCTTGCAGAAGCCCTAAGTACGGTTACTTATGCGTGTAGACATTTTATGTCACTT ATTAAGCCGCTGGCACGGCCCTGCGTAGCAGCACCAGCCGGCCCCACCCCTGCTC GCCCCTATCGCTCCAGCCAAGGCGAAGAAGCACGAACGAATGTCGAGAGGGGGTG AAGACATTTCTCAACTTCTCGGCCGGAGTTTGGCTGAGATCGCGGTATTAAATCTGT
GAAAGAAAACAAAACAAAACAA (SEQ ID NO: 12)
MEQRPRGCAAVAAALLLVLLGARAQGGTRSPRCDCAGDFHKKIGLFCCRGCPAGHYLK APCTEPCGNSTCLVCPQDTFLAWENHHNSECARCQACDEQASQVALENCSAVADTRC GCKPGWFVECQVSQCVSSSPFYCQPCLDCGALHRHTRLLCSRRDTDCGTCLPGFYEH GDGCVSCPTPPPSLAGAPWGAVQSAVPLSVAGGRVGVFWVQVLLAGLWPLLLGATLT YTYRHCWPHKPLVTADEAGMEALTPPPATHLSPLDSAHTLLAPPDSSEKICTVQLVGNS WTPGYPETQEALCPQVTWSWDQLPSRALGPAAAPTLSPESPAGSPAMMLQPGPQLYD
VMDAVPARRWKEFVRTLGLREAEIEAVEVEIGRFRDQQYEMLKRWRQQQPAGLGAVYA ALERMGLDGCVEDLRSRLQRGP (SEQ ID NO: 13).
Representative nucleotide and amino acid sequences for human HVEM are set forth in SEQ ID NO:26 (accession no. CR456909) and SEQ ID NO:27, respectively (accession no. CR456909). In embodiments, the sequence of the entire fusion, or just the HVEM component fused to another Fc domain is used:
ATGGAGCCTCCTGGAGACTGGGGGCCTCCTCCCTGGAGATCCACCCCCAAAACCGA CGTCTTGAGGCTGGTGCTGTATCTCACCTTCCTGGGAGCCCCCTGCTACGCCCCAG CTCTGCCGTCCTGCAAGGAGGACGAGTACCCAGTGGGCTCCGAGTGCTGCCCCAAG TGCAGTCCAGGTTATCGTGTGAAGGAGGCCTGCGGGGAGCTGACGGGCACAGTGT GTGAACCCTGCCCTCCAGGCACCTACATTGCCCACCTCAATGGCCTAAGCAAGTGTC TGCAGTGCCAAATGTGTGACCCAGCCATGGGCCTGCGCGCGAGCCGGAACTGCTCC AGGACAGAGAACGCCGTGTGTGGCTGCAGCCCAGGCCACTTCTGCATCGTCCAGGA CGGGGACCACTGCGCCGCGTGCCGCGCTTACGCCACCTCCAGCCCGGGCCAGAGG GTGCAGAAGGGAGGCACCGAGAGTCAGGACACCCTGTGTCAGAACTGCCCCCCGG GGACCTTCTCTCCCAATGGGACCCTGGAGGAATGTCAGCACCAGACCAAGTGCAGC
TGGCTGGTGACGAAGGCCGGAGCTGGGACCAGCAGCTCCCACTGGGTATGGTGGT TTCTCTCAGGGAGCCTCGTCATCGTCATTGTTTGCTCCACAGTTGGCCTAATCATATG TGTGAAAAGAAGAAAGCCAAGGGGTGATGTAGTCAAGGTGATCGTCTCCGTCCAGC GGAAAAGACAGGAGGCAGAAGGTGAGGCCACAGTCATTGAGGCCCTGCAGGCCCC TCCGGACGTCACCACGGTGGCCGTGGAGGAGACAATACCCTCATTCACGGGGAGGA GCCCAAACCATTAA (SEQ ID NO:26)
MEPPGDWGPPPWRSTPKTDVLRLVLYLTFLGAPCYAPALPSCKEDEYPVGSECCPKCS
PGYRVKEACGELTGTVCEPCPPGTYIAHLNGLSKCLQCQMCDPAMGLRASRNCSRTEN
AVCGCSPGHFCIVQDGDHCAACRAYATSSPGQRVQKGGTESQDTLCQNCPPGTFSPN GTLEECQHQTKCSWLVTKAGAGTSSSHWVWWFLSGSLVIVIVCSTVGLIICVKRRKPRG
DWKVIVSVQRKRQEAEGEATVIEALQAPPDVTTVAVEETIPSFTGRSPNH (SEQ ID NO:27).
Representative nucleotide and amino acid sequences for human CD28 are set forth in SEQ ID NO:28 (accession no. NM_006139) and SEQ ID NO:29, respectively. In embodiments, the sequence of the entire fusion, or just the CD28 component fused to another Fc domain is used:
TAAAGTCATCAAAACAACGTTATATCCTGTGTGAAATGCTGCAGTCAGGATGCCTTGT GGTTTGAGTGCCTTGATCATGTGCCCTAAGGGGATGGTGGCGGTGGTGGTGGCCGT GGATGACGGAGACTCTCAGGCCTTGGCAGGTGCGTCTTTCAGTTCCCCTCACACTTC GGGTTCCTCGGGGAGGAGGGGCTGGAACCCTAGCCCATCGTCAGGACAAAGATGC TCAGGCTGCTCTTGGCTCTCAACTTATTCCCTTCAATTCAAGTAACAGGAAACAAGAT TTTGGTGAAGCAGTCGCCCATGCTTGTAGCGTACGACAATGCGGTCAACCTTAGCTG CAAGTATTCCTACAATCTCTTCTCAAGGGAGTTCCGGGCATCCCTTCACAAAGGACT GGATAGTGCTGTGGAAGTCTGTGTTGTATATGGGAATTACTCCCAGCAGCTTCAGGT TTACTCAAAAACGGGGTTCAACTGTGATGGGAAATTGGGCAATGAATCAGTGACATT CTACCTCCAGAATTTGTATGTTAACCAAACAGATATTTACTTCTGCAAAATTGAAGTTA TGTATCCTCCTCCTTACCTAGACAATGAGAAGAGCAATGGAACCATTATCCATGTGAA AGGGAAACACCTTTGTCCAAGTCCCCTATTTCCCGGACCTTCTAAGCCCTTTTGGGT GCTGGTGGTGGTTGGTGGAGTCCTGGCTTGCTATAGCTTGCTAGTAACAGTGGCCTT TATTATTTTCTGGGTGAGGAGTAAGAGGAGCAGGCTCCTGCACAGTGACTACATGAA CATGACTCCCCGCCGCCCCGGGCCCACCCGCAAGCATTACCAGCCCTATGCCCCAC CACGCGACTTCGCAGCCTATCGCTCCTGACACGGACGCCTATCCAGAAGCCAGCCG GCTGGCAGCCCCCATCTGCTCAATATCACTGCTCTGGATAGGAAATGACCGCCATCT CCAGCCGGCCACCTCAGGCCCCTGTTGGGCCACCAATGCCAATTTTTCTCGAGTGA CTAGACCAAATATCAAGATCATTTTGAGACTCTGAAATGAAGTAAAAGAGATTTCCTG TGACAGGCCAAGTCTTACAGTGCCATGGCCCACATTCCAACTTACCATGTACTTAGT GACTTGACTGAGAAGTTAGGGTAGAAAACAAAAAGGGAGTGGATTCTGGGAGCCTCT TCCCTTTCTCACTCACCTGCACATCTCAGTCAAGCAAAGTGTGGTATCCACAGACATT TTAGTTGCAGAAGAAAGGCTAGGAAATCATTCCTTTTGGTTAAATGGGTGTTTAATCT TTTGGTTAGTGGGTTAAACGGGGTAAGTTAGAGTAGGGGGAGGGATAGGAAGACAT ATTTAAAAACCATTAAAACACTGTCTCCCACTCATGAAATGAGCCACGTAGTTCCTATT TAATGCTGTTTTCCTTTAGTTTAGAAATACATAGACATTGTCTTTTATGAATTCTGATCA TATTTAGTCATTTTGACCAAATGAGGGATTTGGTCAAATGAGGGATTCCCTCAAAGCA ATATCAGGTAAACCAAGTTGCTTTCCTCACTCCCTGTCATGAGACTTCAGTGTTAATG
TTCACAATATACTTTCGAAAGAATAAAATAGTTCTCCTACATGAAGAAAGAATATGTCA
GGAAATAAGGTCACTTTATGTCAAAATTATTTGAGTACTATGGGACCTGGCGCAGTG
GCTCATGCTTGTAATCCCAGCACTTTGGGAGGCCGAGGTGGGCAGATCACTTGAGA
TCAGGACCAGCCTGGTCAAGATGGTGAAACTCCGTCTGTACTAAAAATACAAAATTTA
GCTTGGCCTGGTGGCAGGCACCTGTAATCCCAGCTGCCCAAGAGGCTGAGGCATGA
GAATCGCTTGAACCTGGCAGGCGGAGGTTGCAGTGAGCCGAGATAGTGCCACAGCT
CTCCAGCCTGGGCGACAGAGTGAGACTCCATCTCAAACAACAACAACAACAACAACA
ACAACAACAAACCACAAAATTATTTGAGTACTGTGAAGGATTATTTGTCTAACAGTTCA
TTCCAATCAGACCAGGTAGGAGCTTTCCTGTTTCATATGTTTCAGGGTTGCACAGTTG
GTCTCTTTAATGTCGGTGTGGAGATCCAAAGTGGGTTGTGGAAAGAGCGTCCATAGG
AGAAGTGAGAATACTGTGAAAAAGGGATGTTAGCATTCATTAGAGTATGAGGATGAG
TCCCAAGAAGGTTCTTTGGAAGGAGGACGAATAGAATGGAGTAATGAAATTCTTGCC
ATGTGCTGAGGAGATAGCCAGCATTAGGTGACAATCTTCCAGAAGTGGTCAGGCAGA
AGGTGCCCTGGTGAGAGCTCCTTTACAGGGACTTTATGTGGTTTAGGGCTCAGAGCT
CCAAAACTCTGGGCTCAGCTGCTCCTGTACCTTGGAGGTCCATTCACATGGGAAAGT
ATTTTGGAATGTGTCTTTTGAAGAGAGCATCAGAGTTCTTAAGGGACTGGGTAAGGC
CTGACCCTGAAATGACCATGGATATTTTTCTACCTACAGTTTGAGTCAACTAGAATAT
GCCTGGGGACCTTGAAGAATGGCCCTTCAGTGGCCCTCACCATTTGTTCATGCTTCA
GTTAATTCAGGTGTTGAAGGAGCTTAGGTTTTAGAGGCACGTAGACTTGGTTCAAGT
CTCGTTAGTAGTTGAATAGCCTCAGGCAAGTCACTGCCCACCTAAGATGATGGTTCT
TCAACTATAAAATGGAGATAATGGTTACAAATGTCTCTTCCTATAGTATAATCTCCATA
AGGGCATGGCCCAAGTCTGTCTTTGACTCTGCCTATCCCTGACATTTAGTAGCATGC
CCGACATACAATGTTAGCTATTGGTATTATTGCCATATAGATAAATTATGTATAAAAAT
TAAACTGGGCAATAGCCTAAGAAGGGGGGAATATTGTAACACAAATTTAAACCCACTA
CGCAGGGATGAGGTGCTATAATATGAGGACCTTTTAACTTCCATCATTTTCCTGTTTC
TTGAAATAGTTTATCTTGTAATGAAATATAAGGCACCTCCCACTTTTATGTATAGAAAG
AGGTCTTTTAATTTTTTTTTAATGTGAGAAGGAAGGGAGGAGTAGGAATCTTGAGATT
CCAGATCGAAAATACTGTACTTTGGTTGATTTTTAAGTGGGCTTCCATTCCATGGATT
TAATCAGTCCCAAGAAGATCAAACTCAGCAGTACTTGGGTGCTGAAGAACTGTTGGA
TTTACCCTGGCACGTGTGCCACTTGCCAGCTTCTTGGGCACACAGAGTTCTTCAATC
CAAGTTATCAGATTGTATTTGAAAATGACAGAGCTGGAGAGTTTTTTGAAATGGCAGT
GGCAAATAAATAAATACTTTTTTTTAAATGGAAAGACTTGATCTATGGTAATAAATGAT
TTTGTTTTCTGACTGGAAAAATAGGCCTACTAAAGATGAATCACACTTGAGATGTTTCT TACTCACTCTGCACAGAAACAAAGAAGAAATGTTATACAGGGAAGTCCGTTTTCACTA
TTAGTATGAACCAAGAAATGGTTCAAAAACAGTGGTAGGAGCAATGCTTTCATAGTTT
CAGATATGGTAGTTATGAAGAAAACAATGTCATTTGCTGCTATTATTGTAAGAGTCTTA
TAATTAATGGTACTCCTATAATTTTTGATTGTGAGCTCACCTATTTGGGTTAAGCATGC
CAATTTAAAGAGACCAAGTGTATGTACATTATGTTCTACATATTCAGTGATAAAATTAC
TAAACTACTATATGTCTGCTTTAAATTTGTACTTTAATATTGTCTTTTGGTATTAAGAAA
GATATGCTTTCAGAATAGATATGCTTCGCTTTGGCAAGGAATTTGGATAGAACTTGCT
ATTTAAAAGAGGTGTGGGGTAAATCCTTGTATAAATCTCCAGTTTAGCCTTTTTTGAAA
AAGCTAGACTTTCAAATACTAATTTCACTTCAAGCAGGGTACGTTTCTGGTTTGTTTG
CTTGACTTCAGTCACAATTTCTTATCAGACCAATGGCTGACCTCTTTGAGATGTCAGG
CTAGGCTTACCTATGTGTTCTGTGTCATGTGAATGCTGAGAAGTTTGACAGAGATCCA
ACTTCAGCCTTGACCCCATCAGTCCCTCGGGTTAACTAACTGAGCCACCGGTCCTCA
TGGCTATTTTAATGAGGGTATTGATGGTTAAATGCATGTCTGATCCCTTATCCCAGCC
ATTTGCACTGCCAGCTGGGAACTATACCAGACCTGGATACTGATCCCAAAGTGTTAA
ATTCAACTACATGCTGGAGATTAGAGATGGTGCCAATAAAGGACCCAGAACCAGGAT
CTTGATTGCTATAGACTTATTAATAATCCAGGTCAAAGAGAGTGACACACACTCTCTC
AAGACCTGGGGTGAGGGAGTCTGTGTTATCTGCAAGGCCATTTGAGGCTCAGAAAG
TCTCTCTTTCCTATAGATATATGCATACTTTCTGACATATAGGAATGTATCAGGAATAC
TCAACCATCACAGGCATGTTCCTACCTCAGGGCCTTTACATGTCCTGTTTACTCTGTC
TAGAATGTCCTTCTGTAGATGACCTGGCTTGCCTCGTCACCCTTCAGGTCCTTGCTC
AAGTGTCATCTTCTCCCCTAGTTAAACTACCCCACACCCTGTCTGCTTTCCTTGCTTA
TTTTTCTCCATAGCATTTTACCATCTCTTACATTAGACATTTTTCTTATTTATTTGTAGTT
TATAAGCTTCATGAGGCAAGTAACTTTGCTTTGTTTCTTGCTGTATCTCCAGTGCCCA
GAGCAGTGCCTGGTATATAATAAATATTTATTGACTGAGTGAAAAAAAAAAAAAAAAA (SEQ ID NO:28)
MLRLLLALNLFPSIQVTGNKILVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDS
AVEVCWYGNYSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKIEVMYPP
PYLDNEKSNGTIIHVKGKHLCPSPLFPGPSKPFWVLVWGGVLACYSLLVTVAFIIFWVRS KRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS (SEQ ID NO:29).
Representative nucleotide and amino acid sequences for human CD30L are set forth in SEQ ID NQ:30 (accession no. L09753) and SEQ ID NO:31, respectively. In embodiments, the sequence of the entire fusion, or just the CD30L component fused to another Fc domain is used: CCAAGTCACATGATTCAGGATTCAGGGGGAGAATCCTTCTTGGAACAGAGATGGGCC
CAGAACTGAATCAGATGAAGAGAGATAAGGTGTGATGTGGGGAAGACTATATAAAGA
ATGGACCCAGGGCTGCAGCAAGCACTCAACGGAATGGCCCCTCCTGGAGACACAGC
CATGCATGTGCCGGCGGGCTCCGTGGCCAGCCACCTGGGGACCACGAGCCGCAGC
TATTTCTATTTGACCACAGCCACTCTGGCTCTGTGCCTTGTCTTCACGGTGGCCACTA
TTATGGTGTTGGTCGTTCAGAGGACGGACTCCATTCCCAACTCACCTGACAACGTCC
CCCTCAAAGGAGGAAATTGCTCAGAAGACCTCTTATGTATCCTGAAAAGAGCTCCATT
CAAGAAGTCATGGGCCTACCTCCAAGTGGCAAAGCATCTAAACAAAACCAAGTTGTC
TTGGAACAAAGATGGCATTCTCCATGGAGTCAGATATCAGGATGGGAATCTGGTGAT
CCAATTCCCTGGTTTGTACTTCATCATTTGCCAACTGCAGTTTCTTGTACAATGCCCA
AATAATTCTGTCGATCTGAAGTTGGAGCTTCTCATCAACAAGCATATCAAAAAACAGG
CCCTGGTGACAGTGTGTGAGTCTGGAATGCAAACGAAACACGTATACCAGAATCTCT
CTCAATTCTTGCTGGATTACCTGCAGGTCAACACCACCATATCAGTCAATGTGGATAC
ATTCCAGTACATAGATACAAGCACCTTTCCTCTTGAGAATGTGTTGTCCATCTTCTTAT
ACAGTAATTCAGACTGAACAGTTTCTCTTGGCCTTCAGGAAGAAAGCGCCTCTCTAC
CATACAGTATTTCATCCCTCCAAACACTTGGGCAAAAAGAAAACTTTAGACCAAGACA
AACTACACAGGGTATTAAATAGTATACTTCTCCTTCTGTCTCTTGGAAAGATACAGCT
CCAGGGTTAAAAAGAGAGTTTTTAGTGAAGTATCTTTCAGATAGCAGGCAGGGAAGC
AATGTAGTGTGGTGGGCAGAGCCCCACACAGAATCAGAAGGGATGAATGGATGTCC
CAGCCCAACCACTAATTCACTGTATGGTCTTGATCTATTTCTTCTGTTTTGAGAGCCT
CCAGTTAAAATGGGGCTTCAGTACCAGAGCAGCTAGCAACTCTGCCCTAATGGGAAA
TGAAGGGGAGCTGGGTGTGAGTGTTTACACTGTGCCCTTCACGGGATACTTCTTTTA
TCTGCAGATGGCCTAATGCTTAGTTGTCCAAGTCGCGATCAAGGACTCTCTCACACA
GGAAACTTCCCTATACTGGCAGATACACTTGTGACTGAACCATGCCCAGTTTATGCCT
GTCTGACTGTCACTCTGGCACTAGGAGGCTGATCTTGTACTCCATATGACCCCACCC
CTAGGAACCCCCAGGGAAAACCAGGCTCGGACAGCCCCCTGTTCCTGAGATGGAAA
GCACAAATTTAATACACCACCACAATGGAAAACAAGTTCAAAGACTTTTACTTACAGA
TCCTGGACAGAAAGGGCATAATGAGTCTGAAGGGCAGTCCTCCTTCTCCAGGTTACA
TGAGGCAGGAATAAGAAGTCAGACAGAGACAGCAAGACAGTTAACAACGTAGGTAAA
GAAATAGGGTGTGGTCACTCTCAATTCACTGGCAAATGCCTGAATGGTCTGTCTGAA
GGAAGCAACAGAGAAGTGGGGAATCCAGTCTGCTAGGCAGGAAAGATGCCTCTAAG
TTCTTGTCTCTGGCCAGAGGTGTGGTATAGAACCAGAAACCCATATCAAGGGTGACT
AAGCCCGGCTTCCGGTATGAGAAATTAAACTTGTATACAAAATGGTTGCCAAGGCAA
CATAAAATTATAAGAATTC (SEQ ID NO:30) MDPGLQQALNGMAPPGDTAMHVPAGSVASHLGTTSRSYFYLTTATLALCLVFTVATIMV LWQRTDSIPNSPDNVPLKGGNCSEDLLCILKRAPFKKSWAYLQVAKHLNKTKLSWNKD GILHGVRYQDGNLVIQFPGLYFIICQLQFLVQCPNNSVDLKLELLINKHIKKQALVTVCESG MQTKHVYQNLSQFLLDYLQVNTTISVNVDTFQYIDTSTFPLENVLSIFLYSNSD (SEQ ID NO:31).
Representative nucleotide and amino acid sequences for human CD40 are set forth in SEQ ID NO:32 (accession no. NM_001250) and SEQ ID NO:33, respectively. In embodiments, the sequence of the entire fusion, or just the CD40 component fused to another Fc domain is used:
TTTCCTGGGCGGGGCCAAGGCTGGGGCAGGGGAGTCAGCAGAGGCCTCGCTCGGG CGCCCAGTGGTCCTGCCGCCTGGTCTCACCTCGCTATGGTTCGTCTGCCTCTGCAG TGCGTCCTCTGGGGCTGCTTGCTGACCGCTGTCCATCCAGAACCACCCACTGCATG CAGAGAAAAACAGTACCTAATAAACAGTCAGTGCTGTTCTTTGTGCCAGCCAGGACA GAAACTGGTGAGTGACTGCACAGAGTTCACTGAAACGGAATGCCTTCCTTGCGGTGA AAGCGAATTCCTAGACACCTGGAACAGAGAGACACACTGCCACCAGCACAAATACTG CGACCCCAACCTAGGGCTTCGGGTCCAGCAGAAGGGCACCTCAGAAACAGACACCA TCTGCACCTGTGAAGAAGGCTGGCACTGTACGAGTGAGGCCTGTGAGAGCTGTGTC CTGCACCGCTCATGCTCGCCCGGCTTTGGGGTCAAGCAGATTGCTACAGGGGTTTC TGATACCATCTGCGAGCCCTGCCCAGTCGGCTTCTTCTCCAATGTGTCATCTGCTTT CGAAAAATGTCACCCTTGGACAAGCTGTGAGACCAAAGACCTGGTTGTGCAACAGGC AGGCACAAACAAGACTGATGTTGTCTGTGGTCCCCAGGATCGGCTGAGAGCCCTGG TGGTGATCCCCATCATCTTCGGGATCCTGTTTGCCATCCTCTTGGTGCTGGTCTTTAT CAAAAAGGTGGCCAAGAAGCCAACCAATAAGGCCCCCCACCCCAAGCAGGAACCCC AGGAGATCAATTTTCCCGACGATCTTCCTGGCTCCAACACTGCTGCTCCAGTGCAGG AGACTTTACATGGATGCCAACCGGTCACCCAGGAGGATGGCAAAGAGAGTCGCATC TCAGTGCAGGAGAGACAGTGAGGCTGCACCCACCCAGGAGTGTGGCCACGTGGGC AAACAGGCAGTTGGCCAGAGAGCCTGGTGCTGCTGCTGCTGTGGCGTGAGGGTGA GGGGCTGGCACTGACTGGGCATAGCTCCCCGCTTCTGCCTGCACCCCTGCAGTTTG AGACAGGAGACCTGGCACTGGATGCAGAAACAGTTCACCTTGAAGAACCTCTCACTT CACCCTGGAGCCCATCCAGTCTCCCAACTTGTATTAAAGACAGAGGCAGAAGTTTGG TGGTGGTGGTGTTGGGGTATGGTTTAGTAATATCCACCAGACCTTCCGATCCAGCAG TTTGGTGCCCAGAGAGGCATCATGGTGGCTTCCCTGCGCCCAGGAAGCCATATACA CAGATGCCCATTGCAGCATTGTTTGTGATAGTGAACAACTGGAAGCTGCTTAACTGT CCATCAGCAGGAGACTGGCTAAATAAAATTAGAATATATTTATACAACAGAATCTCAA AAACACTGTTGAGTAAGGAAAAAAAGGCATGCTGCTGAATGATGGGTATGGAACTTT TTAAAAAAGTACATGCTTTTATGTATGTATATTGCCTATGGATATATGTATAAATACAAT ATGCATCATATATTGATATAACAAGGGTTCTGGAAGGGTACACAGAAAACCCACAGCT CGAAGAGTGGTGACGTCTGGGGTGGGGAAGAAGGGTCTGGGGG (SEQ ID NO:32)
MVRLPLQCVLWGCLLTAVHPEPPTACREKQYLINSQCCSLCQPGQKLVSDCTEFTETEC LPCGESEFLDTWNRETHCHQHKYCDPNLGLRVQQKGTSETDTICTCEEGWHCTSEACE SCVLHRSCSPGFGVKQIATGVSDTICEPCPVGFFSNVSSAFEKCHPWTSCETKDLWQQ AGTNKTDWCGPQDRLRALWIPIIFGILFAILLVLVFIKKVAKKPTNKAPHPKQEPQEINFP DDLPGSNTAAPVQETLHGCQPVTQEDGKESRISVQERQ (SEQ ID NO:33).
Representative nucleotide and amino acid sequences for human CD70 are set forth in SEQ ID NO:34 (accession no. NM_001252) and SEQ ID NO:35, respectively. In embodiments, the sequence of the entire fusion, or just the CD70 component fused to another Fc domain is used:
CCAGAGAGGGGCAGGCTGGTCCCCTGACAGGTTGAAGCAAGTAGACGCCCAGGAG CCCCGGGAGGGGGCTGCAGTTTCCTTCCTTCCTTCTCGGCAGCGCTCCGCGCCCCC ATCGCCCCTCCTGCGCTAGCGGAGGTGATCGCCGCGGCGATGCCGGAGGAGGGTT CGGGCTGCTCGGTGCGGCGCAGGCCCTATGGGTGCGTCCTGCGGGCTGCTTTGGT CCCATTGGTCGCGGGCTTGGTGATCTGCCTCGTGGTGTGCATCCAGCGCTTCGCAC AGGCTCAGCAGCAGCTGCCGCTCGAGTCACTTGGGTGGGACGTAGCTGAGCTGCA GCTGAATCACACAGGACCTCAGCAGGACCCCAGGCTATACTGGCAGGGGGGCCCA GCACTGGGCCGCTCCTTCCTGCATGGACCAGAGCTGGACAAGGGGCAGCTACGTAT CCATCGTGATGGCATCTACATGGTACACATCCAGGTGACGCTGGCCATCTGCTCCTC CACGACGGCCTCCAGGCACCACCCCACCACCCTGGCCGTGGGAATCTGCTCTCCCG CCTCCCGTAGCATCAGCCTGCTGCGTCTCAGCTTCCACCAAGGTTGTACCATTGCCT CCCAGCGCCTGACGCCCCTGGCCCGAGGGGACACACTCTGCACCAACCTCACTGG GACACTTTTGCCTTCCCGAAACACTGATGAGACCTTCTTTGGAGTGCAGTGGGTGCG CCCCTGACCACTGCTGCTGATTAGGGTTTTTTAAATTTTATTTTATTTTATTTAAGTTCA AGAGAAAAAGTGTACACACAGGGGCCACCCGGGGTTGGGGTGGGAGTGTGGTGGG GGGTAGTGGTGGCAGGACAAGAGAAGGCATTGAGCTTTTTCTTTCATTTTCCTATTAA AAAATACAAAAATCA (SEQ ID NO:34)
MPEEGSGCSVRRRPYGCVLRAALVPLVAGLVICLWCIQRFAQAQQQLPLESLGWDVAE LQLNHTGPQQDPRLYWQGGPALGRSFLHGPELDKGQLRIHRDGIYMVHIQVTLAICSSTT ASRHHPTTLAVGICSPASRSISLLRLSFHQGCTIASQRLTPLARGDTLCTNLTGTLLPSRN TDETFFGVQWVRP(SEQ ID NO: 35). Representative nucleotide and amino acid sequences for human LIGHT are set forth in SEQ ID NO:36 (accession no. CR541854) and SEQ ID NO:37, respectively. In embodiments, the sequence of the entire fusion, or just the LIGHT component fused to another Fc domain is used:
ATGGAGGAGAGTGTCGTACGGCCCTCAGTGTTTGTGGTGGATGGACAGACCGACAT CCCATTCACGAGGCTGGGACGAAGCCACCGGAGACAGTCGTGCAGTGTGGCCCGG GTGGGTCTGGGTCTCTTGCTGTTGCTGATGGGGGCCGGGCTGGCCGTCCAAGGCT GGTTCCTCCTGCAGCTGCACTGGCGTCTAGGAGAGATGGTCACCCGCCTGCCTGAC GGACCTGCAGGCTCCTGGGAGCAGCTGATACAAGAGCGAAGGTCTCACGAGGTCAA CCCAGCAGCGCATCTCACAGGGGCCAACTCCAGCTTGACCGGCAGCGGGGGGCCG CTGTTATGGGAGACTCAGCTGGGCCTGGCCTTCCTGAGGGGCCTCAGCTACCACGA TGGGGCCCTTGTGGTCACCAAAGCTGGCTACTACTACATCTACTCCAAGGTGCAGCT GGGCGGTGTGGGCTGCCCGCTGGGCCTGGCCAGCACCATCACCCACGGCCTCTAC AAGCGCACACCCCGCTACCCCGAGGAGCTGGAGCTGTTGGTCAGCCAGCAGTCACC CTGCGGACGGGCCACCAGCAGCTCCCGGGTCTGGTGGGACAGCAGCTTCCTGGGT GGTGTGGTACACCTGGAGGCTGGGGAGGAGGTGGTCGTCCGTGTGCTGGATGAAC GCCTGGTTCGACTGCGTGATGGTACCCGGTCTTACTTCGGGGCTTTCATGGTGTGA (SEQ ID NO:36)
MEESWRPSVFWDGQTDIPFTRLGRSHRRQSCSVARVGLGLLLLLMGAGLAVQGWFLL QLHWRLGEMVTRLPDGPAGSWEQLIQERRSHEVNPAAHLTGANSSLTGSGGPLLWET QLGLAFLRGLSYHDGALWTKAGYYYIYSKVQLGGVGCPLGLASTITHGLYKRTPRYPEE LELLVSQQSPCGRATSSSRVWWDSSFLGGWHLEAGEEVWRVLDERLVRLRDGTRSY FGAFMV (SEQ ID NO:37).
A representative amino acid sequences for human GITRL are set forth in SEQ ID NO:52 (accession no. Q9UNG2), respectively. In embodiments, the sequence of the entire fusion, or just the GITRL component fused to another Fc domain is used:
MSKYGPPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSQEDPEVQFNW YVDGVEVHNAKTKPREEQFNSTYRWSVLTVLHQDWLSGKEYKCKVSSKGLPSSIEKTIS NATGQPREPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSFFLYSRLTVDKSSWQEGNVFSCSVMHEALHNHYTQKSLSLSLGKIEGRMD MTLHPSPITCEFLFSTALISPKMCLSHLENMPLSHSRTQGAQRSSWKLWLFCSIVMLLFL CSFSWLIFIFLQLETAKEPCMAKFGPLPSKWQMASSEPPCVNKVSDWKLEILQNGLYLIY GQVAPNANYNDVAPFEVRLYKNKDMIQTLTNKSKIQNVGGTYELHVGDTIDLIFNSEHQV LKNNTYWGIILLANPQFIS (SEQ ID NO:52). In embodiments, variants are provided comprising any of the sequences described herein, for instance, a sequence having at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71 %, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81 %, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 92%, or at least about 93%, or at least about 94%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99%) sequence identity with any of the sequences disclosed herein (for example, SEQ ID NOS: 1-13 and 26-37 and 52).
In embodiments, an amino acid sequence is provided that has one or more amino acid mutations relative to any of the protein sequences described herein. In embodiments, the one or more amino acid mutations may be independently selected from conservative or non-conservative substitutions, insertions, deletions, and truncations as described herein.
As defined herein, a "conservative substitution” denotes the replacement of an amino acid residue by another, biologically similar, residue. Typically, biological similarity, as referred to above, reflects substitutions on the wild type sequence with conserved amino acids. For example, conservative amino acid substitutions would be expected to have little or no effect on biological activity, particularly if they represent less than 10% of the total number of residues in the polypeptide or protein. Conservative substitutions may be made, for instance, on the basis of similarity in polarity, charge, size, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the amino acid residues involved. The 20 naturally occurring amino acids can be grouped into the following six standard amino acid groups: (1) hydrophobic: Met, Ala, Vai, Leu, lie; (2) neutral hydrophilic: Cys, Ser, Thr; Asn, Gin; (3) acidic: Asp, Glu; (4) basic: His, Lys, Arg; (5) residues that influence chain orientation: Gly, Pro; and (6) aromatic: Trp, Tyr, Phe. Accordingly, conservative substitutions may be affected by exchanging an amino acid by another amino acid listed within the same group of the six standard amino acid groups shown above. For example, the exchange of Asp by Glu retains one negative charge in the so modified polypeptide. In addition, glycine and proline may be substituted for one another based on their ability to disrupt o-helices. Additional examples of conserved amino acid substitutions, include, without limitation, the substitution of one hydrophobic residue for another, such as isoleucine, valine, leucine, or methionine, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like. The term "conservative substitution” also includes the use of a substituted amino acid residue in place of an un-substituted parent amino acid residue, provided that antibodies raised to the substituted polypeptide also immunoreact with the un-substituted polypeptide. As used herein, "non-conservative substitutions” are defined as exchanges of an amino acid by another amino acid listed in a different group of the six standard amino acid groups (1) to (6) shown above.
In embodiments, the substitutions may also include non-classical amino acids (e.g. selenocysteine, pyrrolysine, N- formylmethionine p-alanine, GABA and 6-Aminolevulinic acid, 4-aminobenzoic acid (PABA), D-isomers of the common amino acids, 2,4-diaminobutyric acid, o-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, y-Abu, s-Ahx, 6-amino hexanoic acid, Alb, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosme, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, p-alanine, fluoro-amino acids, designer amino acids such as p methyl amino acids, C o-methyl amino acids, N o-methyl amino acids, and amino acid analogs in general).
In embodiments, mutations may also be made to the nucleotide sequences of the present fusion proteins by reference to the genetic code, including taking into account codon degeneracy.
In embodiments, the gp96-lg fusion protein and/or a T cell costimulatory fusion protein comprises a linker. In embodiments, the linker may be derived from naturally-occurring multi-domain proteins or are empirical linkers as described, for example, in Chichili et al. (2013) Protein Sci. 22(2): 153-167; Chen et al. (2013), Adv Drug Deliv Rev. 65(10): 1357-1369, the entire contents of which are hereby incorporated by reference. In embodiments, the linker may be designed using linker designing databases and computer programs such as those described in Chen et al. (2013) Adv Drug Deliv Rev. 65(10): 1357-1369 and Crasto et. al. (2000) Protein Eng. 13(5):309-312, the entire contents of which are hereby incorporated by reference.
In embodiments, the linker is a synthetic linker such as PEG.
In embodiments, the linker is a polypeptide. In embodiments, the linker is less than about 100 amino acids long. For example, the linker may be less than about 100, about 95, about 90, about 85, about 80, about 75, about 70, about 65, about 60, about 55, about 50, about 45, about 40, about 35, about 30, about 25, about 20, about 19, about 18, about 17, about 16, about 15, about 14, about 13, about 12, about 11 , about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, or about 2 amino acids long. In embodiments, the linker is flexible. In another embodiment, the linker is rigid. In embodiments, the linker is substantially comprised of glycine and serine residues (e.g. about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 95%, or about 97% glycines and serines).
In embodiments, the linker is a hinge region of an antibody (e.g., of IgG, IgA, IgD, and IgE, inclusive of subclasses (e.g. lgG1, lgG2, lgG3, and lgG4, and lgA1 and lgA2)). The hinge region, found in IgG, IgA, IgD, and IgE class antibodies, acts as a flexible spacer, allowing the Fab portion to move freely in space. In contrast to the constant regions, the hinge domains are structurally diverse, varying in both sequence and length among immunoglobulin classes and subclasses. For example, the length and flexibility of the hinge region varies among the IgG subclasses. The hinge region of lgG1 encompasses amino acids 216-231 and, because it is freely flexible, the Fab fragments can rotate about their axes of symmetry and move within a sphere centered at the first of two inter-heavy chain disulfide bridges. I gG2 has a shorter hinge than lgG1 , with 12 amino acid residues and four disulfide bridges. The hinge region of lgG2 lacks a glycine residue, is relatively short, and contains a rigid poly-proline double helix, stabilized by extra inter-heavy chain disulfide bridges. These properties restrict the flexibility of the lgG2 molecule. lgG3 differs from the other subclasses by its unique extended hinge region (about four times as long as the lgG1 hinge), containing 62 amino acids (including 21 prolines and 11 cysteines), forming an inflexible poly-proline double helix. In lgG3, the Fab fragments are relatively far away from the Fc fragment, giving the molecule a greater flexibility. The elongated hinge in lgG3 is also responsible for its higher molecular weight compared to the other subclasses. The hinge region of lgG4 is shorter than that of lgG1 and its flexibility is intermediate between that of IgG 1 and I gG2. The flexibility of the hinge regions reportedly decreases in the order lgG3>lgG 1 >lgG4>lgG2.
Additional illustrative linkers include, but are not limited to, linkers having the sequence LE, GGGGS (SEQ ID NO: 14), (GGGGS)n (n=1-4) (SEQ ID NO: 15), (Gly)s (SEQ ID NO: 16), (Gly)6 (SEQ ID NO: 17), (EAAAK)n (n=1-3) (SEQ ID NO: 18), A(EAAAK)nA (n = 2-5) (SEQ ID NO: 19), AEAAAKEAAAKA (SEQ ID NO: 20), A(EAAAK)4ALEA(EAAAK)4A (SEQ ID NO: 21), PAPAP (SEQ ID NO: 22), KESGSVSSEQLAQFRSLD (SEQ ID NO: 23), EGKSSGSGSESKST(SEQ ID NO: 24), GSAGSAAGSGEF (SEQ ID NO: 25), and (XP)n, with X designating any amino acid, e.g., Ala, Lys, or Glu.
In embodiments, the linker may be functional. For example, without limitation, the linker may function to improve the folding and/or stability, improve the expression, improve the pharmacokinetics, and/or improve the bioactivity of the present compositions. In another example, the linker may function to target the compositions to a particular cell type or location.
It has been previously discovered that a combination of a vaccination, e.g. gp96-lg vaccination, and T cell costimulation with one or more agonists of 0X40, IGOS, 4-1 BB, TNFRSF25, CD40, CD27, and/or GITR, among others, provides a synergistic anti-tumor benefit, as described in International Application PCT/US2016/016682 (published as WO2016127015) which is incorporated by reference herein in its entirety. An expression vector was engineered to include both the gp96-lg fusion protein and a T cell costimulatory fusion protein, such that a combination immunotherapy could be achieved by vector re-engineering to obviate the need for vaccine/antibody/fusion protein regimens, which may reduce a cost of therapy and the risk of systemic toxicity. In embodiments, a therapy additionally includes a checkpoint inhibitor. Further, the need for using separate different drug products {e.g., a vaccine, a T cell costimulatory protein, and a checkpoint inhibitor) is obviated.
The present disclosure further improves the combination therapy by fusing first and second biological cells including nucleotide sequences encoding a vaccine protein (such as a secretable vaccine protein) and a T cell costimulatory fusion protein, respectively, such that a ratio of a secretable vaccine protein and a T cell costimulatory fusion protein is controlled. The first and second biological cells can be generated to express a known quantity of a vaccine protein and a T cell costimulatory fusion protein, respectively, such that the fused biological cell expresses known, desired quantities of the vaccine protein and the T cell costimulatory fusion protein. In this way, the vaccine protein and the T cell costimulatory fusion protein can be expressed in the fused cell at a desired ratio. This allows creating compositions for treating cancer or an infectious disease with an appropriate dosage and ratio of components.
The vaccine protein can be a secretable vaccine protein, such as a gp96-lg fusion protein, or a native gp96 protein.
In embodiments, the fused biological cell expresses the vaccine protein (e.g., the secretable gp96-lg fusion protein or gp96 in its native form) and the T cell costimulatory fusion protein in a ratio of from about 1:1 to about 1:5. In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1:1, or about 1:1.5, or about 1:2, or about 1:2.5, or about 1:3, or about 1:3.5, or about 1:4, or about 1:4.5, or about 1:5. In embodiments, the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1:3.
In embodiments, the fused biological cell expresses the vaccine protein, such as gp96-lg fusion protein or a native gp96 protein, and OX40L- 1 g fusion protein in a ratio of about 1 :3.
In embodiments, a fused biological cell is created such that an amount of a vaccine protein (e.g., gp96-lg or native gp96) is lower than the expression of a T cell costimulatory fusion protein, (e.g., OX40-lg or any of the other fusion proteins including those described herein). In embodiments, a ratio of an amount of a vaccine protein (e.g., gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is about 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1:25, 1:30, 1:40, 1:50, 1:60, 1:70, 1:80. 1:90, or 1:100. In embodiments, a ratio of an amount of a vaccine protein (e.g., gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is from about 1:6 to about 1:100, inclusive of all endpoints.
In embodiments, secretable vaccine protein (e.g., gp96-lg) secretion is lower than the expression of a T cell costimulatory fusion protein (e.g, OX40-lg). In embodiments, a ratio of secretable vaccine protein (e.g, gp96-lg) secretion to the expression of aT cell costimulatory fusion protein (e.g, OX40-lg) is about 1:6, 1:7, 1:8, 1:9, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:16, 1:17, 1:18, 1:19, 1:20, 1:25, 1:30, 1:40, 1:50, 1:60, 1:70, 1:80. 1:90, or 1:100. In embodiments, a ratio of secretable vaccine protein (e.g, gp96-lg) secretion to the expression of a T cell costimulatory fusion protein (e.g, OX40-lg) is from about 1:6 to about 1:100, inclusive of all endpoints.
Furthermore, In embodiments, an amount of a vaccine protein (e.g, gp96-lg or native gp96) is higher than the expression of a T cell costimulatory fusion protein (e.g, OX40-lg). In embodiments, the ratio of an amount of vaccine protein (e.g, gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g, OX40-lg) is about 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, 11:1, 12:1, 13:1, 14:1, 15:1, 16:1, 17:1, 18:1, 19:1, 20:1, 25:1, 30:1, 40:1, or 50:1. In embodiments, the ratio of an amount of vaccine protein (e.g, gp96-lg or native gp96) to the expression of a T cell costimulatory fusion protein (e.g, OX40-lg) is from about 2:1 to about 50:1, inclusive of all endpoints.
In embodiments, secretable vaccine protein (e.g, gp96-lg) secretion is higher than the expression of a T cell costimulatory fusion protein (e.g., OX40-lg). costimulatory fusion protein (e.g., OX40-lg). In embodiments, the ratio of the vaccine protein (e.g., gp96-lg) secretion to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is about 2:1 , 3:1 , 4: 1 , 5: 1 , 6: 1 , 7:1 , 8: 1, 9: 1 , 10:1 , 11 :1 , 12: 1 , 13:1 , 14: 1 , 15:1 , 16: 1, 17: 1 , 18:1 , 19: 1 , 20:1 , 25:1 , 30: 1 , 40:1 , or 50: 1. In embodiments, the ratio of the vaccine protein (e.g., gp96-lg) secretion to the expression of a T cell costimulatory fusion protein (e.g., OX40-lg) is from about 2: 1 to about 50: 1 , inclusive of all endpoints.
In embodiments, a combination therapy in accordance with embodiments of the present disclosure involves the use of a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein, as well as a checkpoint inhibitor. For example, the fused biological cell can express a gp96-lg fusion protein and a T cell costimulatory fusion protein such as, without limitation, OX40L, and a checkpoint inhibitor such as, without limitation, checkpoint inhibitors that block immune checkpoints such as PD-1 , PD-L1 , PD-L2, IGOS, LAG3, TIM3, or CTLA-4. In embodiments, checkpoint inhibitors are, without limitation, antibodies such as an anti-PD1 antibody, anti-PDL1 antibody, anti-PDL2 antibody, anti-ICOS antibody, anti-CTLA-4 antibody, anti-TIM-3 antibody, and/or anti-LAG-3 antibody.
In embodiments, checkpoint inhibitors include, without limitation, ipilimumab, pembrolizumab, nivolumab, pidilizumab, and others.
Accordingly, In embodiments, a tri-functional immunotherapy is provided that includes the use of a fused biological cell encoding a gp96-lg fusion protein and a T cell costimulatory fusion protein (e.g., without limitation, OX40L), and a checkpoint inhibitor such as, without limitation, a PD1 inhibitor.
In embodiments, the fused biological cell is made by fusing together at least two biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, and the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein. Thus, In embodiments of the present disclosure, a fused biological cell includes both a nucleotide sequence encoding a secretable vaccine protein and a nucleotide sequence encoding a T cell costimulatory fusion protein. The fused biological cell can further include a nucleotide sequence encoding the one or more disease antigens, as a result of a fusion with a third biological cell. In embodiments, the fused biological cell is made by fusing more than three biological cells together. For example, In embodiments, the fused biological cell is made by fusing four, five, six, or more than six biological cells.
In embodiments, the fused biological cell is made by fusing together at least three biological cells - the first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein, the second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein, and a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens. Accordingly, In embodiments, the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens; wherein contacting the first biological cell and the second biological cell with the fusion agent further comprises contacting the third biological cell with the fusion agent, to result in the fused biological cell being created such that the fused biological cell comprises a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
In embodiments, the fused biological cell (created by fusing the first and second biological cells) is further fused with a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens, to thereby result in the fused biological cell (which can also be referred to as "a second fused biological cell”) that can express a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens. Accordingly, In embodiments, the method of making the biological cell comprises obtaining a third biological cell comprising a nucleotide sequence encoding one or more disease antigens, and contacting the third biological cell with the fusion agent, to result in a second fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
In embodiments of the present disclosure, the fused cell increases tumor-specific immune response by simulating innate immunoactivation mechanisms as more types of tumor antigens are introduced into the fused cell. In addition to lymph node homing ability, the fused cell can display antigen presenting capacity to activate T cells due to the presence of antigen peptide MHC class I and class II molecules and costimulatory molecules. The fused cell overexpress cancer tumor antigens in sync with T cell costimulatory molecules. Furthermore, like tumor cells, tumor antigens expressed by the fused cell be recognized by dendritic cells (DCs), triggering DC maturation followed by the T cell activation.
In embodiments, one or more of the first and second biological cells, or a fused biological cell secrete a variety of antigens. Illustrative, but non-limiting, antigens that can be secreted are: MART-1/Melan-A, gp100, Dipeptidyl peptidase IV (DPPIV), adenosine deaminase-binding protein (ADAbp), cyclophilin b, Colorectal associated antigen (CRC)-0017- 1A/GA733, Carcinoembryonic Antigen (CEA) and its immunogenic epitopes CAP-1 and CAP-2, etv6, aml1 , Prostate Specific Antigen (PSA) and its immunogenic epitopes PSA-1 , PSA-2, and PSA-3, prostate-specific membrane antigen (PSMA), T-cell receptor/CD3-zeta chain, MAGE-family of tumor antigens (e.g., MAGE-A1 , MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A5, MAGE-A6, MAGE-A7, MAGE-A8, MAGE-A9, MAGE-A10, MAGE-A11 , MAGE-A12, MAGE-Xp2 (MAGE-B2), MAGE-Xp3 (MAGE-B3), MAGE-Xp4 (MAGE-B4), MAGE-C1 , MAGE-C2, MAGE-C3, MAGE-C4, MAGE- C5), GAGE-family of tumor antigens (e.g., GAGE-1 , GAGE-2, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7, GAGE- 8, GAGE-9), BAGE, RAGE, LAGE-1 , NAG, GnT-V, MUM-1 , CDK4, tyrosinase, p53, MUC family, HER2/neu, p21 ras, RCAS1 , o-fetoprotein, E-cadherin, o-catenin, p-catenin and y-catenin, p120ctn, gp100 Pmell 17, PRAME, NY-ESO-1 , cdc27, adenomatous polyposis coli protein (APC), fodrin, Connexin 37, Ig-idiotype, p15, gp75, GM2 and GD2 gangliosides, viral products such as human papilloma virus proteins, Smad family of tumor antigens, Imp-1 , NA, EBV- encoded nuclear antigen (EBNA)-1 , brain glycogen phosphorylase, SSX-1 , SSX-2 (HCM-MEL-40), SSX-1 , SSX-4, SSX-5, SCP-1 CT-7, c-erbB-2, CD19, CD20, CD22, CD30, CD33, CD37, CD56, CD70, CD74, CD138, AGS16, MUC1 , GPNMB, Ep-CAM, PD-L1 , PD-L2, PMSA, bladder cancer antigens such as ACTL8, ADAM22, ADAM23, ATAD2, ATAD2B, BIRC5, CASC5, CEP290, CEP55, CTAGE5, DCAF12, DDX5, FAM133A, IL13RA2, IMP3, KIAA0100, MAGEA11, MAGEA3, MAGEA6, MPHOSPH10, 0DF2, 0DF2L, 0IP5, PBK, RQCD1, SPAG1 , SPAG4, SPAG9, TMEFF1 , TTK, and prostate cancer antigens such as PRAME, BIRC5, CEP55, ATAD2, ODF2, KIAA0100, SPAG9, GPATCH2, ATAD2B, CEP290, SPAG1, ODF2L, CTAGE5, DDX5, DCAF12, IMP3. In embodiments, the antigens are human endogenous retroviral antigens. Illustrative antigens can also include antigens from human endogenous retroviruses which include, but are not limited to, epitopes derived from at least a portion of Gag, at least a portion of Tat, at least a portion of Rev, a least a portion of Nef, and at least a portion of gp160.
Various fused cells can be made in accordance with embodiments of the present disclosure. In embodiments, one, two, or three biological cells are provided that include the nucleotide sequences in accordance with the present disclosure in various ways. For example, In embodiments, a nucleotide sequence encoding a secretable vaccine protein and a nucleotide sequence encoding a T cell costimulatory fusion protein are present in one biological cell, whereas a nucleotide sequence encoding one or more disease antigens is present on another biological cell.
As another variation, In embodiments, a nucleotide sequence encoding a secretable vaccine protein and a nucleotide sequence encoding one or more disease antigens are present in one biological cell, whereas a nucleotide sequence encoding a T cell costimulatory fusion is present on another biological cell. In an embodiment, a nucleotide sequence encoding a T cell costimulatory fusion and a nucleotide sequence encoding one or more disease antigens are present in one biological cell, whereas a nucleotide sequence encoding a secretable vaccine protein is present on another biological cell. The biological cells are contacted with a fusion agent to result in a fused cell in accordance with embodiments of the present disclosure.
In embodiments, a fused biological cell can be fused with another biological cell. Also, In embodiments, a fused biological cell can be fused with another fused biological cell.
In embodiments, one or more of the biological cells used to make a fused cell are immortalized. In embodiments, the fused biological cell is immortalized.
In embodiments, one or more of biological cells used to make a fused cell are derived from a human tumor cell line. In embodiments, a human tumor cell line is a primary tumor cell line.
In embodiments, one or more of biological cells used to make a fused cell are Expi293F human cells that are derived from the 293 cell line. In embodiments, a first biological cell comprising a nucleotide sequence encoding a vaccine protein (such as a secretable vaccine protein) comprises human kidney cells, e.g., Expi293 cells derived from the 293 cell line. In embodiments, human tumor cells provide antigenic peptides that become non-covalently associated with the expressed gp96-lg fusion proteins. Cell lines derived from a preneoplastic lesion, cancer tissue, or cancer cells also can be used, provided that the cells of the cell line have at least one or more antigenic determinant in common with the antigens on the target cancer cells. Cancer tissues, cancer cells, cells infected with a cancer-causing agent, other preneoplastic cells, and cell lines of human origin can be used. Cancer cells excised from the patient to whom the compositions in accordance with the present disclosure are ultimately to be administered can be particularly useful, although allogeneic cells can also be used.
In embodiments, one or more of the first, second, and third biological cells is fused with a human tumor cell. In embodiments, one or more of the first, second, and third biological cells is a human tumor cell. In embodiments, the human tumor cell is a cell from an established non-small cell lung cancer (NSCLC), bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
In embodiments, the human tumor cell is a cell from an established NSCLC cell line. In embodiments, the NSCLC cell line is a human lung adenocarcinoma cell line (e.g., AD100). In embodiments, the human tumor cell is a cell from a squamous cell carcinoma or large cell lung cancer cell line.
In embodiments, the third biological cell is a trophoblast. In embodiments, the third biological cell is fused with a trophoblast. In embodiments, one or both of the first and second biological cells is a trophoblast or is fused with a trophoblast.
The presence in several cancer cells of syncytin, a trophoblastic fusogen, led to a conclusion that a cancer cell fusion mechanism is similar to the one used by the trophoblasts. See Bastida-Ruiz et al. (2016) Int. J. Mol. Sci.Vf ), 638.
In embodiments, the fused biological cell is immortalized. In embodiments, at least one of the first, second, and third biological cells is immortalized.
In embodiments, two or more biological cells that are fused are wild type, transfected, stable, transduced, or any combinations thereof.
In embodiments, the nucleotide sequence is a mammalian expression vector. The nucleotide sequence from any of the first, second, and third biological cells can be a mammalian expression vector.
In embodiments, the expression vector comprises DNA. In embodiments, the expression vector comprises RNA. In embodiments, the expression vector is incorporated into a virus or virus-like particle.
In embodiments, the expression vector comprises a pCEP4-EGFP plasmid DNA.
Both prokaryotic and eukaryotic vectors can be used for expression of a secretable vaccine protein (e.g., gp96-lg) and T cell costimulatory fusion proteins in biological cells provided herein. Prokaryotic vectors include constructs based on E. coli sequences (see, e.g., Makrides, Microbiol Rev 1996, 60:512-538). Non-limiting examples of regulatory regions that can be used for expression in E. coli include lac, trp, Ipp, phoA, recA, tac, T3, T7 and APL. Non-limiting examples of prokaryotic expression vectors may include the Agt vector series such as Agt11 (Huynh et al., in "DNA Cloning Techniques, Vol. I: A Practical Approach,” 1984, (D. Glover, ed.), pp. 49-78, IRL Press, Oxford), and the pET vector series (Studier et al., Methods Enzymol 1990, 185:60-89). Prokaryotic host-vector systems cannot perform much of the post-translational processing of mammalian cells, however. Thus, eukaryotic host-vector systems may be particularly useful.
In embodiments, various fusion (or fusogenic) agents can be used to allow two or more biological cells to fuse. In embodiments, the fusion agent comprises a protein from Paramyxoviridae Genus paramyxovirus. The protein can be, for example, hemagglutinating virus of Japan (HVJ) envelope (HVJ-E). The HVJ is also known as Sendai virus. See, e.g., Okada (1993) Methods Enzymol. 221 : 18—41 . The HVJ-E is a nonproliferative and noninfectious vesicle purified after inactivation of Sendai virus genomic RNA. See, e.g., Kaneda et al. (2002) Molecular Therapy 6, 219-226; see also Lund et al. (2010) Pharm Res. 27(3):400-20, the entire contents of which are hereby incorporated by reference.
In embodiments, the fusion agent comprises a membrane fusion protein from an envelope of a virus selected from Orthomyxoviridae, Herpesviridae, Hepadnaviridae, and Flaviviridae.
In embodiments, the fusion agent comprises a virosome derived from the influenza virus (Orthomyxovirus) or the Sendai virus. A virosome is defined as a vector derived from an inactive enveloped virus or a reconstituted envelope that contains viral envelope proteins or viral envelope particles. Saga & Kaneda (2013). BioMed research international, 2013, 764706.
In embodiments, the fusion agent comprises a Sendai virosome containing pCEP4-EGFP plasmid.
In embodiments, the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof. See Koido et al. (2009) Immunotherapy 1 :49-62. doi: 10.2217/1750743X.1.1.49; Homma etal. (2003) J. Gastroenterol. 38:989-994. doi: 10.1007/S00535-002-1183-3.
In embodiments, the PEG moiety comprises PEG-1500. In embodiments, the fusion agent comprises PEG-300, PEG- 400, PEG-550, PEG-550, or PEG-1000, and other PEG molecules having an average molecular weight of less than 2000. In embodiments, PEG molecules have an average molecular weight of greater than 2000.
In embodiments, cell fusion is performed using electrofusion. See, e.g., Skelley et al. (2009) Nat. Methods 6: 147-152; Kjaergaard et al. (2003) Cell. Immunol. 225:65-74; Trevor et al. (2004) Cancer Immunol. Immunother. 53:705-714; Scott-Taylor et al. (2000) Biochim. Biophys. Acta. 2000; 1500:265-279.
In embodiments, one or both the first biological cell and the second biological cell comprise at least one tumor antigen. In embodiments, the tumor antigen is a cancer testis (CT) antigen. In embodiments, the fused biological cell expresses one or more CT antigens. In embodiments, one or more of the first, second, and third biological cells expresses one or more CT antigens. In embodiments, the fused biological cell is enriched for genes encoding one or more CT antigens. In embodiments, one or more of the first, second, and third biological cells expresses one or more CT antigens.
As mentioned above, In embodiments, the fused biological cell further comprises a nucleotide sequence encoding one or more disease antigens. In embodiments, the one or more disease antigens comprise one of more infectious disease antigens. This approach can be used in developing compositions (e.g., vaccines) for treatment of disease vaccines, by fusing cancer cells with other biological cells expressing either mini-genes or bacterial or viral proteins capable of driving B and T cell responses. In embodiments, a "mini-gene” is a part of a disease antigen, such as an immunogenic portion of the disease antigen which is sufficient to induce immune response specific to the disease antigen.
In embodiments, the fused cell in accordance with the present disclosure leverages gp96 to effectively present one or more disease antigens and activate the immune system. The gp96-based system utilizes natural immune process to induce long-lasting memory responses and can effectively present multiple antigens and activate the immune system. The described methods and compositions aim to trigger mucosal immunity by activating both B and T cell responses at the point of pathogen entry.
In embodiments, the one or more infectious disease antigens comprise an antigenic fragment of a betacoronavirus protein or an alphacoronavirus protein, optionally wherein the betacoronavirus protein is selected from a SARS-CoV- 2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43 protein, or the alphacoronavirus protein is selected from a HCoV-NL63 and HCoV-229E protein.
In embodiments, the betacoronavirus protein is a SARS-CoV-2 protein, also called 2019-nCoV protein.
In embodiments, the SARS-CoV-2 protein comprises an amino acid encoded by the nucleic acid of SEQ ID NO: 38, or an antigenic fragment thereof. In embodiments, the SARS-CoV-2 protein comprises the amino acid that encompasses an amino acid of sequence of SEQ ID NO: 39, an amino acid of sequence of SEQ ID NO: 40, an amino acid of sequence of SEQ ID NO: 41 , an amino acid of sequence of SEQ ID NO: 42, an amino acid of sequence of SEQ ID NO: 43, an amino acid of sequence of SEQ ID NO: 44, an amino acid of sequence of SEQ ID NO: 45, and an amino acid of sequence of SEQ ID NO: 46, or an antigenic fragment thereof.
In embodiments, the SARS-CoV-2 is selected from a spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N.
In embodiments, the coronavirus protein is a SARS-CoV-2 protein, or an antigenic fragment thereof, selected from the spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N. In embodiments, the spike surface glycoprotein comprises the amino acid sequence of SEQ ID NO: 39, the envelope protein E comprises the amino acid sequence of SEQ ID NO: 41 , the membrane glycoprotein precursor M comprises the amino acid sequence of SEQ ID NO: 42, and the nucleocapsid phosphoprotein N comprises the amino acid sequence of SEQ ID NO: 46, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity with any of the foregoing, or an antigenic fragment of any of the foregoing.
In embodiments, the spike protein comprises the following amino acid sequence:
MFVFLVLLPLVSSQCVNLTTRTQLPPAYTNSFTRGVYYPDKVFRSSVLHSTQDLFLPFFSNVTWFHAIHVSGTNGTKR FDNPVLPFNDGVYFASTEKSNIIRGWIFGTTLDSKTQSLLIVNNATNWIKVCEFQFCNDPFLGVYYHKNNKSWMESEF RVYSSANNCTFEYVSQPFLMDLEGKQGNFKNLREFVFKNIDGYFKIYSKHTPINLVRDLPQGFSALEPLVDLPIGINITR FQTLLALHRSYLTPGDSSSGWTAGAAAYYVGYLQPRTFLLKYNENGTITDAVDCALDPLSETKCTLKSFTVEKGIYQT
SNFRVQPTESIVRFPNITNLCPFGEVFNATRFASVYAWNRKRISNCVADYSVLYNSASFSTFKCYGVSPTKLNDLCFT NVYADSFVIRGDEVRQIAPGQTGKIADYNYKLPDDFTGCVIAWNSNNLDSKVGGNYNYLYRLFRKSNLKPFERDISTEI
YQAGSTPCNGVEGFNCYFPLQSYGFQPTNGVGYQPYRVWLSFELLHAPATVCGPKKSTNLVKNKCVNFNFNGLTG TGVLTESNKKFLPFQQFGRDIADTTDAVRDPQTLEILDITPCSFGGVSVITPGTNTSNQVAVLYQDVNCTEVPVAIHAD QLTPTWRVYSTGSNVFQTRAGCLIGAEHVNNSYECDIPIGAGICASYQTQTNSPRRARSVASQSIIAYTMSLGAENSV AYSNNSIAIPTNFTISVTTEILPVSMTKTSVDCTMYICGDSTECSNLLLQYGSFCTQLNRALTGIAVEQDKNTQEVFAQV
KQIYKTPPIKDFGGFNFSQILPDPSKPSKRSFIEDLLFNKVTLADAGFIKQYGDCLGDIAARDLICAQKFNGLTVLPPLLT DEMIAQYTSALLAGTITSGWTFGAGAALQIPFAMQMAYRFNGIGVTQNVLYENQKLIANQFNSAIGKIQDSLSSTASAL GKLQDWNQNAQALNTLVKQLSSNFGAISSVLNDILSRLDKVEAEVQIDRLITGRLQSLQTYVTQQLIRAAEIRASANLA ATKMSECVLGQSKRVDFCGKGYHLMSFPQSAPHGWFLHVTYVPAQEKNFTTAPAICHDGKAHFPREGVFVSNGTH
WFVTQRNFYEPQIITTDNTFVSGNCDWIGIVNNTVYDPLQPELDSFKEELDKYFKNHTSPDVDLGDISGINASWNIQK EIDRLNEVAKNLNESLIDLQELGKYEQYIKWPWYIWLGFIAGLIAIVMVTIMLCCMTSCCSCLKGCCSCGSCCKFDEDD SEPVLKGVKLHYT (SEQ ID NO: 39).
In embodiments, the envelope protein comprises the following amino acid sequence:
MYSFVSEETGTLIVNSVLLFLAFWFLLVTLAILTALRLCAYCCNIVNVSLVKPSFYVYSRVKNLNSSRVPDLLV (SEQ ID NO: 41).
In embodiments, the membrane protein comprises the following amino acid sequence: MADSNGTITVEELKKLLEQWNLVIGFLFLTWICLLQFAYANRNRFLYIIKLIFLWLLWPVTLACFVLAAVYRINWITGGIAI AMACLVGLMWLSYFIASFRLFARTRSMWSFNPETNILLNVPLHGTILTRPLLESELVIGAVILRGHLRIAGHHLGRCDIK DLPKEITVATSRTLSYYKLGASQRVAGDSGFAAYSRYRIGNYKLNTDHSSSSDNIALLVQ (SEQ ID NO: 42).
In embodiments, the nucleocapsid phosphoprotein comprises the following amino acid sequence:
MSDNGPQNQRNAPRITFGGPSDSTGSNQNGERSGARSKQRRPQGLPNNTASWFTALTQHGKEDLKFPRGQGVPIN
TNSSPDDQIGYYRRATRRIRGGDGKMKDLSPRWYFYYLGTGPEAGLPYGANKDGIIWVATEGALNTPKDHIGTRNPA
NNAAIVLQLPQGTTLPKGFYAEGSRGGSQASSRSSSRSRNSSRNSTPGSSRGTSPARMAGNGGDAALALLLLDRLN QLESKMSGKGQQQQGQTVTKKSAAEASKKPRQKRTATKAYNVTQAFGRRGPEQTQGNFGDQELIRQGTDYKHWP QIAQFAPSASAFFGMSRIGMEVTPSGTWLTYTGAIKLDDKDPNFKDQVILLNKHIDAYKTFPPTEPKKDKKKKADETQA LPQRQKKQQTVTLLPAADLDDFSKQLQQSMSSADSTQA (SEQ ID NO: 46).
In embodiments, the 2019-nCoV protein may comprise an amino acid sequence that has at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71%, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81 %, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 92%, or at least about 93%, or at least about 94%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% sequence identity with any known wild type amino acid sequence of the SARS- CoV-2 protein or a SARS-CoV-2 amino acid sequence disclosed herein (e.g. about 60%, or about 61 %, or about 62%, or about 63%, or about 64%, or about 65%, or about 66%, or about 67%, or about 68%, or about 69%, or about 70%, or about 71%, or about 72%, or about 73%, or about 74%, or about 75%, or about 76%, or about 77%, or about 78%, or about 79%, or about 80%, or about 81%, or about 82%, or about 83%, or about 84%, or about 85%, or about 86%, or about 87%, or about 88%, or about 89%, or about 90%, or about 91 %, or about 92%, or about 93%, or about 94%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99% sequence identity), e.g. relative to an amino acid encoded by SEQ ID NO: 38, or an antigenic portion thereof.
In embodiments, the SARS-CoV-2 protein may comprise an amino acid sequence that has at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71 %, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81%, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 92%, or at least about 93%, or at least about 94%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% sequence identity with any known wild type amino acid sequence of the SARS-CoV-2 protein or a SARS-CoV-2 amino acid sequence disclosed herein (e.g. about 60%, or about 61 %, or about 62%, or about 63%, or about 64%, or about 65%, or about 66%, or about 67%, or about 68%, or about 69%, or about
70%, or about 71 %, or about 72%, or about 73%, or about 74%, or about 75%, or about 76%, or about 77%, or about
78%, or about 79%, or about 80%, or about 81 %, or about 82%, or about 83%, or about 84%, or about 85%, or about
86%, or about 87%, or about 88%, or about 89%, or about 90%, or about 91 %, or about 92%, or about 93%, or about 94%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99% sequence identity), e.g. relative to any one of SEQ ID NOs: 39, 41 , 42, 46, or an antigenic fragment thereof.
In embodiments, a SARS-CoV-2 protein portion can encode an amino acid sequence that differs from any known wild type amino acid sequence of the SARS-CoV-2 protein or a SARS-CoV-2 amino acid sequence disclosed herein having a mutation, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms.
Cell Fusions
In embodiments, a fused biological cell can be made in various ways.
In embodiments, cell fusion between two biological cells can be performed using a method that is similar to conventional method for fusing immune cells and myeloma cells, for example, as described in Milstein et al. Methods Enzymol., 73, 3-46 (1981)).
In embodiments, two or more biological cells that are fused are wild type, transfected, stable, transduced cells, or any combinations thereof.
In embodiments, a fused biological cell is prepared in a conventional nutrient medium in the presence of a fusogenic or fusion agent. The fusion promoter can be polyethylene glycol (PEG), Hemagglutinating virus (HVJ), or another type of a fusion promoter. In embodiments, an auxiliary agent, such as, e.g, dimethylsulfoxide, can be used to enhance fusion efficiency.
In embodiments, a fusion agent comprises a protein suitable to cause a protein-mediated membrane fusion. For example, a protein can be a membrane fusion protein of an inactivated virus, such as an envelope protein of an inactivated virus. Viruses having an envelope comprising a membrane fusion protein include Paramyxoviridae, Orthomyxoviridae, Herpesviridae, Hepadnaviridae and Flaviviridae viruses. These viruses utilize their membrane fusion activity to infect host cells through fusion with the plasma membranes of the host cells. An "inactivated” virus can be defined as a virus with inactivated genes such that the virus can no longer replicate its genes and has lost its proliferation potency and infectivity. A virus can be inactivated using UV irradiation, irradiation of radioactive rays, or treatment with an alkylating agent, or a combination thereof.
A virus envelope generally is formed of small spike-like structures made of spike proteins encoded by a virus gene, and a lipid bilayer derived from the host and that does not have proliferation ability. Spike proteins constituting the small spikes comprise a membrane fusion protein which functions to confer a membrane fusion activity. In embodiments, an inactivated virus envelope having membrane fusion activity can be used for making a fused biological cell.
In embodiments, the fusion agent comprises a membrane fusion protein from an envelope of an inactivated virus selected from Orthomyxoviridae, Herpesviridae, Hepadnaviridae, and Flaviviridae. In embodiments, a fusion agent comprises a protein from Paramyxoviridae Genus paramyxovirus. The protein can be, for example, hemagglutinating virus of Japan (HVJ) envelope (HVJ-E). The HVJ, also known as Sendai virus, an enveloped virus with a nonsegmented negative-strand RNA genome. See, e.g., Okada (1993) Methods Enzymol. 221 :18-41. The HVJ-E is a nonproliferative and noninfectious vesicle purified after inactivation of Sendai virus genomic RNA. See, e.g., Kaneda ef a/. (2002) Molecular Therapy 6, 219-226; see a/so Lund etal. (2010) Pharm Res. 27(3):400- 20, the entire contents of which are hereby incorporated by reference.
In embodiments, the fusion agent comprises inactivated Sendai virus envelope protein.
In embodiments, the fusion agent is inactivated Sendai virus HVJ-E.
In embodiments, the fusion agent comprises a Sendai virosome comprising pCEP4-EGFP plasmid.
Sendai virus can be cultivated, for example, in chicken fertilized eggs or by persistent infection in mammalian cell or tissue cultures, which can include a hydrolase such as trypsin. An inactivated virus envelope having membrane fusion activity can be prepared using ultracentrifugation, isolation by column chromatography, reconstitution and the like. Isolation can be carried out by ion chromatography, as described, for example, in Example 1 (1) and Examples 7 (1) to (5) of U.S. Patent Application Publication No. 20040253272, which is incorporated herein by reference in its entirety.
In embodiments, a freeze-dried composition of an inactivated virus envelope having membrane fusion activity can be prepared, e.g., as described in U.S. Patent No. 8,043,610, which is incorporated herein by reference in its entirety. As described in U.S. Patent No. 8,043,610, the freeze-dried composition can be prepared, for example, by adding a protein hydrolysate, an amino acid or a polysaccharide at a predetermined concentration, and optionally adding a known pH adjusting agent, a known isotonicity agent, a known stabilizer or a known preservative, to the virus envelope having membrane fusion activity above. The protein hydrolysate, the amino acid or the polysaccharide may be dissolved in aqueous solvent(s) {e.g., distilled water, a buffer solution, etc.) respectively before added at predetermined concentrations to the virus envelope having membrane fusion activity. The resulting mixture can be freeze-dried by ordinary methods. For the freeze-drying, a known method such as tray freeze-drying, spray freeze-drying or vial freeze- drying under ordinarily used conditions may be used.
In embodiments, an antibiotic-containing media is used for cell fusion of biological cells. In embodiments, the media comprises one or more of G418 (Geneticin), Puromycin, & Hygromycin.
In embodiments, the antibiotic-containing media comprises from about 100 ug/mL (micrograms per milliliter) to about 800 ug/mL of one or more of an antibiotic. In embodiments, the media comprises from about 100 ug/mL to about 1000 ug/mL of one or more antibiotic. In embodiments, the media comprises about 100 ug/mL, or about 200 ug/mL, or about 300 ug/mL, or about 400 ug/mL, or about 500 ug/mL, or about 600 ug/mL, or about 700 ug/mL, or about 800 ug/mL, or about 900 ug/mL, or about 1000 ug/mL of an antibiotic.
In embodiments, the antibiotic-containing media comprises about 500 ug/mL G418 and about 200 ug/mL Hygromycin. In embodiments, a fused biological cell is resuspended in a buffering agent such as, without limitation, PBS (phosphate- buffered saline) or tris-buffered saline (TBS). In embodiments, a buffering agent comprises, without limitation, one or more of PBS, triethanolamine, Tris, bicine, TAPS, tricine, HEPES, TES, MOPS, PIPES, cacodylate, MES, acetate, citrate, succinate, histidine or other pharmaceutically acceptable buffers.
The buffering agent In embodiments includes Bovine serum albumin (BSA). In embodiments, the buffering agent comprises PBS including from about 1 % to about 10% BSA. In embodiments, the buffering agent comprises PBS including about 1 % BSA. In embodiments, the buffering agent comprises TBS including from about 1 % to about 10% BSA. In embodiments, the buffering agent comprises TBS including about 1 % BSA.
In embodiments, the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof. See Koido et al. (2009) Immunotherapy 1 :49-62. doi: 10.2217/1750743X.1.1.49; Homma etal. (2003) J. Gastroenterol. 38:989-994. doi: 10.1007/S00535-002-1183-3.
In embodiments, the PEG moiety comprises PEG-1500. In embodiments, the fusion agent comprises PEG-300, PEG- 400, PEG-550, PEG-550, or PEG-1000, and other PEG molecules having an average molecular weight of less than 2000. In embodiments, PEG molecules have an average molecular weight of greater than 2000.
In embodiments, cell fusion is performed using electrofusion. See, e.g., Skelley et al. (2009) Nat. Methods 6: 147-152; Kjaergaard et al. (2003) Cell. Immunol. 225:65-74; Trevor et al. (2004) Cancer Immunol. Immunother. 53:705-714; Scott-Taylor et al. (2000) Biochim. Biophys. Acta. 2000; 1500:265-279.
In embodiments, a non-limiting example of cell fusion is as follows. AD-100 (the 293 Cell Line) transduced (OX40L, 10E5 cells) cells and transfected human kidney cells (Expi293 cells expressing gp96, 10E5 cells) are seeded in 12- well plates and grown for 24 hours at 37°C. In some cases, AD-100 transduced (OX40L, 10E5 cells) cells or transfected human kidney cells (Expi293 cells expressing gp96, 10E5 cells) are seeded in 12-well plates and grown for 24 hours at 37°C. The gp96 can be native gp96 or a gp96-lg fusion protein. The cells are washed and replaced with antibiotic- free media and then incubated at 37°C in the presence of Sendai virosomes containing pCEP4-EGFP plasmid DNA. After de novo protein expression and a 24-hour time period of transfection, serum-free wells are replaced by antibioticcontaining media (500 ug/mL G418 and 200 ug/mL Hygromycin), and the cells are then incubated further for 48 hours at 37°C and 5% CO2. After 48 hours of incubation, the fused cells are harvested and then resuspended in 500 uL of PBS containing 1 % BSA and evaluated by immunofluorescence.
In embodiments, the disclosure provides sequential fusion combination {e.g., a single cell is transduced with gp96-lg, and a single cell is transduced with T cell costimulatory fusion protein, e.g. OX40L-lg, then the cells are fused together).
In embodiments, the provided methods result in considerably greater expression and/or secretion (e.g. about or at least about 2-fold, or about or at least about 3-fold, or about or at least about 4-fold, or about or at least about 5-fold, or about or at least about 6-fold, or about or at least about 7-fold, or about or at least about 8-fold, or about or at least about 9-fold, or about or at least about 10-fold, or about or at least about 15-fold, or about or at least about 20-fold, or about or at least about 50% greater, or about or at least about 75% greater, or about or at least about 100% greater, or about or at least about 200% greater, or about or at least about 300% greater, or about or at least about 400% greater, or about or at least about 500% greater) of secretable vaccine protein (e.g., without limitation, gp96-l g) and/or T cell costimulatory fusion protein (e.g., without limitation OX40L-lg), as compared to an unfused cell that has been caused to express (e.g transduced with nucleic acids encoding) the secretable vaccine protein (e.g., without limitation, gp96-lg) and/or T cell costimulatory fusion protein (e.g., without limitation OX40L-lg).
Lentiviruses and Lentiviral Packaging Systems
In one aspect, the disclosure relates to a method for generating a cellular therapy, comprising: (a) obtaining a lentivirus, the lentivirus comprising: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject; and (b) introducing into a biological cell the lentivirus of step (a).
In one another aspect, the disclosure relates to a method for generating a cellular therapy, comprising: (a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and (i) introducing the nucleic acid into a first biological cell; (b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject, and (i) introducing the nucleic acid into a second biological cell, and (c) merging together the first biological cell and the second biological cell.
Lentiviruses or lentiviral vectors are powerful tools to ensure stable and long-term gene expression by transfer of genes into dividing and non-dividing cells. In embodiments, lentiviral expression vectors are based on human immunodeficiency virus- 1 (HIV-1). Therapeutic lentiviruses have been modified from the original HIV- 1 genome to avoid replication in host cells. To increase the safety of lentivirus, the components necessary for virus production are split across multiple plasmids. In embodiments, the lentivirus is produced using either a second generation lentiviral packaging system, in which the lentivirus components are split across three plasmids, or a third generation lentiviral packaging system, in which the lentivirus components are split across four plasmids. In embodiments, the lentivirus is produced using a third-generation lentiviral packaging system.
In embodiments, the lentiviral packaging system comprises a transfer plasmid, a packaging plasmid, and an envelope plasmid. In embodiments, the packaging plasmid comprises two plasmids. In embodiments, the packaging plasmid encodes a Gag and Pol gene. In embodiments, the packaging plasmid encodes a Rev gene. In embodiments, the envelope plasmid comprises a VSV-G envelope protein. In embodiments, the transfer plasmid comprises a transgene, or a nucleic acid of interest. In embodiments, the transfer plasmid comprises: (i) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, (ii) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject. In embodiments, the sequence for the transgene or the nucleic acid of interest is flanked by long terminal repeat (LTR) sequences, which facilitate integration of the transfer plasmid sequences into the host genome. Typically, it is the sequences between and including the LTRs that are integrated into the host genome upon viral transduction. For safety reasons, at least two different modifications are made to lentiviruses. In embodiments, the lentivirus is replication incompetent. In embodiments, the lentivirus comprises a deletion in a long terminal repeat (LTR) sequence, rendering it self-inactivating (SIN) after integration. In embodiments, the transfer plasmid is optimized. In embodiments, the transfer plasmid utilizes a hybrid LTR promoter. In embodiments, Tat is eliminated from the third-generation lentiviral packaging system through the addition of a chimeric 5' LTR fused to a heterologous promoter on the transfer plasmid. In embodiments, the expression of the transgene from this promoter is no longer dependent on Tat transactivation. In embodiments, the transfer plasmid comprises additional or specialized promoters, such as a U6 promoter. In embodiments, the transfer plasmid comprises Tet- or Cre-based regulation element. In embodiments, the transfer plasmid comprises fluorescent fusions or reporters.
Lentiviruses are produced using established processes that are well known in the art. In embodiments, the lentivirus is produced by contacting a cell with the third-generation lentiviral packaging system and isolating the lentivirus from the cell. In embodiments, the cell contacted is a host cell, such as an A293T cell. Briefly, In embodiments, the one transfer plasmid, one or two packaging plasmids, and one envelope plasmid are transfected into a host cell. In embodiments, after media change and a brief incubation period, supernatant containing the lentivirus is removed and stored or centrifuged to concentrate the lentivirus. In embodiments, crude or concentrated lentivirus can then be introduced into the cells of interest.
Lentiviruses can be used to make stable cell lines in the same manner as standard retroviruses. For instance, many lentiviral genomes have selectable markers, such as the puromycin resistance gene, conferring antibiotic resistance to infected host cells. When selection antibiotics are added to the growth medium of the host cells, they kill off any cells that have not incorporated the lentiviral genome and those clonal cells that survive can be expanded to create stable cell lines, which have incorporated the lentiviral genome and harbor the genetic information encoded by that genome.
In embodiments, the lentivirus and/or transfer plasmid comprises one or more selection markers, optionally selected from puromycin, neomycin, zeocin, and hygromycin. In exemplary aspects, the selection marker is a gene product which confers resistance to an antibiotic, including but not limited to ampicillin, kanamycin, neomycin/G418, tetracycline, geneticin, triclosan, puromycin, zeocin, and hygromycin. In exemplary aspects, the selection marker is one or more of kanamycin resistance genes, puromycin resistance genes, zeocin resistance genes, neomycin/G418 resistance genes, hygromycin resistance genes, histidinol resistance genes, tetracycline resistance genes, geneticin resistance genes, triclosan resistance genes, R-fluroorotic acid resistance genes, 5-fluorouracil resistance genes and ampicillin resistance genes. Combination of any of the selection markers described herein is contemplated. In exemplary aspects, when the lentiviral packaging system comprises more than one plasmid, each plasmid comprises a selection marker. In exemplary aspects, each plasmid has the same selection marker. Alternatively, each plasmid within the system comprises a different selection marker. In embodiments, lentiviral transfer plasmids do not have selectable markers conferring resistance to an antibiotic, but do encode another marker, such as GFP. In embodiments, FACS is used to sort cells expressing GFP, wherein these cells are later expanded into a cell line.
In embodiments, high titer lentiviruses can be introduced into cancer cell lines, such as AD-100 or HEK293, to express dominant B and T cell epitopes. In embodiments, techniques suitable for the transfer of nucleic acids into mammalian cells in vitro are used, including the use of liposomes, electroporation, microinjection, cell fusion, polymer-based systems, DEAE-dextran, viral transduction, the calcium phosphate precipitation method, etc. In embodiments, for in vivo gene transfer, a number of techniques and reagents are used, including electroporation, liposomes; natural polymer-based delivery vehicles, such as chitosan and gelatin. In embodiments, viral vectors are used for in vivo transduction. In embodiments, multiplicity of infection (MOI) is optimized to for use in: (I) manufacturing, when many billions of cells are needed for infectious disease vaccination, or (II) generating stable cell lines containing three or more proteins.
In embodiments, the high titer of the lentiviruses is about 0.5x108 transducing units (TU)/ ml. In embodiments, the high titer of the lentiviruses is about 1x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 1.5x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 2x108TU/ml. In embodiments, the high titer of the lentiviruses is about 2.5x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 3x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 3.5x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 4x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 4.5x108 TU/ml. In embodiments, the high titer of the lentiviruses is about 5x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 0.5x108 TU/ml to about 1x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 1x108 TU/ml to about 1.5x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 1.5x108 TU/ml to about 2x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 2x108 TU/ml to about 2.5x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 2.5x108 TU/ml to about 3x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 3x108 TU/ml to about 3.5x108TU/ml. In embodiments, the high titer of the lentiviruses is between about 3.5x108 TU/ml to about 4x108 TU/ml. In embodiments, the high titer of the lentiviruses is between about 4x108 TU/ml to about 4.5x108 TU/ml.
Chaperones and Fusion Proteins In embodiments, the lentivirus and/or transfer plasmid of the present disclosure comprises a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof. In embodiments, chaperone proteins chaperone antigens to the immune system for MHC-l-mediated antigen crosspresentation, leading to a robust cytotoxic CD8+ T cell response. In embodiments, the chaperone protein is selected from the group consisting of: gp96, Hsp70, BiP, and Grp78. In embodiments, the chaperone protein comprises an amino acid sequence of any one of SEQ ID NOs: 1 , 2, 3, and 4, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. The amino acid sequence (SEQ ID NO: 2) and nucleic acid sequence (SEQ ID NO: 1) of chaperone protein, gp96, are provided below:
MRALWVLGLCCVLLTFGSVRADDEVDVDGTVEEDLGKSREGSRTDDEWQRE EEAIQLDGLNASQIRELREKSEKFAFQAEVNRMMKLIINSLYKNKEIFLRELISNA SDALDKIRLISLTDENALSGNEELTVKIKCDKEKNLLHVTDTGVGMTREELVKNL GTIAKSGTSEFLNKMTEAQEDGQSTSELIGQFGVGFYSAFLVADKVIVTSKHNN DTQHIWESDSNEFSVIADPRGNTLGRGTTITLVLKEEASDYLELDTIKNLVKKYS QFINFPIYVWSSKTETVEEPMEEEEAAKEEKEESDDEAAVEEEEEEKKPKTKKV EKTVWDWELMNDIKPIWQRPSKEVEEDEYKAFYKSFSKESDDPMAYIHFTAEG EVTFKSILFVPTSAPRGLFDEYGSKKSDYIKLYVRRVFITDDFHDMMPKYLNFVK GWDSDDLPLNVSRETLQQHKLLKVIRKKLVRKTLDMIKKIADDKYNDTFWKEF GTNIKLGVIEDHSNRTRLAKLLRFQSSHHPTDITSLDQYVERMKEKQDKIYFMA GSSRKEAESSPFVERLLKKGYEVIYLTEPVDEYCIQALPEFDGKRFQNVAKEGV KFDESEKTKESREAVEKEFEPLLNWMKDKALKDKIEKAWSQRLTESPCALVAS QYGWSGNMERIMKAQAYQTGKDISTNYYASQKKTFEINPRHPLIRDMLRRIKED EDDKTVLDLAWLFETATLRSGYLLPDTKAYGDRIERMLRLSLNIDPDAKVEEEP EEEPEETAEDTTEDTEQDEDEEMDVGTDEEEETAKESTAEKDEL (SEQ ID NO: 2) atgagggccctgtgggtgctgggcctctgctgcgtcctgctgaccttcgggtcggtcagagctgacgatgaagtt gatgtggatggtacagtagaagaggatctgggtaaaagtagagaaggatcaaggacggatgatgaagtagt acagagagaggaagaagctattcagttggatggattaaatgcatcacaaataagagaacttagagagaagtc ggaaaagtttgccttccaagccgaagttaacagaatgatgaaacttatcatcaattcattgtataaaaataaaga g attttcctg ag ag aactg atttcaaatgcttctg atgctttag ataagataaggctaatatcactg actg atg aaaa tgctctttctggaaatgaggaactaacagtcaaaattaagtgtgataaggagaagaacctgctgcatgtcacag acaccggtgtaggaatgaccagagaagagttggttaaaaaccttggtaccatagccaaatctgggacaagcg agtttttaaacaaaatgactgaagcacaggaagatggccagtcaacttctgaattgattggccagtttggtgtcgg tttctattccgccttccttgtagcagataaggttattgtcacttcaaaacacaacaacgatacccagcacatctggg agtctgactccaatgaattttctgtaattgctgacccaagaggaaacactctaggacggggaacgacaattacc cttgtcttaaaagaagaagcatctgattaccttgaattggatacaattaaaaatctcgtcaaaaaatattcacagtt cataaactttcctatttatgtatggagcagcaagactgaaactgttgaggagcccatggaggaagaagaagca gccaaagaagagaaagaagaatctgatgatgaagctgcagtagaggaagaagaagaagaaaagaaacc aaagactaaaaaagttgaaaaaactgtctgggactgggaacttatgaatgatatcaaaccaatatggcagag accatcaaaagaagtagaagaagatgaatacaaagctttctacaaatcattttcaaaggaaagtgatgacccc atggcttatattcactttactgctgaaggggaagttaccttcaaatcaattttatttgtacccacatctgctccacgtgg tctgtttgacgaatatggatctaaaaagagcgattacattaagctctatgtgcgccgtgtattcatcacagacgactt ccatgatatgatgcctaaatacctcaattttgtcaagggtgtggtggactcagatgatctccccttgaatgtttcccg cgagactcttcagcaacataaactgcttaaggtgattaggaagaagcttgttcgtaaaacgctggacatgatca agaagattgctgatgataaatacaatgatactttttggaaagaatttggtaccaacatcaagcttggtgtgattgaa gaccactcgaatcgaacacgtcttgctaaacttcttaggttccagtcttctcatcatccaactgacattactagccta gaccagtatgtggaaagaatgaaggaaaaacaagacaaaatctacttcatggctgggtccagcagaaaaga ggctgaatcttctccatttgttgagcgacttctgaaaaagggctatgaagttatttacctcacagaacctgtggatga atactgtattcaggcccttcccgaatttgatgggaagaggttccagaatgttgccaaggaaggagtgaagttcga tgaaagtgagaaaactaaggagagtcgtgaagcagttgagaaagaatttgagcctctgctgaattggatgaaa gataaagcccttaaggacaagattgaaaaggctgtggtgtctcagcgcctgacagaatctccgtgtgctttggtg gccagccagtacggatggtctggcaacatggagagaatcatgaaagcacaagcgtaccaaacgggcaagg acatctctacaaattactatgcg ag tcagaag aaaacatttg aaattaatcccag acacccgctg atcag ag ac atgcttcgacgaattaaggaagatgaagatgataaaacagttttggatcttgctgtggttttgtttgaaacagcaac gcttcggtcagggtatcttttaccagacactaaagcatatggagatagaatagaaagaatgcttcgcctcagtttg aacattgaccctgatgcaaaggtggaagaagagcccgaagaagaacctgaagagacagcagaagacac aacagaagacacagagcaagacgaagatgaagaaatggatgtgggaacagatgaagaagaagaaaca gcaaaggaatctacagctgaaaaagatgaattgtaa (SEQ ID NO: 1)
The amino acid sequences of chaperone proteins, Hsp70 (SEQ ID NO: 47), BiP (SEQ ID NO: 48), and Grp78 (SEQ ID NO: 49), are provided below:
MSWGIDLGFQSCYVAVARAGGIETIANEYSDRCTPACISFGPKNRSIGAAAKS QVISNAKNTVQGFKRFHGRAFSDPFVEAEKSNLAYDIVQWPTGLTGIKVTYMEE ERNFTTEQVTAMLLSKLKETAESVLKKPWDCWSVPCFYTDAERRSVMDATQI AGLNCLRLMNETTAVALAYGIYKQDLPRLEEKPRNVVFVDMGHSAYQVSVCAF NRGKLKVLATAFDTTLGGRKFDEVLVNHFCEEFGKKYKLDIKSKIRALLRLSQEC EKLKKLMSANASDLPLSIECFMNDVDVSGTMNRGKFLEMCNDLLARVEPPLRS VLEQTKLKKEDIYAVEIVGGATRIPAVKEKISKFFGKELSTTLNADEAVTRGCALQ
CAILSPAFKVREFSITDWPYPISLRWNSPAEEGSSDCEVFSKNHAAPFSKVLTF
YRKEPFTLEAYYSSPQDLPYPDPAIAQFSVQKVTPQSDGSSSKVKVKVRVNVH
GIFSVSSASLVEVHKSEENEEPMETDQNAKEEEKMQVDQEEPHVEEQQQQTP
AENKAESEEMETSQAGSKDKKMDQPPQCQEGKSEDQYCGPANRESAIWQID
REMLNLYIENEGKMIMQDKLEKERNDAKNAVEEYVYEMRDKLSGEYEKFVSED
DRNSFTLKLEDTENWLYEDGEDQPKQVYVDKLAELKNLGQPIKIRFQESEERP NYLKN (SEQ ID NO: 47)
MKLSLVAAMLLLLSAARAEEEDKKEDVGTWGIDLGTTYSCVGVFKNGRVEIIAN
DQGNRITPSYVAFTPEGERLIGDAAKNQLTSNPENTVFDAKRLIGRTWNDPSVQ
QDIKFLPFKWEKKTKPYIQVDIGGGQTKTFAPEEISAMVLTKMKETAEAYLGKK
VTHAWTVPAYFNDAQRQATKDAGTIAGLNVMRIINEPTAAAIAYGLDKREGEK
NILVFDLGGGTFDVSLLTIDNGVFEWATNGDTHLGGEDFDQRVMEHFIKLYKK
KTGKDVRKDNRAVQKLRREVEKAKALSSQHQARIEIESFYEGEDFSETLTRAKF
EELNMDLFRSTMKPVQKVLEDSDLKKSDIDEIVLVGGSTRIPKIQQLVKEFFNGK
EPSRGINPDEAVAYGAAVQAGVLSGDQDTGDLVLLHVCPLTLGIETVGGVMTK
LIPSNTWPTKNSQIFSTASDNQPTVTIKVYEGERPLTKDNHLLGTFDLTGIPPAP
RGVPQIEVTFEIDVNGILRVTAEDKGTGNKNKITITNDQNRLTPEEIERMVNDAE
KFAEEDKKLKERIDTRNELESYAYSLKNQIGDKEKLGGKLSSEDKETMEKAVEE
KIEWLESHQDADIEDFKAKKKELEEIVQPIISKLYGSAGPPPTGEEDTAEKDEL (SEQ ID NO: 48)
MKLSLVAAMLLLLSAARAEEEDKKEDVGTWGIDLGTTYSCVGVFKNGRVEIIAN
DQGNRITPSYVAFTPEGERLIGDAAKNQLTSNPENTVFDAKRLIGRTWNDPSVQ
QDIKFLPFKWEKKTKPYIQVDIGGGQTKTFAPEEISAMVLTKMKETAEAYLGKK
VTHAWTVPAYFNDAQRQATKDAGTIAGLNVMRIINEPTAAAIAYGLDKREGEK
NILVFDLGGGTFDVSLLTIDNGVFEWATNGDTHLGGEDFDQRVMEHFIKLYKK
KTGKDVRKDNRAVQKLRREVEKAKRALSSQHQARIEIESFYEGEDFSETLTRAK
FEELNMDLFRSTMKPVQKVLEDSDLKKSDIDEIVLVGGSTRIPKIQQLVKEFFNG
KEPSRGINPDEAVAYGAAVQAGVLSGDQDTGDLVLLDVCPLTLGIETVGGVMT
KLIPRNTWPTKKSQIFSTASDNQPTVTIKVYEGERPLTKDNHLLGTFDLTGIPPA
PRGVPQIEVTFEIDVNGILRVTAEDKGTGNKNKITITNDQNRLTPEEIERMVNDA
EKFAEEDKKLKERIDTRNELESYAYSLKNQIGDKEKLGGKLSSEDKETMEKAVE EKIEWLESHQDADIEDFKAKKKELEEIVQPIISKLYGSAGPPPTGEEDTAEKDEL
(SEQ ID NO: 49)
In embodiments, the chaperone protein is gp96. The coding region of human gp96 is 2,412 bases in length, and encodes an 803 amino acid protein that includes a 21 amino acid signal peptide at the amino terminus, a potential transmembrane region rich in hydrophobic residues, and an ER retention peptide sequence at the carboxyl terminus (GEN BANK® Accession No. X15187; see, Maki ef a/., Proc Natl Acad Sc/ USA 1990, 87:5658-5562). In embodiments, the chaperone protein is gp96 comprising the amino acid sequence of SEQ ID NO: 2. In embodiments, the chaperone protein is gp96 comprising the nucleic acid sequence of SEQ ID NO: 1. In embodiments, any of the nucleic acid sequences described herein may be codon optimized.
In embodiments, the chaperone protein of the secretable fusion protein is a secretable gp96-lg fusion protein which optionally lacks the gp96 KDEL (SEQ ID NO: 3) sequence. In embodiments, gp96, genetically fused to an immunoglobulin domain (e.g., an lgG1 , lgG2, lgG3, lgG4, IgM, IgA, or IgE molecule), activates TLR2 and TLR4 on professional antigen-presenting cells (APCs).
In embodiments, the gp96 portion of the secretable fusion protein comprises an amino acid sequence that has at least about 60%, or at least about 61 %, or at least about 62%, or at least about 63%, or at least about 64%, or at least about 65%, or at least about 66%, or at least about 67%, or at least about 68%, or at least about 69%, or at least about 70%, or at least about 71%, or at least about 72%, or at least about 73%, or at least about 74%, or at least about 75%, or at least about 76%, or at least about 77%, or at least about 78%, or at least about 79%, or at least about 80%, or at least about 81 %, or at least about 82%, or at least about 83%, or at least about 84%, or at least about 85%, or at least about 86%, or at least about 87%, or at least about 88%, or at least about 89%, or at least about 90%, or at least about 91 %, or at least about 92%, or at least about 93%, or at least about 94%, or at least about 95%, or at least about 96%, or at least about 97%, or at least about 98%, or at least about 99% sequence identity with any known wild type amino acid sequences of gp96 or a gp96 amino acid sequence disclosed herein (e.g. about 60%, or about 61%, or about 62%, or about 63%, or about 64%, or about 65%, or about 66%, or about 67%, or about 68%, or about 69%, or about 70%, or about 71 %, or about 72%, or about 73%, or about 74%, or about 75%, or about 76%, or about 77%, or about 78%, or about 79%, or about 80%, or about 81 %, or about 82%, or about 83%, or about 84%, or about 85%, or about 86%, or about 87%, or about 88%, or about 89%, or about 90%, or about 91 %, or about 92%, or about 93%, or about 94%, or about 95%, or about 96%, or about 97%, or about 98%, or about 99% sequence identity).
Thus, In embodiments, the gp96 portion of nucleic acid encoding a gp96-lg fusion polypeptide can encode an amino acid sequence that differs from the wild type gp96 polypeptide at one or more amino acid positions, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms as described previously. In embodiments, the chaperone protein may be a heat shock protein. In embodiments, the heat shock protein is one or more of hsp40, hsp60, hsp70, hsp90, and hsp110 family members, inclusive of fragments, variants, mutants, derivatives or combinations thereof (Hickey, et al., 1989, Mol. Cell. Biol. 9:2615-2626; Jindal, 1989, Mol. Cell. Biol. 9:2279-2283).
In aspects, the secretable fusion protein comprises an immunoglobulin or antibody. The antibody may be any type of antibody, i.e., immunoglobulin, known in the art. In illustrative embodiments, the antibody is an antibody of class or isotype IgA, IgD, IgE, IgG, or IgM. In illustrative embodiments, the antibody described herein comprises one or more alpha, delta, epsilon, gamma, and/or mu heavy chains. In illustrative embodiments, the antibody described herein comprises one or more kappa or light chains. In illustrative aspects, the antibody is an IgG antibody and optionally is one of the four human subclasses: lgG1, lgG2, lgG3 and lgG4. Also, the antibody In embodiments is a monoclonal antibody. In other embodiments, the antibody is a polyclonal antibody. In embodiments, the antibody is structurally similar to or derived from a naturally-occurring antibody, e.g, an antibody isolated and/or purified from a mammal, e.g., mouse, rabbit, goat, horse, chicken, hamster, human, and the like. In this regard, the antibody may be considered as a mammalian antibody, e.g., a mouse antibody, rabbit antibody, goat antibody, horse antibody, chicken antibody, hamster antibody, human antibody, and the like. In illustrative aspects, the antibody comprises sequence of only mammalian antibodies. In embodiments, the antibody is a humanized antibody, such as a humanized mouse, rabbit, goat, horse, chicken, hamster, or human antibody. In embodiments, the antibody is a humanized monoclonal antibody. In embodiments, the antibody is an antibody or antibody-like molecule such as a single-domain antibody, a recombinant heavy-chain-only antibody (VHH), a single-chain antibody (scFv), a shark heavy-chain-only antibody (VNAR), a microprotein (cysteine knot protein, knottin), a DARPin; a Tetranectin; an Affibody; a Transbody; an Anticalin; an AdNectin; an Affilin; a Microbody; a peptide aptamer; an alterase; a plastic antibody; a phylomer; a stradobody; a maxibody; an evibody; a fynomer, an armadillo repeat protein, a Kunitz domain, an avimer, an atrimer, a probody, an immunobody, a triomab, a troybody; a pepbody; a vaccibody, a UniBody; Affimers, a DuoBody, a Fv, a Fab, a Fab', and/or a F(ab')2.
In embodiments, the secretable fusion protein comprises a scaffold protein. In embodiments, the scaffold protein is Affilin, Min-23, and/or Kunitz domain.
In illustrative aspects, the fusion protein comprises a fragment of an immunoglobulin or antibody. Antibody fragments include, but are not limited to, the F(ab')2 fragment which may be produced by pepsin digestion of the antibody molecule; the Fab' fragments which may be generated by reducing the disulfide bridges of the F(ab')2 fragment, and the two Fab' fragments which may be generated by treating the antibody molecule with papain and a reducing agent.
In embodiments, a gp96 peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen et al., J Immunol 1996, 156:442-449). This region of the lgG1 molecule contains three cysteine residues that normally are involved in disulfide bonding with other cysteines in the Ig molecule. Since none of the cysteines are required for the peptide to function as a tag, one or more of these cysteine residues can be substituted by another amino acid residue, such as, for example, serine.
In embodiments, the secretable fusion protein comprises an Fc fragment of an immunoglobulin. In embodiments, the Fc fragment comprises the amino acid sequence of SEQ ID NO: 51 , or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. In embodiments, the immunoglobulin is an lgG1 immunoglobulin. In embodiments, the immunoglobulin comprises an Ig tag of the gp96-lg fusion protein comprising the Fc region of human I gG 1 , lgG2, 1 gG3, lgG4, IgM, IgA, or Ig E. Such Ig tags typically include at least functional CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. In embodiments, a T cell costimulatory peptide can be fused to the hinge, CH2 and CH3 domains of murine lgG1 (Bowen etal., J Immunol 1996, 156:442-449). The Ig tag can be from a mammalian (e.g, human, mouse, monkey, or rat) immunoglobulin, but human immunoglobulin can be particularly useful when the fusion protein is intended for in vivo use for humans. In embodiments, the secretable fusion protein comprises the amino acid sequence of SEQ ID NO: 51 , or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto. The amino acid sequences of an Fc fragment of an lgG1 antibody (SEQ ID NO: 50) and of gp96 fused to an Fc fragment of an lgG1 antibody (SEQ ID NO: 51) are provided below:
VPRDSGSKPSISTVPEVSSVFIFPPKPKDVLTITLTPKVTCVWDISKDDPEVQFS WFVDDVEVHTAQTKPREEQFNSTFRSVSELPIMHQDWLNGKEFKCRVNSAAF PAPIEKTISKTKGRPKAPQVYTIPPPKEQMAKDKVSLTCMITDFFPEDITVEWQW NGQPAENYKNTQPIMDTDGSYFVYSKLNVQKSNWEAGNTFTCSVLHEGLHNH HTEKSLSHSPGK (SEQ ID NO: 50)
MMKLIINSLYKNKEIFLRELISNASDALDKIRLISLTDENALSGNEELTVKIKCDKEK NLLHVTDTGVGMTREELVKNLGTIAKSGTSEFLNKMTEAQEDGQSTSELIGQFG VGFYSAFLVADKVIVTSKHNNDTQHIWESDSNEFSVIADPRGNTLGRGTTITLVL KEEASDYLELDTIKNLVKKYSQFINFPIYVWSSKTETVEEPMEEEEAAKEEKEES DDEAAVEEEEEEKKPKTKKVEKTVWDWELMNDIKPIWQRPSKEVEEDEYKAFY KSFSKESDDPMAYIHFTAEGEVTFKSILFVPTSAPRGLFDEYGSKKSDYIKLYVR RVFITDDFHDMMPKYLNFVKGWDSDDLPLNVSRETLQQHKLLKVIRKKLVRKT LDMIKKIADDKYNDTFWKEFGTNIKLGVIEDHSNRTRLAKLLRFQSSHHPTDITSL DQYVERMKEKQDKIYFMAGSSRKEAESSPFVERLLKKGYEVIYLTEPVDEYCIQ ALPEFDGKRFQNVAKEGVKFDESEKTKESREAVEKEFEPLLNWMKDKALKDKI EKAWSQRLTESPCALVASQYGWSGNMERIMKAQAYQTGKDISTNYYASQKK TFEINPRHPLIRDMLRRIKEDEDDKTVLDLAWLFETATLRSGYLLPDTKAYGDRI ERMLRLSLNIDPDAKVEEEPEEEPEETAEDTTEDTEQDEDEEMDVGTDEEEET AKESTAEGSVPRDSGSKPSISTVPEVSSVFIFPPKPKDVLTITLTPKVTCVWDIS KDDPEVQFSWFVDDVEVHTAQTKPREEQFNSTFRSVSELPIMHQDWLNGKEF KCRVNSAAFPAPIEKTISKTKGRPKAPQVYTIPPPKEQMAKDKVSLTCMITDFFP EDITVEWQWNGQPAENYKNTQPIMDTDGSYFVYSKLNVQKSNWEAGNTFTCS VLHEGLHNHHTEKSLSHSPGK (SEQ ID NO: 51)
In embodiments, the Fc fragment can have an amino acid sequence that differs from the Fc fragment polypeptide at one or more amino acid positions, such that it contains one or more conservative substitutions, non-conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms as described herein.
In embodiments, the gp96-lg fusion protein further comprises a linker. In embodiments, the linker may be derived from naturally-occurring multi-domain proteins or are empirical linkers as described, for example, in Chichili et al., (2013), Protein Sci. 22(2): 153-167, Chen et al., (2013), Adv Drug Deliv Rev. 65(10):1357-1369, the entire contents of which are hereby incorporated by reference. In embodiments, the linker may be designed using linker designing databases and computer programs such as those described in Chen et al., (2013), Adv Drug Deliv Rev. 65(10): 1357-1369 and Crasto et. al., (2000), Protein Eng. 13(5):309-312, the entire contents of which are hereby incorporated by reference. In embodiments, the linker is a synthetic linker such as PEG. In other embodiments, the linker is a polypeptide. In embodiments, the linker is less than about 100 amino acids long. For example, the linker may be less than about 100, about 95, about 90, about 85, about 80, about 75, about 70, about 65, about 60, about 55, about 50, about 45, about 40, about 35, about 30, about 25, about 20, about 19, about 18, about 17, about 16, about 15, about 14, about 13, about 12, about 11 , about 10, about 9, about 8, about 7, about 6, about 5, about 4, about 3, or about 2 amino acids long. In embodiments, the linker is flexible. In another embodiment, the linker is rigid. In embodiments, the linker is substantially comprised of glycine and serine residues (e.g. about 30%, or about 40%, or about 50%, or about 60%, or about 70%, or about 80%, or about 90%, or about 95%, or about 97% glycines and serines).
In embodiments, the linker is a hinge region of an antibody (e.g., of IgG, IgA, IgD, and IgE, inclusive of subclasses (e.g. lgG1, lgG2, lgG3, and lgG4, and lgA1 and lgA2)). The hinge region, found in IgG, IgA, IgD, and IgE class antibodies, acts as a flexible spacer, allowing the Fab portion to move freely in space. In contrast to the constant regions, the hinge domains are structurally diverse, varying in both sequence and length among immunoglobulin classes and subclasses. For example, the length and flexibility of the hinge region varies among the IgG subclasses. The hinge region of lgG1 encompasses amino acids 216-231 and, because it is freely flexible, the Fab fragments can rotate about their axes of symmetry and move within a sphere centered at the first of two inter-heavy chain disulfide bridges. I gG2 has a shorter hinge than lgG1 , with 12 amino acid residues and four disulfide bridges. The hinge region of lgG2 lacks a glycine residue, is relatively short, and contains a rigid poly-proline double helix, stabilized by extra inter-heavy chain disulfide bridges. These properties restrict the flexibility of the lgG2 molecule. lgG3 differs from the other subclasses by its unique extended hinge region (about four times as long as the lgG1 hinge), containing 62 amino acids (including 21 prolines and 11 cysteines), forming an inflexible poly-proline double helix. In lgG3, the Fab fragments are relatively far away from the Fc fragment, giving the molecule a greater flexibility. The elongated hinge in lgG3 is also responsible for its higher molecular weight compared to the other subclasses. The hinge region of lgG4 is shorter than that of lgG1 and its flexibility is intermediate between that of IgG 1 and I gG2. The flexibility of the hinge regions reportedly decreases in the order lgG3>lgG 1 >lgG4>lgG2.
Additional illustrative linkers include, but are not limited to, linkers having the sequence LE, GGGGS (SEQ ID NO:14), (GGGGS)n (n=1-4) (SEQ ID NO: 15), (Gly)s (SEQ ID NO: 16), (Gly)6 (SEQ ID NO: 17), (EAAAK)n (n=1-3) (SEQ ID NO: 18), A(EAAAK)nA (n = 2-5) (SEQ ID NO: 19), AEAAAKEAAAKA (SEQ ID NO: 20), A(EAAAK)4ALEA(EAAAK)4A (SEQ ID NO: 21), PAPAP (SEQ ID NO: 22), KESGSVSSEQLAQFRSLD (SEQ ID NO: 23), EGKSSGSGSESKST(SEQ ID NO: 24), GSAGSAAGSGEF (SEQ ID NO: 25), and (XP)n, with X designating any amino acid, e.g., Ala, Lys, or Glu.
In embodiments, the linker may be functional. For example, without limitation, the linker may function to improve the folding and/or stability, improve the expression, improve the pharmacokinetics, and/or improve the bioactivity of the present compositions. In another example, the linker may function to target the compositions to a particular cell type or location.
Methods of Treatment
Fused biological cells generated in accordance with embodiments of the present disclosure can be used in various methods of treatment of cancer and infections.
The infection can be, for example, an acute infection or a chronic infection. In embodiments, the infection can be an infection by coronavirus, hepatitis C virus, hepatitis B virus, human immunodeficiency virus, or malaria. The methods can include administering to a subject a fused biological cell, under conditions wherein the progression or a symptom of a clinical condition in the subject is reduced in a therapeutic manner.
In embodiments, the present disclosure pertains to treatment of cancers and/or tumors. Cancers or tumors refer to an uncontrolled growth of cells and/or abnormal increased cell survival and/or inhibition of apoptosis which interferes with the normal functioning of the bodily organs and systems. Included are benign and malignant cancers, polyps, hyperplasia, as well as dormant tumors or micrometastases. Also, included are cells having abnormal proliferation that is not impeded by the immune system {e.g. virus infected cells). The cancer may be a primary cancer or a metastatic cancer. The primary cancer may be an area of cancer cells at an originating site that becomes clinically detectable, and may be a primary tumor. In contrast, the metastatic cancer may be the spread of a disease from one organ or part to another non-adjacent organ or part. The metastatic cancer may be caused by a cancer cell that acquires the ability to penetrate and infiltrate surrounding normal tissues in a local area, forming a new tumor, which may be a local metastasis. The cancer may also be caused by a cancer cell that acquires the ability to penetrate the walls of lymphatic and/or blood vessels, after which the cancer cell is able to circulate through the bloodstream (thereby being a circulating tumor cell) to other sites and tissues in the body. The cancer may be due to a process such as lymphatic or hematogeneous spread. The cancer may also be caused by a tumor cell that comes to rest at another site, repenetrates through the vessel or walls, continues to multiply, and eventually forms another clinically detectable tumor. The cancer may be this new tumor, which may be a metastatic (or secondary) tumor.
The cancer may be caused by tumor cells that have metastasized, which may be a secondary or metastatic tumor. The cells of the tumor may be like those in the original tumor. As an example, if a breast cancer or colon cancer metastasizes to the liver, the secondary tumor, while present in the liver, is made up of abnormal breast or colon cells, not of abnormal liver cells. The tumor in the liver may thus be a metastatic breast cancer or a metastatic colon cancer, not liver cancer.
The cancer may have an origin from any tissue. The cancer may originate from melanoma, colon, breast, or prostate, and thus may be made up of cells that were originally skin, colon, breast, or prostate, respectively. The cancer may also be a hematological malignancy, which may be lymphoma. The cancer may invade a tissue such as liver, lung, bladder, or intestinal.
Illustrative cancers that may be treated include, but are not limited to, carcinomas, e.g. various subtypes, including, for example, adenocarcinoma, basal cell carcinoma, squamous cell carcinoma, and transitional cell carcinoma), sarcomas (including, for example, bone and soft tissue), leukemias (including, for example, acute myeloid, acute lymphoblastic, chronic myeloid, chronic lymphocytic, and hairy cell), lymphomas and myelomas (including, for example, Hodgkin and non-Hodgkin lymphomas, light chain, non-secretory, MGUS, and plasmacytomas), and central nervous system cancers (including, for example, brain {e.g. gliomas {e.g. astrocytoma, oligodendroglioma, and ependymoma), meningioma, pituitary adenoma, and neuromas, and spinal cord tumors {e.g. meningiomas and neurofibroma).
In embodiments, the compositions and methods in accordance with embodiments of the present disclosure activate an innate, humoral (i.e. antibody response), and/or cellular (i.e. T cell) response in a subject receiving the present compositions.
In aspects, methods of treatments using cell-fusion based immune agents in accordance with the present disclosure are provided.
In embodiments, a method is suitable for increasing the subject's T-cell response as compared to the T-cell response of a subject that was not administered the compositions in accordance with embodiments of the present disclosure. In embodiments, the method is suitable for increasing the subject's antibody response as compared to the antibody response of a subject that was not administered the present compositions. In embodiments, the method is suitable for increasing the subject's innate immune response as compared to the innate immune response of a subject that was not administered the present compositions. In embodiments, the method is suitable for increasing the subject's T-cell response, antibody response, and innate immune response as compared to the T-cell response, antibody response, and innate immune responses of a subject that was not administered the present compositions.
In embodiments, the method is suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell population(s) of a subject that was not administered the present compositions. The subject's T cells include T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells. In embodiments, the method is suitable for increasing and/or restoring the subject's CD4+ helper cells population(s) as compared to the CD4+ helper cells population(s) of a subject that was not administered the present compositions.
In embodiments, the method is suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell population(s) of a subject that was administered another composition. The subject's T cells include T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells. In embodiments, the method is suitable for increasing and/or restoring the subject's CD4+ helper cells population(s) as compared to the CD4+ helper cells population(s) of a subject that was administered another composition.
Accordingly, in some aspects, a method of treating cancer is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In embodiments, the cancer comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia.
In embodiments, the cancer is selected from one or more of the basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer; glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer; melanoma; myeloma; neuroblastoma; oral cavity cancer; ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulval cancer; Hodgkin's lymphoma; non-Hodgkin's lymphoma; B-cell lymphoma; small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; Waldenstrom's Macroglobulinemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); and Hairy cell leukemia.
In embodiments, the cancer is selected from one or more of basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and central nervous system cancer; breast cancer; cancer of the peritoneum; cervical cancer; choriocarcinoma; colon and rectum cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); melanoma; myeloma; neuroblastoma; oral cavity cancer (lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulvar cancer; lymphoma including Hodgkin's and non-Hodgkin's lymphoma, as well as B-cell lymphoma (including low grade/follicular non-Hodgkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; and Waldenstrom's Macroglobulinemia; chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); Hairy cell leukemia; chronic myeloblastic leukemia; as well as other carcinomas and sarcomas; and post-transplant lymphoproliferative disorder (PTLD), as well as abnormal vascular proliferation associated with phakomatoses, edema (e.g. that associated with brain tumors), and Meigs syndrome.
In aspects, a method of treating or preventing an infectious disease is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In embodiments, the infectious disease comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection. In embodiments, the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae.
In embodiments, the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E. In embodiments, the infectious disease comprises a coronavirus infection 2019 (COVID-19).
In aspects, a fused biological cell in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell is used to eliminate intracellular pathogens. In aspects, the fused biological cells are used to treat one or more infections. In embodiments, methods of treating viral infections are provided, including, for example, HIV and HCV; parasitic infections (including, for example, malaria); and bacterial infections. In embodiments, the infections induce immunosuppression. For example, HIV infections often result in immunosuppression in the infected subjects. Accordingly, as described elsewhere herein, the treatment of such infections may involve, in embodiments, modulating the immune system with the present fused biological cell to favor immune stimulation over immune inhibition. Alternatively, embodiments of the present disclosure provide methods for treating infections that induce immunoactivation. For example, intestinal helminth infections have been associated with chronic immune activation. In these embodiments, the treatment of such infections may involve modulating the immune system with the present fused biological cell to favor immune inhibition over immune stimulation.
In embodiments, methods of treating viral infections are provided, wherein the viral infections include, without limitation, acute or chronic viral infections, for example, of the respiratory tract, of papilloma virus infections, of herpes simplex virus (HSV) infection, of human immunodeficiency virus (HIV) infection, and of viral infection of internal organs such as infection with hepatitis viruses. In embodiments, the viral infection is caused by a virus of family Flaviviridae. In embodiments, the virus of family Flaviviridae is selected from Yellow Fever Virus, West Nile virus, Dengue virus, Japanese Encephalitis Virus, St. Louis Encephalitis Virus, and Hepatitis C Virus. In other embodiments, the viral infection is caused by a virus of family Picornaviridae, e.g, poliovirus, rhinovirus, coxsackievirus. In other embodiments, the viral infection is caused by a member of Orthomyxoviridae, e.g., an influenza virus. In other embodiments, the viral infection is caused by a member of Retroviridae, e.g, a lentivirus. In other embodiments, the viral infection is caused by a member of Paramyxoviridae, e.g., respiratory syncytial virus, a human parainfluenza virus, rubulavirus {e.g., mumps virus), measles virus, and human metapneumovirus. In other embodiments, the viral infection is caused by a member of Bunyaviridae, e.g., hantavirus. In other embodiments, the viral infection is caused by a member of Reoviridae, e.g., a rotavirus.
In embodiments, methods of treating parasitic infections, such as, e.g., protozoan or helminths infections, are provided. In embodiments, the parasitic infection is by a protozoan parasite. In embodiments, the oritiziab parasite is selected from intestinal protozoa, tissue protozoa, or blood protozoa. Illustrative protozoan parasites include, but are not limited to, Entamoeba hystolytica, Giardia lamblia, Cryptosporidium muris, Trypanosomatida gambiense, Trypanosomatida rhodesiense, Trypanosomatida crusi, Leishmania mexicana, Leishmania braziliensis, Leishmania tropica, Leishmania donovani, Toxoplasma gondii, Plasmodium vivax, Plasmodium ovale, Plasmodium malariae, Plasmodium falciparum, Trichomonas vaginalis, and Histomonas meleagridis. In embodiments, the parasitic infection is by a helminthic parasite such as nematodes (e.g., Adenophorea). In embodiments, the parasite is selected from Secementea (e.g., Trichuris trichiura, Ascaris lumbricoides, Enterobius vermicularis, Ancylostoma duodenale, Necator americanus, Strongyloides stercoralis, Wuchereria bancrofti, Dracunculus medinensis). In embodiments, the parasite is selected from trematodes (e.g. blood flukes, liver flukes, intestinal flukes, and lung flukes). In embodiments, the parasite is selected from: Schistosoma mansoni, Schistosoma haematobium, Schistosoma japonicum, Fasciola hepatica, Fasciola gigantica, Heterophyes, Paragonimus westermani. In embodiments, the parasite is selected from cestodes (e.g, Taenia solium, Taenia saginata, Hymenolepis nana, and Echinococcus granulosus).
In embodiments, methods of treating bacterial infections are provided. In embodiments, the bacterial infection is by a gram-positive bacteria, gram-negative bacteria, aerobic and/or anaerobic bacteria. In embodiments, the bacteria is selected from, but not limited to, Staphylococcus, Lactobacillus, Streptococcus, Sarcina, Escherichia, Enterobacter, Klebsiella, Pseudomonas, Acinetobacter, Mycobacterium, Proteus, Campylobacter, Citrobacter, Nisseria, Baccillus, Bacteroides, Peptococcus, Clostridium, Salmonella, Shigella, Serratia, Haemophilus, Brucella and other organisms. In embodiments, the bacteria is selected from, but not limited to, Pseudomonas aeruginosa, Pseudomonas fluorescens, Pseudomonas acidovorans, Pseudomonas alcaligenes, Pseudomonas putida, Stenotrophomonas maltophilia, Burkholderia cepacia, Aeromonas hydrophilia, Escherichia coli, Citrobacter freundii, Salmonella typhimurium, Salmonella typhi, Salmonella paratyphi, Salmonella enteritidis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Enterobacter cloacae, Enterobacter aerogenes, Klebsiella pneumoniae, Klebsiella oxytoca, Serratia marcescens, Francisella tularensis, Morganella morganii, Proteus mirabilis, Proteus vulgaris, Providencia alcalifaciens, Providencia rettgeri, Providencia stuartii, Acinetobacter baumannii, Acinetobacter calcoaceticus, Acinetobacter haemolyticus, Yersinia enterocolitica, Yersinia pestis, Yersinia pseudotuberculosis, Yersinia intermedia, Bordetella pertussis, Bordetella parapertussis, Bordetella bronchiseptica, Haemophilus influenzae, Haemophilus parainfluenzae, Haemophilus haemolyticus, Haemophilus parahaemolyticus, Haemophilus ducreyi, Pasteurella multocida, Pasteurella haemolytica, Branhamella catarrhalis, Helicobacter pylori, Campylobacter fetus, Campylobacter jejuni, Campylobacter coli, Borrelia burgdorferi, Vibrio cholerae, Vibrio parahaemolyticus, Legionella pneumophila, Listeria monocytogenes, Neisseria gonorrhoeae, Neisseria meningitidis, Kingella, Moraxella, Gardnerella vaginalis, Bacteroides fragilis, Bacteroides distasonis, Bacteroides 3452A homology group, Bacteroides vulgatus, Bacteroides ovalus, Bacteroides thetaiotaomicron, Bacteroides uniformis, Bacteroides eggerthii, Bacteroides splanchnicus, Clostridium difficile, Mycobacterium tuberculosis, Mycobacterium avium, Mycobacterium intracellulare, Mycobacterium leprae, Corynebacterium diphtheriae, Corynebacterium ulcerans, Streptococcus pneumoniae, Streptococcus agalactiae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus saprophyticus, Staphylococcus intermedius, Staphylococcus hyicus subsp. hyicus, Staphylococcus haemolyticus, Staphylococcus hominis, or Staphylococcus saccharolyticus. The fused biological cell made in accordance with embodiments of the present disclosure, which is to be administered to a patient, can be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecular structures, or mixtures of compounds such as, for example, liposomes, receptor or cell targeted molecules, or oral, topical or other formulations for assisting in uptake, distribution and/or absorption. In some cases, an expression vector can be contained within a cell that is administered to a subject, or contained within a virus or virus-like particle.
In embodiments, the fused biological cell can be included in a pharmaceutical composition or can be in the form of a pharmaceutical composition. In embodiments, the pharmaceutical composition comprises a checkpoint inhibitor. In embodiments, the checkpoint inhibitor is an anti-PD1 antibody.
The fused biological cell to be administered can be in combination with a pharmaceutically acceptable carrier.
In embodiments, the pharmaceutically acceptable carrier is a physiologically and pharmaceutically acceptable carrier. The physiologically and pharmaceutically acceptable carrier can include any of the well-known components useful for immunization. The carrier can facilitate or enhance an immune response to antigen(s) expressed by the fused biological cell. The cell formulations can contain buffers to maintain a preferred pH range, salts or other components that present antigen(s) to a subject in a composition that stimulates an immune response to the antigen(s). The physiologically acceptable carrier also can include one or more adjuvants that enhance the immune response to the antigens. Pharmaceutically acceptable carriers include, for example, pharmaceutically acceptable solvents, suspending agents, or any other pharmacologically inert vehicles for delivering compounds to a subject. Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties, when combined with one or more therapeutic compounds and any other components of a given pharmaceutical composition. Typical pharmaceutically acceptable carriers include, without limitation: water, saline solution, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose or dextrose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g, starch or sodium starch glycolate), and wetting agents (e.g, sodium lauryl sulfate). Compositions can be formulated for subcutaneous, intramuscular, or intradermal administration, or in any manner acceptable for delivery of a fused cell to a subject.
An adjuvant refers to a substance which, when added to an immunogenic agent such as a fused cell expressing secreted vaccine protein (e.g, gp96-lg), a T cell costimulatory fusion polypeptide, and optionally one or more disease antigens, nonspecifical ly enhances or potentiates an immune response to the agent in the recipient host upon exposure to the mixture. Adjuvants can include, for example, oil-in-water emulsions, water-in oil emulsions, alum (aluminum salts), liposomes and microparticles, such as, polysytrene, starch, polyphosphazene and poly lactide/polyglycosides.
Adjuvants can also include, for example, squalene mixtures (SAF-I), muramyl peptide, saponin derivatives, mycobacterium cell wall preparations, monophosphoryl lipid A, mycolic acid derivatives, nonionic block copolymer surfactants, Quil A, cholera toxin B subunit, polyphosphazene and derivatives, and immunostimulating complexes (ISCOMs) such as those described by Takahashi et al., Nature 1990, 344:873-875. For veterinary use and for production of antibodies in animals, mitogenic components of Freund's adjuvant (both complete and incomplete) can be used. In humans, Incomplete Freund's Adjuvant (IFA) is a useful adjuvant. Various appropriate adjuvants are well known in the art (see, for example, Warren and Chedid, CRC Critical Reviews in Immunology 1988, 8:83; and Allison and Byars, in Vaccines: New Approaches to Immunological Problems, 1992, Ellis, ed., Butterworth-Heinemann, Boston). Additional adjuvants include, for example, bacille Calmett-Guerin (BCG), DETOX (containing cell wall skeleton of Mycobacterium phlei (CWS) and monophosphoryl lipid A from Salmonella minnesota (MPL)), and the like (see, for example, Hoover etal., J Clin Oncol 1993, 11 :390; and Woodlock etal., J Immunother 1999, 22:251-259).
In embodiments, a fused biological cell can be administered to a subject one or more times (e.g, once, twice, two to four times, three to five times, five to eight times, six to ten times, eight to 12 times, or more than 12 times). A fused biological cell as provided herein can be administered one or more times per day, one or more times per week, every other week, one or more times per month, once every two to three months, once every three to six months, or once every six to 12 months. A fused biological cell can be administered over any suitable period of time, such as a period from about 1 day to about 12 months. In embodiments, for example, the period of administration can be from about 1 day to 90 days; from about 1 day to 60 days; from about 1 day to 30 days; from about 1 day to 20 days; from about 1 day to 10 days; from about 1 day to 7 days. In embodiments, the period of administration can be from about 1 week to 50 weeks; from about 1 week to 50 weeks; from about 1 week to 40 weeks; from about 1 week to 30 weeks; from about 1 week to 24 weeks; from about 1 week to 20 weeks; from about 1 week to 16 weeks; from about 1 week to 12 weeks; from about 1 week to 8 weeks; from about 1 week to 4 weeks; from about 1 week to 3 weeks; from about 1 week to 2 weeks; from about 2 weeks to 3 weeks; from about 2 weeks to 4 weeks; from about 2 weeks to 6 weeks; from about 2 weeks to 8 weeks; from about 3 weeks to 8 weeks; from about 3 weeks to 12 weeks; or from about 4 weeks to 20 weeks.
In embodiments, after an initial dose (sometimes referred to as a "priming” dose) of a fused biological cell has been administered and a maximal antigen-specific immune response has been achieved, one or more boosting doses of a fused biological cell as provided herein can be administered. For example, a boosting dose can be administered about 10 to 30 days, about 15 to 35 days, about 20 to 40 days, about 25 to 45 days, or about 30 to 50 days after a priming dose.
In embodiments, the methods provided herein can be used for controlling solid tumor growth (e.g., breast, prostate, melanoma, renal, colon, or cervical tumor growth) and/or metastasis. The methods can include administering an effective amount of a fused biological cell as described herein to a subject in need thereof. In embodiments, the subject is a mammal (e.g., a human).
The methods provided herein can be useful for stimulating an immune response against a tumor. Such immune response is useful in treating or alleviating a sign or symptom associated with the tumor. As used herein, "treating” is defined as reducing, preventing, and/or reversing the symptoms in the individual to which a fused biological cell in accordance with embodiments of the present disclosure has been administered, as compared to the symptoms of an individual not being treated. A practitioner will appreciate that the methods described herein are to be used in concomitance with continuous clinical evaluations by a skilled practitioner (physician or veterinarian) to determine subsequent therapy. Such evaluations will aid and inform in evaluating whether to increase, reduce, or continue a particular treatment dose, mode of administration, etc.
The methods provided herein can thus be used to treat a tumor, including, for example, a cancer. The methods can be used, for example, to inhibit the growth of a tumor by preventing further tumor growth, by slowing tumor growth, or by causing tumor regression. Thus, the methods can be used, for example, to treat a cancer such as a lung cancer. It will be understood that the subject to which a compound is administered need not suffer from a specific traumatic state. Indeed, the fused biological cells described herein may be administered prophylactically, prior to development of symptoms (e.g., a patient in remission from cancer). The terms "therapeutic” and "therapeutically,” and permutations of these terms, are used to encompass therapeutic, palliative, and prophylactic uses. Thus, as used herein, by "treating or alleviating the symptoms” is meant reducing, preventing, and/or reversing the symptoms of the individual to which a therapeutically effective amount of a composition has been administered, as compared to the symptoms of an individual receiving no such administration.
As used herein, the terms "effective amount” and "therapeutically effective amount” refer to an amount sufficient to provide the desired therapeutic (e.g., anti-cancer, anti-tumor, or anti-infection) effect in a subject (e.g., a human diagnosed as having cancer or an infection). Anti-tumor and anti-cancer effects include, without limitation, modulation of tumor growth (e.g, tumor growth delay), tumor size, or metastasis, the reduction of toxicity and side effects associated with a particular anti-cancer agent, the amelioration or minimization of the clinical impairment or symptoms of cancer, extending the survival of the subject beyond that which would otherwise be expected in the absence of such treatment, and the prevention of tumor growth in an animal lacking tumor formation prior to administration, i.e., prophylactic administration. In embodiments, administration of an effective amount of a fused biological cell or a composition including the fused biological cell can increase the activation or proliferation of tumor antigen specific T cells in a subject. For example, the activation or proliferation of tumor antigen specific T cells in the subject can be is increased by at least 10 percent (e.g, at least 25 percent, at least 50 percent, or at least 75 percent) as compared to the level of activation or proliferation of tumor antigen specific T cells in the subject prior to the administration.
Anti-infection effects include, for example, a reduction in the number of infective agents (e.g, viruses or bacteria). When the clinical condition in the subject to be treated is an infection, administration of a vector as provided herein can stimulate the activation or proliferation of pathogenic antigen specific T cells in the subject. For example, administration of the vector can lead to activation of antigen-specific T cells in the subject to a level greater than that achieved by gp96-lg vaccination alone. One of skill in the art will appreciate that an effective amount of a fused biological cell may be lowered or increased by fine tuning and/or by administering more than one dose. This document therefore provides a method for tailoring the administration/treatment to the particular exigencies specific to a given mammal. Therapeutically effective amounts can be determined by, for example, starting at relatively low amounts and using step-wise increments with concurrent evaluation of beneficial effects. The methods provided herein thus can be used alone or in combination with other well- known tumor therapies, to treat a patient having a tumor. One skilled in the art will readily understand advantageous uses of the fused biological cells and methods provided herein, for example, in prolonging the life expectancy of a cancer patient and/or improving the quality of life of a cancer patient (e.g., a lung cancer patient).
In aspects, a method of stimulating a patient's antibody response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In aspects, a method of stimulating a patient T cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In aspects, a method of stimulating one or both a patient's antibody response and T cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In embodiments, the T cell-driven cellular immune response induces a CD8+ T cell response in the patient. In embodiments, the T cell-driven cellular immune response induces a CD4+ T cell response in the patient.
In embodiments, a method induces an immune response, selected from a CD8+ T cell, a CD4+ T cell, a cytotoxic T lymphocyte (CTL), a TH1 response, a TH2 response, or a combination thereof.
In embodiments, a method results in reduction of a number of regulatory T cells (Tregs). The depletion of Tregs during an acute phase of infection was shown to result in enhanced effector T cell function and decreased viral loads. See Dietze et al. (2013) PLoS pathogens vol. 9: 12, e1003798. doi:10.1371/journal.ppat.1003798; see also Zelinskyy et al. (2009). Blood 114: 3199-3207. The role of Treg depletion in antitumor immunity has also been observed. See, e.g., Dannull et al. (2005) J Clin Invest 115: 3623-3633. It was shown that I L-2-induced antitumor immunity is enhanced by Treg-cell depletion, due to expansion of the tumor-infiltrating cytotoxic CD8(+) T-cell population. Imai et al. (2007). Cancer Sol. 98, 416-423 (2007).
In embodiments, a method results in the decrease in the functionality of Tregs. In embodiments, the method does not result in the decrease in the number of Tregs but results in the decrease in the functionality of Tregs. In embodiments, the method results in a decrease in the frequency of Tregs. In embodiments, the method results in the decrease in the number of functional Tregs.
In aspects, a method of stimulating one or both a patient's antibody response and B cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with embodiments of the present disclosure, or a pharmaceutical composition comprising the fused biological cell.
In aspects, a method of stimulating a patient B cell-driven cellular immune response is provided that comprises administering to a patient in need thereof the fused biological cell generated in accordance with any of the embodiments of the present disclosure.
The method of stimulating or enhancing one or both a patient's antibody response and B cell-driven cellular immune response is performed without depleting the T cell population in the patient.
Compositions
Embodiments of the present disclosure also provide a composition comprising a fused biological cell as described herein, and an excipient, carrier, or diluent. In exemplary aspects, the composition is a pharmaceutical composition. In illustrative aspects, the composition may comprise virus particles containing the fused biological cell.
Embodiments of the present disclosure also provide a composition comprising a lentivirus and/or transfer plasmid or a biological or host cell or a population of cells, as described herein, and an excipient, carrier, or diluent. In exemplary aspects, the composition is a pharmaceutical composition.
In embodiments, a cellular therapy in accordance with embodiments of the present disclosure may be used as a standalone cellular therapy or as a cellular therapy in combination with other cellular therapies that drive humoral immunity, to provide an added layer of cellular immunity. In embodiments, the cellular therapy comprises a therapeutic biological cell (e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease). The present cellular therapy can be used in combination with one or cellular therapies or any other type.
In illustrative aspects, the composition is a sterile composition. In embodiments, the composition is suitable for administration to a human. In illustrative aspects, the composition comprises a unit dose of fused biological cells. In embodiments, the unit dose is about 105, about 106, about 107, about 108, about 109, about 1010, about 1011, about 1012, about 1013, about 1014, about 1015, or more fused biological cells. In embodiments, the composition comprises at least or about 106 fused cells.
In illustrative aspects, the composition may comprise lentivirus particles containing a transfer plasmid. In illustrative aspects, the composition is a sterile composition. In embodiments, the composition is suitable for administration to a human. In illustrative aspects, the composition comprises a unit dose of biological or host cells. In embodiments, the unit dose is about 105, about 106, about 107, about 108, about 109, about 1010, about 1011 , about 1012, about 1013, about 1014, about 1015, or more biological or host cells into which the lentivirus and/or transfer plasmid is introduced. In embodiments, the composition comprises at least or about 106 cells into with the lentivirus and/or transfer plasmid is introduced.
The pharmaceutical composition can comprise any pharmaceutically acceptable ingredient, including, for example, acidifying agents, additives, adsorbents, aerosol propellants, air displacement agents, alkalizing agents, anticaking agents, anticoagulants, antimicrobial preservatives, antioxidants, antiseptics, bases, binders, buffering agents, chelating agents, coating agents, coloring agents, desiccants, detergents, diluents, disinfectants, disintegrants, dispersing agents, dissolution enhancing agents, dyes, emollients, emulsifying agents, emulsion stabilizers, fillers, film forming agents, flavor enhancers, flavoring agents, flow enhancers, gelling agents, granulating agents, humectants, lubricants, mucoadhesives, ointment bases, ointments, oleaginous vehicles, organic bases, pastille bases, pigments, plasticizers, polishing agents, preservatives, sequestering agents, skin penetrants, solubilizing agents, solvents, stabilizing agents, suppository bases, surface active agents, surfactants, suspending agents, sweetening agents, therapeutic agents, thickening agents, tonicity agents, toxicity agents, viscosity-increasing agents, water-absorbing agents, water-miscible cosolvents, water softeners, or wetting agents.
The pharmaceutical compositions may be formulated to achieve a physiologically compatible pH. In embodiments, the pH of the pharmaceutical composition may be at least 5, at least 5.5, at least 6, at least 6.5, at least 7, at least 7.5, at least 8, at least 8.5, at least 9, at least 9.5, at least 10, or at least 10.5 up to and including pH 11, depending on the formulation and route of administration, for example between 4 and 7, or 4.5 and 5.5. In illustrative embodiments, the pharmaceutical compositions may comprise buffering agents to achieve a physiological compatible pH. The buffering agents may include any compounds capable of buffering at the desired pH such as, for example, phosphate buffers (e.g., PBS), triethanolamine, Tris, bicine, TAPS, tricine, HEPES, TES, MOPS, PIPES, cacodylate, MES, acetate, citrate, succinate, histidine or other pharmaceutically acceptable buffers.
In embodiments, a buffering agent comprises PBS (phosphate-buffered saline) including a blocking buffer, such as, e.g., Bovine serum albumin (BSA). In embodiments, the buffering agent comprises PBS including from about 1 % to about 10% BSA. In embodiments, the buffering agent comprises PBS including about 1 % BSA.
In embodiments, a buffering agent comprises tris-buffered saline (TBS) including a blocking buffer, such as, e.g, BSA. In embodiments, the buffering agent comprises TBS including from about 1 % to about 10% BSA. In embodiments, the buffering agent comprises TBS including about 1 % BSA.
In embodiments, the present disclosure therefore provides compositions including pharmaceutical compositions including a fused biological cell as described herein, in combination with a physiologically and pharmaceutically acceptable carrier. In embodiments, the present disclosure therefore provides compositions including pharmaceutical compositions containing packaged lentivirus or a cell containing the lentivirus and/or transfer plasmid as described herein, in combination with a physiologically and pharmaceutically acceptable carrier. In embodiments, the physiologically and pharmaceutically acceptable carrier can include any of the well-known components useful for immunization. The carrier can facilitate or enhance an immune response to an antigen administered in a vaccine. The cell formulations can contain buffers to maintain a preferred pH range, salts or other components that present an antigen to an individual in a composition that stimulates an immune response to the antigen. The physiologically acceptable carrier also can contain one or more adjuvants that enhance the immune response to an antigen. Pharmaceutically acceptable carriers include, for example, pharmaceutically acceptable solvents, suspending agents, or any other pharmacologically inert vehicles for delivering compounds to a subject. Pharmaceutically acceptable carriers can be liquid or solid, and can be selected with the planned manner of administration in mind so as to provide for the desired bulk, consistency, and other pertinent transport and chemical properties, when combined with one or more therapeutic compounds and any other components of a given pharmaceutical composition. Typical pharmaceutically acceptable carriers include, without limitation: water, saline solution, binding agents (e.g., polyvinylpyrrolidone or hydroxypropyl methylcellulose); fillers (e.g., lactose or dextrose and other sugars, gelatin, or calcium sulfate), lubricants (e.g., starch, polyethylene glycol, or sodium acetate), disintegrates (e.g, starch or sodium starch glycolate), and wetting agents (e.g, sodium lauryl sulfate). Compositions can be formulated for subcutaneous, intramuscular, or intradermal administration, or in any manner acceptable for administration.
An adjuvant refers to a substance which, when added to an immunogenic agent such as a cell containing an expression vector system, nonspecifically enhances or potentiates an immune response to the agent in the recipient host upon exposure to the mixture. Adjuvants can include, for example, oil-in-water emulsions, water-in oil emulsions, alum (aluminum salts), liposomes and microparticles, such as, polysytrene, starch, polyphosphazene and polylactide/polyglycosides.
Adjuvants can also include, for example, squalene mixtures (SAF-I), muramyl peptide, saponin derivatives, mycobacterium cell wall preparations, monophosphoryl lipid A, mycolic acid derivatives, nonionic block copolymer surfactants, Quil A, cholera toxin B subunit, polyphosphazene and derivatives, and immunostimulating complexes (ISCOMs) such as those described by Takahashi et al., Nature 1990, 344:873-875. For veterinary use and for production of antibodies in animals, mitogenic components of Freund's adjuvant (both complete and incomplete) can be used. In humans, Incomplete Freund's Adjuvant (IFA) is a useful adjuvant. Various appropriate adjuvants are well known in the art (see, for example, Warren and Chedid, CRC Critical Reviews in Immunology 1988, 8:83; and Allison and Byars, in Vaccines: New Approaches to Immunological Problems, 1992, Ellis, ed., Butterworth-Heinemann, Boston). Additional adjuvants include, for example, bacille Calmett-Guerin (BCG), DETOX (containing cell wall skeleton of Mycobacterium phlei (CWS) and monophosphoryl lipid A from Salmonella minnesota (MPL)), and the like (see, for example, Hoover et al., J Clin Oncol 1993, 11 :390; and Woodlock et al., J Immunother 1999, 22:251-259).
Routes of Administration
Methods of administering cells to a subject are well-known, and include, but not limited to perfusions, infusions, and injections. See, e.g, Burch et al., Clin Cancer Res 6(6): 2175-2182 (2000), Dudley et al., J Clin Oncol 26(32): 5233- 5239 (2008); Khan et al., Cell Transplant 19:409-418 (2010); Gridelli et al., Liver Transpl 18:226-237 (2012).
In embodiments, a fused biological cell prepared in accordance with embodiments of the present disclosure is administered by injection. In embodiments, the fused biological cell (In embodiments in combination with a checkpoint inhibitor and/or another agent) is administered in a pharmaceutically acceptable formulation, including a formulation suitable for one or more of injection, e.g., subcutaneous injection, intradermal injection (including to the dermis or epidermis), subdermal injection, intratumoral injection, intramuscular injection, intraocular injection, intravitreal injection, intra-articular injection, intracardiac injection, intravenous injection, epidural injection, intrathecal injection, and intraportal injection.
Combination Therapies and Conjugation
In embodiments, methods in accordance with the present disclosure comprise administering an additional agent to a subject.
In embodiments, inclusive of, without limitation, cancer applications, embodiments of the present disclosure pertain to chemotherapeutic agents as additional agents. Examples of chemotherapeutic agents include, but are not limited to, alkylating agents such as thiotepa and CYTOXAN cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins {e.g, bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; cally statin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins {e.g, cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB 1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics {e.g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (see, e.g., Agnew, Chem. Inti. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo- 5-oxo-L-norleucine, ADRIAMYCIN doxorubicin (including morpholino- doxorubicin, cyanomorpholino-doxorubicin, 2- pyrrolino-doxorubicin and deoxy doxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as minoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (e.g, T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL paclitaxel (Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANE Cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, 111.), and TAXOTERE doxetaxel (Rhone-Poulenc Rorer, Antony, France); chloranbucil; GEMZAR gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; NAVELBINE, vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (Camptosar, CPT-11) (including the treatment regimen of irinotecan with 5-FU and leucovorin); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; combretastatin; leucovorin (LV); oxaliplatin, including the oxaliplatin treatment regimen (FOLFOX); lapatinib (TYKERB); inhibitors of PKC-o, Raf, H-Ras, EGFR (e.g., erlotinib (Tarceva)) and VEGF-A that reduce cell proliferation and pharmaceutically acceptable salts, acids or derivatives of any of the above. In addition, the methods of treatment can further include the use of radiation. In addition, the methods of treatment can further include the use of photodynamic therapy.
In embodiments, inclusive of, without limitation, infectious disease applications, the present disclosure pertains to anti- infectives as additional agents. In embodiments, the anti-infective is an anti-viral agent including, but not limited to, Abacavir, Acyclovir, Adefovir, Amprenavir, Atazanavir, Cidofovir, Darunavir, Delavirdine, Didanosine, Docosanol, Efavirenz, Elvitegravir, Emtricitabine, Enfuvirtide, Etravirine, Famciclovir, and Foscarnet. In embodiments, the anti- infective is an anti-bacterial agent including, but not limited to, cephalosporin antibiotics (cephalexin, cefuroxime, cefadroxil, cefazolin, cephalothin, cefaclor, cefamandole, cefoxitin, cefprozil, and ceftobiprole); fluoroquinolone antibiotics (cipro, Levaquin, floxin, tequin, avelox, and norflox); tetracycline antibiotics (tetracycline, minocycline, oxytetracycline, and doxycycline); penicillin antibiotics (amoxicillin, ampicillin, penicillin V, dicloxacillin, carbenicillin, vancomycin, and methicillin); monobactam antibiotics (aztreonam); and carbapenem antibiotics (ertapenem, doripenem, imipenem/cilastatin, and meropenem). In embodiments, the anti-infectives include anti-malarial agents (e.g, chloroquine, quinine, mefloquine, primaquine, doxycycline, artemether/lumefantrine, atovaquone/proguanil and sulfadoxine/pyrimethamine), metronidazole, tinidazole, ivermectin, pyrantel pamoate, and albendazole.
In embodiments, additional agents include blocking antibodies targeted to an immune checkpoint molecules. Nonlimiting examples of checkpoint molecules include CTLA4 (cytotoxic T lymphocyte antigen-4), PD1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), LAG-3 (lymphocyte activation gene-3), TIM-3 (T cell immunoglobulin and mucin protein-3), LAG-3 (lymphocyte activation gene-3), and others. See Pardoll, 2012, Nature Reviews Cancer 12:252-64; Nirschl & Drake, 2013, Clin Cancer Res 19:4917-24; He et al., 2018, OncoTargets and therapy, vol. 11 7005-7009; Qin et al., 2019. Mol Cancer 18, 155.
Biological Cells
Also provided by the present disclosure is a biological cell (e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) or host cell comprising any of the lentiviruses as described herein. As used herein, the term "host cell” refers to any type of cell that can contain the lentivirus and/or transfer plasmid, e.g., without limitation, a cell that produces the lentivirus. The host cell can be a eukaryotic cell, e.g, plant, animal, fungi, or algae, or can be a prokaryotic cell, e.g., bacteria or protozoa. The host cell can be a cultured cell or a primary cell, i.e., isolated directly from an organism, e.g., a human. The host cell can be an adherent cell or a suspended cell, i.e., a cell that grows in suspension. In illustrative aspects, the host cell is a mammalian host cell. In illustrative aspects, the host cell is a human host cell. In illustrative aspects, the human host cell is an NIH 3T3 cell or an HEK293 cell. The presently disclosed host cells are not limited to just these two types of cells, however, and may be any cell type described herein. For example, the cells that can be used include, without limitation, epithelial cells, endothelial cells, keratinocytes, fibroblasts, muscle cells, hepatocytes; blood cells such as T lymphocytes, B lymphocytes, monocytes, macrophages, neutrophils, eosinophils, megakaryocytes, or granulocytes, various stem or progenitor cells, such as hematopoietic stem or progenitor cells (e.g., as obtained from bone marrow), umbilical cord blood, peripheral blood, fetal liver, etc., and tumor cells (e.g., human tumor cells). The choice of cell type can be determined by one of skill in the art. In embodiments, the cells are irradiated.
Further provided by the present invention is a population of cells comprising at least one biological cell (e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) or host cell described herein. The population of cells can be a heterogeneous population comprising the host cell comprising any of the lentiviruses described, in addition to at least one other cell. Alternatively, the population of cells can be a substantially homogeneous population, in which the population comprises mainly host cells (e.g., consisting essentially of) comprising the lentivirus. The population also can be a clonal population of cells, in which all cells of the population are clones of a single host cell comprising the lentivirus, such that all cells of the population comprise the lentivirus. In one embodiment of the disclosure, the population of cells is a clonal population comprising host cells comprising the lentivirus as described herein. In illustrative aspects, the cell population of the present disclosure is one wherein at least 50% of the cells are host cells as described herein. In illustrative aspects, the cell population of the present disclosure is one wherein at least 60%, at least 70%, at least 80% or at least 90% or more of the cells are host cells as described herein.
In embodiments, the biological cell is selected from AD-100, HEK293, and NIH 3T3. In embodiments, the biological cell is a human tumor cell. In embodiments, the biological cell is an irradiated or live and attenuated human tumor cell. In embodiments, the human tumor cell is a cell from an established NSCLC, bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
In embodiments, there is provided a biological cell comprising a first recombinant protein having an amino acid sequence of at least 95% sequence identity with SEQ ID NO: 2 and a second recombinant protein having an amino acid sequence of at least 95% sequence identity with the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 42, the amino acid sequence of SEQ ID NO: 41 , the amino acid sequence of SEQ ID NO: 46, the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 41 , the amino acid sequence of SEQ ID NO: 43, the amino acid sequence of SEQ ID NO: 45, the amino acid sequence of SEQ ID NO: 42, or an antigenic fragment of any of the foregoing. In embodiments, the first recombinant protein has at least 97% sequence identity with SEQ ID NO: 2 and the second recombinant protein having an amino acid sequence of at least 97% sequence identity with the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 42, the amino acid sequence of SEQ ID NO: 43, or the amino acid sequence of SEQ ID NO: 46, or an antigenic fragment of any of the foregoing. In embodiments, the first recombinant protein has at least 98% sequence identity with SEQ ID NO: 2 and the second recombinant protein having an amino acid sequence of at least 98% sequence identity with the amino acid sequence of SEQ ID NO: 39, the amino acid sequence of SEQ ID NO: 42, the amino acid sequence of SEQ ID NO: 41 , the amino acid sequence of SEQ ID NO: 46, the amino acid sequence of SEQ ID NO: 5, the amino acid sequence of SEQ ID NO: 7, the amino acid sequence of SEQ ID NO: 9, the amino acid sequence of SEQ ID NO: 11 , the amino acid sequence of SEQ ID NO: 13, the amino acid sequence of SEQ ID NO: 27, the amino acid sequence of SEQ ID NO: 13, the amino acid sequence of SEQ ID NO: 15, the amino acid sequence of SEQ ID NO: 17, the amino acid sequence of SEQ ID NO: 19, or the amino acid sequence of SEQ ID NO: 21 , or an antigenic fragment of any of the foregoing. In any of the embodiments described herein, or combination of the embodiments, SEQ ID NO: 2 can lack the terminal KDEL (SEQ ID NO: 3) sequence.
In embodiments, there are provided at least two biological cells, the first biological cell comprising a lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, the second biological cell comprising a lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject; and/or the third biological cell comprising a lentivirus comprising a nucleic acid encoding a protein, or an antigenic portion thereof.
Cellular Therapies
In embodiments, the disclosure relates to a cellular therapy (e.g., a therapeutic biological cell, and/or a lentivirus as disclosed herein) comprising one or more secretable fusion proteins, T cell costimulatory fusion proteins, and/or fragments thereof, which can stimulate immune responses in a subject.
Without being bound to a particular theory, the methods of the present disclosure advantageously rely on the chaperone function of the secreted fusion protein. The fusion protein chaperones the one or more proteins or antigen portions thereof, which are efficiently taken up by activated antigen presenting cells (APCs). The APCs act to cross-present the proteins or antigen portions thereof via MHC I to CD8+ CTLs, whereupon an avid, antigen specific, cytotoxic CD8+ T cell response is stimulated. Without being bound to a particular theory, the proteins of the present disclosure, alone or in combination, are advantageously capable of initiating both an innate, humoral immune response (including, e.g., activation of APCs, pro-inflammatory cytokine release, activation of NK cells), and an adaptive immune response (including, e.g., priming, activation and proliferation of antigen specific CTLs). Such dual-activation leads to successful clearance of the antigen/pathogen. Accordingly, in embodiments, the present disclosure provides a method of administering to the subject the lentivirus as disclosed herein, or a population of cells transfected with the lentivirus.
In embodiments, the present disclosure provides a method of generating a cellular therapy that is suitable for treating or preventing a disease. In embodiments, the cellular therapy generated by the methods of the disclosure prevent, alleviate, and/or treat one or more symptoms associated with a disease. For example, symptoms that may be treated include, but are not limited to fever, cough {e.g., dry cough), shortness of breath and other breathing difficulties, fatigue, diarrhea, upper respiratory symptoms {e.g. sneezing, runny nose, sore throat), and/or pneumonia.
In embodiments, the cellular therapy is suitable for eliciting innate immunity when administered to a subject. In embodiments, the cellular therapy is suitable for eliciting humoral immunity (i.e. antibody response) when administered to a subject. In embodiments, the cellular therapy is suitable for eliciting cell-mediated immunity (i.e. T-cell response) when administered to a subject. In embodiments, the subject is immunocompromised due to one or more factors, including, without limitation, a genetic predisposition, age, immune status, and disease. In embodiments, the cellular therapy stimulates, promotes, or increases one or more of a T-cell response, antibody response, and activation of innate immunity. In embodiments, the cellular therapy stimulates, promotes, or increases all three of the T-cell response, antibody response, and activation of innate immunity, thereby activating all three arms of the subject's immune system.
As mentioned above, In embodiments, the chaperone protein is gp96. Accordingly, in embodiments in accordance with the present disclosure, an expression vector system or a population of cells transfected with the expression vector system is designed to use gp96 so as to trigger mucosal immunity by activating both B and T cell responses at the point of pathogen entry. The gp96-based cellular therapy effectively presents multiple antigens and activates the immune system thereby. The gp96-based cellular therapy utilizes natural and adaptive immune process to induce long- lasting memory responses against a disease. In embodiments, the gp96-based cellular therapy, compositions, and biological cells in accordance with the present disclosure activate all three of innate immune response, cellular immune response (i.e. T cell response), and humoral immune response (i.e. antibody response).
In embodiments, the present vaccine activates innate immunity via Toll-Like Receptor (TLRs), as, without wishing to be bound by the theory, gp96 activates Toll-Like Receptor 4/2 (TLR4 and TLR2) on macrophages and dendritic cells.
Furthermore, the present cellular therapy, adapted to present multiple antigens, in accordance with embodiments of the present disclosure, stimulates, promotes, or increases a cellular immune response via CD4 and CD8 T cells, in addition to the humoral immune response, via neutralizing IgG antibody.
In embodiments, the present cellular therapy is suitable for increasing the subject's T-cell response as compared to the T-cell response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's antibody response as compared to the antibody response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's innate immune response as compared to the innate immune response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's T-cell response, antibody response, and innate immune response as compared to the T-cell response, antibody response, and innate immune responses of a subject that was not administered the cellular therapy.
In embodiments, the cellular therapy is suitable for increasing the subject's innate immune response as compared to the innate immune response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's adaptive immune response as compared to the adaptive immune response of a subject that was not administered the cellular therapy. In embodiments, the cellular therapy is suitable for increasing the subject's innate immune response and adaptive immune response as compared to the innate and adaptive immune responses of a subject that was not administered the cellular therapy. In embodiments, methods and compositions of the present disclosure are for improving and/or increasing vaccine efficacy in a patient and include maintaining and/or increasing the patient's T cell populations (e.g., CD4+ and/or CD8+ T cell populations). In embodiments, methods and compositions of the present disclosure are for improving and/or increasing vaccine efficacy in a patient and include maintaining and/or increasing the patient's antigen-specific antibody titers (e.g., IgG, IgM and IgA). In embodiments, methods of the present disclosure provide for mitigation of age-related immunosenescence as measured by an increase or restoration of a patient's antigen-specific antibody titers (e.g., IgG, IgM and IgA).
In embodiments, the present cellular therapy, and methods for use thereof, are suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell populations of a subject that was not administered the present cellular therapy. In embodiments, the subject's T cells, including T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells, are increased and/or restored as compared to the T cell populations of a subject that was not administered the cellular therapy.
In embodiments, compositions are provided that includes a cellular therapy in accordance with embodiments of the present disclosure, and at least one other cellular therapy. In embodiments, the present cellular therapy can be administered alone or in combination with at least one other cellular therapy.
In embodiments, the method is suitable for increasing and/or restoring the subject's T cell population(s) as compared to the T cell population(s) of a subject that was administered another cellular therapy. In embodiments, the subject's T cells, including T cells selected from one or more of CD4+ effector T cells, CD8+ effector T cells, CD4+ memory T cells, CD8+ memory T cells, CD4+ central memory T cells, CD8+ central memory T cells, natural killer T cells, CD4+ helper cells, and CD8+ cytotoxic cells, are increased and/or restored as compared to the T cell populations of a subject that was administered another cellular therapy.
In embodiments, the subject's CD4+ helper cells population(s) are increased and/or restored as compared to the CD4+ helper cells population(s) of a subject that was not administered the present cellular therapy. In embodiments, without wishing to be bound by the theory, OX40L co-stimulation expands CD4 helper T cells that promote B-cell differentiation and IgG/lgA antibody class switching.
More specifically, In embodiments, the present disclosure provides compositions and methods for improving and/or increasing cellular therapy efficacy in a patient, as measured by an increase and/or restoration of the patient's T cell subsets. In embodiments, the T cells are T helper cells (e.g, Th cells). In embodiments, T helper cells secrete cytokines that attract one or more of macrophages, neutrophils, other lymphocytes, and other cytokines to further direct these cells. In embodiments, CD4+ T helper cells are one of several subsets, including, Th1 , Th2, Th17, Th9, and Tfh, with each subset having a different function. In embodiments, T cells are cytotoxic cells that optionally produce IL-2 and IFNy cytokines. In embodiments, these T cells are cytotoxic CD8+ T cells (also known as Tc cells or T-killer cells).
In embodiments, memory T cells elicited by the compositions and methods of the present disclosure are long-lived and can expand to large numbers of effector T cells when re-exposed to their cognate antigen. For example, the memory T cells elicited by methods of the present disclosure can persist in a subject for at least about 1 year, or at least about 2 years, or at least about 3 years, or at least about 4 years, or at least about 5 years, or at least about 6 years, or at least about 7 years, or at least about 8 years, or at least about 9 years, or at least about 10 years, or at least about 15 years, or at least about 20 years, or at least about 30 years, or at least about 40 years, or at least about 50 years, or at least about 60 years, or at least about 70 years, or at least about 80 years. In embodiments, memory T cells elicited by the compositions and methods of the present disclosure can last for the entire lifespan of a subject.
In embodiments, memory T cells provide a patient's immune system with memory against previously encountered pathogens. In embodiments, memory T cell populations include, but are not limited to, tissue-resident memory T (Trm) cells, stem memory TSCM cells, and virtual memory T cells. In embodiments, memory T cells are classified as CD4+ or CD8+ and express CD45RO. In embodiments, memory T cells are further differentiated into various subsets. For example, In embodiments, memory T cell subsets include: Central memory T cells (TCM cells), which can express CD45RO, C-C chemokine receptor type 7 (CCR7), L-selectin (CD62L), and CD44; Effector memory T cells (TEM cells and T EMRA cells), which express CD45RO and CD44 but lack expression of CCR7 and CD62L; Tissue resident memory T cells (TRM), which is associated with the integrin oep7; and Virtual memory T cells.
As used herein, the term "treat,” as well as words related thereto, do not necessarily imply 100% or complete treatment. Rather, there are varying degrees of treatment of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the methods of treating an infection of the present disclosure can provide any amount or any level of treatment. Furthermore, the treatment provided by the method of the present disclosure may include treatment of one or more conditions or symptoms or signs of the infection, being treated. Also, the treatment provided by the methods of the present disclosure may encompass slowing the progression of the infection. For example, the methods can treat the infection by virtue of eliciting an immune response against the infection, stimulating or activating CD8+ T cells specific for the infection, to proliferate, and the like.
As used herein, the term "prevent” and words stemming therefrom encompasses inhibiting or otherwise blocking an infection. As used herein, the term "inhibit” and words stemming therefrom may not be a 100% or complete inhibition or abrogation. Rather, there are varying degrees of inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the presently disclosed expression vector systems or host cells may inhibit infection to any amount or level. In illustrative embodiments, the inhibition provided by the methods of the present disclosure is at least or about a 10% inhibition (e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition).
The present cellular therapy and biological cells (e.g., therapeutic biological cells capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) may be administered to a subject by any route considered appropriate by a medical practitioner. Illustrative routes of administration include, for example: oral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, by electroporation, or topically. Administration can be local or systemic.
In embodiments, the cellular therapy or the biological cell (e.g., a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) can be administered to a subject one or more times (e.g., once, twice, two to four times, three to five times, five to eight times, six to ten times, eight to 12 times, or more than 12 times). A cellular therapy or biological cell as provided herein can be administered one or more times per day, one or more times per week, every other week, one or more times per month, once every two to three months, once every three to six months, or once every six to 12 months. A cellular therapy or biological cell can be administered over any suitable period of time, such as a period from about 1 day to about 12 months. In embodiments, for example, the period of administration can be from about 1 day to 90 days; from about 1 day to 60 days; from about 1 day to 30 days; from about 1 day to 20 days; from about 1 day to 10 days; from about 1 day to 7 days. In embodiments, the period of administration can be from about 1 week to 50 weeks; from about 1 week to 50 weeks; from about 1 week to 40 weeks; from about 1 week to 30 weeks; from about 1 week to 24 weeks; from about 1 week to 20 weeks; from about 1 week to 16 weeks; from about 1 week to 12 weeks; from about 1 week to 8 weeks; from about 1 week to 4 weeks; from about 1 week to 3 weeks; from about 1 week to 2 weeks; from about 2 weeks to 3 weeks; from about 2 weeks to 4 weeks; from about 2 weeks to 6 weeks; from about 2 weeks to 8 weeks; from about 3 weeks to 8 weeks; from about 3 weeks to 12 weeks; or from about 4 weeks to 20 weeks.
To establish if a subject is suited for the present treatments and/or to evaluate if the present methods are beneficial, in embodiments, techniques are used for detecting an infection in patient samples. For example, In embodiments, RT reverse transcription PCR (RT-PCR) techniques can be used to detect an infection in a patient sample.
In embodiments, the cellular therapy of the present disclosure is co-administered in conjunction with additional therapeutic agent(s), including vaccines. Co-administration can be simultaneous or sequential. In embodiments, the additional therapeutic agent is an agent that is used to provide relief to symptoms of infections. Such agents include remdesivir; favipiravir; galidesivir; prezcobix; lopinavir and/or ritonavir and/or arbidol; mRNA-1273; MSCs-derived exosomes; lopinavir/ ritonavir and/or ribavirin and/or I FN-beta; xiyanping; anti-VEGF-A (e.g. Bevacizumab); fingolimod; carrimycin; hydroxychloroquine; darunavir and cobicistat; methylprednisolone; brilacidin; leronlimab (PRO 140); and thalidomide. In embodiments, the additional therapeutic agent is chloroquine, including chloroquine phosphate. In an embodiment, the additional therapeutic agent is a composition comprising one or more HIV drugs. In embodiments, the composition comprises a combination of one or more of lopinavir and/or ritonavir and/or arbidol.
In embodiments, the additional therapeutic agent comprises one or more vaccines. In embodiments, the additional therapeutic agent comprises one or more vaccines
In embodiments, the cellular therapy in accordance with embodiments of the present disclosure, which employs gp- 96- (e.g., without limitation, a gp96-based cellular therapy), may be delivered alone (e.g., as a standalone cellular therapy) or in combination with other cellular therapies that drive humoral immunity, to provide an added layer of cellular immunity. The present cellular therapy can be administered in combination with one or more other cellular therapies, e.g., without limitation, flu vaccines, and other vaccines. In a combination approach, the vaccine in accordance with embodiments of the present disclosure, in combination with other vaccines (including conventional vaccines), induces effective and durable immune responses.
In embodiments, a combination of the present cellular therapy and other cellular therapies may boost immunity in certain types of patients, including elderly patients, patents with comorbidities, and patients with compromised immune system. The present cellular therapy enhances effect of other vaccines by providing an added layer of T-cell immunity boost to generate an effective and long-term immune response.
Various v cellular therapies can be co-administered with the present cellular therapy. In embodiments, the present cellular therapy is administered in combination with a cellular therapy either simultaneously or sequentially. In embodiments, the cellular therapy is in the exploratory, preclinical, clinical, post-clinical, or approved stage. In embodiments, the cellular therapy comprises one or more of: a live attenuated virus, an inactivated virus, a nonreplicating viral vector, a replicating viral vector, a recombinant protein, a peptide, a virus-like particle, DNA, RNA, mRNA, another macromolecule, and a fragment thereof.
Subjects and/or Animals
In embodiments, the subject and/or animal is a mammal, e.g., a human, mouse, rat, guinea pig, dog, cat, horse, cow, pig, rabbit, sheep, or non-human primate, such as a monkey, chimpanzee, or baboon. In other embodiments, the subject and/or animal is a non-mammal, such, for example, a zebrafish. In embodiments, the subject and/or animal may comprise fluorescently-tagged cells (with e.g. GFP).
In embodiments, the subject and/or animal is a human. In embodiments, the human is a pediatric human. In other embodiments, the human is an adult human. In other embodiments, the human is a geriatric human. In other embodiments, the human may be referred to as a patient.
In certain embodiments, the human has an age in a range of from about 0 months to about 6 months old, from about 6 to about 12 months old, from about 6 to about 18 months old, from about 18 to about 36 months old, from about 1 to about 5 years old, from about 5 to about 10 years old, from about 10 to about 15 years old, from about 15 to about 20 years old, from about 20 to about 25 years old, from about 25 to about 30 years old, from about 30 to about 35 years old, from about 35 to about 40 years old, from about 40 to about 45 years old, from about 45 to about 50 years old, from about 50 to about 55 years old, from about 55 to about 60 years old, from about 60 to about 65 years old, from about 65 to about 70 years old, from about 70 to about 75 years old, from about 75 to about 80 years old, from about 80 to about 85 years old, from about 85 to about 90 years old, from about 90 to about 95 years old or from about 95 to about 100 years old.
In other embodiments, the subject is a non-human animal, and therefore the disclosure pertains to veterinary use. In a specific embodiment, the non-human animal is a household pet. In another specific embodiment, the non-human animal is a livestock animal.
Patient Selection
In embodiments, methods for selecting patients who can benefit from compositions and methods in accordance with embodiments of the present disclosure are provided. In embodiments, the cellular therapy, compositions, cells, and methods employ a cellular therapy (e.g, a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease) which can be, without limitation, a gp96/OX40L-lg cellular therapy, and which can activate robust T-cell immunity along with humoral immunity. It should be appreciated however that a gp96-based cellular therapy in accordance with embodiments of the present disclosure can have any other T cell costimulatory fusion protein, as the cellular therapy is not limited to the OX40L-I g T cell costimulatory fusion protein.
In embodiments, the present cellular therapy is useful for harnessing natural antigen presentation and T-cell activation pathways in, without limitations, elderly patients (e.g., patients over the age of 65), patients with comorbidities, and/or in patients with a compromised immune system. Accordingly, the patient can be selected for treatment in accordance with embodiments of the present disclosure based on one or more of that patient's age, the status of the patient's immune system, and based on whether or not the patient has a comorbidity. The comorbidity can be defined as the simultaneous presence of two or more chronic diseases or conditions in the patient.
Kits
Kits comprising a fused biological cell or a composition comprising any one of the foregoing of the present disclosure are also provided. In illustrative aspects, an exemplary kit of the disclosure comprises any composition described herein in a unit dosage form. In one embodiment, the unit dosage form is a container, such as a pre-filled syringe, which can be sterile, containing any agent described herein and a pharmaceutically acceptable carrier, diluent, excipient, or vehicle. In illustrative aspects, the kit comprises a sterile, GMP-grade unit dose of the cells. In illustrative aspects, a unit dose of cells comprises 105, 106, 107, 108, 109, 1010, 1011, 1012 1013, or more than 1015 cells made in accordance with embodiments of the present disclosure. In illustrative aspects, the unit dose of cells are packaged in an intravenous bag. In illustrative aspects, the unit dose of cells are provided in a cryogenic form. In illustrative aspects, the unit dose of cells are ready to use. In illustrative aspects, the unit dose of cells are provided in a tube, a flask, a dish, or like container.
Kits can simplify the administration of any agent described herein. The kit can further comprise a label or printed instructions instructing the use of any agent described herein. In an embodiment, the kit comprises a container containing an effective amount of a composition of the disclosure and an effective amount of another composition, such those described herein.
In illustrative aspects, the cells are cryopreserved. In illustrative aspects, the cells are not frozen.
Definitions
The following definitions are used in connection with embodiments of the present disclosure. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of skill in the art to which this invention belongs.
The use of the terms "a” and "an” and "the” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.
The term "about,” when used in connection with a referenced numeric indication, means the referenced numeric indication plus or minus up to 10% of that referenced numeric indication. For example, the language "about 50” covers the range of 45 to 55.
An "effective amount,” when used in connection with medical uses is an amount that is effective for providing a measurable treatment, prevention, or reduction in the rate of pathogenesis of a disease of interest.
As used herein, something is "decreased” if a read-out of activity and/or effect is reduced by a significant amount, such as by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, or more, up to and including at least about 100%, in the presence of an agent or stimulus relative to the absence of such modulation. As will be understood by one of ordinary skill in the art, In embodiments, activity is decreased and some downstream read-outs will decrease but others can increase.
Conversely, activity is "increased” if a read-out of activity and/or effect is increased by a significant amount, for example by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, or more, up to and including at least about 100% or more, at least about 2-fold, at least about 3-fold, at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, at least about 10-fold, at least about 50-fold, at least about 100-fold, in the presence of an agent or stimulus, relative to the absence of such agent or stimulus.
As referred to herein, all compositional percentages are by weight of the total composition, unless otherwise specified.
The terms "comprising,” "having,” "including,” and "containing” are to be construed as open-ended terms (i.e. , meaning "including, but not limited to,”) unless otherwise noted.
As used herein, the word "include,” and its variants, is intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that may also be useful in the compositions and methods of this technology. Similarly, the terms "can” and "may” and their variants are intended to be non-limiting, such that recitation that an embodiment can or may comprise certain elements or features does not exclude other embodiments of the present technology that do not contain those elements or features.
Although the open-ended term "comprising,” as a synonym of terms such as including, containing, or having, is used herein to describe and claim the invention, the present invention, or embodiments thereof, may alternatively be described using alternative terms such as "consisting of' or "consisting essentially of.”
As used herein, the words "preferred” and "preferably” refer to embodiments of the technology that afford certain benefits, under certain circumstances. However, other embodiments may also be preferred, under the same or other circumstances. Furthermore, the recitation of one or more preferred embodiments does not imply that other embodiments are not useful, and is not intended to exclude other embodiments from the scope of the technology.
The amount of compositions described herein needed for achieving a therapeutic effect may be determined empirically in accordance with conventional procedures for the particular purpose. Generally, for administering therapeutic agents (and/or additional agents) described herein) for therapeutic purposes, the therapeutic agents are given at a pharmacologically effective dose. A "pharmacologically effective amount,” "pharmacologically effective dose,” "therapeutically effective amount,” or "effective amount” refers to an amount sufficient to produce the desired physiological effect or amount capable of achieving the desired result, particularly for treating the disorder or disease. An effective amount as used herein would include an amount sufficient to, for example, delay the development of a symptom of the disorder or disease, alter the course of a symptom of the disorder or disease (e.g, slow the progression of a symptom of the disease), reduce or eliminate one or more symptoms or manifestations of the disorder or disease, and reverse a symptom of a disorder or disease. For example, administration of therapeutic agents to a patient suffering from cancer provides a therapeutic benefit not only when the underlying condition is eradicated or ameliorated, but also when the patient reports a decrease in the severity or duration of the symptoms associated with the disease, e.g., a decrease in tumor burden, a decrease in circulating tumor cells, an increase in progression free survival. Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized. Effective amounts, toxicity, and therapeutic efficacy can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to about 50% of the population) and the ED50 (the dose therapeutically effective in about 50% of the population). The dosage can vary depending upon the dosage form employed and the route of administration utilized. The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50. In embodiments, compositions and methods that exhibit large therapeutic indices are preferred. A therapeutically effective dose can be estimated initially from in vitro assays, including, for example, cell culture assays. Also, a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 as determined in cell culture, or in an appropriate animal model. Levels of the described compositions in plasma can be measured, for example, by high performance liquid chromatography. The effects of any particular dosage can be monitored by a suitable bioassay. The dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.
In certain embodiments, the effect will result in a quantifiable change of at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 70%, or at least about 90%. In embodiments, the effect will result in a quantifiable change of about 10%, about 20%, about 30%, about 50%, about 70%, or even about 90% or more. Therapeutic benefit also includes halting or slowing the progression of the underlying disease or disorder, regardless of whether improvement is realized.
In certain embodiments, a pharmacologically effective amount that will treat cancer will modulate the symptoms typically by at least about 10%, at least about 20%, at least about 30%, at least about 40%, or at least about 50%. In exemplary embodiments, such modulations will result in, for example, statistically significant and quantifiable changes in the numbers of cancerous cells.
Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range and each endpoint, unless otherwise indicated herein, and each separate value and endpoint is incorporated into the specification as if it were individually recited herein.
All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or illustrative language {e. , "such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.
This invention is further illustrated by the following non-limiting examples.
EXAMPLES
Example 1 The experiments of this example demonstrate methods for generating a cellular therapy using a lentivirus transduced with DNA encoding for gp96-lg or CX40L-lg, or both gp96-lg and OX40L-lg. In these experiments, stable expressing cells were transduced with a replication incompetent lentivirus vectors having DNA encoding for either gp96-lg or CX40L-lg, or both gp96-lg and OX40L-lg. As shown in FIG. 1, the expression levels of individual clones varied from low to high across cells transduced with gp96-lg or CX40L-lg, or both gp96-lg and OX40L-lg. In the far right bar graph of FIG. 1, individual stable expressing cells were transduced with either gp96-lg or CX40L-lg, and the individual clones were then subsequently fused together to engineer multiple target expressor clones. The results of these experiments show that the expression levels can be varied depending on the clone type (e.g., a single cell transduced with either gp96-lg or CX40L-lg, or both gp96-lg and CX40L-lg), and/or the sequential fusion combination (e.g., a single cell transduced with gp96-lg, and a single cell transduced with OX40L-I g, then fused together), and how these engineered clones can be used for a cellular therapy to treat oncology, infectious diseases, and/or autoimmune related diseases.
Example 2
FIG. 2 illustrates cell fusion expression profile of a fusion of a HEK293 cell and an HS112 cell (which is an AD-100 cell expressing gp96). In this example, the HEK293 cell expresses such CTAs as LIN28B, SRSF12, TEX15, TEX19, SYNGR4 and GAPDHS; and the HS112 cell expresses COX7B2, MAGEA6, and MAGEC1 CTAs, and gp96-lg. In embodiments, RPS18 and GAPDH, which are housekeeping genes (translation and metabolism, respectively), were used as endogenous control, to normalize the quantitative real-time PCR (qPCR) data.
CTAs RPS18 and GAPDH are expressed by both HEK293 cell and the HS112 cell. In this example, in the fusion, the HEK293 to HS112 cells ratio was 1 :10 (1 repetiion) and 9:10 (2 repetitions). The expression of genes in the HEK293 cell was tested via one repetition, and the expression of genes in the HS112 cell was tested via two repetitions.
FIG. 2 illustrates dCt for various genes - COX7B2, MAGEA6, gp-96lg, LIN28B, SRSF12, TEX15, TEX19, SYNGR4, MAGEC1 , and GAPDHS. For each gene, the expression is shown, in this order, in six bars - HEK293 (11/13), HS112 (5/19), HS112 (6/16/), Fusion 9: 10 (12/28), Fusion 9: 10 (12/30), and Fusion 1 :10 (1/14). FIG. 2 shows that, in the HEK293-HS112 fused cell, the expression (where a higher dCt means a lower expression) of LIN28B, SRSF12, and TEX15 CTAs shows significant activation as compared to the parent HS112 cell; and the expression of COX7B2, MAGEA6, and gp96-lg genes shows significant activation as compared to the parent HEK293 cell. Also, in the HEK293- HS112 fused cell, TEX19, SYNGR4, MAGEC1, GAPDHS CTAs genes unexpectedly did not display differential expression at a baseline.
Accordingly, FIG. 2 shows that CTAs of interest in the HEK293 cell line show strong activation after fusion with the HS112 cell line, without sacrificing the pre-existing HS112 CTA profile.
Example 3 Two successive rounds of transduction were performed in AD100 cells 24 hours apart. One million cells were seeded into 6-well dishes and allowed to adhere. 1 ml of fresh media was added for 24 hours, collected, and used to assess levels of the two factors produced by the clones (FIG. 3). Cells were transduced with lentivirus encoding human GP96- IgG (vGP96) or human OX40L-lgG (vOX40L) at an MOI of 100. 72 hours after the second round, the cells were passaged into a larger culture vessel and allowed to recover and expand for another 72 hours. After sufficient cell expansion, selection began using the respective antibiotic encoded by the lentivirus construct. After 7 days of selection, the remaining cells underwent single cell isolation by limiting dilution. Single-cell clones were further expanded up and screened for levels of either hGP96-lgG or hOX40L-lgG. Protein levels were measured by ELISA using media conditioned for 24 hours by 1 million cells. The highest expressing clone for hGP96-lgG and hOX40L was selected for cell fusion to generate a hybrid cell that expresses both factors.
Cellular fusion was achieved using the GenomeONETM -CF EX kit and utilized the viral HV J-envelope. The cell fusion was stochastic, but different antibiotic resistance markers allowed for selection ef fusion events containing DNA of both cells. The fusions were allowed to recover and expand for 72 hours followed by dual drug treatment to eliminate cells that do not contain both antibiotic resistance genes. After 7 days of selection, the remaining cells underwent another round of single cell isolation by limiting dilution. These clones were expanded up and screened for high level expression of both hGP96-lgG and hOX40L-lgG. These data highlight unprecedented levels of GP96-lg and OX40L-lg expression in the AD-100 cell line.
EQUIVALENTS
While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth and as follows in the scope of the appended claims.
Those skilled in the art will recognize, or be able to ascertain, using no more than routine experimentation, numerous equivalents to the specific embodiments described specifically herein. Such equivalents are intended to be encompassed in the scope of the following claims.
INCORPORATION BY REFERENCE
All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.
As used herein, all headings are simply for organization and are not intended to limit the disclosure in any manner. The content of any individual section may be equally applicable to all sections.

Claims

CLAIMS What is claimed is:
1 . A method of making a fused biological cell, comprising: obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein; obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein; and contacting the first biological cell and the second biological cell with a fusion agent, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
2. The method of claim 1 , wherein the secretable vaccine protein is a secretable gp96-lg fusion protein, which optionally lacks the gp96 KDEL sequence.
3. The method of claim 2, wherein the Ig tag in the gp96-lg fusion protein comprises the Fc region of human lgG1, lgG2, lgG3, lgG4, IgM, IgA, or IgE.
4. The method of claim 1, wherein the T cell costimulatory fusion protein is selected from OX40L-lg, ICOSL-lg, 4-1 BBL-lg, LAG3-lg, CD40L-lg, TL1 A-lg, CD70-lg, GITRL-lg, and CD28-lg.
5. The method of claim 1, wherein the T cell costimulatory fusion protein is selected from OX40L-lg or a portion thereof that binds specifically to 0X40, ICOSL-lg or a portion thereof that binds specifically to IGOS, 4-1 BBL-lg, or a portion thereof that binds specifically to 4-1 BBR, CD 40L-lg, or a portion thereof that binds specifically to CD40, CD70- Ig, or a portion thereof that binds specifically to CD27, TL1 A-lg or a portion thereof that binds specifically to TNFRSF25, or GITRL-lg or a portion thereof that binds specifically to GITR.
6. The method of claim 5, wherein the T cell costimulatory fusion protein is OX40L-lg or a portion thereof that binds specifically to 0X40.
7. The method of any one of claims 4 to 6, wherein the Ig tag in the T cell costimulatory fusion protein comprises the Fc region of human IgG 1 , 1 gG2, lgG3, 1 gG4, IgM, IgA, or IgE.
8. The method of any one of the above claims, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to the patient.
9. The method of claim 1 , wherein the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of from about 1 : 1 to about 1 :5.
86
10. The method of claim 9, wherein the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1 : 1.
11. The method of claim 6, wherein the fused biological cell expresses the secretable vaccine protein and the T cell costimulatory fusion protein in a ratio of about 1 : 1.
12. The method of any one of claims 1 to 11 , wherein the fused biological cell further comprises a nucleotide sequence encoding one or more disease antigens.
13. The method of claim 12, comprising: obtaining a third biological cell comprising a nucleotide sequence encoding the one or more disease antigens; wherein contacting the first biological cell and the second biological cell with a fusion agent further comprises contacting the third biological cell with the fusion agent, to result in the fused biological cell being created such that the fused biological cell comprises a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
14. The method of any one of claims 1 to 11 , further comprising: obtaining a third biological cell comprising a nucleotide sequence encoding one or more disease antigens; and contacting the third biological cell with the fusion agent, to result in the fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein, a nucleotide sequence encoding the T cell costimulatory fusion protein, and a nucleotide sequence encoding the one or more disease antigens.
15. The method of claim 13 or claim 14, wherein one or more of the first, second, and third biological cells is a human tumor cell.
16. The method of claim 15, wherein the human tumor cell is a cell from an established non-small cell lung cancer (NSCLC), bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
17. The method of claim 16, wherein the human tumor cell is a cell from an established NSCLC cell line.
18. The method of any one of claims 12 to 17, wherein the third biological cell is a trophoblast.
87
19. The method of claim 13 or claim 14, wherein one or more of the first, second, and third biological cells is a non-tumor cell.
20. The method of claim 19, wherein the non-tumor cell is a fibroblast or a fibroblast-like cell.
21. The method of any one of claims 1 to 20, wherein the fused biological cell is immortalized.
22. The method of any one of claims 1 to 21, wherein at least one of the first, second, and third biological cells is immortalized.
23. The method of any one of the above claims, wherein the nucleotide sequence is a mammalian expression vector.
24. The method of claim 23, wherein the expression vector comprises DNA.
25. The method of claim 23, wherein the expression vector comprises RNA.
26. The method of claim 23, wherein the expression vector is incorporated into a virus or virus-like particle.
27. The method of any one of claims 1 to 26, wherein the fusion agent comprises a protein from Paramyxoviridae
Genus paramyxovirus.
28. The method of claim 27, wherein the protein is inactivated hemagglutinating virus of Japan envelope (HVJ- E).
29. The method of any one of claims 1 to 26, wherein the fusion agent comprises a poly(ethyleneglycol) (PEG) moiety or derivatives thereof.
30. The method of claim 29, wherein the PEG moiety comprises PEG-1500.
31. The method of any one of claims 12 to 18 or 21 to 30, wherein one or more of the first biological cell, the second biological cell, and the third biological cell comprise at least one tumor antigen.
32. The method of claim 31, wherein the tumor antigen is a cancer testis (CT) antigen.
33. The method of any one of claims 1 to 18 or 21 to 30, wherein the fused biological cell expresses one or more cancer testis (CT) antigens.
34. The method of any one of claims 1 to 18 or 21 to 30, the fused biological cell is enriched for genes encoding one or more cancer testis (CT) antigens.
88
35. The method of any one of claims 12 to 34, wherein the one or more disease antigens comprise one or more infectious disease antigens.
36. The method of claim 35, wherein the one or more infectious disease antigens comprise an antigenic fragment of a betacoronavirus protein or an alphacoronavirus protein, optionally wherein the betacoronavirus protein is selected from a SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1 , and HCoV-OC43 protein, or the alphacoronavirus protein is selected from a HCoV-NL63 and HCoV-229E protein.
37. The method of claim 36, wherein a presence of the one or more infectious disease antigens comprise an antigenic fragment of the betacoronavirus protein.
38. The method of claim 37, wherein the betacoronavirus protein is a SARS-CoV-2 protein.
39. The method of claim 38, wherein the SARS-CoV-2 protein is selected from spike surface glycoprotein, membrane glycoprotein M, envelope protein E, and nucleocapsid phosphoprotein N.
40. A method of treating cancer comprising administering to a patient in need thereof the fused biological cell generated using the method of any one of claims 1 to 34 or a pharmaceutical composition comprising the fused biological cell generated using the method of any one of claims 1 to 34.
41. The method of claim 40, wherein the cancer comprises an adrenal cancer, a biliary track cancer, a bladder cancer, a bone/bone marrow cancer, a brain cancer, a breast cancer, a cervical cancer, a colorectal cancer, a cancer of the esophagus, a gastric cancer, a head/neck cancer, a hepatobiliary cancer, a kidney cancer, a liver cancer, a lung cancer, an ovarian cancer, a pancreatic cancer, a pelvis cancer, a pleura cancer, a prostate cancer, a renal cancer, a skin cancer, a stomach cancer, a testis cancer, a thymus cancer, a thyroid cancer, a uterine cancer, a lymphoma, a melanoma, a multiple myeloma, or a leukemia.
42. A method of treating or preventing an infectious disease comprising administering to a patient in need thereof the fused biological cell generated using the method of any one of claims 1 to 39 or a pharmaceutical composition comprising the fused biological cell generated using the method of any one of claims 1 to 39.
43. The method of claim 42, wherein the infectious disease comprises a disease comprising a viral infection, a parasitic infection, or a bacterial infection.
44. The method of claim 43, wherein the viral infection is caused by a virus of family Flaviviridae, a virus of family Picornaviridae, a virus of family Orthomyxoviridae, a virus of family Coronaviridae, a virus of family Retroviridae, a virus of family Paramyxoviridae, a virus of family Bunyaviridae, or a virus of family Reoviridae.
89
45. The method of claim 44, wherein the virus of family Coronaviridae comprises a betacoronavirus or an alphacoronavirus, optionally wherein the betacoronavirus is selected from SARS-CoV-2, SARS-CoV, MERS-CoV, HCoV-HKU1, and HCoV-OC43, or the alphacoronavirus is selected from a HCoV-NL63 and HCoV-229E
46. The method of claim 45, wherein the infectious disease comprises a coronavirus infection 2019 (COVID-19).
47. A method of stimulating a patient antibody response, comprising administering to a patient in need thereof the fused biological cell generated using the method of any one of claims 1 to 39 or a pharmaceutical composition comprising the fused biological cell generated using the method of any one of claims 1 to 39.
48. A method of stimulating a patient T cell-driven cellular immune response, comprising administering to a patient in need thereof the fused biological cell generated using the method of any one of claims 1 to 39 or a pharmaceutical composition comprising the fused biological cell generated using the method of any one of claims 1 to 39.
49. A method of stimulating a patient B cell-driven cellular immune response, comprising administering to a patient in need thereof the fused biological cell generated using the method of any one of claims 1 to 39 or a pharmaceutical composition comprising the fused biological cell generated using the method of any one of claims 1 to 39.
50. A method of stimulating one or both a patient's antibody response and T cell-driven cellular immune response, comprising administering to a patient in need thereof the fused biological cell generated using the method of any one of claims 1 to 39 or a pharmaceutical composition comprising the fused biological cell generated using the method of any one of claims 1 to 39.
51. The method of claim 48 or claim 49, wherein the T cell-driven cellular immune response induces a CD8+ T cell response in the patient.
52. The method of claim 48 or claim 49, wherein the T cell-driven cellular immune response induces a CD4+ T cell response in the patient.
53. The method of any one of claims 40 to 52, further comprising administering to the patient an inhibitor of an immune checkpoint molecule.
54. The method of claim 53, wherein the immune checkpoint molecule is selected from PD-1, PD-L1, PD-L2, CTLA-4, IGOS, LAG3, 0X40, OX40L, and TIM3.
55. The method of claim 53 or 54, wherein the immune checkpoint molecule is PD-1.
90
56. The method of any one of the above claims, wherein a lentivirus vector encodes the nucleotide sequence encoding the secretable vaccine protein, and a lentivirus vector encodes the nucleotide sequence encoding the T cell costimulatory fusion protein.
57. The method of claim 56, further comprising merging together the first biological cell and the second biological cell.
58. A method for generating a cellular therapy, comprising:
(a) obtaining a lentivirus, the lentivirus comprising:
(I) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof and/or,
(II) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein is capable of enhancing activation of antigen-specific T cells when administered to a subject; and
(b) introducing into a biological cell the lentivirus of step (a).
59. A method for generating a cellular therapy, comprising:
(a) obtaining a first lentivirus, the lentivirus comprising a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and
(I) introducing the nucleic acid into a first biological cell;
(b) obtaining a second lentivirus, the lentivirus comprising a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein is capable of enhancing activation of antigen-specific T cells when administered to a subject, and
(I) introducing the nucleic acid into a second biological cell, and
(c) merging together the first biological cell and the second biological cell.
60. The method of claim 58 or 59, wherein the biological cell is a therapeutic biological cell capable of harnessing the subject's immune response for treating a cancer and/or an infectious disease.
61. The method of claim 60, wherein the lentivirus is produced using a third-generation lentiviral packaging system.
62. The method of claim 61 , wherein the lentiviral packaging system comprises a transfer plasmid, a packaging plasmid, and an envelope plasmid.
63. The method of claim 62, wherein the packaging plasmid comprises two plasmids.
91
64. The method of claim 62 or claim 63, wherein the packaging plasmid encodes a Gag and Pol gene.
65. The method of any one of claims 62-64, wherein the packaging plasmid encodes a Rev gene.
66. The method of any one of claims 62-65, wherein the envelope plasmid comprises a VSV-G envelope protein.
67. The method of any one of claims 60-64, wherein the transfer plasmid comprises:
(I) a nucleic acid encoding a secretable fusion protein comprising a chaperone protein and an immunoglobulin, or a fragment thereof, and/or
(II) a nucleic acid encoding a T cell costimulatory fusion protein, wherein the T cell costimulatory fusion protein enhances activation of antigen-specific T cells when administered to a subject.
68. The method of any one of claims 58-67, wherein the lentivirus is replication incompetent.
69. The method of any one of claims 58-68, wherein the lentivirus comprises a deletion in a long terminal repeat (LTR) sequence, rendering it self-inactivating (SIN) after integration.
70. The method of any one of claims 58-69, wherein the lentivirus is produced by contacting a cell with the third- generation lentiviral packaging system and isolating the lentivirus from the cell.
71. The method of any one of claims 58-70, wherein the lentivirus comprises one or more selection markers, optionally selected from puromycin, neomycin, zeocin, hygromycin.
72. The method of any one of claims 58-71 , wherein the chaperone protein is selected from the group consisting of: gp96, Hsp70, BiP, and Grp78.
73. The method of any one of claims 58-71 , wherein the chaperone protein comprises an amino acid sequence of any one of SEQ ID NOs: 1 , 2, 3, and 4, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
74. The method of any one of claims 58-71, wherein the chaperone protein is gp96 comprising the amino acid sequence of SEQ ID NO: 2.
75. The method of any one of claims 58-71 , wherein the chaperone protein of the secretable fusion protein is a secretable gp96-lg fusion protein which optionally lacks the gp96 KDEL sequence.
92
76. The method of any one of claims 58-71, wherein the secretable fusion protein comprises the amino acid sequence of SEQ ID NO: 6, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
77. The method of any one of claims 58-76, wherein the secretable fusion protein comprises an Fc fragment of an immunoglobulin.
78. The method of claim 77, wherein the Fc fragment comprises the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence having at least about 90%, or at least about 95%, or at least about 97%, or at least about 98%, or at least about 99% identity thereto.
79. The method of any one of claims 58-78, wherein the immunoglobulin is an lgG1 immunoglobulin.
80. The method of any one of claims 58-79, wherein the immunoglobulin comprises an Ig tag of the gp96-lg fusion protein comprising the Fc region of human lgG1, lgG2, lgG3, lgG4, IgM, IgA, or IgE.
81. The method of any one of claims 58-80, wherein the T cell costimulatory fusion protein is selected from OX40L-lg or a portion thereof that binds specifically to 0X40, ICOSL-lg or a portion thereof that binds specifically to IGOS, 4-1 BBL-lg, or a portion thereof that binds specifically to 4-1 BBR, CD40L-lg, or a portion thereof that binds specifically to CD40, CD70-lg, or a portion thereof that binds specifically to CD27, TL1 A-lg or a portion thereof that binds specifically to TNFRSF25, or GITRL-lg or a portion thereof that binds specifically to GITR.
82. The method of any one of claims 58-81, wherein the biological cell is selected from AD-100, HEK293, and NIH 3T3.
83. The method of any one of claims 58-81, wherein the biological cell is a human tumor cell.
84. The method of claim 83, wherein the biological cell is an irradiated or live and attenuated human tumor cell.
85. The method of claim 83 or claim 84, wherein the human tumor cell is a cell from an established NSCLC, bladder cancer, melanoma, ovarian cancer, renal cell carcinoma, prostate carcinoma, sarcoma, breast carcinoma, squamous cell carcinoma, head and neck carcinoma, hepatocellular carcinoma, pancreatic carcinoma, or colon carcinoma cell line.
86. The method of any one of claims 58-85, wherein the cellular therapy or therapeutic biological cell is suitable for eliciting humoral immunity when administered to a subject.
93
87. The method of any one of claims 56-84, wherein the cellular therapy or therapeutic biological cell is suitable for eliciting cell-mediated immunity when administered to a subject.
88. The method of any one of claims 56-85, wherein the cellular therapy or therapeutic biological cell is suitable for eliciting innate immunity when administered to a subject.
89. The method any one of claims 56-86, wherein the cellular therapy or therapeutic biological cell is suitable for administration in combination with one or more additional therapies.
90. A biological cell produced by the method of one or more of claims 1-56.
91 . A cellular therapy produced by the method of one or more of claims 57-89.
92. A method of making a fused biological cell, comprising, in order:
(a) obtaining a first biological cell comprising a nucleotide sequence encoding a secretable vaccine protein;
(b) obtaining a second biological cell comprising a nucleotide sequence encoding a T cell costimulatory fusion protein; and
(c) fusing the first biological cell and the second biological cell, to result in a fused biological cell comprising a nucleotide sequence encoding the secretable vaccine protein and a nucleotide sequence encoding the T cell costimulatory fusion protein.
93. The method of claim 92, wherein the secretable vaccine protein is gp96-lg and the T cell costimulatory fusion protein is OX40L-lg.
94. The method of claim 93, wherein the method results in greater expression of gp96-lg and/or OX40L-lg, as compared to an unfused cell that has been transduced with a nucleic acids encoding gp96-lg and OX40L-lg.
PCT/US2022/012136 2021-01-13 2022-01-12 Cell-fusion based immune agents WO2022155212A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163136842P 2021-01-13 2021-01-13
US63/136,842 2021-01-13
US202163222503P 2021-07-16 2021-07-16
US63/222,503 2021-07-16

Publications (1)

Publication Number Publication Date
WO2022155212A1 true WO2022155212A1 (en) 2022-07-21

Family

ID=82448648

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/012136 WO2022155212A1 (en) 2021-01-13 2022-01-12 Cell-fusion based immune agents

Country Status (1)

Country Link
WO (1) WO2022155212A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200031901A1 (en) * 2017-04-04 2020-01-30 Heat Biologics, Inc. Intratumoral vaccination
WO2020072395A1 (en) * 2018-10-01 2020-04-09 Heat Biologics, Inc. Combination cell-based therapies
US20200330596A1 (en) * 2016-11-22 2020-10-22 Alloplex Biotherapeutics, Inc. Allogenic tumor cell vaccine

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200330596A1 (en) * 2016-11-22 2020-10-22 Alloplex Biotherapeutics, Inc. Allogenic tumor cell vaccine
US20200031901A1 (en) * 2017-04-04 2020-01-30 Heat Biologics, Inc. Intratumoral vaccination
WO2020072395A1 (en) * 2018-10-01 2020-04-09 Heat Biologics, Inc. Combination cell-based therapies

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
BROWNING, MJ: "Antigen presenting cell /tumor cell fusion vaccines for cancer immunotherapy", HUMAN VACCINES & IMMUNOTHERAPEUTICS, vol. 9, no. 7, 8 March 2013 (2013-03-08), pages 1545 - 1548, XP055279915, DOI: 10.4161/hv.24235 *
LIU WEN-LONG, ZOU MEI-ZHEN, LIU TAO, ZENG JIN-YUE, LI XUE, YU WU-YANG, LI CHU-XIN, YE JING-JIE, SONG WEN, FENG JUN, ZHANG XIAN-ZHE: "Cytomembrane nanovaccines show therapeutic effects by mimicking tumor cells and antigen presenting cells", NATURE COMMUNICATIONS, vol. 10, no. 1, 1 December 2019 (2019-12-01), XP055939119, DOI: 10.1038/s41467-019-11157-1 *
MILONE ET AL.: "Clinical use of lentiviral vectors", LEUKEMIA, vol. 32, 22 March 2018 (2018-03-22), pages 1529 - 1541, XP036541149, DOI: 10.1038/s41375-018-0106-0 *
ROSENBLATT ET AL.: "Vaccination with dendritic cell /tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma", BLOOD, vol. 117, no. 2, 13 January 2011 (2011-01-13), pages 393 - 402, XP055280630, DOI: 10.1182/blood-2010-04-277137 *

Similar Documents

Publication Publication Date Title
US20210000945A1 (en) Vector co-expressing vaccine and costimulatory molecules
JP6963051B2 (en) Compositions and methods for immunotherapy
US10988517B2 (en) Heterodimeric proteins for modulating gamma delta T cells
US20230303659A1 (en) Intratumoral vaccination
US11643447B2 (en) Heterodimeric proteins for modulating gamma delta T cells
US20210346486A1 (en) Combination cell-based therapies
WO2022155212A1 (en) Cell-fusion based immune agents
US20210100896A1 (en) Methods and compositions for stimulating the immune system
US20220298252A1 (en) Methods of treating cancer using tnfrsf25 antibodies
US20220177548A1 (en) Methods and Compositions for Treating Melanoma

Legal Events

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

Ref document number: 22739983

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22739983

Country of ref document: EP

Kind code of ref document: A1