Chikungunya virus (CHIKV) or Venezuelan equine encephalitis virus (VEEV) virus-like particles comprising heterologous antigens inserted into the envelope protein

Information

  • Patent Grant
  • 11345726
  • Patent Number
    11,345,726
  • Date Filed
    Tuesday, December 8, 2015
    8 years ago
  • Date Issued
    Tuesday, May 31, 2022
    a year ago
  • Inventors
  • Original Assignees
    • VLP Theranentics. Inc. (Wilmington, DE, US)
  • Examiners
    • Parkin; Jeffrey S
    Agents
    • Sughrue Mion, PLLC
Abstract
The present invention provides a particle comprising a polypeptide and at least one antigen, and a composition comprising thereof.
Description
TECHNICAL FIELD

The present invention relates to a particle comprising a polypeptide and at least one antigen, and a composition comprising thereof.


BACKGROUND

Virus-like particles (VLPs) are multiprotein structures that mimic the organization and conformation of authentic native viruses but lack the viral genome, potentially yielding safer and cheaper vaccine candidates. A handful of prophylactic VLP-based vaccines is currently commercialized worldwide: GlaxoSmithKline's Engerix® (hepatitis B virus) and Cervarix® (human papillomavirus), and Merck and Co., Inc.'s Recombivax HB® (hepatitis B virus) and Gardasil® (human papillomavirus) are some examples. Other VLP-based vaccine candidates are in clinical trials or undergoing preclinical evaluation, such as, influenza virus, parvovirus, Norwalk and various chimeric VLPs. Many others are still restricted to small-scale fundamental research, despite their success in preclinical tests. The implications of large-scale VLP production are discussed in the context of process control, monitorization and optimization. The main up- and down-stream technical challenges are identified and discussed accordingly. Successful VLP-based vaccine blockbusters are briefly presented concomitantly with the latest results from clinical trials and the recent developments in chimeric VLP-based technology for either therapeutic or prophylactic vaccination (Expert Rev. Vaccines 9(10), 1149-1176, 2010).


Chikungunya virus (CHIKV) has infected millions of people in Africa, Europe and Asia since this alphavirus reemerged from Kenya in 2004. The severity of the disease and the spread of this epidemic virus present a serious public health threat in the absence of vaccines or antiviral therapies. It is reported that a VLP vaccine for epidemic Chikungunya virus protects non-human primates against infection (Nat Med. 2010 March; 16(3): 334-338). US patent publication No. 2012/0003266 discloses a virus-like particle (VLP) comprising one or more Chikungunya virus structural polypeptides which is useful for formulating a vaccine or antigenic composition for Chikungunya that induces immunity to an infection or at least one symptom thereof. WO2012/106356 discloses modified alphavirus or flavivirus virus-like particles (VLPs) and methods for enhancing production of modified VLPs for use in the prevention or treatment of alphavirus and flavivirus-mediated diseases. (these cited references are herein incorporated by reference).


SUMMARY OF THE INVENTION

In the first aspect, the present invention provides a particle which is capable of being self-assembled, comprising a polypeptide and at least one antigen, wherein said polypeptide comprises at least one first attachment site and said at least one antigen comprises at least one second attachment site, and wherein said polypeptide and said antigen are linked through said at least one first and said at least one second attachment site.


In the second aspect, the present invention provides a nucleic acid molecule comprising a nucleotide sequence that encodes a particle provided in the first aspect of the present invention.


In the third aspect, the present invention provides a composition comprising the particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention.


In the fourth aspect, the present invention provides a method of producing an antibody, comprising contacting the particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention to a mammal.


In the fifth aspect, the present invention provides a method of immunomodulation, a method of treating an autoimmune disease, a method of inducing and/or enhancing immune response against an antigen in a mammal, and a method of treating cancer comprising administering the composition provided in the third aspect of the present invention to a mammal.


In sixth aspect, the present invention provides a method of passive immunization, comprising administering the antibody provided in the fourth aspect of the present invention to a mammal.


In seventh aspect, the present invention provides a method of presenting an antigen on macrophage, comprising contacting the particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention to a mammal.


In eighth aspect, the present invention provides a method for producing the particle provided in the first aspect of the present invention, comprising preparing a gene comprising a nucleotide sequence encoding said particle; culturing a cell which is transfected with said gene to express said particle; and recovering said particle.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows modification of TNF alpha sequence to be inserted into Venezuelan Equine Encephalitis virus (VEEV) structural polypeptide.



FIG. 2 shows results of Western Blot which indicates that TNF alpha conjugated VLP was expressed.



FIG. 3 shows VLP_CHI 512 vector.



FIG. 4 shows VLP_CHI 532 vector.



FIG. 5 shows VLP_CHI 520 vector.



FIG. 6 shows VLP_VEEV VLP 518 vector.



FIG. 7 shows VLP_VEEV VLP 519 vector.



FIG. 8 shows VLP_VEEV VLP 538 vector.



FIG. 9 shows detection of anti-TNF alpha antibodies induced by TNF alpha derived peptide-conjugated virus like particle.



FIG. 10 shows detection of anti-human CD20 antibodies induced by CD20 derived peptide-conjugated virus like particle.



FIG. 11 shows detection of anti-mouse CD20 antibodies induced by CD20 derived peptide-conjugated virus like particle.





DETAILED DESCRIPTION OF THE INVENTION

(1) A Particle Comprising a Polypeptide and at Least One Antigen


In the first aspect, the present invention provides a particle which is capable of being self-assembled, comprising a polypeptide and at least one antigen, wherein said polypeptide comprises at least one first attachment site and said at least one antigen comprises at least one second attachment site, and wherein said polypeptide and said antigen are linked through said at least one first and said at least one second attachment site.


As used herein, “a particle which is capable of being self-assembled” refers to a particle formed by at least one constituent which is spontaneously assembled. The constituent may be a polypeptide or non-peptide chemical compound. In one embodiment, “a particle which is capable of being self-assembled” may be a particle comprising or consisting of at least one polypeptide. The at least one polypeptide consists of one or more kinds of peptide. In one embodiment, said particle has a diameter of at least 10 nm, for example, at least 20 nm, preferably at least 50 nm. In one embodiment, molecular weight of said particle is from 100 kDa to 100,000 kDa, preferably from 400 kDa to 30,000 kDa.


A polypeptide used for the present invention is not limited as long as it is spontaneously assembled. The polypeptide may be a virus structural polypeptide. Thus, the particle provided by the present invention may be a virus like particle.


A virus structural polypeptide may be a naturally occurring viral polypeptide or modified polypeptide thereof. In one embodiment, the modified polypeptide has at least 70%, 75%, 80%, 85%, 90%, 95% or 98% amino acid sequence identity to a naturally occurring viral structural polypeptide including capsid and envelope protein. In one embodiment, the modified polypeptide is a mutant where at most 10% of the amino acids are deleted, substituted, and/or added to a naturally occurring viral structural polypeptide including capsid and envelope protein.


In one embodiment, virus structural polypeptide used for the present invention consists of or comprises capsid and/or envelope protein or fragment thereof. For example, an envelope protein comprises at least one selected from the group consisting of E3, E2, 6K and E1. Virus structural polypeptide used for the present invention may be derived from Alphavirus or Flavivirus. Thus, the particle provided by the present invention may be a virus like particle derived from Alphavirus or Flavivirus. Examples of Alphavirus and Flavivirus include, but not limited to, Aura virus, Babanki virus, Barmah Forest virus (BFV), Bebaru virus, Cabassou virus, Chikungunya virus (CHIKV), Eastern equine encephalitis virus (EEEV), Eilat virus, Everglades virus, Fort Morgan virus, Getah virus, Highlands J virus, Kyzylagach virus, Mayaro virus, Me Tri virus, Middelburg virus, Mosso das Pedras virus, Mucambo virus, Ndumu virus, O'nyong-nyong virus, Pixuna virus, Rio Negro virus, Ross River virus (RRV), Salmon pancreas disease virus, Semliki Forest virus, Sindbis virus, Southern elephant seal virus, Tonate virus, Trocara virus, Una virus, Venezuelan equine encephalitis virus (VEEV), Western equine encephalitis virus (WEEV), Whataroa virus, West Nile virus, dengue virus, tick-borne encephalitis virus and yellow fever virus.


As used herein, the term “antigen” refers to a molecule capable of being bound by an antibody or a T cell receptor (TCR) if presented by MHC molecules. The term “antigen”, as used herein, also encompasses T-cell epitopes. A T-cell epitope is recognized by a T-cell receptor in the context of a MHC class I, present on all cells of the body except erythrocytes, or class II, present on immune cells and in particular antigen presenting cells. This recognition event leads to activation of T-cells and subsequent effector mechanisms such as proliferation of the T-cells, cytokine secretion, perforin secretion etc. An antigen is additionally capable of being recognized by the immune system and/or being capable of inducing a humoral immune response and/or cellular immune response leading to the activation of B- and/or T-lymphocytes. This may, however, require that, at least in certain cases, the antigen contains or is linked to a TH cell epitope and is given in adjuvant. An antigen can have one or more epitopes (B- and T-epitopes). The specific reaction referred to above is meant to indicate that the antigen will preferably react, typically in a highly selective manner, with its corresponding antibody or TCR and not with the multitude of other antibodies or TCRs which may be evoked by other antigens. Antigens as used herein may also be mixtures of several individual antigens. Antigens, as used herein, include but are not limited to allergens, self antigens, haptens, cancer antigens (i.e. tumor antigens) and infectious disease antigens as well as small organic molecules such as drugs of abuse (like nicotine) and fragments and derivatives thereof. Furthermore, antigens used for the present invention can be peptides, proteins, domains, carbohydrates, alkaloids, lipids or small molecules such as, for example, steroid hormones and fragments and derivatives thereof, autoantibody and cytokine itself.


Examples of cytokines include, but are not limited to, interleukin (IL) including over 30 type such as IL-1α, IL-1β, IL-2, -3, -4, -5, -6, -7, -8, -9, -10, -11 to -37; interferon (IFN) such as IFN-α, IFN-β and IFN-γ; tumor necrosis factor (TNF) such as TNF-α and TNF-β; transforming growth factor (TGF) such as TGF-α and TGF-β; colony stimulating factor (CSF) such as granulocyte-colony-stimulating factor (G-CSF), granulocyte-macrophage-colony-stimulating factor (GM-CSF), macrophage-colony Stimulating factor (M-CSF), erythropoietin (EPO), stem cell factor (SCF) and monocyte chemotactic and activating factor (MCAF); growth factor (GF) such as epidermal growth factor (EGF), fibroblast growth factor (FGF), insulin like growth factor (IGF), nerve growth factor (NGF), Brain-derived neurotrophic factor (BDNF), platelet derived growth factor (PDGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), thrombopoietin (TPO), and bone morphogenic protein (BMP); and other polypeptide factors including LIF, kit ligand (KL), MPO (Myeloperoxidase) and CRP (C-reactive protein); COX (Cyclooxygenase) such as COX-1, COX-2 and COX-3, NOS (Nitric oxide synthase) such as NOS-1, NOS-2 and NOS-3; and so on.


Cytokines also includes chemokines which are cytokines that induce chemotaxis. There are two major classes of chemokines, CXC and CC. The CXC chemokines, such as neutrophil-activating protein-2 (NAP-2) and melanoma growth stimulatory activity protein (MGSA) are chemotactic primarily for neutrophils and T lymphocytes, whereas the CC chemokines, such as RANTES, Macrophage inflammatory protein (MIP) including MIP-1α and MIP-1β, keratinocyte-derived chemokine (KC), the monocyte chemotactic proteins (MCP-1, MCP-2, MCP-3, MCP-4, and MCP-5) and the eotaxins (-1 and -2) are chemotactic for, among other cell types, macrophages, T lymphocytes, eosinophils, neutrophils, dendritic cells, and basophils. There also exist the chemokines lymphotactin-1, lymphotactin-2 (both C chemokines), and fractalkine (a CX3C chemokine) that do not fall into either of the major chemokine subfamilies.


As used herein, the term “antigenic determinant” is meant to refer to that portion of an antigen that is specifically recognized by either B- or T-lymphocytes. B-lymphocytes respond to foreign antigenic determinants via antibody production, whereas T-lymphocytes are the mediator of cellular immunity. Thus, antigenic determinants or epitopes are those parts of an antigen that are recognized by antibodies, or in the context of an MHC, by T-cell receptors. An antigenic determinant contains one or more epitopes.


As used herein, the term “antibody” refers to molecules which are capable of binding an epitope or antigenic determinant. The term is meant to include whole antibodies and antigen-binding fragments thereof, including single-chain antibodies. Such antibodies include human antigen binding antibody fragments and include, but are not limited to, Fab, Fab′ and F(ab′)2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain. The antibodies can be from any animal origin including birds and mammals. Preferably, the antibodies are mammalian e.g. human, murine, rabbit, goat, guinea pig, camel, horse and the like, or other suitable animals e.g. chicken. As used herein, “human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries or from animals transgenic for one or more human immunoglobulins and that do not express endogenous immunoglobulins, as described, for example, in U.S. Pat. No. 5,939,598, the disclosure of which is incorporated herein by reference in its entirety.


Antigen may be a substance (e.g. protein) which is not derived from virus (e.g. Chikungunya virus, Venezuelan equine encephalitis virus).


In one embodiment, antigen which is used for the present invention is at least one target or a polypeptide therefrom as listed in Table 1.










TABLE 1





Target
Use







GD2
neuroblastoma


CA-125 (imitation)
ovarian cancer


CD41 (integrin alpha-IIb)
platelet aggregation inhibitor


TNF-α
rheumatoid arthritis etc.


EpCAM
prostate and breast cancer


TNF-α
sepsis


CD20
lymphoma


VEGFR2
cancer


IL-6
rheumatoid arthritis


CD52
CLL, CTCL


CEA
colorectal cancer


TAG-72
non-small cell lung carcinoma


HLA-DR
hematological cancers


CEA
gastrointestinal cancers


L-selectin (CD62L)
severely injured patients


IL-6 receptor
rheumatoid arthritis


Rhesus factor
hemolytic disease of the newborn


beta amyloid
Alzheimer's disease


CD25 (α chain of IL-2 receptor)
prevention of organ transplant rejections


phosphatidylserine
cancer, viral infections


CD22
non-Hodgkin's lymphoma


BAFF
non-Hodgkin lymphoma etc.


CD125
asthma


CCL11 (eotaxin-1)
severe allergic disorders


CEA-related antigen
inflammatory lesions and metastases


VEGF-A
metastatic cancer


fibrin II, beta chain
thromboembolism


CD44 v6
squamous cell carcinoma


CD19
cancer


CD30 (TNFRSF8)
hematologic cancers


IL-12, IL-23
psoriasis, rheumatoid arthritis, inflammatory bowel diseases, multiple sclerosis


IL-1
rheumatoid arthritis


mucin CanAg
colorectal cancer etc.


prostatic carcinoma cells
prostate cancer


EpCAM, CD3
ovarian cancer, malignant ascites, gastric cancer


TAG-72
tumor detection


CD4
prevention of organ transplant rejections, treatment of autoimmune diseases


TNF-α
Crohn's disease


EGFR
metastatic colorectal cancer and head and neck cancer


EpCAM
ovarian cancer and other solid tumors


IGF-1 receptor
solid tumors


CD4
rheumatoid arthritis


MUC1
pancreatic cancer


TRAIL-R2
cancer


Influenza A hemagglutinin
infectious disease/influenza A


CD40
hematologic cancers


CD25 (α chain of IL-2 receptor)
prevention of organ transplant rejections


RANKL
osteoporosis, bone metastases etc.


B-lymphoma cell
lymphoma


GD3 ganglioside
malignant melanoma


C5
peroxysmal nocturnal hemoglobinuria


endotoxin
sepsis caused by Gram-negative bacteria


EpCAM
colorectal carcinoma


LFA-1 (CD11a)
psoriasis (blocks T-cell migration)


Hsp90
invasive Candida infection


SLAMF7
multiple myeloma


CD22
cancer, SLE


ITGB2 (CD18)
heart attack, stroke, traumatic shock


HER2/neu. CD3
breast cancer etc.


integrin αvβ3
melanoma, prostate cancer, ovarian cancer etc.


hepatitis B surface antigen
hepatitis B


CD15
appendicitis


folate receptor 1
ovarian cancer


respiratory syncytial virus
respiratory syncytial virus infection


IL-22
rheumatoid arthritis, psoriasis


IGF-1 receptor
adrenocortical carcinoma, non-small cell lung carcinoma etc.


IFN-γ
Crohn's disease etc.


rabies virus glycoprotein
rabies (prophylaxis)


TGF-β
idiopathic pulmonary fibrosis, focal segmental glomerulosclerosis, cancer


CD80
B-cell lymphoma


beta amyloid
Alzheimer's disease


CD147 (basigin)
graft versus host disease


CD33
acute myelogenous leukemia


carbonic anhydrase 9 (CA-IX)
clear cell renal cell carcinoma


GPNMB
melanoma, breast cancer


TNF-α
rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis


CD23 (IgE receptor)
allergic asthma


CD4
HIV infection


CD20
non-Hodgkin's lymphoma


CA-125
ovarian cancer


cardiac myosin
cardiac imaging


TNF-α
rheumatoid arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis, Crohn's



disease, ulcerative colitis


CD51
solid tumors (prostate cancer, melanoma)


CD25 (α chain of IL-2 receptor)
graft versus host disease


CD22
cancer


CD152
melanoma


CD30 (TNFRSF8)
Hodgkin's lymphoma


CD4
chronic asthma


CEA
colorectal cancer


IL-13
asthma


NCA-90 (granulocyte antigen)
diagnostic agent


TGF beta 2
reduction of scarring after glaucoma surgery


TRAIL-R2
cancer


hepatitis B surface antigen
hepatitis B


CD33
cancer


CD56
cancer


CD40
multiple myeloma, non-Hodgkin's lymphoma, Hodgkin's lymphoma


CD23 (IgE receptor)
chronic lymphocytic leukemia


TRAIL-R1
cancer


EGFR
colorectal, lung and stomach cancer


IL-5
asthma and white blood cell diseases


TGF beta 1
systemic scleroderma


CD74
multiple myeloma and other hematological malignancies


CD3 ganglioside
small cell lung carcinoma


respiratory syncytial virus
respiratory syncytial virus (prevention)


CD3
prevention of organ transplant rejections


C242 antigen
colorectal cancer


5T4
non-small cell lung carcinoma, renal cell carcinoma


integrin α4
multiple sclerosis, Crohn's disease


endotoxin
sepsis


EGFR
non-small cell lung carcinoma


EGFR
squamous cell carcinoma, head and neck cancer, nasopharyngeal cancer, glioma


CD20
rheumatoid arthritis, lupus erythematosus etc.


LFA-1 (CD11a)
prevention of organ transplant rejections, immunological diseases


CD20
chronic lymphocytic leukemia etc.


PDGF-R α
cancer


IgE, Fc region
allergic asthma


EpCAM
cancer


CA-125
ovarian cancer


CD3
diabetes mellitus type 1


lipoteichoic acid
sepsis (Staphylococcus)


respiratory syncytial virus
respiratory syncytial virus (prevention)


EGFR
colorectal cancer


respiratory syncytial virus
respiratory syncytial virus (prevention)


EGFR
colorectal cancer



Pseudomonas aeruginosa


Pseudomonas aeruginosa infection



IL-4
asthma


MUC1
cancer


HER2/neu
cancer


C5
reduction of side effects of cardiac surgery


adenocarcinoma antigen
adenocarcinoma


CD4
Crohn's disease, multiple sclerosis


vimentin
brain cancer


CCR5
HIV infection


rabies virus glycoprotein
rabies (prophylaxis)


VEGFR2
solid tumors


VEGF-A
macular degeneration (wet form)


anthrax toxin, protective antigen
anthrax (prophylaxis and treatment)


cytomegalovirus glycoprotein B
cytomegalovirus infection


IL-5
inflammations of the airways, skin and gastrointestinal tract


HGF
solid tumors


CD20
lymphomas, leukemias, some autoimmune disorders


IGF-1 receptor
cancer


IFN-α
systemic lupus erythematosus


CD11, CD18
haemorrhagic shock etc.


CD154 (CD40L)
rheumatic diseases


TAG-72
cancer


cytomegalovirus
cytomegalovirus infection


FAP
cancer


IFN-α
SLE, dermatomyositis, polymyositis


CD2
psoriasis, graft-versus-host disease (prevention)


beta amyloid
Alzheimer's disease


sphingosine-1-phosphate
choroidal and retinal neovascularization


myostatin
muscular dystrophy


NCA-90 (granulocyte antigen)
osteomyelitis


alpha-fetoprotein
cancer


integrin αIIbβ3
percutaneous coronary intervention


IgE
allergic reaction


NGF
pain


CD19
cancer


clumping factor A

Staphylococcus aureus infection



tenascin C
cancer


CD3
diabetes mellitus type 1


CD28
chronic lymphocytic leukemia, rheumatoid arthritis


CTLA-4
cancer


TRAIL-R2
cancer


IL-13
Hodgkin's lymphoma


IL-6 receptor
rheumatoid arthritis


CD154 (CD40L)
rheumatoid arthritis, lupus nephritis etc.


CD20
follicular lymphoma


HER2/neu
breast cancer


CTLA-4
cancer


EpCAM
cancer


hepatitis B virus
chronic hepatitis B



Escherichia coli

diarrhoea caused by E. coli


IL-12, IL-23
multiple sclerosis, psoriasis, psoriatic arthritis


integrin α4β7
Crohn's disease, ulcerative colitis


CD20
non-Hodgkin's lymphoma


AOC3 (VAP-1)
inflammation


CD3
Crohn's disease, ulcerative colitis


integrin α5β1
solid tumors


tumor antigen CTAA16.88
colorectal tumors


EGFR
squamous cell carcinoma of the head and neck


CD4
rheumatoid arthritis, psoriasis, T-cell lymphoma


CD5
systemic lupus erythematosus, graft-versus-host disease









In one embodiment, antigen which is used for the present invention is at least one protein or a polypeptide therefrom selected from the group consisting of CTLA-4, PD-1, TIM-3, BILA, VISTA, LAG-3, CD28, OX40, GITR, CD137, CD27 and HVEM. CTLA-4, PD-1, TIM-3, BTLA, VISTA and LAG-3 are inhibitory receptors for T-cell stimulation, and CD28, OX40, GITR, CD137, CD27 and HVEM are activating receptors for T-cell stimulation (see Mellman et al., Nature 480, 480-489 (2011)).


The antigen used for the present invention can be modified polypeptide derived from a naturally occurring protein. The modified polypeptide may be a fragment of the naturally occurring protein. In one embodiment, the modified polypeptide has at least 70%, 75%, 80%, 85%, 90%, 95% or 98% amino acid sequence identity to a polypeptide derived from a naturally occurring protein. In one embodiment, the modified polypeptide derived is a mutant where at most 10% of the amino acids are deleted, substituted, and/or added based on a polypeptide derived from naturally occurring protein.


In the particle as provided by the present invention, a polypeptide and an antigen may be linked through at least one first attachment site which is present in the polypeptide and at least one second attachment site which is present in the antigen.


As used herein, each of “a first attachment site” and “a second attachment site” refers to a site where more than one substance is linked each other.


In one embodiment, the polypeptide and the antigen are directly fused. Alternatively, one or two linkers may intervene between N-terminal residue of the antigen and the polypeptide and/or between C-terminal residue of the antigen and the polypeptide.


The antigen or the polypeptide can be truncated and replaced by short linkers. In some embodiments, the antigen or the polypeptide include one or more peptide linkers. Typically, a linker consists of from 2 to 25 amino acids. Usually, it is from 2 to 15 amino acids in length, although in certain circumstances, it can be only one, such as a single glycine residue.


In one embodiment, a nucleic acid molecule, in which polynucleotide encoding the polypeptide is genetically fused with polynucleotide encoding the antigen, is expressed in a host cell so that the first attachment site and the second attachment site are linked through a peptide bond. In this case, the polypeptide and the antigen are linked through a peptide bond. Relating to this embodiment, the first attachment site and/or the second attachment site may be genetically modified from the original polypeptide or antigen. For example, the first attachment site is modified from the polypeptide so that through a linker peptide including SG, GS, SGG, GGS and SGSG, the polypeptide is conjugated with the antigen.


When the polypeptide are chemically conjugated with the antigen, the first attachment site and the second attachment site may be linked through a chemical cross-linker which is a chemical compound.


Examples of the cross-linker include, but are not limited to, SMPH, Sulfo-MBS, Sulfo-EMCS, Sulfo-GMBS, Sulfo-SIAB, Sulfo-SMPB, Sulfo-SMCC, SVSB, SIA and other cross-linkers available from the Pierce Chemical Company.


In one embodiment, the particle provided by the present invention comprises a polypeptide linked to an antigen, wherein spatial distance between the N-terminal residue and C-terminal residue of the antigen is 30 custom character or less when the distance is determined in a crystal of the antigen or a naturally occurring protein containing the antigen or modified protein therefrom.


The antigen used for the present invention can be designed by a person skilled in the art. For example, the antigen used for the present invention may be a naturally occurring protein or a fragment thereof. Alternatively, the antigen used for the present invention may be a protein modified from a naturally occurring protein or a fragment thereof. A person skilled in the art can design the antigen so that spatial distance between the N-terminal residue and C-terminal residue of the antigen is 30 custom character or less when the distance is determined in a crystal of the antigen or a naturally occurring protein containing the antigen or modified protein therefrom. For example, the antigen used for the particle provided by the present invention can be designed using a free software including PyMOL (e.g. PyMOL v0.99: www.pymol.org). In one embodiment, the spatial distance between the N-terminal residue and C-terminal residue of the antigen is 30 custom character (angstrom) or less, 20 custom character or less, or 10 custom character or less (e.g. from 5 custom character to 15 custom character, from 5 custom character to 12 custom character, from 5 custom character to 11 custom character, from 5 custom character to 10 custom character, from 5 custom character to 8 custom character, from 8 custom character to 15 custom character, from 8 custom character to 13 custom character, from 8 custom character to 12 custom character, from 8 custom character to 11 custom character, from 9 custom character to 12 custom character, from 9 custom character to 11 custom character, from 9 custom character to 10 custom character or from 10 custom character to 11 custom character).


Chikungunya Virus Like Particle or a Venezuelan Equine Encephalitis Virus Like Particle


In one embodiment, the present invention provides a Chikungunya virus like particle or a Venezuelan equine encephalitis virus like particle comprising a Chikungunya or Venezuelan equine encephalitis virus structural polypeptide and at least one antigen, wherein said Chikungunya virus structural polypeptide or said Venezuelan equine encephalitis virus structural polypeptide comprises at least one first attachment site and said at least one antigen comprises at least one second attachment site, and wherein said Chikungunya or Venezuelan equine encephalitis virus structural polypeptide and said at least one antigen are linked through said at least one first and said at least one second attachment site.


In one embodiment, a spatial distance between the N-terminal residue and C-terminal residue of the antigen may be 30 custom character or less; 25 custom character or less; 20 custom character or less; 15 custom character or less; 14 custom character or less; 13 custom character or less; 12 custom character or less; 11 custom character or less; 10 custom character or less; 9 custom character or less; or 8 custom character or less (e.g. from 5 custom character to 15 custom character, from 5 custom character to 12 custom character, from 5 custom character to 11 custom character, from 5 custom character to 10 custom character, from 5 custom character to 8 custom character, from 8 custom character to 15 custom character, from 8 custom character to 13 custom character, from 8 custom character to 12 custom character, from 8 custom character to 11 custom character, from 9 custom character to 12 custom character, from 9 custom character to 11 custom character, from 9 custom character to 11 custom character or from 10 custom character to 11 custom character) when the distance is determined in a crystal of the antigen or a naturally occurring protein containing the antigen or modified protein therefrom.


In one embodiment, the antigen is linked to the Chikungunya or Venezuelan equine encephalitis virus structural polypeptide by way of chemical cross-linking or as a fusion protein produced by way of genetic engineering.


A Chikungunya or Venezuelan equine encephalitis virus structural polypeptide used in the present invention may comprise a Chikungunya or Venezuelan equine encephalitis virus envelope protein and/or a capsid.


Examples of Chikungunya virus include, but are not limited to, strains of 37997 and LR2006 OPY-1.


Examples of Venezuelan equine encephalitis virus include, but are not limited to, TC-83.


Chikungunya or Venezuelan equine encephalitis virus structural polypeptide used in the present invention may naturally occurring virus structural polypeptide or modified polypeptide thereof. The modified polypeptide may be a fragment of the naturally occurring virus structural polypeptide. In one embodiment, the modified polypeptide has at least 70%, 75%, 80%, 85%, 90%, 95% or 98% amino acid sequence identity to a naturally occurring viral capsid and/or envelope protein. In one embodiment, the modified polypeptide is a mutant where at most 10% of the amino acids are deleted, substituted, and/or added based on a naturally occurring viral capsid and/or envelope protein. For example, K64A or K64N mutation may be introduced into a capsid of Venezuelan equine encephalitis virus structural polypeptide used in the present invention.


Chikungunya or Venezuelan equine encephalitis virus envelope protein may comprise at least one selected from the group consisting of E3, E2, 6K and E1.


Examples of Chikungunya virus structural polypeptide include, but are not limited to, E3-E2-6K-E1 of Chikungunya virus Strain 37997, Capsid-E3-E2-6K-E1 of Chikungunya virus Strain 37997, E3-E2-6K-E1 of Chikungunya virus Strain LR2006 OPY-1 and Capsid-E3-E2-6K-E1 of Chikungunya virus LR2006 OPY-1.


Examples of Venezuelan equine encephalitis virus structural polypeptide include, but are not limited to, E3-E2-6K-E1 of Venezuelan equine encephalitis virus Strain TC-83 and Capsid-E3-E2-6K-E1 of Venezuelan equine encephalitis virus Strain TC-83.


An exemplary Chikungunya virus structural polypeptide sequence is provided at Genbank Accession No. ABX40006.1, which is described below (SEQ ID No.:1):











MEFIPTQTFYNRRYQPRPWTPRPTIQVIRPRPR







PQRQAGQLAQLISAVNKLTMRAVPQQKPRRNR







KNKKQKQKQQAPQNNTNQKKQPPKKKPAQK







KKKPGRRERMCMKIENDCIFEVKHEGKVTGY







ACLVGDKVMKPAHVKGTIDNADLAKLAFKRSS







KYDLECAQIPVHMKSDASKFTHEKPEGYYNW







HHGAVQYSGGRFTIPTGAGKPGDSGRPIFDNK







GRVVAIVLGGANEGARTALSVVTWNKDIVTKIT







PEGAEEWSLAIPVMCLLANTTFPCSQPPCTPCC







YEKEPEETLRMLEDNVMRPGYYQLLQASLTCS







PHRQRRSTKDNFNVYKATRPYLAHCPDCGEG







HSCHSPVALERIRNEATDGTLKIQVSLQIGIKTD







DSHDWTKLRYMDNHMPADAERAGLFVRTSAP







CTITGTMGHFILARCPKGETLTVGFTDSRKISH







SCTHPFHHDPPVIGREKFHSRPQHGKELPCST







YVQSTAATTEEIEVHMPPDTPDRTLMSQQSGN







VKITVNGQTVRYKCNCGGSNEGLTTTDKVINN







CKVDQCHAAVTNHKKWQYNSPLVPRNAELGD







RKGKIHIPFPLANVTCRVPKARNPTVTYGKNQ







VIMLLYPDHPTLLSYRNMGEEPNYQEEWVMH







KKEVVLTVPTEGLEVTWGNNEPYKYWPQLST







NGTAHGHPHEIILYYYELYPTMTVVVVSVATFI







LLSMVGMAAGMCMCARRRCITPYELTPGATVP







FLLSLICCIRTAKAATYQEAAIYLWNEQQPLFW







LQALIPLAALIVLCNCLRLLPCCCKTLAFLAVM







SVGAHTVSAYEHVTVIPNTVGVPYKTLVNRPG







YSPMVLEMELLSVTLEPTLSLDYITCEYKTVIPS







PYVKCCGTAECKDKNLPDYSCKVFTGVYPFM







WGGAYCFCDAENTQLSEAHVEKSESCKTEFAS







AYRAHTASASAKLRVLYQGNNITVTAYANGDH







AVTVKDAKFIVGPMSSAWTPFDNKIVVYKGDV







YNMDYPPFGAGRPGQFGDIQSRTPESKDVYAN







TQLVLQRPAVGTVHVPYSQAPSGFKYWLKERG







ASLQHTAPFGCQIATNPVRAVNCAVGNMPISID







IPEAAFTRVVDAPSLTDMSCEVPACTHSSDFGG







VAIIKYAASKKGKCAVHSMTNAVTIREAEIEVE







GNSQLQISFSTALASAEFRVQVCSTQVHCAAEC







HPPKDHIVNYPASHTTLGVQDISATAMSWVQK







ITGGVGLVVAVAALILIVVLCVSFSRH






Another exemplary Chikungunya virus structural polypeptide sequence is provided at Genbank Accession No. ABX40011.1, which is described below (SEQ ID No.:2):











MEFIPTQTFYNRRYQPRPWAPRPTIQVIRPRPR







PQRQAGQLAQLISAVNKLTMRAVPQQKPRRN







RKNKKQRQKKQAPQNDPKQKKQPPQKKPAQ







KKKKPGRRERMCMKIENDCIFEVKHEGKVM







GYACLVGDKVMKPAHVKGTIDNADLAKLAFK







RSSKYDLECAQIPVHMKSDASKFTHEKPEGYY







NWHHGAVQYSGGRFTIPTGAGKPGDSGRPIF







DNKGRVVAIVLGGANEGARTALSVVTWNKDI







VTKITPEGAEEWSLALPVLCLLANTTFPCSQPP







CTPCCYEKEPESTLRMLEDNVMRPGYYQLLK







ASLTCSPHRQRRSTKDNFNVYKATRPYLAHCP







DCGEGHSCHSPIALERIRNEATDGTLKIQVSLQ







IGIKTDDSHDWTKLRYMDSHTPADAERAGLL







VRTSAPCTITGTMGHFILARCPKGETLTVGFT







DSRKISHTCTHPFHHEPPVIGRERFHSRPQHG







KELPCSTYVQSTAATAEEIEVHMPPDTPDRTL







MTQQSGNVKITVNGQTVRYKCNCGGSNEGLT







TTDKVINNCKIDQCHAAVTNHKNWQYNSPLV







PRNAELGDRKGKIHIPFPLANVTCRVPKARNP







TVTYGKNQVTMLLYPDHPTLLSYRNMGQEPN







YHEEWVTHKKEVTLTVPTEGLEVTWGNNEPY







KYWPQMSTNGTAHGHPHEIILYYYELYPTMT







VVIVSVASFVLLSMVGTAVGMCVCARRRCITP







YELTPGATVPFLLSLLCCVRTTKAATYYEAAA







YLWNEQQPLFWLQALIPLAALIVLCNCLKLLP







CCCKTLAFLAVMSIGAHTVSAYEHVTVIPNTV







GVPYKTLVNRPGYSPMVLEMELQSVTLEPTLS







LDYITCEYKTVIPSPYVKCCGTAECKDKSLPDY







SCKVFTGVYPFMWGGAYCFCDAENTQLSEAH







VEKSESCKTEFASAYRAHTASASAKLRVLYQG







NNITVAAYANGDHAVTVKDAKFVVGPMSSAW







TPFDNKIVVYKGDVYNMDYPPFGAGRPGQFG







DIQSRTPESKDVYANTQLVLQRPAAGTVHVPY







SQAPSGFKYWLKERGASLQHTAPFGCQIATNP







VRAVNCAVGNIPISIDIPDAAFTRVVDAPSVTD







MSCEVPACTHSSDFGGVAIIKYTASKKGKCAV







HSMTNAVTIREADVEVEGNSQLQISFSTALAS







AEFRVQVCSTQVHCAAACHPPKDHIVNYPAS







HTTLGVQDISTTAMSWVQKITGGVGLIVAVAA







LILIVVLCVSFSRH






An exemplary Venezuelan equine encephalitis virus structural protein is described below (SEQ ID No.:3):











MFPFQPMYPMQPMPYRNPFAAPRRPWFPRT







DPFLAMQVQELTRSMANLTFKQRRDAPPEGP







SAAKPKKEASQKQKGGGQGKKKKNQGKKK







AKTGPPNPKAQNGNKKKTNKKPGKRQRMV







MKLESDKTFPIMLEGKINGYACVVGGKLFRP







MHVEGKIDNDVLAALKTKKASKYDLEYADVP







QNMRADTFKYTHEKPQGYYSWHHGAVQYE







NGRFTVPKGVGAKGDSGRPILDNQGRVVAIV







LGGVNEGSRTALSVVMWNEKGVTVKYTPEN







CEQWSLVTTMCLLANVTFPCAQPPICYDRKP







AETLAMLSVNVDNPGYDELLEAAVKCPGRKR







RSTEELFNEYKLTRPYMARCIRCAVGSCHSPI







AIEAVKSDGHDGYVRLQTSSQYGLDSSGNLK







GRTMRYDMHGTIKEIPLHQVSLYTSRPCHIV







DGHGYFLLARCPAGDSITMEFKKDSVRHSCS







VPYEVKFNPVGRELYTHPPEHGVEQACQVYA







HDAQNRGAYVEMHLPGSEVDSSLVSLSGSSV







TVTPPDGTSALVECECGGTKISETINKTKQFS







QCTKKEQCRAYRLQNDKWVYNSDKLPKAAG







ATLKGKLHVPFLLADGKCTVPLAPEPMITFG







FRSVSLKLHPKNPTYLITRQLADEPHYTHELI







SEPAVRNFTVTEKGWEFVWGNHPPKRFWAQ







ETAPGNPHGLPHEVITHYYHRYPMSTILGLSI







CAAIATVSVAASTWLFCRSRVACLTPYRLTPN







ARIPFCLAVLCCARTARAETTWESLDHLWNN







NQQMFWIQLLIPLAALIVVTRLLRCVCCVVPF







LVMAGAAGAGAYEHATTMPSQAGISYNTIVN







RAGYAPLPISITPTKIKLIPTVNLEYVTCHYKT







GMDSPAIKCCGSQECTPTYRPDEQCKVFTGV







YPFMWGGAYCFCDTENTQVSKAYVMKSDDC







LADHAEAYKAHTASVQAFLNITVGEHSIVTTV







YVNGETPVNFNGVKITAGPLSTAWTPFDRKI







VQYAGEIYNYDFPEYGAGQPGAFGDIQSRTVS







SSDLYANTNLVLQRPKAGAIHVPYTQAPSGFE







QWKKDKAPSLKFTAPFGCEIYTNPIRAENCA







VGSIPLAFDIPDALFTRVSETPTLSAAECTLNE







CVYSSDFGGIATVKYSASKSGKCAVHVPSGTA







TLKEAAVELTEQGSATIHFSTANIHPEFRLQI







CTSYVTCKGDCHPPKDHIVTHPQYHAQTFTA







AVSKTAWTWLTSLLGGSAVIIIIGLVLATIVAM







YVLTNQKHN






In one embodiment, a first attachment site comprises an amino group, preferably an amino group of a lysine residue. In one embodiment, the second attachment site comprises sulfhydryl group, preferably, a sulfhydryl group of a cysteine.


In one embodiment, a conjugation of more than two substances (e.g. antigen and Chikungunya or Venezuelan equine encephalitis virus structural polypeptide) through a first attachment site or a second attachment site is achieved using chemical cross linker. Examples of the cross-linker include, but are not limited to, SMPH, Sulfo-MBS, Sulfo-EMCS, Sulfo-GMBS, Sulfo-SIAB, Sulfo-SMPB, Sulfo-SMCC, SVSB, SIA and other cross-linkers available from the Pierce Chemical Company.


According to the present invention, a Chikungunya or Venezuelan equine encephalitis virus like particle comprising a Chikungunya or Venezuelan equine encephalitis virus structural polypeptide and an antigen, wherein said Chikungunya or Venezuelan equine encephalitis virus structural polypeptide and said antigen are expressed as a fusion protein can be provided.


In one embodiment, the antigen can be fused with any site of the Chikungunya or Venezuelan equine encephalitis virus structural polypeptide. For example, the antigen may be directly or indirectly linked to N- or C-terminal of the Chikungunya or Venezuelan equine encephalitis virus structural polypeptide (e.g. capsid, E3, E2, 6K or E1), or the antigen may be inserted into Chikungunya or Venezuelan equine encephalitis virus structural protein (e.g. capsid, E3, E2, 6K, or E1).


In one embodiment, at least one antigen is inserted into E2 of Chikungunya or Venezuelan equine encephalitis virus structural protein. For example, regarding Chikungunya virus structural protein, at least one antigen is inserted between residues 519 and 520 of SEQ ID Nos.1 or (i.e. between G at 519-position and Q at 520-position of SEQ ID Nos.1 or 2); between residues 530 and 531 of SEQ ID Nos.1 or 2 (i.e. between G at 530-position and S at 531-position of SEQ ID Nos.1 or 2); between residues 531 and 532 of SEQ ID Nos.1 or 2 (i.e. between S at 531-position and N at 532-position of SEQ ID Nos.1 or 2); between residues 529 and 530 of SEQ ID Nos.1 or 2 (i.e. between G at 529-position and G at 530-position of SEQ ID Nos.1 or 2); or between residues 510 and 511 of SEQ ID Nos.1 or 2 (i.e. between S at 510-position and G at 511-position of SEQ ID Nos.1 or 2); or between residues 511 and 512 of SEQ ID Nos.1 or 2 (i.e. between G at 511-position and N at 512-position of SEQ ID Nos.1 or 2); or between residues 509 and 510 of SEQ ID Nos.1 or 2 (i.e. between Q at 509-position and S at 510-position of SEQ ID Nos.1 or 2).


For example, regarding Venezuelan equine encephalitis virus structural protein, at least one antigen is inserted between residues 517 and 518 of SEQ ID No.3 (i.e. between G at 517-position and S at 518-position of SEQ ID No.3); between residues 518 and 519 of SEQ ID No.3 (i.e. between S at 518-position and S at 519-position of SEQ ID No.3); between residues 519 and 520 of SEQ ID No.3 (i.e. between S at 519-position and V at 520-position of SEQ ID No.3); between residues 515 and 516 of SEQ ID No.3 (i.e. between L at 515-position and S at 516-position of SEQ ID No.3); between residues 516 and 517 of SEQ ID No.3 (i.e. between S at 516-position and G at 517-position of SEQ ID No.3); between residues 536 and 537 of SEQ ID No.3 (i.e. between C at 536-position and G at 537-position of SEQ ID No.3); between residues 537 and 538 of SEQ ID No.3 (i.e. between G at 537-position and G at 538-position of SEQ ID No.3); between residues 538 and 539 of SEQ ID No.3 (i.e. between G at 538-position and T at 539-position of SEQ ID No.3).


The fusion protein may be expressed using a conventional technique in the art. A variety of expression systems can be used for the expression of the fusion protein. For example, the fusion protein can be expressed in 293 cells, Sf9 cells or E. coli.


In one embodiment, antigen is a substance (e.g. protein) which is not derived from Chikungunya or Venezuelan equine encephalitis virus. Antigen may be at least one selected from the group consisting of self antigens and cancer antigens. For example, antigen is a polypeptide derived from TNF-α, CD20 or CTLA4. Thus, examples of combinations of the polypeptide and the antigen used for the present invention include, but are not limited to, i) a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide derived from TNF-α;


ii) a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide derived from CD20;


iii) a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from TNF-α; or


iv) a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from CD20


v) a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from CTLA4.


A polypeptide derived from Chikungunya virus (CHIKV) or Venezuelan equine encephalitis virus (VEEV) may be a naturally occurring viral polypeptide or modified polypeptide thereof. In addition, a polypeptide derived from TNF-α, CD20 or CTLA4 may be a naturally occurring polypeptide or modified polypeptide of the naturally occurring polypeptide or a fragment of the naturally occurring polypeptide or the modified peptide. The modified polypeptide may be a fragment of the naturally occurring virus structural polypeptide.


In one embodiment, the modified polypeptide derived from TNF-α, CD20 or CTLA4 has at least 70%, 75%, 80%, 85%, 90%, 95% or 98% amino acid sequence identity to a naturally occurring polypeptide. In one embodiment, the modified peptide derived from TNF-α, CD20 or CTLA4 is a mutant where at most 10% of the amino acids are deleted, substituted, and/or added based on a naturally occurring polypeptide derived from TNF-α, CD20 or CTLA4.


When a polypeptide derived from a virus is conjugated with a polypeptide derived from an antigen, a linker peptide including SG, GS, SGG, GGS SGSG (SEQ ID NO: 51) and TRGGS (SEQ ID NO: 52) may be used. Examples of conjugation of the polypeptide derived from a virus (referred to as “PFV” below) with the polypeptide derived from the antigen (referred to as “PFA” below) include, but not limited to: PFV-SG-PFA-GS-PFV; PFV-SG-PFA-GGS-PFV; PFV-SSG-PFA-GS-PFV; PFV-SGG-PFA-GGS-PFV; PFV-SGSG(SEQ ID NO: 51)-PFA-GS-PFV; and PFA-SGG-PFA-TRGGS(SEQ ID NO: 52)-PFV.


In one embodiment, the present invention provides a virus like particle comprising


i) a fusion protein of a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide derived from TNF-α, which consists of an amino acid sequence represented by SEQ ID No.4;


ii) a fusion protein of a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide derived from CD20, which consists of an amino acid sequence represented by SEQ ID No.5;


iii) a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from TNF-α, which consists of an amino acid sequence represented by SEQ ID No.6; or


iv) a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from CD20, which consists of an amino acid sequence represented by SEQ ID No.7;


v) a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from CTLA4, which consists of an amino acid sequence represented by SEQ ID No.8.


In one embodiment, the present invention provides a virus like particle comprising a fusion protein which is modified from the fusion protein having an amino acid sequence represented by any one of SEQ ID Nos.4-8. The modified fusion protein may have at least 70%, 75%, 80%, 85%, 90%, 95% or 98% amino acid sequence identity to the fusion protein having an amino acid sequence represented by any one of SEQ ID Nos.4-8. Also, the modified fusion protein may be a mutant where at most 10% of the amino acids are deleted, substituted, and/or added based on the fusion protein having an amino acid sequence represented by any one of SEQ ID Nos.4-8.


(2) Nucleotide, Vector, Host Cell


In the second aspect, the present invention provides a nucleic acid molecule comprising a nucleotide sequence that encodes a particle as provided in the first aspect of the present invention.


In one embodiment, the present invention provides a nucleic acid molecule comprising a nucleotide sequence that encodes the Chikungunya or Venezuelan equine encephalitis virus like particle as described above.


Examples of the nucleotide sequence that encodes the Chikungunya or Venezuelan equine encephalitis virus like particle include, but are not limited to, a nucleotide sequence encoding E3-E2-6K-E1 of Chikungunya virus Strain 37997, a nucleotide sequence encoding Capsid-E3-E2-6K-E1 of Chikungunya virus Strain 37997, a nucleotide sequence encoding E3-E2-6K-E1 of Chikungunya virus Strain LR2006 OPY-1, a nucleotide sequence encoding Capsid-E3-E2-6K-E1 of Chikungunya virus LR2006 OPY-1, a nucleotide sequence encoding E3-E2-6K-E1 of Venezuelan equine encephalitis virus Strain TC-83 and a nucleotide sequence encoding Capsid-E3-E2-6K-E1 of Venezuelan equine encephalitis virus TC-83.


Regarding Chikungunya virus, an exemplary nucleotide sequence that encodes E3-E2-6K-E1 is described below (SEQ ID No.:9):









Atgagcctcgccctcccggtcttgtgcctgttggcaaacactacattccc





tgctctcagccgccttgcacaccctgctgctacgaaaaggaaccggaaag





caccttgcgcatgcttgaggacaacgtgatgagacccggatactaccagc





tactaaaagcatcgctgacttgctctccccaccgccaaagacgcagtact





aaggacaattttaatgtctataaagccacaagaccatatctagctcattg





tcctgactgcggagaagggcattcgtgccacagccctatcgcattggagc





gcatcagaaatgaagcaacggacggaacgctgaaaatccaggtctctttg





cagatcgggataaagacagatgacagccacgattggaccaagctgcgcta





tatggatagccatacgccagcggacgcggagcgagccggattgcttgtaa





ggacttcagcaccgtgcacgatcaccgggaccatgggacactttattctc





gcccgatgcccgaaaggagagacgctgacagtgggatttacggacagcag





aaagatcagccacacatgcacacacccgttccatcatgaaccacctgtga





taggtagggagaggttccactctcgaccacaacatggtaaagagttacct





tgcagcacgtacgtgcagagcaccgctgccactgctgaggagatagaggt





gcatatgcccccagatactcctgaccgcacgctgatgacgcagcagtctg





gcaacgtgaagatcacagttaatgggcagacggtgcggtacaagtgcaac





tgcggtggctcaaacgagggactgacaaccacagacaaagtgatcaataa





ctgcaaaattgatcagtgccatgctgcagtcactaatcacaagaattggc





aatacaactcccctttagtcccgcgcaacgctgaactcggggaccgtaaa





ggaaag





atccacatcccattcccattggcaaacgtgacttgcagagtgccaaaagc





aagaaaccctacagtaacttacggaaaaaaccaagtcaccatgctgctgt





atcctgaccatccgacactcttgtcttaccgtaacatgggacaggaacca





aattaccacgaggagtgggtgacacacaagaaggaggttaccttgaccgt





gcctactgagggtctggaggtcacttggggcaacaacgaaccatacaagt





actggccgcagatgtctacgaacggtactgctcatggtcacccacatgag





ataatcttgtactattatgagctgtaccccactatgactgtagtcattgt





gtcggtggcctcgttcgtgcttctgtcgatggtgggcacagcagtgggaa





tgtgtgtgtgcgcacggcgcagatgcattacaccatatgaattaacacca





ggagccactgttcccttcctgctcagcctgctatgctgcgtcagaacgac





caaggcggccacatattacgaggctgcggcatatctatggaacgaacagc





agcccctgttctggttgcaggctcttatcccgctggccgccttgatcgtc





ctgtgcaactgtctgaaactcttgccatgctgctgtaagaccctggcttt





tttagccgtaatgagcatcggtgcccacactgtgagcgcgtacgaacacg





taacagtgatcccgaacacggtgggagtaccgtataagactcttgtcaac





agaccgggttacagccccatggtgttggagatggagctacaatcagtcac





cttggaaccaacactgtcacttgactacatcacgtgcgagtacaaaactg





tcatcccctccccgtacgtgaagtgctgtggtacagcagagtgcaaggac





aagagcctaccagactacagctgcaaggtctttactggagtctacccatt





tatgtggggcggcgcctactgcttttgcgacgccgaaaatacgcaattga





gcgaggcacatgtagagaaatctgaatcttgcaaaacagagtttgcatcg





gcctacagagcccacaccgcatcggcgtcggcgaagctccgcgtccttac





caaggaaacaacattaccgtagctgcctacgctaacggtgaccatgccgt





cacagtaaaggacgccaagtttgtcgtgggcccaatgtcctccgcctgga





caccttttgacaacaaaatcgtggtgtacaaaggcgacgtctacaacatg





gactacccaccttttggcgcaggaagaccaggacaatttggtgacattca





aagtcgtacaccggaaagtaaagacgtttatgccaacactcagttggtac





tacagaggccagcagcaggcacggtacatgtaccatactctcaggcacca





tctggcttcaagtattggctgaaggaacgaggagcatcgctacagcacac





ggcaccgttcggttgccagattgcgacaaacccggtaagagctgtaaatt





gcgctgtggggaacataccaatttccatcgacataccgatgcggccttta





ctagggttgtcgatgcaccctctgtaacggacatgtcatgcgaagtacca





gcctgcactcactcctccgactttgggggcgtcgccatcatcaaatacac





agctagcaagaaaggtaaatgtgcagtacattcgatgaccaacgccgtta





ccattcgagaagccgacgtagaagtagaggggaactcccagctgcaaata





tccttctcaacagccctggcaagcgccgagtttcgcgtgcaagtgtgctc





cacacaagtacactgcgcagccgcatgccaccctccaaaggaccacatag





tcaattacccagcatcacacaccacccttggggtccaggatatatccaca





acggcaatgtcttgggtgcagaagattacgggaggagtaggattaattgt





tgctgttgctgccttaattttaattgtggtgctatgcgtgtcgtttagca





ggcac.






Regarding Chikungunya virus, another exemplary nucleotide sequence that encodes E3-E2-6K-E1 is described below (SEQ ID No.:10):









Atgagtcttgccatcccagttatgtgcctgttggcaaacaccacgttccc





ctgctcccagcccccttgcacgccctgctgctacgaaaaggaaccggagg





aaaccctacgcatgcttgaggacaacgtcatgagacctgggtactatcag





ctgctacaagcatccttaacatgttctccccaccgccagcgacgcagcac





caaggacaacttcaatgtctataagccacaagaccatacttagctcactg





tcccgactgtggagaagggcactcgtgccatagtcccgtagcactagaac





gcatcagaaatgaagcgacagacgggacgctgaaaatccaggtctccttg





caaatcggaataaagacggatgacagccacgattggaccaagctgcgtta





tatggacaaccacatgccagcagacgcagagagggcggggctatttgtaa





gaacatcagcaccgtgtacgattactggaacaatgggacacttcatcctg





gcccgatgccaaaaggggaaactctgacggtgggattcactgacagtagg





aagattagtcactcatgtacgcacccatttcaccacgaccctcctgtgat





aggtcgggaaaaattccattcccgaccgcagcacggtaaagagctacctt





gcagcacgtacgtgcagagcaccgccgcaactaccgaggagatagaggta





cacatgcccccagacacccctgatcgcacattaatgtcacaacagtccgg





caacgtaaagatcacagtcaatggccagacggtgcggtacaagtgtaatt





gcggtggctcaaatgaaggactaacaactacagacaagtgattaataact





gcaaggttgatcaatgtcatgccgcggtcaccaatcacaaaagtggcagt





ataactcccctctggtcccgcgtaatgctgaacttggggaccgaaaagga





aaaattcacatcccgtttccgctggcaaatgtaacatgcagggtgcctaa





agcaaggaaccccaccgtgacgtacgggaaaaaccaagtcatcatgctac





tgtatcctgaccacccaacactcctgtcctaccggaatatgggagaagaa





ccaaactatcaagaagagtgggtgatgcataagaaggaagtcgtgctaac





cgtgccgactgaagggctcgaggtcacgtggggcaacaacgagccgtata





agtattggccgcagttatctacaaacggtacagcccatggccacccgcat





gagataattctgtattattatgagctgtaccccactatgactgtagtagt





tgtgtcagtggccacgttcatactcctgtcgatggtgggtatggcagcgg





ggatgtgcatgtgtgcacgacgcagatgcatcacaccgtatgaactgaca





ccaggagctaccgtccctttcctgcttagcctaatatgctgcatcagaac





agctaaagcggccacataccaagaggctgcgatatacctgtggaacgagc





agcaacctttgttttggctacaagcccttattccgctggcagccctgatt





gttctatgcaactgtctgagactcttaccatgctgctgtaaaacgttgct





tttttagccgtaatgagcgtcggtgcccacactgtgagcgcgtacgaaca





cgtaacagtgatcccgaacacggtgggagtaccgtataagactctagtca





atagacctggctacagccccatggtattggagatggaactactgtcagtc





actttggagccaacactatcgcttgattacatcacgtgcgagtacaaaac





cgtcatcccgtctccgtacgtgaagtgctgcggtacagcagagtgcaagg





acaaaaacctacctgactacagctgtaaggtcttcaccggcgtctaccca





tttatgtggggcggcgcctactgcttctgcgacgctgaaaacacgcagtt





gagcgaagcacacgtggagaagtccgaatcatgcaaaacagaatttgcat





cagcatacagggctcataccgcatctgcatcagctaagctccgcgtcctt





taccaaggaaataacatcactgtaactgcctatgcaaacggcgaccatgc





cgtcacagttaaggacgccaaattcattgtggggccaatgtcttcagcct





ggacacctttcgacaacaaaattgtggtgacaaaggtgacgtctataaca





tggactacccgccctttggcgcaggaagaccaggacaatttggcgatatc





caaagtcgcacacctgagagtaaagacgtctatgctaatacacaactggt





actgcagagaccggctgtgggtacggtacacgtgccatactctcaggcac





catctggcttaagtattggctaaaagaacgcggggcgtcgctgcagcaca





cagcaccatttggctgccaaatagcattcaaacccggtaagagcggtgaa





ctgcgccgtaggaacatgccatctccatcgacataccggaagcggccttc





actagggtcgtcgacgcgccctctttaacggacatgtcgtgcgaggtacc





agcctgcacccattcctcagactttgggggcgtcgccattattaaatatg





cagccagcaagaaaggcaagtgtgcggtgcattcgatgactaacgccgtc





actattcgggaagctgagatagaagttgaagggaattctcagctgcaaat





ctctttctcgacggccttagccagcgccgaattccgcgtacaagtctgtt





ctacacaagtacactgtgcagccgagtgccaccccccgaaggaccacata





gtcaatacccggcgtcacataccaccctcggggtccaggacatctccgct





acggcgatgtcatgggtgcagaagatcacgggaggtgtgggactggttgt





tgctgttgccgcactgattctaatcgtggtgctatgcgtgtcgttcagca





ggcac.






Regarding Chikungunya virus, an exemplary nucleotide sequence that encodes a Capsid-E3-E2-6K-E1 is described below (SEQ ID No.:11):









atggagttcatcccgacgcaaactttctataacagaaggtaccaaccccg





accctgggccccacgccctacaattcaagtaattagacctagaccacgtc





cacagaggcaggctgggcaactcgcccagctgatctccgcagtcaacaaa





ttgaccatgcgcgcggtacctcaacagaagcctcgcagaaatcggaaaaa





caagaagcaaaggcagaagaagcaggcgccgcaaaacgacccaaagcaaa





agaagcaaccaccacaaaagaagccggctcaaaagaagaagaaaccaggc





cgtagggagagaatgtgcatgaaaattgaaaatgattgcatcttcgaagt





caagcatgaaggcaaagtgatgggctacgcatgcctggtgggggataaag





taatgaaaccagcacatgtgaagggaactatcgacaatgccgatctggct





aaactggcctttaagcggtcgtctaaatacgatcttgaatgtgcacagat





accggtgcacatgaagtctgatgcctcgaagtttacccacgagaaacccg





aggggtactataactggcatcacggagcagtgcagtattcaggaggccgg





ttcactatcccgacgggtgcaggcaagccgggagacagcggcagaccgat





cttcgacaacaaaggacgggtggtggccatcgtcctaggaggggccaacg





aaggtgcccgcacggccctctccgtggtgacgtggaacaaagacatcgtc





acaaaaattacccctgagggagccgaagagtggagcctcgccctcccggt





cttgtgcctgttggcaaacactacattcccctgctctcagccgccttgca





caccctgctgctacgaaaaggaaccggaaagcaccttgcgcatgcttgag





gacaacgtgatgagacccggatactaccagctactaaaagcatcgctgac





ttgctctccccaccgccaaagacgcagtactaaggacaattttaatgtct





ataaagccacaagaccatatctagctcattgtcctgactgcggagaaggg





cattcgtgccacagccctatcgcattggagcgcatcagaaatgaagcaac





ggacggaacgctgaaaatccaggtctctttgcagatcgggataaagacag





atgacagccacgattggaccaagctgcgctatatggatagccatacgcca





gcggacgcggagcgagccggattgcttgtaaggacttcagcaccgtgcac





gatcaccgggaccatgggacactttattctcgcccgatgcccgaaaggag





agacgctgacagtgggatttacggacagcagaaagatcagccacacatgc





acacacccgttccatcatgaaccacctgtgataggtagggagaggttcca





ctctcgaccacaacatggtaaagagttaccttgcagcacgtacgtgcaga





gcaccgctgccactgctgaggagatagaggtgcatatgcccccagatact





cctgaccgcacgctgatgacgcagcagtctggcaacgtgaagatcacagt





taatgggcagacggtgcggtacaagtgcaactgcggtggctcaaacgagg





gactgacaaccacagacaaagtgatcaataactgcaaaattgatcagtgc





catgctgcagtcactaatcacaagaattggcaatacaactcccctttagt





cccgcgcaacgctgaactcggggaccgtaaaggaaagatccacatcccat





tcccattggcaaacgtgacttgcagagtgccaaaagcaagaaaccctaca





gtaacttacggaaaaaaccaagtcaccatgctgctgtatcctgaccatcc





gacactcttgtcttaccgtaacatgggacaggaaccaaattaccacgagg





agtgggtgacacacaagaaggaggttaccttgaccgtgcctactgagggt





ctggaggtcacttggggcaacaacgaaccatacaagtactggccgcagat





gtctacgaacggtactgctcatggtcacccacatgagataatcttgtact





attatgagctgtaccccactatgactgtagtcattgtgtcggtggcctcg





ttcgtgcttctgtcgatggtgggcacagcagtgggaatgtgtgtgtgcgc





acggcgcagatgcattacaccatatgaattaacaccaggagccactgttc





ccttcctgctcagcctgctatgctgcgtcagaacgaccaaggcggccaca





tattacgaggctgcggcatatctatggaacgaacagcagcccctgttctg





gttgcaggctcttatcccgctggccgccttgatcgtcctgtgcaactgtc





tgaaactcttgccatgctgctgtaagaccctggcttttttagccgtaatg





agcatcggtgcccacactgtgagcgcgtacgaacacgtaacagtgatccc





gaacacggtgggagtaccgtataagactcttgtcaacagaccgggttaca





gccccatggtgttggagatggagctacaatcagtcaccttggaaccaaca





ctgtcacttgactacatcacgtgcgagtacaaaactgtcatcccctcccc





gtacgtgaagtgctgtggtacagcagagtgcaaggacaagagcctaccag





actacagctgcaaggtctttactggagtctacccatttatgtggggcggc





gcctactgcttttgcgacgccgaaaatacgcaattgagcgaggcacatgt





agagaaatctgaatcttgcaaaacagagtttgcatcggcctacagagccc





acaccgcatcggcgtcggcgaagctccgcgtcctttaccaaggaaacaac





attaccgtagctgcctacgctaacggtgaccatgccgtcacagtaaagga





cgccaagtttgtcgtgggcccaatgtcctccgcctggacaccttttgaca





acaaaatcgtggtgtacaaaggcgacgtctacaacatggactacccacct





tttggcgcaggaagaccaggacaatttggtgacattcaaagtcgtacacc





ggaaagtaaagacgtttatgccaacactcagttggtactacagaggccag





cagcaggcacggtacatgtaccatactctcaggcaccatctggcttcaag





tattggctgaaggaacgaggagcatcgctacagcacacggcaccgttcgg





ttgccagattgcgacaaacccggtaagagctgtaaattgcgctgtgggga





acataccaatttccatcgacataccggatgcggcctttactagggttgtc





gatgcaccctctgtaacggacatgtcatgcgaagtaccagcctgcactac





atcctccgactttgggggcgtcgccatcatcaaatacacagctagcaaga





aaggtaaatgtgcagtacattcgatgaccaacgccgttaccattcgagaa





gccgacgtagaagtagaggggaactcccagctgcaaatatccttctcaac





agccctggcaagcgccgagtttcgcgtgcaagtgtgctccacacaagtac





actgcgcagccgcatgccaccctccaaaggaccacatagtcaattaccca





gcatcacacaccacccttggggtccaggatatatccacaacggcaatgtc





ttgggtgcagaagattacgggaggagtaggattaattgttgctgttgctg





ccttaattttaattgtggtgctatgcgtgtcgtttagcaggcactaa.






Regarding Chikungunya virus, another exemplary nucleotide sequence that encodes a Capsid-E3-E2-6K-E1 is described below (SEQ ID No.:12):









atggagttcatcccaacccaaactttttacaataggaggtaccagcctcg





accctggactccgcgccctactatccaagtcatcaggcccagaccgcgcc





ctcagaggcaagctgggcaacttgcccagctgatctcagcagttaataaa





ctgacaatgcgcgcggtaccacaacagaagccacgcaggaatcggaagaa





taagaagcaaaagcaaaaacaacaggcgccacaaaacaacacaaatcaaa





agaagcagccacctaaaaagaaaccggctcaaaagaaaaagaagccgggc





cgcagagagaggatgtgcatgaaaatcgaaaatgattgtattttcgaagt





caagcacgaaggtaaggtaacaggttacgcgtgcctggtgggggacaaag





taatgaaaccagcacacgtaaaggggaccatcgataacgcggacctggcc





aaactggcctttaagcggtcatctaagtatgaccttgaatgcgcgcagat





acccgtgcacatgaagtccgacgcttcgaagttcacccatgagaaaccgg





aggggtactacaactggcaccacggagcagtacagtactcaggaggccgg





ttcaccatccctacaggtgctggcaaaccaggggacagcggcagaccgat





cttcgacaacaagggacgcgtggtggccatagtcttaggaggagctaatg





aaggagcccgtacagccctctcggtggtgacctggaataaagacattgtc





actaaaatcacccccgagggggccgaagagtggagtcttgccatcccagt





tatgtgcctgttggcaaacaccacgttcccctgctcccagcccccttgca





cgccctgctgctacgaaaaggaaccggaggaaaccctacgcatgcttgag





gacaacgtcatgagacctgggtactatcagctgctacaagcatccttaac





atgttctccccaccgccagcgacgcagcaccaaggacaacttcaatgtct





ataaagccacaagaccatacttagctcactgtcccgactgtggagaaggg





cactcgtgccatagtcccgtagcactagaacgcatcagaaatgaagcgac





agacgggacgctgaaaatccaggtctccttgcaaatcggaataaagacgg





atgacagccacgattggaccaagctgcgttatatggacaaccacatgcca





gcagacgcagagagggcggggctatttgtaagaacatcagcaccgtgtac





gattactggaacaatgggacacttcatcctggcccgatgtccaaaagggg





aaactctgacggtgggattcactgacagtaggaagattagtcactcatgt





acgcacccatttcaccacgaccctcctgtgataggtcgggaaaaattcca





ttcccgaccgcagcacggtaaagagctaccttgcagcacgtacgtgcaga





gcaccgccgcaactaccgaggagatagaggtacacatgcccccagacacc





cctgatcgcacattaatgtcacaacagtccggcaacgtaaagatcacagt





caatggccagacggtgcggtacaagtgtaattgcggtggctcaaatgaag





gactaacaactacagacaaagtgattaataactgcaaggttgatcaatgt





catgccgcggtcaccaatcacaaaaagtggcagtataactcccctctggt





cccgcgtaatgctgaacttggggaccgaaaaggaaaaattcacatcccgt





ttccgctggcaaatgtaacatgcagggtgcctaaagcaaggaaccccacc





gtgacgtacgggaaaaaccaagtcatcatgctactgtatcctgaccaccc





aacactcctgtcctaccggaatatgggagaagaaccaaactatcaagaag





agtgggtgatgcataagaaggaagtcgtgctaaccgtgccgactgaaggg





ctcgaggtcacgtggggcaacaacgagccgtataagtattggccgcagtt





atctacaaacggtacagcccatggccacccgcatgagataattctgtatt





attatgagctgtaccccactatgactgtagtagttgtgtcagtggccacg





ttcatactcctgtcgatggtgggtatggcagcggggatgtgcatgtgtgc





acgacgcagatgcatcacaccgtatgaactgacaccaggagctaccgtcc





ctttcctgcttagcctaatatgctgcatcagaacagctaaagcggccaca





taccaagaggctgcgatatacctgtggaacgagcagcaacctttgttttg





gctacaagcccttattccgctggcagccctgattgttctatgcaactgtc





tgagactcttaccatgctgctgtaaaacgttggcttttttagccgtaatg





agcgtcggtgcccacactgtgagcgcgtacgaacacgtaacagtgatccc





gaacacggtgggagtaccgtataagactctagtcaatagacctggctaca





gccccatggtattggagatggaactactgtcagtcactttggagccaaca





ctatcgcttgattacatcacgtgcgagtacaaaaccgtcatcccgtctcc





gtacgtgaagtgctgcggtacagcagagtgcaaggacaaaaacctacctg





actacagctgtaaggtcttcaccggcgtctacccatttatgtggggcggc





gcctactgcttctgcgacgctgaaaacacgcagttgagcgaagcacacgt





ggagaagtccgaatcatgcaaaacagaatttgcatcagcatacagggctc





ataccgcatctgcatcagctaagctccgcgtcctttaccaaggaaataac





atcactgtaactgcctatgcaaacggcgaccatgccgtcacagttaagga





cgccaaattcattgtggggccaatgtcttcagcctggacacctttcgaca





acaaaattgtggtgtacaaaggtgacgtctataacatggactacccgccc





tttggcgcaggaagaccaggacaatttggcgatatccaaagtcgcacacc





tgagagtaaagacgtctatgctaatacacaactggtactgcagagaccgg





ctgtgggtacggtacacgtgccatactctcaggcaccatctggctttaag





tattggctaaaagaacgcggggcgtcgctgcagcacacagcaccatttgg





ctgccaaatagcaacaaacccggtaagagcggtgaactgcgccgtaggga





acatgcccatctccatcgacataccggaagcggccttcactagggtcgtc





gacgcgccctctttaacggacatgtcgtgcgaggtaccagcctgcaccca





ttcctcagactttgggggcgtcgccattattaaatatgcagccagcaaga





aaggcaagtgtgcggtgcattcgatgactaacgccgtcactattcgggaa





gctgagatagaagttgaagggaattctcagctgcaaatctctttctcgac





ggccttagccagcgccgaattccgcgtacaagtctgttctacacaagtac





actgtgcagccgagtgccaccccccgaaggaccacatagtcaactacccg





gcgtcacataccaccctcggggtccaggacatctccgctacggcgatgtc





atgggtgcagaagatcacgggaggtgtgggactggttgttgctgttgccg





cactgattctaatcgtggtgctatgcgtgtcgttcagcaggcactaa.






In one embodiment, the present invention provides a vector comprising the nucleic acid molecule as described above, wherein the vector optionally comprises an expression control sequence operably linked to the nucleic acid molecule.


Examples of an expression control sequence include, but are not limited to, promoter such as CMV promoter, phage lambda PL promoter, the E. coli lac, phoA and tac promoters, the SV40 early and late promoters, and promoters of retroviral LTRs.


The expression vectors can be prepared by a person skilled in the art based on WO/2012/006180, the entire contents of which are incorporated by reference herein. Examples of vectors which can be used for expressing a fusion protein of a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide of antigen include a vector shown in VLP_CHI 512 vector (SEQ ID No.:23) containing CHIKV VLP polynucleotide (SEQ ID No. 28; corresponding amino acid sequence represented by SEQ ID No.:29); and VLP_CHI 532 vector (SEQ ID No.: 24) containing CHIKV VLP polynucleotide (SEQ ID No. 30; corresponding amino acid sequence represented by SEQ ID No.:31).


The expression vectors can be prepared by a person skilled in the art based on US2012/0003266, the entire contents of which are incorporated by reference herein. Examples of vectors which can be used for expressing a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide of antigen include a vector shown in VLP_VEEV VLP 518 vector (SEQ ID No.:25) containing VEEV VLP polynucleotide (SEQ ID No. 32; corresponding amino acid sequence represented by SEQ ID No.:33); VLP_VEEV VLP 519 vector (SEQ ID No.26) containing VEEV VLP polynucleotide (SEQ ID No. 34; corresponding amino acid sequence represented by SEQ ID No.:35); and VLP_VEEV VLP 538 vector (SEQ ID No.: 27) containing VEEV VLP polynucleotide (SEQ ID No. 36; corresponding amino acid sequence represented by SEQ ID No.:37).


In one embodiment, the present invention provides i) a nucleic acid molecule encoding a fusion protein of a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide derived from TNF-α, which consists of a nucleotide sequence represented by SEQ ID No.13;


ii) a nucleic acid molecule encoding a fusion protein of a polypeptide derived from Chikungunya virus (CHIKV) and a polypeptide derived from CD20, which consists of a nucleotide sequence represented by SEQ ID No.14;


iii) a nucleic acid molecule encoding a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from TNF-α, which consists of a nucleotide sequence represented by SEQ ID No.15;


iv) a nucleic acid molecule encoding a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from CD20, which consists of a nucleotide sequence represented by SEQ ID No.16; or


v) a nucleic acid molecule encoding a fusion protein of a polypeptide derived from Venezuelan equine encephalitis virus (VEEV) and a polypeptide derived from CTLA4, which consists of a nucleotide sequence represented by SEQ ID No.17.


In one embodiment, the present invention provides a nucleic acid molecule which is modified from the nucleic acid molecule having a nucleotide sequence represented by any one of SEQ ID Nos.13-17. The modified nucleic acid molecule may have at least 70%, 75%, 80%, 85%, 90%, 95% or 98% nucleotide sequence identity to the nucleic acid molecule having a nucleotide sequence represented by any one of SEQ ID Nos.13-17. Also, the modified nucleic acid molecule may be a mutant where at most 10% of the amino acids are deleted, substituted, and/or added based on the nucleic acid molecule having a nucleotide sequence represented by any one of SEQ ID Nos.13-17.


(3) Composition


In the third aspect, the present invention provides a composition comprising the particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention.


In one embodiment, the present invention provides a composition comprising the Chikungunya or Venezuelan equine encephalitis virus like particle as described above or the nucleic acid molecule as described above.


The composition may further comprise a pharmaceutical acceptable carrier and/or adjuvant. Examples of adjuvant include, but are not limited to Ribi solution (Sigma Adjuvant system, Sigma-Aldrich).


The pharmaceutical composition of the present invention may contain a single active ingredient or a combination of two or more active ingredients, as far as they are not contrary to the objects of the present invention. For example, cytokines including chemokines, anti-body of cytokines such as anti TNF antibody (e.g. infliximab, adalimumab), anti-VEGF antibody (e.g. bevacizumab and ranibizumab), cytokine receptor antagonist such as anti HER2 antibody (e.g. Trastuzumab), anti EGF receptor antibody (e.g. Cetuximab), anti VEGF aptamer (e.g. Pegaptanib) and immunomodulator such as cyclosporine, tacrolimus, ubenimex may be used for the combination therapy.


In a combination of plural active ingredients, their respective contents may be suitably increased or decreased in consideration of their therapeutic effects and safety.


The term “combination” used herein means two or more active ingredient are administered to a patient simultaneously in the form of a single entity or dosage, or are both administered to a patient as separate entities either simultaneously or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two components in the body, preferably at the same time.


In one embodiment, the composition is a vaccine composition including a DNA vaccine. In one embodiment, the DNA vaccine provided by the present invention comprises CpG containing oligonucleotide.


(4) Method of Producing an Antibody, Method of Immunomodulation, Method of Treating an Autoimmune Disease, Method of Inducing and/or Enhancing Immune Response Against an Antigen in a Mammal, Method of Treating Cancer, Method of Passive Immunization, Method of Presenting an Antigen on Macrophage, and Method for Producing a Particle


In the fourth aspect, the present invention provides a method of producing an antibody, comprising contacting the particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention to a mammal.


The antibody produced in the fourth aspect of the present invention may be humanized using a conventional technique. Thus, in one embodiment, the method provided in the fourth aspect of the invention further comprises a step of humanizing non-human mammal produced antibody.


The particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention may be administered directly into the patient, into the affected organ or systemically, or applied ex vivo to cells derived from the patient or a human cell line which are subsequently administered to the patient, or used in vitro to select a subpopulation from immune cells such as B-cell and T-cell derived from the patient, which are then re-administered to the patient.


According to the present invention, the virus like particle can be applied for the immune therapy.


In the fifth aspect, the present invention provides a method of immunomodulation, a method of treating an autoimmune disease, a method of inducing and/or enhancing immune response against an antigen in a mammal, and a method of treating cancer comprising administering the composition provided in the third aspect of the present invention to a mammal.


In sixth aspect, the present invention provides a method of passive immunization, comprising administering the antibody provided in the fourth aspect of the present invention to a mammal.


In seventh aspect, the present invention provides a method of presenting an antigen on macrophage, comprising contacting the particle provided in the first aspect of the present invention and/or the nucleic acid molecule provided in the second aspect of the present invention to a mammal.


In eighth aspect, the present invention provides a method for producing the particle provided in the first aspect of the present invention, comprising preparing a gene comprising a nucleotide sequence encoding said particle; culturing a cell which is transfected with said gene to express said particle; and recovering said particle.


In one embodiment, the present invention provides a method of producing an antibody, comprising contacting the Chikungunya or Venezuelan equine encephalitis virus like particle as described above and/or the nucleic acid molecule as described above to a mammal. The produced antibody may be an antibody which can specifically bind to the antigen comprised in the Chikungunya or Venezuelan equine encephalitis virus like particle or the antigen encoded by the nucleic acid molecule. The method of producing an antibody provided by the present invention may be a useful method for producing a monoclonal or polyclonal antibody against an antigen (e.g. TNF α, CD20 and CLTA4).


In one embodiment, the antibody obtained by the method of producing an antibody according to the present invention is used for passive immunization. The method of passive immunization may comprise administering the obtained antibody to a mammal.


According to the present invention, the composition of the present invention is useful for immunomodulation. Especially said immunomodulation is for the treatment of autoimmune disease, neural disease, inflammatory disease such as inflammatory lung disease, including the acute respiratory distress syndrome, chronic obstructive pulmonary disease and asthma, angiogenesis associated diseases including neoplasm.


In one preferred embodiment, the immunomodulation provided by the present invention is inducing and/or enhancing immune response against an antigen in a mammal. Thus, in one embodiment, the present invention provides a method of inducing and/or enhancing immune response against an antigen in a mammal, comprising administering an effective amount of the composition as described above to the mammal. Examples of mammal include, but are not limited to, a human.


Since many antibodies are useful for the treatment of disease, the method and the composition which are provided by the present invention can be useful for the treatment of diseases. For example, an antibody which specifically binds the target as listed in Table 1 or an antibody which binds an epitope on the target as listed in Table 1 is useful for the treatment of the disease as listed in Table 1.


In one embodiment, at least one antigen which is used for the present invention is at least one target as listed in Table 1. When at least one antigen which is used for the present invention is at least one target as listed in Table 1, the particle, the isolated nucleic acid, the vector, the composition and the method provided by the present invention can be useful for the treatment of the disease or the condition as listed in Table 1 (see “Use” of Table 1).


For example, when at least one antigen used for the present invention is one or more cancer antigen, the particle, the isolated nucleic acid, the vector, the composition and the method provided by the present invention can be useful for the treatment of cancer.


Examples of cancer antigen include, but are not limited to, VEGF, epidermal growth factor receptor, CD33, CD20 and ErbB2. When the composition of the present invention comprising two or more cancer antigens is administered to a mammal, antibodies directed to the two or more cancer antigens can attack the cancer.


For example, when at least one antigen used for the present invention is amyloid β, the isolated nucleic acid, the vector, the composition and the method provided by the present invention can be useful for the treatment of Alzheimer's disease.


For example, when at least one antigen used for the present invention is TNF alpha, the isolated nucleic acid, the vector, the composition and the method provided by the present invention can be useful for the treatment of inflammation; auto immune disease including rheumatoid arthritis; psoriasis, Crohn's disease; ulcerative colitis etc.


For example, when at least one antigen used for the present invention is CD20, the isolated nucleic acid, the vector, the composition and the method provided by the present invention can be useful for the treatment of auto immune disease including rheumatoid arthritis and SLE; cancer including Non-Hodgkin lymphoma etc.


For example, when at least one antigen used for the present invention is CTLA4, the isolated nucleic acid, the vector, the composition and the method provided by the present invention can be useful for the treatment of cancer including melanoma; and useful for activating T cells etc.


Given the symptom of patients infected with Chikungunya or Venezuelan equine encephalitis together with unusual big molecule of Chikungunya or Venezuelan equine encephalitis, this VLP can act effectively and efficiently to target macrophage and its composition such as cytokines and immunomodulative compounds.


In one aspect, the present invention provides a method of presenting an antigen on macrophage, comprising administering the Chikungunya or Venezuelan equine encephalitis virus like particle as described above and/or the nucleic acid molecule as described above to a mammal. The Chikungunya or Venezuelan equine encephalitis virus like particle provided by the present invention is good to target macrophage. In one embodiment, the Chikungunya or Venezuelan equine encephalitis virus like particle provided by the present invention is a kind of delivery system of the at least one antigen, which is comprised in the Chikungunya or Venezuelan equine encephalitis virus like particle, to macrophage.


In one embodiment, the present invention provides a method for producing Chikungunya or Venezuelan equine encephalitis virus like particle provided in the first aspect of the present invention, comprising preparing a gene comprising a nucleotide sequence encoding said particle; culturing a cell which is transfected with said gene to express said particle; and recovering said particle. In this embodiment, transfection can be conducted using a conventional method. Cells using for the transfection may be 293 cells. Recovering VLP may include collecting a conditioned medium after cells are transfected with a plasmid comprising a gene, and may further include purify VLP from the conditioned medium using ultracentrifugation. In one embodiment, further step may be included in the method for producing Chikungunya or Venezuelan equine encephalitis virus like particle provided in the eighth aspect of the present invention, where a polynucleotide encoding an antigen is designed so that spatial distance between the N-terminal residue and C-terminal residue of the antigen is 30 custom character or less (e.g. from 5 custom character to 15 custom character, from 5 to 12 custom character, from 5 custom character to 11 custom character, from 5 custom character to 10 custom character, from 5 custom character to 8 A, from 8 custom character to 15 custom character, from 8 custom character to 13 custom character, from 8 custom character to 12 custom character, from 8 custom character to 11 custom character, from 9 custom character to 12 custom character, from 9 custom character to 11 custom character, from 9 custom character to 10 custom character, or from 10 custom character to 11 custom character) when the distance is determined in a crystal of the antigen or a naturally occurring protein containing the antigen or modified protein therefrom.


Immune system evolves to recognize foreign antigens for killing pathogens such as viruses or bacteria. It also evolves not to recognize self proteins to protect self proteins. It is called immune tolerance system. Therefore it is difficult to induce antibodies against self-antigen by traditional immunization methods. To overcome the immune tolerance, we developed a novel vaccine method using a self-assembly subunit containing an self-antigen. The self-assembly subunit spontaneously assembles and forms a stable organized unit that presents highly repetitive antigens on the surface. Highly repetitive antigens immunogen strengthen signal pathways in B cells and result in stimulation of antibody responses than single antigen immunogen such as traditional immunization methods. Applying this mechanism to vaccine development not only increases antibody responses against target immunogens but also overcome self-antigen tolerance.


The present invention will be described in detail with reference to the following example, which, however, is not intended to limit the scope of the present invention.


EXAMPLES
(1) Preparation of Chikungunya Virus Like Particle Comprising a Virus Structural Polypeptide and a Fragment of Human TNF Alpha

It was expected that a monomer of TNF alpha polypeptide fused with Chikungunya virus structural polypeptide is difficult to be stably expressed because TNF alpha is found as a trimer under natural conditions. However, use of a fusion protein, in which TNF alpha monomer peptide (a fragment of TNF alpha monomer peptide) is fused with the Chikungunya virus structural polypeptide through linkers at N- and C-terminal of TNF alpha-derived peptide for attaching TNF alpha-derived peptide to the Chikungunya virus structural polypeptide, resulted in stable expression of a Chikungunya virus like particle comprising a virus structural polypeptide and TNF alpha monomer-derived peptide.


In detail, polynucleotide encoding the original human TNF alpha was modified to prepare a polynucleotide encoding modified TNF alpha-derived peptide where RTPSD which is N-terminal sequence of the original TNF alpha-derived peptide is replaced with SGG and TRGGS (SEQ ID NO: 52) is attached to C-terminal of the TNF alpha (see SEQ ID Nos.18-20 as shown in FIG. 1). The resulting polynucleotide was inserted between the codons encoding C at 519-position and Q at 520-position of SEQ ID No.2 to construct a plasmid (hereinafter referred to as CHIKV-TNFa4) for expressing Chikungunya virus like particle were the modified TNF alpha-derived peptide is inserted into E2 of Chikungunya virus structural polypeptide (C-E3-E2-6K-E1). Subsequently, 293F cells (1.5×106 cells/ml) was transfected with 250 μg of CHIKV-TNFa4. After culturing for 4 days, the supernatant was collected. The obtained supernatant was overlaid onto Opti Prep (Sigma D1556) followed by being ultracentrifuged (20000 rpm, 120 min) using SW28 rotor to concentrate VLP (i.e. virus like particle). The concentrated VLP was mixed with Opti Prep to form density gradient followed by ultracentrifuged (75000 rpm, 4 hours) using NVT100 rotor. After the ultracentrifugation, purified VLP was collected. The expression of VLP comprising TNF alpha conjugated with Chikungunya virus structural polypeptide was confirmed by Western Blot using an antibody specific for CHIVK (ATCC: VR-1241AF) and an antibody specific for TNF alpha (Cell Signal: #6945).


The spatial distance between the N-terminal residue and C-terminal residue of the TNF alpha-derived peptide is 8.27 custom character when the distance is determined in a crystal of TNF alpha.


(2) Preparation of Venezuelan Equine Encephalitis Virus Like Particle Comprising a Virus Structural Polypeptide and Human TNF Alpha-Derived Peptide (Referred to as “VEEV-TNFa VLPs”)

According to the above-described (1), polynucleotide encoding modified TNF alpha-derived peptide fused with polynucleotide encoding Venezuelan equine encephalitis virus structural polypeptide was prepared (see SEQ ID No.15) to construct an expression vector followed by transfection in 293F cells.


VEEV-TNFa VLPs were purified by density gradient centrifuge. As seen in Lane 4, TNF alpha-derived peptide and VEEV expression was confirmed by Western blot using TNFa monoclonal antibody (top panel) and VEEV polyclonal antibodies (bottom panel), respectively (see FIG. 2).


The spatial distance between the N-terminal residue and C-terminal residue of the TNF alpha-derived peptide is 8.27 custom character when the distance is determined in a crystal of TNF alpha.


(3) Detection of Anti-Human TNF Alpha Antibody in Immunized Mouse

Mice were divided into three groups (n=5 for each group). The Chikungunya virus like particle comprising a virus structural polypeptide and human TNF alpha-derived polypeptide prepared according to the above-described (1) (referred to as “CHIKV-TNF alpha” below), Chikungunya virus like particle without comprising human TNF alpha-derived polypeptide (referred to as “CHIKV-VLP” below), Venezuelan equine encephalitis virus like particle comprising a virus structural polypeptide and human TNF alpha-derived polypeptide prepared according to the above-described (2)(referred to as “VEEV-TNF alpha” below), Venezuelan equine encephalitis virus like particle without comprising human TNF alpha-derived polypeptide (referred to as “VEEV-VLP” below) or vehicle (i.e. PBS) were intramuscularly administered to each group of mice. The mice were administered at the beginning of the experiment (referred to as “0 week” below) and three weeks after the first administration (referred to as “3 week” below) as described below: Group 1: VEEV-TNF alpha (0 week), CHIKV-TNF alpha (3 week); Group 2: VEEV-VLP (0 week), CHIKV-VLP (3 week); and Group 3: PBS (0 week), PBS (3 week).


6 weeks after the beginning of the experiment, blood sample was obtained from each mouse and serum was prepared. Produced anti-human TNF alpha antibody was detected using ELISA where TNF alpha protein was coated on ELISA plate. The results show that the virus like particle comprising a virus structural polypeptide and human TNF alpha-derived polypeptide induced anti-human TNF alpha antibodies in mouse (see FIG. 9).


(4) Preparation of Venezuelan Equine Encephalitis Virus Like Particle Comprising a Virus Structural Polypeptide and a Fragment of Human CD20

According to the above-described (1) and (2), VEEV-CD20 VLPs were purified by density gradient centrifuge, and a fragment of CD20 and VEEV expression was confirmed by Western blot. IYNCEPANPSEKNSPSTQYCYSIQ (SEQ ID No.: 21), which is a fragment of CD20, was used as an antigen fused with Venezuelan equine encephalitis virus structural polypeptide.


The spatial distance between the N-terminal residue and C-terminal residue of the CD20 fragment is 10.07 custom character when the distance is determined in a crystal of CD20.


(5) Preparation of Venezuelan Equine Encephalitis Virus Like Particle Comprising a Virus Structural Polypeptide and a Fragment of Human CTLA4

A fragment of CTLA4: CKVELMYPPPYYLGIG(SEQ ID No.: 22) was selected based on the full-length CTLA4 amino acid sequence so that spatial distance between the N-terminal residue and the C-terminal residue of the fragment, which is fused into Venezuelan equine encephalitis virus structural polypeptide E2, is about 5.6 custom character when the distance is determined in a crystal of CLTA4.


A polynucleotide encoding the fragment of CTLA4 was introduced into VLP_VEEV VLP 518 vector to construct a plasmid for the expression of the fragment of CTLA4 fused with Venezuelan equine encephalitis virus structural polypeptide consisting of the amino acid sequence by SEQ ID No.:8.


(6) Preparation of Venezuelan Equine Encephalitis Virus Like Particle Comprising a Virus Structural Polypeptide and Full Length Human CTLA4

A polynucleotide encoding full length human CTLA4 was introduced into VLP_VEEV VLP 518 vector to construct a plasmid for the expression of CTLA4 fused with Venezuelan equine encephalitis virus structural polypeptide.


The spatial distance between the N-terminal residue and the C-terminal residue of full length human CTLA4 is about 39.6 custom character when the distance is determined in a crystal of CLTA4.


Expression of CTLA4 fused with Venezuelan equine encephalitis virus structural polypeptide was not be able to be detected by ELISA after transfecting 293 cells with the prepared plasmid.


(7) Preparation of Chikungunya Virus Like Particle Comprising a Virus Structural Polypeptide and a Fragment of Human or Mouse CD20 and Detection of Anti-Human or Mouse CD20 Antibody

Chikungunya virus like particles comprising a virus structural polypeptide and a fragment of human or mouse CD20 were prepared using VLP_CHI 532 vector and a fragment of human or mouse TNF alpha antibody. The fragment of human and mouse TNF alpha antibody are described below:











(SEQ ID No.: 21)










CD20 Human
IYNCEPANPSEKNSPSTQYCYSIQ;













(SEQ ID No.: 41)










CD20 Mouse
YDCEPSNSSEKNSPSTQYCNSI.







Linkers (e.g. SGG, SG, GS or GGS) were used to insert the fragment of CD20 as described below between G at 519-position and Q at 520-position of SEQ ID No.2:











CD20 Human:



(SEQ ID No.: 42)



SGGIYNCEPANPSEKNSPSTQYCYSIQGS







CD20 Mouse version2:



(SEQ ID No.: 43)



SGYDCEPSNSSEKNSPSTQYCNSIGGS







CD20 Mouse version3:



(SEQ ID No.: 44)



SGGYDCEPSNSSEKNSPSTQYCNSIGS.






Plasmids: VLP_CHI VLP 532 CD20H, VLP_CHI VLP 532 CD20-2 mouse and VLP_CHI VLP 532 CD20-3 mouse were used for the expression of Chikungunya virus like particles comprising a virus structural polypeptide and a fragment of human or mouse CD20 (SEQ ID No.: 45 (the amino acid sequence of the expressed polypeptide is represented by SEQ ID No.: 46), SEQ ID No.: 47 (the amino acid sequence of the expressed polypeptide is represented by SEQ ID No.: 48) and SEQ ID No.: 49 (the amino acid sequence of the expressed polypeptide is represented by SEQ ID No.: 50).


Virus like particles were purified according to the method as described in (1). Mice were immunized once with 100 μg of the purified VLPs; 100 μg of the fragment of human or mouse CD20; or PBS (control). Ten days after the immunization, blood sample were obtained from the mice and serum was prepared. Anti-human CD20 antibody induced by the immunization was detected by ELISA coated with the fragment of human CD20, and anti-mouse CD20 antibody induced by the immunization was detected by ELISA coated with the fragment of mouse CD20. The results showed that anti-human CD20 antibodies and anti-mouse CD20 antibodies were adequately induced by administration of the Chikungunya virus like particle comprising the fragment of human or mouse CD20 fused with virus structural polypeptide. Also, the results showed that antibody specific for a self antigen can be adequately induced by administering Chikungunya virus like particle comprising a fragment of the self antigen fused with virus structural polypeptide (see FIG. 10 and FIG. 11).

Claims
  • 1. A Chikungunya virus (CHIKV) or Venezuelan equine encephalitis virus (VEEV) virus-like particle which is capable of being self-assembled, wherein said virus-like particle comprises capsid and envelope proteins,wherein said virus-like particle comprises a Chikungunya virus (CHIKV) or Venezuelan equine encephalitis virus (VEEV) envelope protein with at least one antigen inserted therein,and wherein the spatial distance between the N-terminal residue and the C-terminal residue of the antigen is 30 Å or less.
  • 2. The virus-like particle according to claim 1, wherein said virus-like particle is a Chikungunya virus (CHIKV) virus-like particle.
  • 3. The virus-like particle according to claim 1, wherein said virus-like particle comprises capsid and envelope proteins E2 and E1.
  • 4. The virus-like particle according to claim 3, wherein said at least one antigen is inserted into said envelope protein E2.
  • 5. The virus-like particle according to claim 1, wherein said at least one antigen is a polypeptide derived from TNF-α, CD20 or CTLA4.
  • 6. The virus-like particle according claim 1, wherein said virus-like particle is selected from the group consisting of: i) a Chikungunya virus (CHIKV) virus-like particle, wherein said at least one antigen is a polypeptide derived from TNF-α;ii) a Chikungunya virus (CHIKV) virus-like particle, wherein said at least one antigen is a polypeptide derived from CD20;iii) a Venezuelan equine encephalitis virus (VEEV) virus-like particle, wherein said at least one antigen is a polypeptide derived from TNF-α;iv) a Venezuelan equine encephalitis virus (VEEV) virus-like particle, wherein said at least one antigen is a polypeptide derived from CD20; andv) a Venezuelan equine encephalitis virus (VEEV) virus-like particle, wherein said at least one antigen is a polypeptide derived from CTLA4.
  • 7. The virus-like particle according to claim 1, wherein said at least one antigen is fused to said envelope protein to form a fusion protein.
  • 8. The virus-like particle according to claim 7, wherein in said fusion protein one or two linkers intervene between the N-terminal residue of said at least one antigen and said envelope protein, and/or between the C-terminal residue of said at least one antigen and said envelope protein.
  • 9. The virus-like particle according to claim 4, wherein said at least one antigen is inserted into E2 of a CHIKV structural protein that comprises the amino acid sequence of SEQ ID NO: 1 or 2, and wherein said at least one antigen is inserted between residues 519 and 520 of SEQ ID NO: 1 or 2, between residues 530 and 531 of SEQ ID NO: 1 or 2, between residues 531 and 532 of SEQ ID NO: 1 or 2 or between residues 532 and 533 of SEQ ID NO: 1 or 2.
  • 10. The virus-like particle according to claim 7, wherein said fusion protein is a protein consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs. 4, 5, 6, 7 and 8.
  • 11. A composition comprising the virus-like particle according to claim 1.
  • 12. A vaccine composition comprising the virus-like particle according to claim 1.
  • 13. The virus-like particle according to claim 1, wherein said at least one antigen is selected from the group consisting of self antigens and cancer antigens.
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. patent application Ser. No. 13/768,801, filed Feb. 15, 2013, which claims the benefit of U.S. Provisional Patent Application No. 61/599,746 filed on Feb. 16, 2012. The entire disclosures of the prior applications are considered part of the disclosure of the accompanying continuation application, and are hereby incorporated by reference.

US Referenced Citations (27)
Number Name Date Kind
5439809 Haynes et al. Aug 1995 A
5580773 Kang Dec 1996 A
5629204 Honjo et al. May 1997 A
5698520 Honjo et al. Dec 1997 A
5939598 Kucherlapati et al. Aug 1999 A
7101550 Wood et al. Sep 2006 B2
7595048 Honjo et al. Sep 2009 B2
7790181 Platteborze et al. Sep 2010 B2
9249191 Ueno Feb 2016 B2
9363353 Chik Jun 2016 B1
20030108521 Calatrava Jun 2003 A1
20050214321 Rasochova et al. Sep 2005 A1
20070122378 Freeman et al. May 2007 A1
20090298955 Handa et al. Dec 2009 A1
20090305950 Minato et al. Dec 2009 A1
20090312190 Chinea Santiago et al. Dec 2009 A1
20110081341 Honjo et al. Apr 2011 A1
20110207223 Tang et al. Aug 2011 A1
20110262389 Mosca Oct 2011 A1
20120003266 Nable et al. Jan 2012 A1
20130251744 Ueno et al. Sep 2013 A1
20140120125 Ella et al. May 2014 A1
20140127247 Dubensky, Jr. et al. May 2014 A1
20140363458 Ueno et al. Dec 2014 A1
20150017194 Akahata et al. Jan 2015 A1
20160040134 Akahata et al. Feb 2016 A1
20160200775 Akahata et al. Jul 2016 A1
Foreign Referenced Citations (36)
Number Date Country
102321639 Jan 2012 CN
106085974 Nov 2016 CN
4-506301 Nov 1992 JP
2007-512842 May 2007 JP
2008-543774 Dec 2008 JP
9310152 May 1993 WO
9712048 Apr 1997 WO
WO-9712048 Apr 1997 WO
9941383 Aug 1999 WO
2002096939 Dec 2002 WO
2004043399 May 2004 WO
2006040334 Apr 2006 WO
2006088229 Aug 2006 WO
2007003384 Jan 2007 WO
2007059715 May 2007 WO
2007100098 Sep 2007 WO
2008025067 Mar 2008 WO
2009079185 Jun 2009 WO
2010062396 Jun 2010 WO
2011035004 Mar 2011 WO
2012006180 Jan 2012 WO
2012023995 Feb 2012 WO
2012106356 Aug 2012 WO
2012123755 Sep 2012 WO
2012172574 Dec 2012 WO
2013063248 May 2013 WO
2013122262 Aug 2013 WO
2013151764 Oct 2013 WO
2015005500 Jan 2015 WO
2015139784 Sep 2015 WO
2016021209 Feb 2016 WO
2016109792 Jul 2016 WO
2016199936 Dec 2016 WO
2016210127 Dec 2016 WO
2017009873 Jan 2017 WO
2017015463 Jan 2017 WO
Non-Patent Literature Citations (82)
Entry
Gorchakov, R., et al., 2007, Comparative analysis of the alphavirus-based vectors expressing Rift Valley fever virus glycoproteins, Virol. 366:212-225.
Akahata, W, and G. J. Nabel, 2012, A specific domain of the Chikungunya virus E2 protein regulates particle formation in human cells: Implications for alphavirus vaccine design, J. Virol. 86(16):8879-8883.
Kuo, S.-C., et al., 2012, Cell-based analysis of Chikungunya virus E1 protein in membrane fusion, J. Biomed. Sci. 19(44):1-12.
Urakami, A., et al., Jul. 2017, Development of a novel virus-like particle vaccine platform that mimics the immature form of alphavirus, Clin. Vacc. Immunol. 24(7):1-14.
Communication, dated Jun. 20, 2017, issued by the Japanese Patent Office in Japanese Patent Application No. 2014-557308.
Hevey et al., “Marburg Virus Vaccines Based upon Alphavirus Replicons Protect Guinea Pigs and Nonhuman Primates”, Virology, vol. 251, 1998, pp. 28-37. (11 pages total).
Communication, dated Jun. 6, 2017, issued by the United States Patent and Trademark Office in U.S. Appl. No. 15/299,859.
Bonaldo et al., “Surface Expression of an Immunodominant Malaria Protein B Cell Epitope by Yellow Fever Virus”, J. Mol. Biol., vol. 315, No. 4, Jan. 25, 2002, pp. 873-885. (13 pages total).
Vuola et al. “Differential Immunogenicity of Various Heterologous Prime-Boost Vaccine Regimes Using DNA and Viral Vectors in Healthy Volunteers”, The Journal of Immunology, vol. 174, No. 1, Jan. 1, 2005, pp. 449-455. (7 pages total).
Antonio Roldao et al., “Virus-like particles in vaccine development”, Expert Rev. Vaccines, 2010, 9(10): 1149-1176.
Bryce Chackerian et al., “Determinants of autoantibody induction by conjugated Papillomavirus virus-like particles”, The Journal of Immunology, 2002, 169: 6120-6126.
Communication dated Oct. 2, 2015 from the European Patent Office in counterpart European Patent Application No. 13749307.8.
Communication dated Sep. 16, 2015 from the European Patent Office in counterpart European Patent Application No. 13749307.8.
Elizabeth V.L. Grgacic et al., “Virus-like particles: Passport to immune recognition”, Methods, 2006, 40: 60-65.
Gary T. Jennings et al., “Immunodrugs: Therapeutic VLP-Based vaccines for chronic diseases”, Annu. Rev. Pharmacol. Toxicol., 2009, 49: 303-326.
Gregory J. Atkins et al., “Therapeutic and prophylactic applications of alphavirus vectors”, Expert Reviews in Molecular Medicine, 2008, 10(e33): 1-17.
Gunther Spohn et al., “A virus-like particle-based vaccine selectively targeting soluble TNF-alpha protects from arthritis without inducing reactivation of latent tuberculosis”, The Journal of Immunology, 2007, 178: 7450-7457.
Heinz Leibl et al., “Adjuvant/carrier activity of inactivated tick-borne encephalitis virus”, Vaccine, 1998, 16(4): 340-345.
Ira Mellman et al., “Cancer immunotherapy comes of age”, Nature, 2011, 480: 480-489.
Kathy D. McCoy et al., “Cytotoxic T Lymphocyte-associated Antigen 4 (CTLA-4) Can Regulate Dendritic Cell-induced Activation and Cytotoxicity of CD8+ T Cells Independently of CD4+ T Cell Help”, J. Exp. Med., 1999, 189(7): 1157-1162.
Maria Lia Palomba et al., “CD8+ T-Cell-Dependent Immunity Folling Xenogeneic DNA Immunization against CD20 in a Tumor Challenge Model of B-Cell Lymphoma”, Clinical Cancer Research, 2005, 370(11): 370-379.
Siyang Sun et al: “Structural analyses at pseudo atomic resolution of Chikungunya virus and antibodies show mechanisms of neutralization”, eLIFE, vol. 2, Apr. 2, 2013 (Apr. 2, 2013), xpnwi 194∧ D0I: 10.7554/eLife.00435 * the whole document *.
Wataru Akahata et al., “A VLP vaccine for epidemic Chikungunya virus protects non-human primates against infection”, Nat. Med., 2010, 16(3): 334-338.
Wendy K. Roberts et al., “Vaccination with CD20 peptides induces a biologically active, specific immune response in mice”, Blood, 2002, 99: 3748-3755.
Communication issued from the International Searching Authority dated May 28, 2013 from counterpart International Application No. PCT/JP2013/054422.
Communication, dated Dec. 20, 2017, issued by the European Patent Office in counterpart European Patent Application No. 15829311.8.
Urakami et al., “Development of a Novel Virus-Like Particle Vaccine Platform That Mimics the Immature Form of Alphavirus,” Clinical and Vaccine Immunology, 24(7): e00090-17 (pp. 1-14).
Adams et al. “The expression of hybrid HIV:Ty virus-like particles in yeast”, Nature. Sep. 3-9, 1987;329(6134):68-70.
Agata Y et al., “Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes”, International Immunology, 1996, vol. 8, No. 5, pp. 765-772.
Allsopp CE et al., “Comparison of numerous delivery systems for the induction of cytotoxic T lymphocytes by immunization”, Eur. J. Immunol., 1996, vol. 26, No. 8, pp. 1951-1959.
Arora U et al., “Virus-like particles displaying envelope domain III of dengue virus type 2 induce virus-specific antibody response in mice”, Vaccine, Jan. 2013, vol. 31, No. 6, p. 873-878.
Birkett A et al. “A Modified Hepatitis B Virus Core Particle Containing Multiple Epitopes of the Plasmodium falciparum Circumsporozoite Protein Provides a Highly Immunogenic Malaria Vaccine in Preclinical Analyses in Rodent and Primate Hosts”. Infection and Immunity. American Society for Microbiology. US. vol. 70. No. 12: Dec. 1, 2002. pp. 6860-6870.
Calvo-Calle et al., “A Linear Peptide Containing Minimal T- and B-Cell Epitopes of Plasmodium falciparum Circumsporozoite Protein Elicits Protection against Transgenic Sporozoite Challenge”. Infection and Immunity. Dec. 2006. p. 6929-6939. vol. 74, No. 12.
Carvalho et al. “Malaria Vaccine: Candidate Antigens, Mechanisms, Constraints and Prospects”, Scand. J. Immunol., Blackwell Science Ltd., Jul. 1, 2002, vol. 56, pp. 327-343.
Charoensri N et al. “An optimized expression vector for improving the yield of dengue virus-like particles from transfected insect cells” Journal of Virological Methods, vol. 205, 2014 (pp. 116-123).
Cox, Bryan et al. “Predicting Zika virus structural biology: Challenges and opportunities for intervention” Antiviral Chemistry and Chemotherapy, vol. 24 (3-4), 2015 (pp. 118-126).
Crompton et al, “Advances and challenges in malaria vaccine development”, Science in medicine, The Journal of Clinical Investigation, Dec. 2010, vol. 120, No. 12, pp. 4168-4178.
De Wispelaere, Melisanne, et al., “Mutagenesis of the DI/DIII Linker in Dengue Virus Envelope Protein Impairs Viral Particle Assembly”, Journal of Virology, 2012, vol. 86, No. 13, pp. 7072-7083, ISSN:0022-538X, Abstract, Fig.1, Fig.8-9, p. 7073.
Dobano C et al., “Alphavirus replicon particles are highly immunogenic in the murine Malaria model by homologous or heterologous immunization”, Open Vaccine Journal, vol. 1, 2008, pp. 27-37.
Elshuber S et al., “Cleavage of protein prM is necessary for Infection of BHK-21 cells by tick-borne encephalitis virus”, Journal of General Virology, (2003), vol. 84, pp. 183-191.
Elshuber S et al., “Resuscitating Mutations in a Furin Cleavage-Deficient Mutant of the Flavivirus Tick-Borne Encephalitis Virus”, Journal of Virology, vol. 79, No. 18, Sep. 2005, pp. 11813-11823.
António Roldão et al., “Virus-like particles in vaccine development”, Expert Reviews, Vaccines 9(10), 1149-1176, 2010.
Federico M., “Virus-like particles show promise as candidates for new vaccine strategies”, Future Virology, (2010) 5(4), 371-374.
GenBank: AAB02517.1, “Structural polyprotein precursor, Venezuelan equine encephalitis virus,” dated Nov. 17, 2004, retrieved from https://www.ncbi.nlm.nih.gov/protein/AAB02517.1.
GenBank: ADG95942.1 structural polyprotein Chikungunya virus http://www.ncbi.nlm.nih.gov/protein/296124572?report=genbank&log$=protalign&blast_rank=2&FilD=PBR7NTOU015. Dec. 28, 2010.
GenBank: AAW78190.1. circumsporozoite protein, partial Plasmodium falciparum. Dec. 29, 2006. http://www.ncbi.nlm.nih.gov/brotein/58429573?.
GenBank “Zika virus strain MR 766, complete genome” AY632535.2, Nov. 23, 2010 (6 pages total) Retrieved on May 16, 2017 Retrieved from the Internet, URL: <https://www.ncbi.nlm.nih.gov/nuccore/AY632535>.
Ghasparian A et al., “Engineered synthetic virus-like particles and their use in vaccine delivery”, Chembiochem, 2011, vol. 12, No. 1, pp. 100-109.
Gilbert SC et al., “A protein particle vaccine containing multiple Malaria epitopes”, Nat. Biotechnol., 1997, vol. 15, No. 12, pp. 1280-1284.
Gorchakov R et al.,“Comparative analysis of the alphavirus-based vectors expressing Rift Valley fever virus glycoproteins”, Virology, vol. 366, (2007), pp. 212-225.
Gregson et al., “Phase 1 Trial of an Alhydrogel Adjuvanted Hepatitis B Core Virus-Like Particle Containing Epitopes of Plasmodium falciparum Circumsperozoite Protein”. PLoS One. Feb. 2008, vol. 3, issue 2, e1556.
Haddow A. D. et al., “Genetic Characterization of Zika Virus Strains: Geographic Expansion of the Asian Lineage”, PLoS Neglected Tropical Disease, Feb. 2012, vol. 6, Issue 2, e1477 (7 pages total).
Hsieh Szu-Chia, et al., “A strong endoplasmic reticulum retention signal in the stem-anchor region envelope glycoprotein of dengue virus type 2 affects the production of virus-like particles”, Virology, 2008, vol. 374, No. 2, pp. 338-350, ISSN: 0042-6822.
Hsieh Szu-Chia, et al. “The length of and nonhydrophobic residues in the transmembrane domain of dengue virus envelope protein are critical for its retention and assembly in the endoplasmic reticulum”, Journal of Virology, vol. 84 No. 9, Apr. 2010 (pp. 4782-4797).
http://www.who.int/immunization/research/development/dengue_vaccines/en/ (3 pages).
Huang Claire Y.H., et al., “The dengue virus type 2 envelope protein fusion peptide is essential for membrane fusion”, Virology, 2010, vol. 396, No. 2, pp. 305-315, ISSN: 0042-6822, Table I, Fig. 5, pp. 310-313.
Jones RM et al., “A plant-produced Pfs25 VLP Malaria Vaccine Candidate Induces Persistent Transmission Blocking Antibodies against Plasmodium falciparum in Immunized Mice”, PLoS One, Nov. 18, 2013, vol. 8, No. 11, e79538, doi:10.1371/journal.pone.0079538.
Khetarpal Niyati, et al., “Dengue-specific subviral nanoparticles: design, creation and characterization”, Journal of Nanobiotechnology, 2013, vol. 11, No. 15, 8 pages, ISSN: 1477-3155.
Kostyuchenko V et al., “Structure of the thermally stable Zika virus,” Nature, May 19, 2016, vol. 533, pp. 425-428.
Larocca RA, et al., “Vaccine Protection Against Zika Virus from Brazil”, Nature, Aug. 25, 2016, 536(7617), 474-478, doi:10.1038/nature18952 (24 pages total).
Lechner F et al., “Virus-like particles as a modular system for novel vaccines,” Intervirology, 2002, vol. 45, No. 4-6, pp. 212-217.
Lin et al., “Analysis of Epitopes on Dengue Virus Envelope Protein Recognized by Monoclonal Antibodies and Polyclonal Human Sera by a High Throughput Assay”, PLoS, Jan. 2012, 6(1):e1447, 12 pages.
Malaria Vaccine Program, http://www.globalvaccines.org/content/malaria+vaccine+program/19614, 4 pages total (2012).
Milich D R et al. “Conversion of poorly immunogenic malaria repeat sequences into a highly immunogenic vaccine candidate”. Vaccine. Elsevier Ltd. GB: vol. 20. No. 5-6: 2002: pp. 771-788.
Notka F et al., “Accelerated clearance of SHIV in rhesus monkeys by virus-like particle vaccines is dependent on induction of neutralizing antibodies”, Vaccine, 2000, vol. 18, No. 3-4, p. 291-301.
Oliveira G et al., “Safety and enhanced immunogenicity of a Hepatitis B core practical Plasmodium falciparum Malaria vaccine formulated in adjuvant montanide ISA 720 in a Phase I Trial”, Infect. Immun., 2005, vol. 73, No. 6, pp. 3587-3597.
Oliveira-Ferreira et al., “Immunogenicity of Ty-VLP bearing a CD8+ T cell epitope of the CS protein of P. yoelii: enhanced memory response by boosting with recombinant vaccinia virus”. Vaccine. Mar. 6, 2000;18(17):1863-9.
Ozden S et al., “Inhibition of Chikungunya Virus Infection in Cultured Human Muscle Cells by Furin Inhibitors”, Journal of Biological Chemistry, vol. 283, No. 32, Aug. 8, 2008. (10 pages total).
Pfeiffer B et al., “A virosome-mimotope approach to synthetic vaccine design and optimization: synthesis, conformation, and immune recognition of a potential Malaria-vaccine candidate”, Angew. Chem. Int. Ed., 2003, vol. 42, No. 21, pp. 2368-2371.
Purdy D et al., “Secretion of noninfectious dengue virus-like particles and identification of amino acids in the stem region involved in intracellular retention of envelope protein”, Virology, 2005, vol. 333, No. 2, pp. 239-250, ISSN: 0042-6822, Abstract, Fig. 1-4. Table 1, pp. 240, 247-248.
Pushko et al., “Replicon-Helper Systems from Attenuated Venezuelan Equine Encephalitis Virus: Expression of Heterologous Genes in Vitro and Immunization against Heterologous Pathogens in Vivo”, Virology, 1997, vol. 239, pp. 389-401.
Richner Justin et al. “Modified mRNA vaccines protect against Zika Virus infection” Cell, vol. 168., Mar. 9, 2017 , pp. 1114-1125, (23 pages total).
Rodrigues M et al., “Influenza and Vaccinia viruses expressing Malaria CD8+ T and B Cell epitopes. Comparison of their immunogenicity and capacity to induce protective immunity”, J. Immunol., 1994, vol. 153, No. 10, pp. 4636-4648.
Rodriguez D et al., “Vaccine Efficacy against malaria by the Combination of Porcine Parvovirus-Like Particles and Vaccinia Virus Vectors Expressing CS of Plasmodium”, PLoS One, Apr. 17, 2012, vol. 7, No. 4, e34445.
Seligman S, “Constancy and diversity in the flavivirus fusion peptide”, http://www.virologyj.com/content/5/1/27.
Shiratsuchi T. et al. “Replacing adenoviral vector HVR1 with a malaria B cell epitope improves immunogenicity and circumvents preexisting immunity to adenovirus in mice”. Journal of Clinical Investigation: vol. 120. No. 10: Oct. 2010: pp. 3688-3701.
Taylor TJ et al. “Production of immunogenic West Nile virus-like particles using a herpes simplex virus 1 recombinant vector” Virology, vol. 496, 2016 (pp. 186-193).
Tsai et al., “Complexity of Neutralizing Antibodies against Multiple Dengue Virus Serotypes after Heterotypic Immunization and Secondary Infection Revealed by In-Depth Analysis of Cross-Reactive Antibodies”, Journal of Virology, 2015 89: 7348-7362.
Heinz et al., “Flaviviruses and flavivirus vaccines”, Vaccine 30 (2012) 4301-4306.
Yamaji H et al. “Efficient production of Japanese encephalitis virus-like particles by recombinant lepidopteran insect cells” Appl. Microbiol Biotechnol, vol. 97, 2013 (pp. 1071-1079).
Zhang et al., “Vaccination with dengue virus-like particles induces humoral and cellular immune responses in mice”, Virology Journal, 2011, 8:333, 9 pages.
Zika virus fact sheet, updated Sep. 6, 2016; URL:http://www.who.int/mediacentre/factsheets/zika/en/ ( 5 pages total).
Related Publications (1)
Number Date Country
20160090403 A1 Mar 2016 US
Provisional Applications (1)
Number Date Country
61599746 Feb 2012 US
Divisions (1)
Number Date Country
Parent 13768801 Feb 2013 US
Child 14962805 US