Antigen delivery vectors and constructs

Information

  • Patent Grant
  • 11066353
  • Patent Number
    11,066,353
  • Date Filed
    Friday, May 23, 2014
    10 years ago
  • Date Issued
    Tuesday, July 20, 2021
    2 years ago
Abstract
The present invention relates to fluorocarbon vectors for the delivery of antigens to immunoresponsive target cells. It further relates to fluorocarbon vector-antigen constructs and the use of such vectors associated with antigens as vaccines and immunotherapeutics in animals.
Description
FIELD OF THE INVENTION

The present invention relates to novel antigen delivery constructs and their use in immunisation methods. In particular, the invention relates to constructs useful in immunising against human immunodeficiency virus.


BACKGROUND OF THE INVENTION

Recent advances in our comprehension of mammalian immunological responses have led to the prevention of certain diseases in man through prophylactic vaccination and the control and treatment of diseases by the use of immunotherapeutics. The types of diseases which may be addressed through immunological intervention include those caused by infectious agents, cancers, allergies and autoimmune diseases. In these cases, most commonly, the premise of the medical treatment is the efficient delivery of antigens to appropriate immune recognition cells. For example, prophylactic vaccination has successfully eradicated smallpox worldwide through the administration of a live attenuated strain of the virus bearing all the antigens of the wild type virus. Similarly infections due to the Haemophilus influenzae serotype b bacterium have been significantly reduced in Western countries following the development of a vaccine based upon the polysaccharide antigen from the bacterial cell wall. Moreover, some cancers such as human melanoma respond to immunotherapy using autologous dendritic cells (DC) as a cellular adjuvant and defined peptides derived from the melanosomal protein gp100 as the source of tumour-specific antigen to generate a cell-mediated immune response.


Self-tolerance to autoantigen can be restored in the treatment of experimental autoimmune encephalomyelitis by injection of a specific neuroantigen that is the target of the destructive immune response. Hence specificity can be afforded by such treatment without the need for long-term immunosuppression.


For infectious diseases, the most rapid progress in disease control has occurred where antibody raised to the administered antigen is capable of neutralising the infectious agent or toxin secreted therefrom, whether this be mediated through IgM, IgG or IgA. Likewise, autoimmune diseases have been treated with antigens that can ameliorate the action of auto-antibodies. However, for the eradication of virus-infected cells, cancer cells and cells harbouring intracellular bacteria, cellular immune responses are also required. For example, intracellular viruses (e.g. retroviruses, oncornaviruses, orthomyxoviruses, paramyxoviruses, togaviruses, rhabdoviruses, arenaviruses, adenoviruses, herpesviruses, poxviruses, papovaviruses and rubella viruses) are able to replicate and spread to adjacent cells without becoming exposed to antibody. The importance of cell-mediated immunity is emphasised by the inability of children with primary T-cell deficiency to clear these viruses, whilst patients with immunoglobulin deficiency but intact cell-mediated immunity do not suffer this handicap. A small, but important, number of bacteria, fungi, protozoa and parasites survive and replicate inside host cells. These organisms include Mycobacteria (tuberculosis and leprosy), Legionella (Legionnaires Disease), Rickettsiae (Rocky Mountain spotted fever), Chlamydiae, Listeria monocytogenes, Brucella, Toxoplasma gondii, Leishmania, Trypanosoma, Candida albicans, Cryptococcus, Rhodotorula and Pneumocystis. By living inside cells, these organisms are inaccessible to circulating antibodies. Innate immune responses are also ineffective. The major immune defense against these organisms is cell-mediated immunity; involving both CD8+ cytolytic T Lymphocytes and CD4 helper T Lymphocytes.


The development of vaccines and immunotherapeutics capable of eliciting an effective and sustained cell-mediated immune response remains one of the greatest challenges in vaccinology. In particular the development of a safe and efficacious vaccine for the prevention and treatment of Human Immunodeficiency Virus (HIV) infection has been hindered by the inability of vaccine candidates to stimulate robust, durable and disease-relevant cellular immunity.


The host cell-mediated immune response responsible for eradicating intracellular pathogens or cancer cells is termed the Th1 response. This is characterised by the induction of cytotoxic T-lymphocytes (CTL) and T-helper lymphocytes (HTL) leading to the activation of immune effector mechanisms as well as immunostimulatory cytokines such as IFN-gamma and IL-2. The importance of Th 1 responses in the control of viral infection has recently been shown by Lu et al. (Nature Medicine (2004)). This clinical study with chronically HIV-1 infected individuals demonstrated a positive correlation between the suppression of viral load and both the HIV-1-specific IL-2- or IFN-gamma-expressing CD4+ T cells and specific HIV-1 CD8+ effector cell responses. Current immunological strategies to improve the cellular immunity induced by vaccines and immunotherapeutics include the development of live attenuated versions of the pathogen and the use of live vectors to deliver appropriate antigens or DNA coding for such antigens. Such approaches are limited by safety considerations within an increasingly stringent regulatory environment. Furthermore, issues arising from the scalability of manufacturing processes and cost often limit the commercial viability of products of biological origin.


In this context, rationally defined synthetic vaccines based on the use of peptides have received considerable attention as potential candidates for the development of novel prophylactic vaccines and immunotherapeutics. T cell and B cell epitopes represent the only active part of an immunogen that are recognized by the adaptive immune system. Small peptides covering T or B cell epitope regions can be used as immunogens to induce an immune response that is ultimately cross-reactive with the native antigen from which the sequence was derived. Peptides are very attractive antigens as they are chemically well-defined, highly stable and can be designed to contain T and B cell epitopes. T cell epitopes, including CTL and T helper epitopes, can be selected on the basis of their ability to bind MHC molecules in such a way that broad population coverage can be achieved (The HLA Factsbook, Marsh, S., Academic Press. 2000). Moreover, the ability to select appropriate T and B cell epitopes enable the immune response to be directed to multiple conserved epitopes of pathogens which are characterised by high sequence variability (such as HIV, hepatitis C virus (HCV), and malaria).


In order to stimulate T lymphocyte responses, synthetic peptides contained in a vaccine or an immunotherapeutic product should preferably be internalized by antigen presenting cells and especially dendritic cells. Dendritic cells (DCs) play a crucial role in the initiation of primary T-cell mediated immune responses. These cells exist in two major stages of maturation associated with different functions. Immature dendritic cells (iDCs) are located in most tissues or in the circulation and are recruited into inflamed sites. They are highly specialised antigen-capturing cells, expressing large amounts of receptors involved in antigen uptake and phagocytosis. Following antigen capture and processing, iDCs move to local T-cell locations in the lymph nodes or spleen. During this process, DCs lose their antigen-capturing capacity turning into immunostimulatory mature Dcs (mDCs).


Dendritic cells are efficient presenting cells that initiate the host's immune response to peptide antigen associated with class I and class II MHC molecules. They are able to prime naïve CD4 and CD8 T-cells. According to current models of antigen processing and presentation pathways, exogeneous antigens are internalised into the endocytic compartments of antigen presenting cells where they are degraded into peptides, some of which bind to MHC class II molecules. The mature MHC class II/peptide complexes are then transported to the cell surface for presentation to CD4 T-lymphocytes. In contrast, endogenous antigen is degraded in the cytoplasm by the action of the proteosome before being transported into the cytoplasm where they bind to nascent MHC class I molecules. Stable MHC class I molecules complexed to peptides are then transported to the cell surface to stimulate CD8 CTL. Exogenous antigen may also be presented on MHC class I molecules by professional APCs in a process called cross-presentation. Phagosomes containing extracellular antigen may fuse with reticulum endoplasmic and antigen may gain the machinery necessary to load peptide onto MHC class I molecules. It is well recognised, however, that free peptides are often poor immunogens on their own (Fields Virology, Volume 1, Third Edition, 1996).


To optimise the efficacy of peptide vaccines or therapeutics, various vaccine strategies have been developed to direct the antigens into the antigen-presenting cell in order to target the MHC class I pathway and to elicit cytotoxic T-lymphocyte (CTL) responses. As an example of a synthetic delivery system, fatty acyl chains have been covalently to linked to peptides as a means of delivering an epitope into the MHC class I intracellular compartment in order to induce CTL activity. Such lipopeptides, for example with a monopalmitoyl chain linked to a peptide representing an epitope from HIV Env protein are described in the U.S. Pat. No. 5,871,746. Other technologies have been delivered that aim to deliver epitopes into the intracellular compartment and thereby induce CTLs. These include vectors such as Penetratin, TAT and its derivatives, DNA, viral vectors, virosomes and liposomes. However, these systems either elicit very weak CTL responses, have associated toxicity issues or are complicated and expensive to manufacture at the commercial scale.


There is therefore a recognised need for improved vectors to direct the intracellular delivery of antigens in the development of vaccines and drugs intended to elicit a cellular immune response. A vector in the context of immunotherapeutics or vaccines is any agent capable of transporting or directing an antigen to immune responsive cells in a host. Fluorinated surfactants have been shown to have lower critical micellar concentrations than their hydrogenated counterparts and thus self-organise into micelle structures at a lower concentration than the equivalent hydrocarbon molecule. This physicochemical property is related to the strong hydrophobic interactions and low Van der Waal's interactions associated with fluorinated chains which dramatically increase the tendency of fluorinated amphiphiles to self-assemble in water and to collect at interfaces. The formation of such macromolecular structures facilitates their endocytic uptake by cells, for example antigen-presenting cells (Reichel F. et al. J. Am. Chem. Soc. 1999, 121, 7989-7997). Furthermore haemolytic activity is strongly reduced and often suppressed when fluorinated chains are introduced into a surfactant (Riess, J. G.; Pace, S.; Zarif, L. Adv. Mater. 1991, 3, 249-251) thereby leading to a reduction in cellular toxicity.


SUMMARY OF THE INVENTION

This invention seeks to overcome the problem of delivering antigens to immune responsive cells by using a novel fluorocarbon vector in order to enhance the immunogenicity of administered antigens. The fluorocarbon vector may comprise one or more chains derived from perfluorocarbon or mixed fluorocarbon/hydrocarbon radicals, and may be saturated or unsaturated, each chain having from 3 to 30 carbon atoms. In order to link the vector to the antigen through a covalent linkage, a reactive group, or ligand, is incorporated as a component of the vector, for example —CO—, —NH—, S, O or any other suitable group is included; the use of such ligands for achieving covalent linkages are well-known in the art. The reactive group may be located at any position on the fluorocarbon molecule. Coupling of the fluorocarbon vector to the antigen may be achieved through functional groups such as —OH, —SH, —COOH, —NH2 naturally present or introduced onto any site of the antigen. Examples of such linkages include amide, hydrazone, disulphide, thioether and oxime bonds. Alternatively, non-covalent linkages can be used, for example an ionic interaction may be formed via a cation linking together a histidine residue of a peptide antigen and a carboxylic acid on the fluorocarbon vector. Optionally, a spacer element (peptidic or non-peptidic) may be incorporated to permit cleavage of the antigen from the fluorocarbon element for processing within the antigen-presenting cell and to optimise steric presentation of the antigen. The spacer may also be incorporated to assist in the synthesis of the molecule and to improve its stability and/or solubility. Examples of spacers include polyethylene glycol (PEG), amino acids such as lysine or arginine that may be cleaved by proteolytic enzymes and hydrocarbons.


Thus, in a first aspect, the present invention provides a fluorocarbon vector having a chemical structure or derivatives thereof, where m=3 to 30, n<=2m+1, y=0 to 15, x<=2y, (m+y)=3 −30 and L is a ligand to facilitate covalent attachment to an antigen.


In the context of the present invention “derivatives” refers to relatively minor modifications of the fluorocarbon compound such that the compound is still capable of delivering the antigen as described herein. Thus, for example, a number of the fluorine moieties can be replaced with other halogen moieties such as Cl, Br or I. In addition it is possible to replace a number of the fluorine moieties with methyl groups and still retain the properties of the molecule as discussed herein.


In a particular embodiment of the above formula the vector may be perfluoroundecanoic acid of the following formula (I):




embedded image



or alternatively 2H, 2H, 3H, 3H-perfluoroundecanoic acid of the following formula (II):




embedded image



or heptadecafluoro-pentadecanoic acid of the following formula (III):




embedded image


In a second aspect the invention provides a vector-antigen construct CmFn-CyHx-(Sp)-R where Sp is an optional chemical spacer moiety and R is an antigen.


The antigen associated with the vector may be any antigen capable of inducing an immune response in an animal, including humans Preferably the immune response will have a beneficial effect in the host. Antigens may be derived from a virus, bacterium or mycobacterium, parasite, fungus, or any infectious agent or an autologous antigen or allergen.


Examples of viruses include, but are not limited to, Human Immunodeficiency Virus-1 (HIV-1) or -2, influenza virus, Herpes virus HSV-1 and HSV-2), hepatitis A virus (HAV), hepatitis B virus (HBV), or hepatitis C virus (HCV).


Examples of bacteria and mycobacteria include, but are not limited to Mycobacterium tuberculosis, Legionella, Rickettsiae, Chlamydiae, and Listeria monocytogenes.


Examples of parasites include, but are not limited to Plasmodium falciparum and other species of the Plasmodial family.


Examples of fungi include, but are not limited to Candida albicans, Cryptococcus, Rhodotorula and Pneumocystis.


Autologous or self-antigens include, but are not limited to the following antigens associated with cancers, HER-2/neu expressed in breast cancer, gp 100 or MAGE-3 expressed in melanoma, P53 expressed in colorectal cancer, and NY-ESO-1 or LAGE-1 expressed by many human cancers.


Allergens include, but are not limited to. phospholipase A2 (API ml) associated with severe reactions to bee, Derp-2, Der p 2, Der f , Der p 5 and Der p 7 associated with reaction against the house-dust mite Dermatophagoides pteronyssinus, the cockroach allergen Bla g 2 and the major birch pollen allergen Bet v 1.


Thus in a embodiment, the present invention provides a vector-antigen construct where the antigen is, or represents, an antigen from a virus, bacterium, mycobacterium, parasite, fungus, autologous protein or allergen.


Antigens may be proteins, protein subunits, peptides, carbohydrates, lipid or combinations thereof, provided they present an immunologically recognisable epitope. Such antigens may be derived by purification from the native protein or produced by recombinant technology or by chemical synthesis. Methods for the preparation of antigens are well-known in the art. Furthermore antigens also include DNA or oligonucleotide encoding an antigenic peptide or protein.


Thus in yet a further embodiment, the present invention provides a vector-antigen construct where the antigen is a protein, protein subunit, peptide, carbohydrate or lipid or combinations thereof.


For the construct to be immunologically active the antigen must comprise one or more epitopes. Peptides or proteins used in the present invention preferably contain a sequence of at least seven, more preferably between 9 and 100 amino-acids and most preferably between around 15 to 35 amino acids. Preferably, the amino acid sequence of the epitope(s) bearing peptide is selected to enhance the solubility of the molecule in aqueous solvents. Furthermore, the terminus of the peptide which does not conjugate to the vector may be altered to promote solubility of the construct via the formation of multimolecular structures such as micelles, lamellae, tubules or liposomes. For example, a positively charged amino acid could be added to the peptide in order to promote the spontaneous assembly of micelles. Either the N-terminus or the C-terminus of the peptide can be coupled to the vector to create the construct. To facilitate large scale synthesis of the construct, the N- or C-terminal amino acid residues of the peptide can be modified. When the desired peptide is particularly sensitive to cleavage by peptidases, the normal peptide bond can be replaced by a noncleavable peptide mimetic; such bonds and methods of synthesis are well known in the art.


As a specific example, the peptide NNTRKRIRIQRGPGRAFVTIGK-NH2 (SEQ ID NO: 37) represents an epitope from the Env (301-322) protein of HIV-1, which has been shown to be immunologically active. This represents yet another embodiment of the present invention. (Reference http://www.hiv.lanl.gov/content/immunology/index.html).


More than one antigen may be linked together prior to attachment to the ligand. One such example is the use of fusion peptides where a promiscuous T helper epitope can be covalently linked to one or multiple CTL epitopes or one or multiple B cell epitope which can be a peptide, a carbohydrate, or a nucleic acid. As an example, the promiscuous T helper epitope could be the PADRE peptide, tetanus toxoid peptide (830-843) or influenza haemagglutinin, HA (307-319).


In another embodiment therefore, the vector-antigen construct is one where R is more than one epitope or antigen linked together. Epitopes may also be linear overlapping thereby creating a cluster of densely packed multi-specific epitopes.


Due to the strong non-covalent molecular interactions characteristic to fluorocarbons, the antigen may also be non-covalently associated with the vector and still achieve the aim of being favourably taken up by antigen-presenting cells


The present invention also provides vaccines and immunotherapeutics comprising one or more fluorocarbon vector-antigen constructs. Multi-component products of this type are desirable since they are likely to be more effective in eliciting appropriate immune responses. For example, the optimal formulation of an HIV immunotherapeutic may comprise a number of epitopes from different HIV proteins. In this case each epitope may be linked to a common fluorocarbon vector or each epitope could be bound to a dedicated vector. Alternatively, multiple epitopes may be incorporated into a formulation in order to confer immunity against a range of pathogens. A multi-component product may contain one or more vector-antigen construct, more preferably 2 to about 20, more preferably 3 to about 8 such constructs.


Compositions of the invention comprise fluorocarbon vectors associated to antigens optionally together with one or more pharmaceutically acceptable carriers and/or adjuvants. Such adjuvants, capable of further potentiating the immune response, may include, but are not limited to, muramyldipeptide (MDP) derivatives, CpG, monophosphoryl lipid A, oil in water adjuvants, water-in-oil adjuvants, aluminium salts, cytokines, immunostimulating complex (ISCOMs), liposomes, microparticules, saponins, cytokines, or bacterial toxins and toxoids. Other useful adjuvants will be well-known to one skilled in the art. The choice of carrier if required is frequently a function of the route of delivery of the composition. Within this invention, compositions may be formulated for any suitable route and means of administration. Pharmaceutically acceptable carriers or diluents include those used in formulations suitable for oral, ocular, rectal, nasal, topical (including buccal and sublingual), vaginal or parenteral (including subcutaneous, intramuscular, intravenous, intradermal) administration.


The formulation may be administered in any suitable form, for example as a liquid, solid, aerosol, or gas. For example, oral formulations may take the form of emulsions, syrups or solutions or tablets or capsules, which may be enterically coated to protect the active component from degradation in the stomach. Nasal formulations may be sprays or solutions. Transdermal formulations may be adapted for their particular delivery system and may comprise patches. Formulations for injection may be solutions or suspensions in distilled water or another pharmaceutically acceptable solvent or suspending agent. Thus in a further aspect, the present invention provides a prophylactic or therapeutic formulation comprising the vector-antigen construct with or without a suitable carrier and/or adjuvant.


The appropriate dosage of the vaccine or immunotherapeutic to be administered to a patient will be determined in the clinic. However, as a guide, a suitable human dose, which may be dependent upon the preferred route of administration, may be from 1 to 1000 μg. Multiple doses may be required to achieve an immunological effect, which, if required, will be typically administered between 2 to 12 weeks apart. Where boosting of the immune response over longer periods is required, repeat doses 3 months to 5 years apart may be applied.


The formulation may combine the vector-antigen construct with another active component to effect the administration of more than one vaccine or drug. A synergistic effect may also be observed through the co-administration of the two or more actives. In the treatment of HIV infection, an example of one such drug is Highly Active Anti-Retroviral Therapy (HAART).


In other aspects the invention provides:


i) Use of the immunogenic construct as described herein in the preparation of a medicament for treatment or prevention of a disease or symptoms thereof.


ii) A method of treatment through the induction of an immune response following administration of the constructs or formulations described herein;


iii) The use of the fluorocarbon vectors and fluorocarbon vector-antigen constructs in medicine.





BRIEF DESCRIPTION OF THE DRAWINGS

The examples refer to the figures in which:



FIG. 1: shows HPLC chromatograms of various peptides and constructs at T=0;



FIG. 2: shows HPLC chromatograms of various peptides and constructs stored at 40° C. for 27 days;



FIG. 3: shows critical micelle concentration evaluation for two peptides, FAVS-3-ENV and FAVS-1-ENV;



FIG. 4: shows particle size analysis by quasi light scattering spectrometry after 20 hours standing for various peptide constructs;



FIG. 5: shows cellular immune response assessed by ex vivo IFN-gamma ELISPOT assay in mice after single immunisation (A,B), first boost (C,D) and second boost (E,F);



FIG. 6 shows nature of T lymphocytes primed in vivo by various fluorocarbon-peptide constructs;



FIG. 7: shows cellular immune response assessed by ex vivo IFN-g ELISPOT assay in mice after three immunisations with FAVS-1-ENV alone or in combination with murabutide;



FIG. 8: cytokine measurement after three injections with FAVS-1-ENV alone or in combination with murabutide; and



FIG. 9: shows cellular immune response assessed by ex vivo IFN-g ELISPOT assay in mice after two intranasal administrations with FAVS-1-ENV alone or in combination with murabutide.





DETAILED DESCRIPTION
Example 1
Synthesis of Fluorocarbon-Vectored Peptides

The following fluorocarbon-vector peptides were synthesised:









FAVS-1-ENV:


(SEQ ID NO: 38)


NNTRKRIRIQRGPGRAFVTIGK-C8F17(CH2)2CO-K-NH2





FAVS-2-ENV: 


(SEQ ID NO: 38)


NNTRKRIRIQRGPGRAFVTIGK-C8F17(CH2)6CO-K-NH2





FAVS-3-ENV: 


(SEQ ID NO: 39)


IRIQRGPGRAFVTIGKK-CO(CH2)2-(PEG)4-C8F17(CH2)6CO-K-NH2







Where the standard amino acid one letter code is utilised and PEG is CH2-CH2-O. NNTRKRIRIQRGPGRAFVTIGK (SEQ ID NO: 37) is the ENV(301-322) peptide of the Human Immunodeficiency Virus.


Peptide synthesis was carried out on an ABI 430 or ABI 433 automatic peptide synthesizer, on Rink amide resin (0.38 mmol/g loading) using Nsc (2-(4-nitrophenylsulfonyl)ethoxycarbonyl), or Fmoc ((9-fluorenylmethylcarbonyl) amino acids. Coupling was promoted with HOCt (6-Chloro-1-oxybenzotriazole) and DIC (1,3-diisopropylcarbodiimide), and Fmoc/Nsc deprotection was carried out using 20% piperidine in DMF (Dimethylformamide). Uncoupled N-termini were capped with acetic anhydride as part of each cycle. Cleavage of the peptide from resin and concomitant side-chain deprotection was achieved using TFA, water and TIS (Diisopropylsilane) (95:3:2), with crude isolation of product by precipitation into cold diethyl ether. Purification was performed by preparative HPLC using Jupiter C5 or Luna C18 (2) columns (250×22 mm) and peptide mass was verified by mass spectrometry.


Peptide purity was verified prior to conducting the experiments by HPLC(HP 1050) using a column from Supelco (C5, 250×4.6 mm, 300 A, 5 μm) under gradient elution. Solvent A (90% Water, 10% Acetonitrile, 0.1% TFA), Solvent B (10% Water, 90% Acetonitrile, 0.1% TFA). A gradient 0 to 100% of B in 30 minutes was used and column temperature was 40° C. The wavelength of the UV detector was set up at 215 nm. Purity of the fluorocarbon-vector peptides in each case was greater than 90%.


The chemical stability of hermetically sealed samples containing lyophilised vector-peptides was assessed at 4° C., 20° C. and 40° C. together with the unvectored peptide as a comparator (NNTRKRIRIQRGPGRAFVTIGK-NH2 (SEQ ID NO: 37)). The stability over the time was monitored by HPLC using the conditions described above. The data is shown in FIGS. 1 and 2.


For each peptide conjugate, no sign of degradation was observed after 27 days at 40° C. incubation, with a single peak eluting at the same retention time as found at T=0.


Example 2

Physicochemical analysis of Fluorocarbon-vectored peptides


(i) Solubility


The solubility of the fluorocarbon-vector peptides in aqueous solution at concentrations useful for a pharmaceutical formulation was confirmed. Solutions of peptides were prepared at 20° C. by dissolving the lyophilised peptide powder with PBS (0.01M, pH 7.2) across a range of concentrations. Preparations were then vortexed for one minute. An aliquot was collected and the remainder of the solution was centrifuged for 10 minutes at 12,000 rpm. To a 96-well flat bottom plate containing 25 μl aliquots of serial dilutions of each peptide was added 200 μl of the BCA working reagent (Pierce, UK) containing the solution A (bicichoninic acid, sodium carbonate, sodium tartrate in a sodium hydroxyde 0.1M solution, 50 vol,) and B (4% cupric sulphate solution, 1 vol.). After incubating for 45 minutes at 37° C. and cooling for 10 minutes, the absorbance was measured at 570 nm. The plates were analysed by a Wallac Victor multilabel counter (Perkin Elmer). For each peptide a calibration curve was plotted and used to determine the peptide concentration in the soluble fraction, expressed in nmol/ml. Data are presented Table 1. All the peptides were found to be fully soluble at the concentration of antigen used for murine immunisation studies.









TABLE 1







Summary of the solubility assay performed by the protein assay method








Peptide
Solubility





Free peptide
>3300 nmol/ml


FAVS-1-ENV
>4000 nmol/ml


FAVS-2-ENV
 >500 nmol/ml


FAVS-3-ENV
>3000 nmol/ml










(ii) Critical Micelle Concentration [CMC]


The Critical Micelle Concentration of the fluorocarbon-vectored peptides in physiological phosphate buffered saline was determined by dye bonding with 8-anilino-1-naphthalene-sulphonic acid (ANS). Starting from 300 μg peptide/ml solutions, serial two-fold dilutions of the peptide and peptide-vector solutions in PBS (0.01M, pH 7.2) were prepared at 20° C., from which 200 μl were added to the wells of a microplate. 40 μl of freshly dissolved ANS in PBS was then added to each well. After two minutes the plate was excited at 355 nm and scanned at 460 nm on a Victor microplate fluorimeter. The ratio (Intensity of fluorescence of the sample/Intensity of fluorescence of the blank) was plotted on a linear scale versus the concentration on a logarithmic scale. Data are presented FIG. 3.


(iii) Particle Size Analysis


Particle size analysis was performed on a Malvern 4700C Quasi Light Scattering spectrometer (Malvern Ltd, UK) equipped with an Argon laser (Uniphase Corp., San Jose, Calif.) tuned at 488 nm. Samples were maintained at a temperature of 25° C. The laser has variable detector geometry for angular dependence measurement. Measurements were performed at angles of 90° and 60°. Solutions were prepared by dissolving the peptide in filtered 0.01M phosphate buffered saline to a concentration of 500 nmol/ml and vortexing for 1 minute. Solutions were then dispensed into cuvettes (working volume of 1 ml). Measurements were taken after 15 minutes at an angle of 90° (FIG. 4). The Kcount value output is proportional to the number of particles detected; in all cases the Kcount was >10 in order to ensure that reliable size distribution measurements were obtained.









TABLE 2







Particle size of micellar solution in PBS.













Standing







Time

size (nm)
Average













ITS reference
(h)
Kcount
Population1
Population2
size (nm)
Polydispersity
















FAVS-1-ENV
0.25
177
28

28.3
0.151



20
230
32

32.7
0.180


FAVS-2-ENV
0.25
190
15
120
28.5
0.450



20
245
20
300
68.4
0.539


FAVS-3-ENV
0.25
201
70
400
209
0.659



20
225
105
800
207
0.647









Example 3
(i) Immunogenicity of Fluorocarbon-Vectored Peptides

Specific-pathogen-free mice (6-8 week female Balb/c) were purchased from Harlan (UK). Peptides ENV, FAVS-1-ENV, FAVS-2-ENV or FAVS-3-ENV were dissolved in PBS (0.01M, pH 7.2). Each dose was normalised to 50 nmol peptide per ml based on the net peptide content obtained from amino-acid analysis. Mice (3 per group) were immunized subcutaneously under the skin of the interscapular area with 50 nmol peptide in a volume of 100 μl PBS, pH 7.2. Three doses were administered at ten day intervals. A mouse group receiving a priming dose of free peptide admixed with Complete Freund's adjuvant (50 nmol peptide in PBS emulsified in an equal volume of adjuvant) and booster doses of Incomplete Freund's adjuvant served as a positive control. Ten days after the final immunisation mice were sacrificed and spleens removed to assess the cellular immune response to the peptide. To determine the progress of the immune response development, groups of mice receiving a single and two doses of peptide were also set up.


The in vivo cellular response primed by the vectored peptides was monitored by IFN-gamma ELISPOT on fresh spleen cells in order to enumerate the ex-vivo frequency of peptide-specific IFN-gamma producing cells and more specifically peptide-specific CD8+T lymphocytes primed following immunisation. Spleen cells were restimulated in vitro with the ENV(301-322) NNTRKRIRIQRGPGRAFVTIGK (SEQ ID NO: 37) peptide containing a well-known T-helper epitope and ENV(311-320) RGPGRAFVTI (SEQ ID NO: 40) a shorter peptide corresponding to the CD8 epitope (MHC class I H-2Dd-restricted known as P18-I10) in order to cover both components of the cellular immune response (T Helper and CD8 T cell activity).


The spleens from each group of mice were pooled and spleen cells isolated. Cells were washed three times in RPMI-1640 before counting. Murine IFN-g Elispot assays were performed using Diaclone Kit (Diaclone, France) according to the manufacturer's instructions with the following modifications. Duplicate culture of spleen cells at cell density of 5×105/well were distributed in anti-IFN-gamma antibody coated PVDF bottomed-wells (96-well multiscreen™-IP microplate-Millipore) with the appropriate concentration of peptide (10μ, 1, 0 mg/ml of T helper EN-(301-322) or P18-I10 CTL epitope) in culture medium (RPMI-1640), 5 μM β-mercaptoethanol, 5 mM glutamine supplemented with 10% Foetal Calf Serum during 18 hours at 37° C. under 5% CO2 atmosphere. The spots were counted using a Carl Zeiss Vision ELIspot reader unit. The results correspond to mean values obtained with each conditions after background subtraction. Results are expressed as spot forming units (SFC) per million input spleen cells (FIG. 5).


(ii) Nature of T Lymphocytes Primed in vivo by the Fluorocarbon-Peptides (CD4 and CD8 T Cell Separation)

Spleen Cells from immunized mice were distributed in 48-well microplates at cell density of 2.5×106/well with 1 μg/ml of T helper ENV(301-322) or P18-I10 CTL peptides. At day 3, 5 ng/ml of recombinant murine IL-2 was added to each well. At day 7, pre-stimulated spleen cells were harvested, washed three times in RPMI 1640, counted and separated by magnetic cell sorting using magnetic beads conjugated with monoclonal rat anti-mouse CD8a and CD4 antibodies (MACS, Microbeads Miltenyi Biotec, UK) according to manufacturer's instructions. CD4 and CD8+ T cells were distributed at cell density of 2.5×105/well in duplicate in antibody coated PVDF bottomed-wells (96-well multiscreen™-IP microplate, Millipore) with 1 mg/ml of peptide in culture medium (RPMI-1640, 5 μM β-mercaptoethanol, Glutamine, non-essential amino-acids, sodium pyruvate supplemented with 10% Foetal Calf Serum for 12 hours at 37° C. under 5% CO2 atmosphere. The spots were counted using a Carl Zeiss Vision ELIspot reader unit. The results correspond to mean values obtained with each conditions after background subtraction (<10 spots). Results are expressed as spot forming units (SFC) per million input spleen cells.


According to the ex vivo IFN-γ ELISPOT assays, the FAVS-peptide constructs were able to prime a strong cellular immune response against both the long (ENV301-322) and the short ENV peptides (P18-I10 CTL epitope) after a single in vivo exposure to the antigen (FIGS. 5 A and B). FIG. 6 demonstrates that both CD4+ and CD8+ ENV-specific T cells were efficiently primed in vivo.


The intensity of the response after priming with the FAVS-peptides was in the same range as the responses obtained from mice immunized with the native peptide emulsified in Freund's adjuvant. ENV-specific T cell responses are clearly amplified after a first and a second boost with the FAVS-1-ENV formulation (FIG. 5C, D, E, F) as summarized in FIG. 6.


This clearly demonstrates the ability of the FAVS-peptides to be taken up by antigen presenting cells in vivo in order to reach the MHC class I and MHC class II pathways and thereby prime strong cellular immune responses.


Example 4
Immunogenicity of Fluorocarbon-Vectored Peptides Co-Administered with Synthetic Adjuvant

In order to assess the potential impact of a synthetic immunostimulant on the quantitative and qualitative immunogenicity of the FAVS-peptides, FAVS-1-ENV was injected alone and in combination with Murabutide. Murabutide (N-acetyl-muramyl-L-alanyl-D-glutamine-O-n-butyl-ester; a synthetic derivative of muramyl dipeptide and NOD-2 agonist) is a synthetic immune potentiator that activates innate immune mechanisms and is known to enhance both cellular and humoral responses when combined with immunogens (“Immune and antiviral effects of the synthetic immunomodulator murabutide: Molecular basis and clinical potential”, G. Bahr, in: “Vaccine adjuvants: Immunological and Clinical Principles”, eds Hacket and Harn (2004), Humana Press).


Specific-pathogen-free mice (6-8 week female Balb/c) were purchased from Harlan (UK). The FAVS-1-ENV construct was used at two different dose levels, one group of mice receiving 50 nmoles and a second group received 5 nmoles of construct. Mice (3 per group) were immunized subcutaneously under the skin of the interscapular area with FAVS-1-ENV either alone or in combination with 100 μg of Murabutide in a total volume of 100 μl PBS, pH 7.2. Three doses were administered at ten day intervals. A control group receiving murabutide alone was also set up.


Ten days after the final immunisation mice were sacrificed and spleens removed to assess the cellular immune response to the T helper ENV(301-322) or P18-I10 CTL epitope peptides. Interferon-gamma Elispot and Th-1 and Th-2 cytokine measurements were performed on the isolated spleens as described in Example 3. Briefly, spleen cells were cultured with the appropriate concentration of peptide (10 or 0 μg/ml of T helper ENV (301-322) or P18-I10 CTL epitope) in culture medium during 18 hours at 37° C. under 5% CO2 atmosphere. IFN-g Elispot assay was then performed. The spots were counted using a Carl Zeiss Vision Elispot reader unit. The results correspond to mean values obtained with each conditions after background subtraction (<10 spots). Results are expressed as spot forming units (SFC) per million input spleen cells (FIG. 7).


Multiplex cytokine measurements (IL-2, IFN-g, IL4, IL5, IL-10, IL-13) were performed on fresh spleen cells re-stimulated with the ENV (301-322) peptide from mice immunised with the 5 nmol dose of FAVS-1-ENV. Supernatants were collected at 24 hours and 48 hours. Levels of cytokines (IL2, IL4, IL-5, IL-10, IL-13, IFN-γ) in cell culture supernatant samples were measured using the Cytokine specific Sandwich ELISA according to the mutiplex format developed by SearchLight™ Proteomic Arrays (Pierce Biotechnology, Woburn, Mass.). Results were expressed in pg cytokine/ml.


FAVS-1-ENV administered alone was shown to induce predominantly Th-1 cytokine production (i.e. IL-2 and IFN-g) with low levels of Th-2 cytokines also being produced. The inclusion of murabutide within the formulation led to the induction of a more balanced Th-1/Th-2 response with higher levels of Th-2 cytokines such as IL-5, IL-10 and IL-13 (FIG. 8).


Example 5
Immunogenicity of Fluorocarbon-Vectored Peptides Administered Mucosally

Specific-pathogen-free mice (6-8 week female Balb/c) were purchased from Harlan (UK).


FAVS-1-ENV (50 nmoles per mouse) was administered twice intranasally in 0.01M PBS alone or in combination with 100 μg of Murabutide with 10 days interval between both administration. Mice were slightly anaesthetised with Isoflurane (Isoflo, Solvay, UK). 20 μl of soluble peptide solution (10 μl/nostril) was administered using a micropipette. A control group received PBS only. Each dosing group comprised six animals. Mice were sacrificed 10 days after the last administration by carbon dioxide asphyxiation. Spleens were removed, pooled for each group of mice and spleen cells were isolated. Cells were washed three times with RPMI-1640 before counting. Counting was performed using a Thomas counting slide. Spleen cells from individual mice were cultured with the appropriate concentration of peptide (10 or 0 μg/ml of T helper ENV (301-322) or P18-I10 CTL epitope) in culture medium during 18 hours at 37° C. under 5% CO2 atmosphere. IFN-g Elispot assay was then performed using the Diaclone Kit as described in Example 3. The spots were counted using a Carl Zeiss Vision Elispot reader unit. The results correspond to mean values obtained with each conditions after background subtraction (<10 spots). Results are expressed as spot forming units (SFC) per million input spleen cells. The data represent the average for 6 mice.


All six mice per group immunised intranasally either with FAVS-1-ENV alone or in combination with murabutide produced a robust systemic T-cell response. Combination with murabutide led to modest increases in the frequency of IFN-gamma producing T cells (FIG. 9).


Example 6
Example HIV Peptides

Candidate peptides for attachment to the fluorocarbon vector to produce a prophylactic or therapeutic vaccine for HIV may include the following one or more peptides or fragments thereof, or homologues (including the corresponding consensus, ancestral or central tree sequences from HIV-1 representing different clades such as but not limited to clades A, B, C, D, F, G and H as referred to in the 2004 Los Alamos National Laboratory database) or natural and non-natural variants thereof, but not necessarily exclusively. The standard one letter and three-letter amino acid codes have been utilised. Homologues have at least a 50% identity compared to a reference sequence. Preferably a homologue has 80, 85, 90, 95, 98 or 99% identity to a naturally occurring sequence. The sequences provided below are 35 amino acids in length. Fragments of these sequences that contain one or more epitopes are also candidate peptides for attachment to the fluorocarbon vector.









SEQ ID No 1


WKGEGAVVIQDNSDIKVVPRRKAKIIRDYGKQMAG


Trp-Lys-Gly-Glu-Gly-Ala-Val-Val-Ile-Gln-Asp-Asn-


Ser-Asp-Ile-Lys-Val-Val-Pro-Arg-Arg-Lys-Ala-Lys-


Ile-Ile-Arg-Asp-Tyr-Gly-Lys-Gln-Met-Ala-Gly





SEQ ID No 2


EIYKRWIILGLNKIVRMYSPTSILDIRQGPKEPFR


Glu-Ile-Tyr-Lys-Arg-Trp-Ile-Ile-Leu-Gly-Leu-Asn-


Lys-Ile-Val-Arg-Met-Tyr-Ser-Pro-Thr-Ser-Ile-Leu-


Asp-Ile-Arg-Gln-Gly-Pro-Lys-Glu-Pro-Phe-Arg





SEQ ID No 3


EHLKTAVQMAVFIHNFKRKGGIGGYSAGERIVDII


Glu-His-Leu-Lys-Thr-Ala-Val-Gln-Met-Ala-Val-Phe-


Ile-His-Asn-Phe-Lys-Arg-Lys-Gly-Gly-Ile-Gly-Gly-


Tyr-Ser-Ala-Gly-Glu-Arg-Ile-Val-Asp-Ile-Ile





SEQ ID No 4


WEFVNTPPLVKLWYQLEKEPIVGAETFYVDGAANR


Trp-Glu-Phe-Val-Asn-Thr-Pro-Pro-Leu-Val-Lys-Leu-


Trp-Tyr-Gln-Leu-Glu-Lys-Glu-Pro-Ile-Val-Gly-Ala-


Glu-Thr-Phe-Tyr-Val-Asp-Gly-Ala-Ala-Asn-Arg





SEQ ID No 5


GERIVDIIATDIQTKELQKQITKIQNFRVYYRDSR


Gly-Glu-Arg-Ile-Val-Asp-Ile-Ile-Ala-Thr-Asp-Ile-


Gln-Thr-Lys-Glu-Leu-Gln-Lys-Gln-Ile-Thr-Lys-Ile-


Gln-Asn-Phe-Arg-Val-Tyr-Tyr-Arg-Asp-Ser-Arg





SEQ ID No 6


FRKYTAFTIPSINNETPGIRYQYNVLPQGWKGSPA


Phe-Arg-Lys-Tyr-Thr-Ala-Phe-Thr-Ile-Pro-Ser-Ile-


Asn-Asn-Glu-Thr-Pro-Gly-Ile-Arg-Tyr-Gln-Tyr-Asn-


Val-Leu-Pro-Gln-Gly-Trp-Lys-Gly-Ser-Pro-Ala





SEQ ID No 7


NWFDITNWLWYIKIFIMIVGGLIGLRIVFAVLSIV


Asn-Trp-Phe-Asp-Ile-Thr-Asn-Trp-Leu-Trp-Tyr-Ile-


Lys-Ile-Phe-Ile-Met-Ile-Val-Gly-Gly-Leu-Ile-Gly-


Leu-Arg-Ile-Val-Phe-Ala-Val-Leu-Ser-Ile-Val





SEQ ID No 8


ENPYNTPVFAIKKKDSTKWRKLVDFRELNKRTQDF


Glu-Asn-Pro-Tyr-Asn-Thr-Pro-Val-Phe-Ala-Ile-Lys-


Lys-Lys-Asp-Ser-Thr-Lys-Trp-Arg-Lys-Leu-Val-Asp-


Phe-Arg-Glu-Leu-Asn-Lys-Arg-Thr-Gln-Asp-Phe





SEQ ID No 9


VASGYIEAEVIPAETGQETAYFLLKLAGRWPVKTI


Val-Ala-Ser-Gly-Tyr-Ile-Glu-Ala-Glu-Val-Ile-Pro-


Ala-Glu-Thr-Gly-Gln-Glu-Thr-Ala-Tyr-Phe-Leu-Leu-


Lys-Leu-Ala-Gly-Arg-Trp-Pro-Val-Lys-Thr-Ile





SEQ ID No 10


PDKSESELVSQIIEQLIKKEKVYLAWVPAHKGIGG


Pro-Asp-Lys-Ser-Glu-Ser-Glu-Leu-Val-Ser-Gln-Ile-


Ile-Glu-Gln-Leu-Ile-Lys-Lys-Glu-Lys-Val-Tyr-Leu-


Ala-Trp-Val-Pro-Ala-His-Lys-Gly-Ile-Gly-Gly





SEQ ID No 11


NRWQVMIVWQVDRMRIRTWKSLVKHHMYISRKAKG


Asn-Arg-Trp-Gln-Val-Met-Ile-Val-Trp-Gln-Val-Asp-


Arg-Met-Arg-Ile-Arg-Thr-Trp-Lys-Ser-Leu-Val-Lys-


His-His-Met-Tyr-Ile-Ser-Arg-Lys-Ala-Lys-Gly





SEQ ID No 12


HPDKWTVQPIVLPEKDSWTVNDIQKLVGKLNWASQ


His-Pro-Asp-Lys-Trp-Thr-Val-Gln-Pro-Ile-Val-Leu-


Pro-Glu-Lys-Asp-Ser-Trp-Thr-Val-Asn-Asp-Ile-Gln-


Lys-Leu-Val-Gly-Lys-Leu-Asn-Trp-Ala-Ser-Gln





SEQ ID No 13


PAIFQSSMTKILEPFRKQNPDIVIYQYMDDLYVGS


Pro-Ala-Ile-Phe-Gln-Ser-Ser-Met-Thr-Lys-Ile-Leu-


Glu-Pro-Phe-Arg-Lys-Gln-Asn-Pro-Asp-Ile-Val-Ile-


Tyr-Gln-Tyr-Met-Asp-Asp-Leu-Tyr-Val-Gly-Ser





SEQ ID No 14


MRGAHTNDVKQLTEAVQKIATESIVIWGKTPKFKL


Met-Arg-Gly-Ala-His-Thr-Asn-Asp-Val-Lys-Gln-Leu-


Thr-Glu-Ala-Val-Gln-Lys-Ile-Ala-Thr-Glu-Ser-Ile-


Val-Ile-Trp-Gly-Lys-Thr-Pro-Lys-Phe-Lys-Leu





SEQ ID No 15


EKAFSPEVIPMFSALSEGATPQDLNTMLNTVGGHQ


Glu-Lys-Ala-Phe-Ser-Pro-Glu-Val-Ile-Pro-Met-Phe-


Ser-Ala-Leu-Ser-Glu-Gly-Ala-Thr-Pro-Gln-Asp-Leu-


Asn-Thr-Met-Leu-Asn-Thr-Val-Gly-Gly-His-Gln





SEQ ID No 16


NLLRAIEAQQHLLQLTVWGIKQLQARVLAVERYLK


Asn-Leu-Leu-Arg-Ala-Ile-Glu-Ala-Gln-Gln-His-Leu-


Leu-Gln-Leu-Thr-Val-Trp-Gly-Ile-Lys-Gln-Leu-Gln-


Ala-Arg-Val-Leu-Ala-Val-Glu-Arg-Tyr-Leu-Lys





SEQ ID No 17


ASVLSGGELDRWEKIRLRPGGKKKYKLKHIVWASR


Ala-Ser-Val-Leu-Ser-Gly-Gly-Glu-Leu-Asp-Arg-Trp-


Glu-Lys-I1e-Arg-Leu-Arg-Pro-Gly-Gly-Lys-Lys-Lys-


Tyr-Lys-Leu-Lys-His-Ile-Val-Trp-Ala-Ser-Arg





SEQ ID No 18


ELYKYKVVKIEPLGVAPTKAKRRVVQREKRAVGIG


Glu-Leu-Tyr-Lys-Tyr-Lys-Val-Val-Lys-Ile-Glu-Pro-


Leu-Gly-Val-Ala-Pro-Thr-Lys-Ala-Lys-Arg-Arg-Val-


Val-Gln-Arg-Glu-Lys-Arg-Ala-Val-Gly-Ile-Gly





SEQ ID No 19


FPISPIETVPVKLKPGMDGPKVKQWPLTEEKIKAL


Phe-Pro-Ile-Ser-Pro-Ile-Glu-Thr-Val-Pro-Val-Lys-


Leu-Lys-Pro-Gly-Met-Asp-Gly-Pro-Lys-Val-Lys-Gln-


Trp-Pro-Leu-Thr-Glu-Glu-Lys-Ile-Lys-Ala-Leu





SEQ ID No 20


QIYQEPFKNLKTGKYARMRGAHTNDVKQLTEAVQK


Gln-Ile-Tyr-Gln-Glu-Pro-Phe-Lys-Asn-Leu-Lys-Thr-


Gly-Lys-Tyr-Ala-Arg-Met-Arg-Gly-Ala-His-Thr-Asn-


Asp-Val-Lys-Gln-Leu-Thr-Glu-Ala-Val-Gln-Lys





SEQ ID No 21


NLLRAIEAQQHLLQLTVWGIKQLQARVLAVERYLK


Asn-Leu-Leu-Arg-Ala-Ile-Glu-Ala-Gln-Gln-His-Leu-


Leu-Gln-Leu-Thr-Val-Trp-Gly-Ile-Lys-Gln-Leu-Gln-


Ala-Arg-Val-Leu-Ala-Val-Glu-Arg-Tyr-Leu-Lys





SEQ ID No 22


AGLKKKKSVTVLDVGDAYFSVPLDKDFRKYTAFTI


Ala-Gly-Leu-Lys-Lys-Lys-Lys-Ser-Val-Thr-Val-Leu-


Asp-Val-Gly-Asp-Ala-Tyr-Phe-Ser-Val-Pro-Leu-Asp-


Lys-Asp-Phe-Arg-Lys-Tyr-Thr-Ala-Phe-Thr-Ile





SEQ ID No 23


TTNQKTELQATHLALQDSGLEVNIVTDSQYALGII


Thr-Thr-Asn-Gln-Lys-Thr-Glu-Leu-Gln-Ala-Ile-His-


Leu-Ala-Leu-Gln-Asp-Ser-Gly-Leu-Glu-Val-Asn-Ile-


Val-Thr-Asp-Ser-Gln-Tyr-Ala-Leu-Gly-Ile-Ile





SEQ ID No 24


VSQNYPIVQNLQGQMVHQAISPRTLNAWVKVVEEK


Val-Ser-Gln-Asn-Tyr-Pro-Ile-Val-Gln-Asn-Leu-Gln-


Gly-Gln-Met-Val-His-Gln-Ala-Ile-Ser-Pro-Arg-Thr-


Leu-Asn-Ala-Trp-Val-Lys-Val-Val-Glu-Glu-Lys





SEQ ID No 25


EAELELAENREILKEPVHGVYYDPSKDLIAEIQKQ


Glu-Ala-Glu-Leu-Glu-Leu-Ala-Glu-Asn-Arg-Glu-Ile-


Leu-Lys-Glu-Pro-Val-His-Gly-Val-Tyr-Tyr-Asp-Pro-


Ser-Lys-Asp-Leu-Ile-Ala-Glu-Ile-Gln-Lys-Gln





SEQ ID No 26


TPDKKHQKEPPFLWMGYELHPDKWTVQPIVLPEKD


Thr-Pro-Asp-Lys-Lys-His-Gln-Lys-Glu-Pro-Pro-Phe-


Leu-Trp-Met-Gly-Tyr-Glu-Leu-His-Pro-Asp-Lys-Trp-


Thr-Val-Gln-Pro-Ile-Val-Leu-Pro-Glu-Lys-Asp





SEQ ID No 27


EPFRDYVDRFYKTLRAEQASQEVKNWMTETLLVQN


Glu-Pro-Phe-Arg-Asp-Tyr-Val-Asp-Arg-Phe-Tyr-Lys-


Thr-Leu-Arg-Ala-Glu-Gln-Ala-Ser-Gln-Glu-Val-Lys-


Asn-Trp-Met-Thr-Glu-Thr-Leu-Leu-Val-Gln-Asn





SEQ ID No 28


NEWTLELLEELKSEAVRHFPRIWLHGLGQHIYETY


Asn-Glu-Trp-Thr-Leu-Glu-Leu-Leu-Glu-Glu-Leu-Lys-


Ser-Glu-Ala-Val-Arg-His-Phe-Pro-Arg-Ile-Trp-Leu-


His-Gly-Leu-Gly-Gln-His-Ile-Tyr-Glu-Thr-Tyr





SEQ ID No 29


EGLIYSQKRQDILDLWVYHTQGYFPDWQNYTPGPG


Glu-Gly-Leu-Ile-Tyr-Ser-Gln-Lys-Arg-Gln-ASp-Ile-


Leu-Asp-Leu-Trp-Val-Tyr-His-Thr-Gln-Gly-Tyr-Phe-


Pro-Asp-Trp-Gln-Asn-Tyr-Thr-Pro-Gly-Pro-Gly





SEQ ID No 30


HFLKEKGGLEGLIYSQKRQDILDLWVYHTQGYFPD


His-Phe-Leu-Lys-Glu-Lys-Gly-Gly-Leu-Glu-Gly-Leu-


Ile-Tyr-Ser-Gln-Lys-Arg-Gln-Asp-Ile-Leu-Asp-Leu-


Trp-Val-Tyr-His-Thr-Gln-Gly-Tyr-Phe-Pro-Asp





SEQ ID No 31


FPVRPQVPLRPMTYKAAVDLSHFLKEKGGLEGLIY


Phe-Pro-Val-Arg-Pro-Gln-Val-Pro-Leu-Arg-Pro-Met-


Thr-Tyr-Lys-Ala-Ala-Val-Asp-Leu-Ser-His-Phe-Leu-


Lys-Glu-Lys-Gly-Gly-Leu-Glu-Gly-Leu-Ile-Tyr





SEQ ID No 32


FPQITLWQRPLVTIKIGGQLKEALLDTGADDTVLE


Phe-Pro-Gln-Ile-Thr-Leu-Trp-Gln-Arg-Pro-Leu-Val-


Thr-Ile-Lys-Ile-Gly-Gly-Gln-Leu-Lys-Glu-Ala-Leu-


Leu-Asp-Thr-Gly-Ala-Asp-Asp-Thr-Val-Leu-Glu





SEQ ID No 33


LVITTYWGLHTGERDWHLGQGVSIEWRKKRYSTQV


Leu-Val-Ile-Thr-Thr-Tyr-Trp-Gly-Leu-His-Thr-Gly-


Glu-Arg-Asp-Trp-His-Leu-Gly-Gln-Gly-Val-Ser-Ile-


Glu-Trp-Arg-Lys-Lys-Arg-Tyr-Ser-Thr-Gln-Val





SEQ ID No 34


APPEESFRFGEETTTPSQKQEPIDKELYPLASLRS


Ala-Pro-Pro-Glu-Glu-Ser-Phe-Arg-Phe-Gly-Glu-Glu-


Thr-Thr-Thr-Pro-Ser-Gln-Lys-Gln-Glu-Pro-Ile-Asp-


Lys-Glu-Leu-Tyr-Pro-Leu-Ala-Ser-Leu-Arg-Ser





SEQ ID No 35


KRRVVQREKRAVGIGAMFLGFLGAAGSTMGAASMT


Lys-Arg-Arg-Val-Val-Gln-Arg-Glu-Lys-Arg-Ala-Val-


Gly-Ile-Gly-Ala-Met-Phe-Leu-Gly-Phe-Leu-Gly-Ala-


Ala-Gly-Ser-Thr-Met-Gly-Ala-Ala-Ser-Met-Thr





SEQ ID No 36


GLGQHIYETYGDTWAGVEAIIRILQQLLFIHFRIG


Gly-Leu-Gly-Gln-His-Ile-Tyr-Glu-Thr-Tyr-Gly-Asp-


Thr-Trp-Ala-Gly-Val-Glu-Ala-Ile-Ile-Arg-Ile-Leu-


Gln-Gln-Leu-Leu-Phe-Ile-His-Phe-Arg-Ile-Gly






Candidate peptides for inclusion into a prophylactic or therapeutic vaccine for HIV may be peptides from any of the structural or functional domains Gag, Pol, Nef, Env, Vif, Vpr, Vpu, Tat or Rev in any such combination.


INCORPORATION BY REFERENCE

The entire disclosure of each of the publications, web sites and patent documents referred to herein is incorporated by reference in its entirety for all purposes to the same extent as if each individual publication, web site or patent document were so individually denoted.


EQUIVALENTS

The invention may be embodied in other specific forms without departing form the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.

Claims
  • 1. A multi-component pharmaceutical composition configured for intracellular delivery of T cell epitopes to induce a cell-mediated immune response, the pharmaceutical composition comprising: two to about 20 different types of fluorocarbon peptide constructs, wherein each different type of fluorocarbon peptide construct comprises: a fluorocarbon chain from 3 to 30 carbon atoms, wherein one or more fluorine moieties is optionally substituted with chlorine, bromine or iodine or a methyl group; and,a peptide comprising one or more CD8+T cell epitopes including at least two MHC class I or II epitopes, the peptide being coupled to the fluorocarbon vector via either an N-terminus or C-terminus of the peptide and the fluorocarbon vector;wherein the fluorocarbon peptide constructs are comprised within micelles, each different type of fluorocarbon peptide construct comprises a different peptide sequence, and the fluorocarbon chain enhances CD8+T cell response against the peptide; and,one or more pharmaceutical acceptable carriers, excipients, diluents or adjuvants.
  • 2. The composition of claim 1 wherein the fluorocarbon peptide construct is according to structure
  • 3. The composition of claim 1 wherein the fluorocarbon peptide construct is according to structure
  • 4. The composition of claim 1 wherein the fluorocarbon peptide construct is according to structure
  • 5. The composition of claim 1 wherein the peptide comprises one or more immunogenic epitopes from a viral protein.
  • 6. The composition of claim 1 wherein the peptide comprises one or more immunogenic epitopes from an influenza or hepatitis B virus protein.
  • 7. The composition of claim 1 wherein the peptide comprises between 7 to 70 amino acids.
  • 8. The composition of claim 1 wherein the peptide comprises two or more overlapping T cell epitopes.
  • 9. The composition of claim 1 wherein the peptide is an HIV epitope.
  • 10. The composition of claim 1 wherein the peptide comprises multiple overlapping viral T cell epitopes and/or fusion peptides.
  • 11. The composition of claim 1, wherein the fluorocarbon peptide construct is according to structure CmFn-CyHx-Sp)-R, where m=3 to 30, n<=2m+1, y=0 to 15, x<=2y, (m+y)=3−30 and Sp is an optional chemical spacer moiety and R is the peptide.
  • 12. A combination of different types of fluorocarbon peptide constructs configured for intracellular delivery of T cell epitopes to induce a cell-mediated immune response, each different type of fluorocarbon peptide construct comprising: a fluorocarbon chain from 3 to 20 carbon atoms, wherein one or more fluorine moieties is optionally substituted with chlorine, bromine or iodine or a methyl group, andan in vivo immunogenic peptide of between 7 to 70 amino acids in length, comprising one or more CD8+T cell epitopes, at least two MHC class I or MHC class II epitopes, and being covalently linked via either an N-terminus or C-terminus terminal lysine to the fluorocarbon chain,wherein each different type of fluorocarbon peptide construct comprises a different in vivo immunogenic peptide sequence and the fluorocarbon chain enhances CD8+T cell response against the peptide; and,wherein the combination of different types of the fluorocarbon constructs is lyophilized.
  • 13. The fluorocarbon peptide construct of claim 12, wherein the peptide is between 15 to 35 amino acids in length.
  • 14. The fluorocarbon peptide construct of claim 12, wherein the fluorocarbon peptide construct is according to structure CmFn—CyHx-Sp-R, where m=3 to 30, n<=2m+1, y=0 to 15, x<=2y, (m+y)=3−30 and Sp is an optional chemical spacer moiety and R is the peptide.
  • 15. The fluorocarbon peptide construct of claim 12, wherein the fluorocarbon peptide construct is according to structure
  • 16. The fluorocarbon peptide construct of claim 12, wherein the peptide comprises one or more epitopes from an influenza or hepatitis B virus protein.
  • 17. A multi-component pharmaceutical composition configured for intracellular delivery of T cell epitopes to induce a cell-mediated immune response, the pharmaceutical composition comprising: two to about 20 different types of fluorocarbon peptide constructs, wherein each different type of fluorocarbon peptide construct comprises: a fluorocarbon chain from 3 to 30 carbon atoms, wherein one or more fluorine moieties is substituted with chlorine, bromine or iodine or a methyl group; and,a peptide comprising one or more CD8+T cell epitopes including at least two MHC class I or II epitopes, the peptide being coupled to the fluorocarbon vector via either an N-terminus or C-terminus of the peptide and the fluorocarbon vector;wherein the fluorocarbon peptide constructs are comprised within micelles, and wherein each different type of fluorocarbon peptide construct comprises a different peptide sequence, and the fluorocarbon chain enhances CD8+T cell response against the peptide; and,one or more pharmaceutical acceptable carriers, excipients, diluents or adjuvants.
  • 18. The composition of claim 17, wherein the peptide comprises multiple overlapping viral T cell epitopes and/or fusion peptides.
  • 19. The composition of claim 17 wherein the peptide comprises between 7 to 70 amino acids.
Priority Claims (1)
Number Date Country Kind
0408164 Apr 2004 GB national
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 13/354,871, filed Jan. 20, 2012, which is a continuation of U.S. application Ser. No. 12/700,526, filed Feb. 4, 2010, now U.S. Pat. No. 8,110,540, which is a continuation of U.S. patent application Ser. No. 11/096,725, filed Apr. 1, 2005, now U.S. Pat. No. 7,687,455, which claims the benefit of and priority to Great Britain Patent Application Serial No. 0408164.2, filed Apr. 13, 2004, the disclosures of each of which is are incorporated herein by reference.

US Referenced Citations (59)
Number Name Date Kind
3065141 Gessler et al. Nov 1962 A
3843443 Fishman Oct 1974 A
4332787 Homcy et al. Jun 1982 A
4689398 Wu et al. Aug 1987 A
4954444 Eveleigh Sep 1990 A
5021551 Allen et al. Jun 1991 A
5055562 Koganty Oct 1991 A
5401634 Milbrath Mar 1995 A
5635181 Harwood et al. Jun 1997 A
5728578 Jahn et al. Mar 1998 A
5817318 Sia et al. Oct 1998 A
5858374 Levy Jan 1999 A
5871746 Boutillon et al. Feb 1999 A
5882645 Toth Mar 1999 A
5919459 Nacy et al. Jul 1999 A
6069232 Malikayl et al. May 2000 A
6121123 Lyons et al. Sep 2000 A
6174532 Campo et al. Jan 2001 B1
6270778 Kawakami et al. Aug 2001 B1
6413516 Chang et al. Jul 2002 B1
6491926 Morton Dec 2002 B1
6537560 Kawakami et al. Mar 2003 B1
6541009 Inglis et al. Apr 2003 B1
6548046 Lanza et al. Apr 2003 B1
6676963 Lanza et al. Jan 2004 B1
6710035 Feigner et al. Mar 2004 B2
6884414 Palese et al. Apr 2005 B1
7476386 Gras-Masse et al. Jan 2009 B1
7687455 Bonnet Mar 2010 B2
8110540 Bonnet Feb 2012 B2
8110541 Bonnet Feb 2012 B2
8129333 Bonnet Mar 2012 B2
8642531 Bonnet Feb 2014 B2
8759281 Bonnet Jun 2014 B2
9119811 Brown et al. Sep 2015 B2
9446143 Bonnet Sep 2016 B2
10155049 Bonnet et al. Dec 2018 B2
20050009008 Robinson et al. Jan 2005 A1
20060013820 Bonnet et al. Jan 2006 A1
20070059799 Sette et al. Mar 2007 A1
20070172929 Maassab et al. Jul 2007 A1
20070202100 Wood et al. Aug 2007 A1
20080145383 Zauner et al. Jun 2008 A1
20090023895 Miyakawa et al. Jan 2009 A1
20090191233 Bonnet et al. Jul 2009 A1
20100047275 Stoloff et al. Feb 2010 A1
20100183650 Bonnet et al. Jul 2010 A1
20100183708 Bonnet et al. Jul 2010 A1
20110014221 Kang et al. Jan 2011 A1
20120034259 Bonnet et al. Feb 2012 A1
20120251569 Martin et al. Oct 2012 A1
20120315293 Bonnet et al. Dec 2012 A1
20130295641 Gee Nov 2013 A1
20130330382 Brown et al. Dec 2013 A1
20140234356 Bonnet Aug 2014 A1
20150112042 Bonnet et al. Apr 2015 A1
20160051661 Brown Feb 2016 A1
20160324834 Ahmed et al. Nov 2016 A1
20200016263 Georges Jan 2020 A1
Foreign Referenced Citations (26)
Number Date Country
2070254 Dec 1993 CA
0 327 070 Aug 1989 EP
2190474 Jun 2010 EP
1991563 Nov 2011 EP
2383284 Nov 2011 EP
2383285 Nov 2011 EP
2752161 Feb 1998 FR
2883563 Sep 2006 FR
1193378 May 1970 GB
2465733 Dec 2010 GB
2218175 Dec 2003 RU
WO-1994026903 Nov 1994 WO
WO-1999021541 May 1999 WO
WO-1999045954 Sep 1999 WO
WO-2001000225 Jan 2001 WO
WO-2001070772 Sep 2001 WO
WO-2002072627 Sep 2002 WO
WO-2003040165 May 2003 WO
WO-2004031211 Apr 2004 WO
WO-2005094891 Oct 2005 WO
WO-2005099752 Oct 2005 WO
WO-2005120564 Dec 2005 WO
WO-2006074024 Jun 2006 WO
WO-2007091030 Aug 2007 WO
WO-2009027688 Mar 2009 WO
WO-2012090002 Jul 2012 WO
Non-Patent Literature Citations (134)
Entry
Sommer, Stefanie et al, “Covalent inhibition of sumo and upbiquitin specific systeine proteases by an in situ thio-alkyne addition.” Bioorg. Medicinal Chem. (2013)21 p. 2511-2517.
Gripon, P. et al, “Myristylation of the hepatitis b virus large surface protein is essential for viral infectivity.” Virology (1995) 213 p. 292-299.
Boutin, Jean A., “Myristoylation.” Cell Signal. (1997) 9(1) p. 15-35.
Chai, Ning et al, “Assembly of hepatits b virus envelope proteins into a lentivirus pseudotype that infects primary human hepatocytes.” J. Virology (2007) 81 p. 10897-10904.
Mishra, A. et al, “Immune response to hepatitis b virus surface antigen peptides during hbv infection.” Clin. Exp. Immunol (1992) 90 p. 194-198.
Weber, Bernard et al, “Improved detection of hepatits b virus surface antigen by a new rapid automated assay.” J. Clin. Microbiol. (1999) 37(8) p. 2639-2647.
Leenaars, Marlies et al, “The production of polyclonal antibodies in laboratory animals.” Alternatives to Laboratory Animals (1999) p. 1-28.
Pierce biotechnology pamphlet on affinity purification, available Jan. 2004.
Cui, Zhengrong et al, “Novel enthanol in fluorocarbon microemulsions for topical genetic immunization.” Pharmaceut. Res. (2003) 20(1 )p. 16-23.
Yamaguchi, Shigehiko et al, “O/w emulsion as formed by cholesterol-pearing pullulan.” Bull. Chem. Soc. Japan. (1992) 2 p. 186-190.
Translation of Yamaguchi et al, Bull. Chem. Soc. Japan. (1992) 2 p. 186-190.
Chang, Byeong S. et al, “Physical factors affecting the storage stability of freeze dried interleukin 1 receptor antagonist: glass transition temperature.” Arch. Biochem. Biophys. (1996) 331(2) p. 249-258.
MacKay, J. Andrew and Szoka, Francis C., “Hiv tat protein transduction domain mediated cell binding and intracellular delivery of nanoparticles.” J. Dispers. Sci. Technol. (2003) 24(3) p. 465-473.
Pardridge, William M., “Drug and gene targeting to the brain with molecular trojan horses.” Nat. Rev. (2002) 1 p. 131-139.
Yamaguchi, Shigehiko et al, “O/w-emulsion as formed by cholesterol-bearing pullulan.” Bull. Chem. Soc. Japan (1992) 2 p. 186-190. This reference is in Japanese; please note that an English language translation is attached.
Baczko, Krystyna et al, “Synthesis and surfactant properties of symmetric and unsymmetric sulfosuccinic diesters, aerosol-ot homologues.” J. Chem. Soc. Perkin Trans. 2 (2001) 2179-2188.
Harris, D. P. etal, “Immunogenicity of peptides forb cells is not impaired by overlapping t-cell epitope topology.” Immunol. (1996) 88 p. 348-354.
Knoll, Gerd et al, “Fusion of liposomes with the plasma membrane of epithelial cells: fate of inocrporated lipids as follwed by freeze fracture and autoradiography of plastic sections.” J. Cell Biol. (1988) 107 (6 pt2) p. 2511-2521.
The IEDB analysis resource, T-cell epitope prediction tools (http://tools.iedb.org/main/tcell/, accessed Feb. 27, 2020.
Reddehause, Matthias J. et al; “A pentapeptide as minimal antigenic determinant for mhc class I restricted t lymphocytes.” Nat. (1989) 337 p. 651-653.
Sanches-Triincado, Jose L. et al; “Fundamentals and methods fort-and b-cell epitope prediction.” J. Immunol. Res. (2017) article ID 2380160.
Alatrakchi et al. (2002) “Strong CD4 Th1 responses to HIV and hepatitis C virus in HIV-infected long-term non-progressors co-infected with hepatitis C virus,” AIDS 16(5):713-717.
Alexander et al. (1997) “Derivation of HLA-A11/Kb Transgenic Mice: Functional CTL Repertoire and Recognition of Human A11-Restricted CTL Epitopes,” J. Immunol. 159(10):4753-4761.
Alix (1999) “Predictive estimation of protein linear epitopes by using the program PEOPLE,” Vaccine 18(3-4):311-314.
Bastin et al. (1987) “Use of Synthetic Peptides of Influenza Nucleoprotein to Define Epitopes Recognized by Class I-Restricted Cytotoxic T Lymphocytes,” J. Exp. Med. 165:1508-1523.
Beebe et al. (2008) “Formulation and characterization of a ten-peptide single vial vaccine, EP-2101, designed to induce cytotoxic T-lymphocyte responses for cancer immunotherapy” Human Vaccines, 4(3):210-218.
BenMohamed et al. (2003) “Identification of novel immunodominant CD4+ THL-type T-cell peptide epitopes from herpes simplex virus glycoprotein D that confer protective immunity,” J. of Virology 77(17):9463-9473.
Berkhoff et al. (2005) “Functional Constraints of Influenza A Virus Epitopes Limit Escape from Cytotoxic T Lymphocytes,” J. Virol.. 79(17): 11239-11246.
Boaz et al. (2002) “Presence of HIV-1 Gag-specific IFN-gamma+IL-2+ and CD28+IL-2+ CD4 T cell responses is associated with nonprogression in HIV-1 infection,” J. of Immunology 169:6376-6385.
Bodmer et al. (1989) “Class I cross-restricted T cells reveal low responder allele due to processing of viral antigen,” Nature 337:653-655.
Bonnet et al. (2005) “Effect of Glycoamphiphiles on the Solubilization and Dendritic Cell Uptake of a Lipopeptide: A Preliminary Study” Mol. Pharm., 2(5):420-427.
Boon et al. (2005) “Functional profile of human influenza virus-specific cytotoxic T lymphocyte activity is influenced by interleukin-2 concentration and epitope specificity,” Clin. Exp. Immunol. 142:45-52.
Cheuk et al. (2005) “Strong memory CD8+T cell responses against immunodominant and three new subdominant HLA-B27-restricted influenza a CTL epitopes following secondary infection of HLA-B27 transgenic mice,” Cellular Immunology 234:110-123.
Crowe et al. (2006) “Identification of protective and non-protective T cell epitopes in influenza,” Vaccine 24:452-456.
Database Uniprot Nov. 1, 1996 (Nov. 1, 1996), Castrucci M et al.: “Genetic reassortment between avian and human influenza A virus” XP002442455 accession No. Q67194.
Database WPI Week 200416; Thomson Scientific, London, GB; AN 2004-165290; XP002508268 (2 Pages).
De Groot et al. (1997) “An Interactive Web Site Providing Major Histocompatibility Ligand Predictions: Applications to HIV Research,” AIDS Res. and Human Retroviruses 13(7):529-531.
Deckert, C.A. (1980) “Pattern Etching of CVD S13N4/5102 Composites in HF/Glycerol Mixtures,” Journal of the Electrochemical Society 127(2):2433-2438.
Deliyannis et al. (2002) “Induction of Long-Term Memory CD8+ T Cells for Recall of Viral Clearing Responses against Influenza Virus,” J. of Virology 76(9):4212-4221.
Deres et al. (1989) “In vivo priming of virus-specific cytotoxic T lymphocytes with synthetic lipopeptide vaccine” Nature, 342:561-564.
Do et al. (2008) “Broad T cell immunity to the LcrV virulence protein is induced by targeted delivery to DEC-205/CD205-positive mouse dendritic cells” Eur. J. Immunol., 38:20-29.
Doolan et al. (2000) “HLA-DR-Promiscuous T Cell Epitopes from Plasmodium falciparum Pre-Erythrocytic-Stage Antigens Restricted by Multiple HLA Class II Alleles,” J. Immunol. 165:1123-1137.
English Translation and Original Search Report dated Feb. 8, 2012 for Taiwanese Patent Application No. 097133417 (Taiwanese counterpart of PCT/GB2005/001279, Immune Targeting Systems, 2 pages).
EP Search Report for Application No. EP 05 72 9595 dated Jan. 26, 2009 (5 pages).
Europharmatoday website (http://www.europharmatoday.com/2009/08/startup-profile-immune-targeting-systems-ltd-synthetic-vaccines-against-mutating-viruses.html, accessed May 11, 2011).
Faroux-Corlay et al. (2000) “Synthesis of single- and double-chain fluorocarbon and hydrocarbon galactosyl amphiphiles and their anti-HIV-1 activity,” Carbohydrate Res. 327:223-260.
Fayolle et al. (2004) “Bordetella pertussis adenylate cyclase delivers chemically coupled CD8+ T-cell epitopes to dendritic cells and elicits CTL in vivo” Vaccine, 23:604-614.
Felix et al. (1995) “Pegylated peptides. IV. Enhanced biological activity of site-directed pegylated GRF analogs” Int. J. Pept. Protein Res., 46:253-264.
Filippov et al. (2002) “Use of benzloxycarbonyl (Z)-based fluorophillic taging reagents in the purification of synthetic peptide,” Tet. Let., 43, 7809-7812.
Frahm et al. (2007) “Extensive HLA class I allele promiscuity among viral CTL epitopes,” Eur. J. Immunol. 37:2419-2433.
Gahery-Segard et al. (2000) “Multiepitopic B- and T-Cell Responses Induced in Humans by a Human Immunodeficiency Virus Type 1 Lipopeptide Vaccine,” J. of Virology 74(4):1694-1703.
Gahery-Segard et al. (2003) “Long-Term Specific Immune Responses Induced in Humans by a Human Immunodeficiency Virus Type 1 Lipopeptide Vaccine: Characterization of CD8+-T-Cell Epitopes Recognized,” J. of Virology 77(20):11220-11231.
Genbank accession No. AAB62620. Oct. 2005.
Genbank accession No. AAL32169. Nov. 2001.
Genbank accession No. NP_626519. Apr. 2006.
Gianfrani et al. (2000) “Human Memory CTL Response Specific for Influenza A Virus is Broad and Multispecific,” Hum. Immunol. 61:438-452.
Gileadi et al. (1999) “Effect of epitope flanking residues on the presentation of N-terminal cytotoxic T lymphocyte epitopes,” Eur J Immunol. 29(7):2213-22.
Gileadi et al. (1999) “Generation of an Immunodominant CTL Epitope Is Affected by Proteasome Subunit Composition and Stability of the Antigenic Protein,” J. Immunol. 163:6045-6052.
Gotch et al. (1987) “Cytotoxic T lymphocytes recognize a fragment of influenza virus matrix protein in association with HLA-A2,” Nature 326:881-882.
Hackett et al. (2005) “Vaccine Adjuvants,” Chapter 10 pp. 193-219.
Hall et al. (1981) “Variation in Nucleotide Sequences Coding for the N-terminal Regions of the Matrix and Nonstructural Proteins of Influenza a Viruses,” J. Virol. 38(1):1-7.
HIV Databases at Los Alamos National Laboratory (http://www.hiv.lanl.gov/content index) printed Oct. 3, 2007.
HIV Immunology Database: Compendia at Los Alamos National Laboratory (http://www.hiv.lanl.gov/content/immunology/compendium.html) printed Oct. 3, 2007.
HIV Molecular Immunology Database at Los Alamos National Laboratory (http://www.hiv.lanl.gov/content/immunology/index.html) printed Oct. 3, 2007.
Idemyor (2003) “Human immunodeficiency virus: scientific challenges impeding candidate vaccines,” HIV Clin. Trial, 4:421-424.
International Search Report for PCT/GB11/001781 (Immune Targeting Systems), dated Apr. 10, 2012 (4 pages).
International Search Report for PCT/GB2005/001279, dated Sep. 12, 2005 (7 pages).
International Search Report for PCT/GB2008/002930 dated Dec. 16, 2008 (3 pages).
Internet Search Engine search for “Los Alamos National Laboratory Database” at Los Alamos National Laboratory (http://www.google.com) printed Oct. 3, 2007.
Jameson et al. (1998) “Human cytotoxic T-lymphocyte repertoire to influenza A viruses,” Journal of Virology 72(11):8682-8689.
Jin et al. (2005) “Two residues in the hemagglutinin of A/Fujian/411/02-like influenza viruses are responsible for antigenic drift from A/Panama/2007/99,” Virology 336:113-119.
Jones et al. (2005) “Characterisation of cell-penetrating peptide-mediated peptide delivery” Br. J. Pharmacol., 145:1093-1102.
Koch et al. (2005) “The crystal structure of human CD1d with and without alpha-galactosylceramide,” Nature Immunology 6(8):819-826.
Korber et al. (2003) “HIV Immunology and HIV/SIV Vaccine Databases” 1:343, 508, 661, 1042-1044.
Krafft et al. (2009) “Chemistry, Physical Chemistry, and Uses of Molecular Fluorocarbon-Hydrocarbon Diblocks, Triblocks, and Related Compounds—Unique “Apolar” Components for Self-Assembled Colloid and Interface Engineering” Chem. Rev., 109:1714-1792.
Lawson et al. (1992) “Nucleotide Sequence Changes in the Polymerase Basic Protein 2 Gene of Temperture-Sensitive Mutants of Influenza A Virus,” Virology 191:506-510.
Linnemann et al. (2000) “Detection and Quantification of CD4+T Cells with Specificity for a New Major Histocompatibility Complex Class II-Restricted Influenza A Virus Matrix Protein Epitope in Peripheral Blood of Influenza Patients,” J. Virol. 74(18):8740-8743.
Lu et al. (2004) “Therapeutic dendritic-cell vaccine for chronic HIV-1 infection,” Nature Medicine 10(12):1359-1365.
Macken et al. (2001) “The value of a database in surveillance and vaccine selection” Congress Series 1219:103-106.
Maksyutov et al. (1993) “ADEPT: a computer program for prediction of protein antigenic determinants,” Comput. Appl. Biosci. 9(3):291-297.
Marsh et al. (2000) “The HLA Facts Book,” Chapter 11 pp. 61-72.
Mortara et al. (1999) “Type 1 CD4+ T-Cell Help is Required for Induction of Antipeptide Multispecific Cytotoxic T Lymphocytes by a Lipopeptidic Vaccine in Rhesus Macaques,” J. of Virology 73(5):4447-4451.
Nijman et al. (1993) “Identification of peptide sequences that potentially trigger HLA-A2.1—restricted cytotoxic T lymphocytes,” Eur. J. Immunol. 23:1215-1219.
Notice of Opposition and Statement of Facts and Arguments in Support of Opposition filed on Aug. 23, 2012 in EP Patent No. 1991563 owned by PepTcell Limited (corresponds to PepTcell's PCT/GB2007/000383) (31 pages).
Odorico et al. (2003) “BEPITOPE: predicting the location of continuous epitopes and patterns in proteins,” J. Mol. Recognit. 16(1):20-22.
Opposition statement against related Chilean Application No. 2559-2008 by Asociacion Nacional de Laboratorios Farmaceuticos AG (in Spanish) and its English translation (7 pages).
Ozer et al. (1999) “Synthesis of perfluoroalkyltated beta-alanine and some peptide derivatives: an access to original surfactants” Amino Acids, 16:381-389.
Panina-Bordignon et al. (1989) “Universally immunogenic T cell epitopes: promiscuous binding to human MHC class II and promiscuous recognition by T cells,” Eur. J. Immunol. 19:2237-2242.
Parker et al. (1992) “Sequence Motifs Important for Peptide Binding to the Human MHC Class I Molecule, HLA-A2,” J. Immunol. 149(11):3580-3587.
Pellequer et al. (1993) “PREDITOP: A program for antigenicity prediction,” J. Mol. Graph. 11(3):204-10.
Perrie et al.(2007) “Recent developments in particulate-based vaccines” Recent Pat. Drug Deliv. Formul., 1(2):117-129.
Plotnicky et al. (2003) “The immunodominant influenza matrix T cell epitope recognized in human induces influenza protection in HLA-A2/Kb transgenic mice,” Virology 309:320-329.
Rammensee (1995) “Chemistry of Peptides Associated with MHC Class I and Class II Molecules,” Current Opinion in Immunology 7:85-96.
Rammensee et al. (1999) “SYFPEITHI: database for MHC ligands and peptide motifs,” Immunogenetics 50:213-219.
Reichel, et al. (1999) “Stereochemical Dependence of the Self-Assembly of the Imunoadjuvants Pam3Cys and Pam3Cys-Ser” J. Am. Chem. Soc. 121:7989-7997.
Reyes et al. (1991) “Binding of Radioiodinated Influenza Virus Peptides to Class I MHC Molecules and to Other Cellular Proteins As Analyzed by Gel Filtration and Photoaffinity Labeling,” Mol. Immunol. 28(4-5):341-348.
Riess et al. (1991) “Highly Effective Surfactants with Low Hemolytic Activity,” Adv. Mater. 3(5):249-251.
Rosenberg et al. (1997) “Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia,” Science 278:1447-1450.
Samad et al. (2007) “Liposomal drug delivery systems: an update review” Current Drug Deliv., 4:297-305.
Scheibenbogen et al. (1997) “A Sensitive Elispot Assay for Detection of CD8+ T Lymphocytes Specific for HLA Class I-binding Peptide Epitopes Derived from Influenza Proteins in the Blood of Healthy Donors and Melanoma Patients,” Clin. Cancer Res. 3:221-226.
Schlaphoff et al. (2007) “Functional and Phenotypic Characterization of Peptide-Vaccine-Induced HCV-specific CD8+ T Cells in Healthy Individuals and Chronic Hepatitis C Patients,” Vaccine 25 (37-38): 6793-6806.
Schmittel et al. (2001) “Application of the IFN-γ Elispot assay to quantify T cell responses against proteins,” J. Immunol. Methods 247:17-24.
Sette et al. (1998) “HLA supertypes and supermotifs: a functional perspective on HLA polymorphism,” Current Opinion in Immunology 10:478-482.
Speiser et al. (2005) “Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909,” J. of Clin. Investigation 115(3):739-746.
Sugimoto et al. (1993) “Peptide Vaccine,” Cell Technology 12(1): 58-63, 74. Original article in Japanese with English translation attached (20 pages).
Takeshita (1995) “Molecular Analysis of the Same HIV Peptide Functionally Binding to Both a Class I and a Class II MHC Molecule,” J. Immunol. 154:1973-1986.
Tang et al. (2005) “Isolation and Characterization of H3N2 Influenza A Virus from Turkeys,” Avian Diseases 49:207-213.
Taubenberger et al. (1997) “Initial Genetic Characterization of the 1918 “Spanish” Influenza Virus,” Science 275:1793-1796.
Thimme et al. (2001) “Determinants of viral clearance and persistence during acute hepatitis C virus infection,” J. Exp. Med. 11:1395-1406.
Third Party Observations submitted in EP07712678.7 owned by PepTcell Limited (EP Patent No. 1991563, corresponds to PepTcell's PCT/GB2007/000383) filed with European Patent Office dated Dec. 17, 2008 (13 pages).
Thomas et al. (2006) “Cell-Mediated Protection in Influenza Infection,” Emerg. Infect. Dis. 12(1):48-54.
Thust et al. (2003) “Protease-catalyzed peptide synthesis for the site-specific incorporation of alpha-fluoroalkyl amino acids into peptides,” J. Organic Chem. 68(6):2290-2296.
Voeten et al. (2001) “Antigen processing for MHC class I restricted presentation of exogenous influenza A virus nucleoprotein by B-lymphoblastoid cells,” Clin. Exp. Immunol. 125:423-431.
Wang et al. (2007) “CTL epitopes for influenza A including the H5N1 bird flu; genome-, pathogen-, and HLA-wide screening,” Vaccine 25(15):2823-2831.
Written Opinion of the International Searching Authority for PCT/GB2007/000383 (PepTcell Limited), dated Aug. 7, 2008 (10 pages).
Written Opinion of the International Searching Authority, International Application No. PCT/GB2008/002930 (Immune Targeting Systems), dated Feb. 28, 2010 (5 pages).
Yang et al. (2005) “Expression of HLA-DP0401 Molecules for Identification of DP0401 Restricted Antigen Specific T Cells,” J. Clin. Immunol. 25(5):428-436.
Yang et al. (2006) “Multiplex mapping of CD4 T cell epitopes using class II tetramers,” Clinical Immunology 120:21-32.
Zhong et al. (2003) “Genome-wide Characterization of a Viral Cytotoxic T Lymphocyte Epitope Repertoire,” J. Biol. Chem. 278(46):45135-45144.
“Solvent Systems and Their Selection in Pharmaceutics and Biopharmaceutics,” Springer (2007) Augustijns, P. And Brewster, M., eds. “Chapter I. Principles of Solubility,” Gong et al., pp. 1-27.
U.S. Appl. No. 11/096,725, Antigen Delivery Vectors and Constructs, filed Apr. 1, 2005, Granted, U.S. Pat. No. 7,687,455.
U.S. Appl. No. 12/207,828, Antigen Delivery Vectors and Constructs, filed Sep. 10, 2008, Granted, U.S. Pat. No. 8,129,333.
U.S. Appl. No. 12/700,526, Antigen Delivery Vectors and Constructs, filed Feb. 4, 2010, Granted, U.S. Pat. No. 8,110,540.
U.S. Appl. No. 12/700,566, Antigen Delivery Vectors and Constructs, filed Feb. 4, 2010, Granted, U.S. Pat. No. 8,110,541.
U.S. Appl. No. 13/354,871, Antigen Delivery Vectors and Constructs, filed Jan. 20, 2012, Granted, U.S. Pat. No. 8,759,281.
U.S. Appl. No. 12/201,894, Influenza Antigen Delivery Vectors and Constructs, filed Aug. 29, 2008, Granted, U.S. Pat. No. 8,642,531.
U.S. Appl. No. 14/139,293, Influenza Antigen Delivery Vectors and Constructs, filed Dec. 23, 2013, Granted, U.S. Pat. No. 9,446,143.
U.S. Appl. No. 15/242,682, Influenza Antigen Delivery Vectors and Constructs, filed Aug. 22, 2016, Granted, U.S. Pat. No. 10,155,049.
U.S. Appl. No. 16/186,558, Influenza Antigen Delivery Vectors and Constructs, filed Nov. 11, 2018, Pending, US 2019- 0142951.
U.S. Appl. No. 13/977,265, Fluorocarbon-Linked Peptide Formulation, filed Aug. 15, 2013, Granted, U.S. Pat. No. 9,119,811.
U.S. Appl. No. 14/807,606, Fluorocarbon-Linked Peptide Formulation, filed Jul. 23, 2015, Granted, U.S. Pat. No. 10,729,759.
U.S. Appl. No. 16/909,040, Fluorocarbon-Linked Peptide Formulation, filed Jun. 23, 2020, Pending, Not yet published.
U.S. Appl. No. 14/655,041, Vaccines Against Hepatitis B Virus, filed Jun. 23, 2015, Granted, U.S. Pat. No. 10,300,132.
U.S. Appl. No. 16/423,149, Vaccines Against Hepatitis B Virus, filed May 27, 2019, Pending, US 2020-0016263.
Related Publications (1)
Number Date Country
20150112042 A1 Apr 2015 US
Continuations (3)
Number Date Country
Parent 13354871 Jan 2012 US
Child 14286531 US
Parent 12700526 Feb 2010 US
Child 13354871 US
Parent 11096725 Apr 2005 US
Child 12700526 US