Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PAMPS, PATHOGEN ASSOCIATED MOLECULAR PATTERNS
Document Type and Number:
WIPO Patent Application WO/2007/011776
Kind Code:
A2
Abstract:
A method for identifying a polypeptide which acts as an adjuvant in a host organism. The invention further provides adjuvant compositions comprising said polypeptides and optionally further comprising an antigen.

Inventors:
ULMER JEFFREY (US)
DONATI CLAUDIO (IT)
COVACCI ANTONELLO (IT)
VALIANTE NICHOLAS (US)
XU FENG (US)
Application Number:
PCT/US2006/027484
Publication Date:
January 25, 2007
Filing Date:
July 15, 2006
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NOVARTIS AG (CH)
ULMER JEFFREY (US)
DONATI CLAUDIO (IT)
COVACCI ANTONELLO (IT)
VALIANTE NICHOLAS (US)
XU FENG (US)
International Classes:
A21D10/02; G16B30/10; G16B20/30; G16B20/50
Domestic Patent References:
WO2001074130A22001-10-11
WO2005000780A12005-01-06
Other References:
See references of EP 1910827A4
Attorney, Agent or Firm:
MORAN, Michael, J. et al. (Corporate Intellectual Property P.O. Box 809, Emeryville CA, US)
Download PDF:
Claims:

CLAIMS

1. A method for identifying a polypeptide which acts as an adjuvant in a host organism comprising the steps of: a) generating protein families by grouping together amino acid sequences from at least a different pathogenic organisms, which sequences share a BLAST alignment with an E-score less than 1E-05; b) selecting a protein family of step a) wherein:

(i) the family includes sequences from at least b different pathogenic organisms, and

(ii) at least one of the proteins in the family does not share a BLAST alignment with an E-score smaller than 1E-05 with amino acid sequences from a chosen non-pathogenic organism; c) determining the sequence motifs from the resulting families of step b) that are conserved within the family; and d) selecting polypeptide sequences that comprise the motifs of step c), wherein: a is at least 60 and b is at least 30, where b_<a.

2. The method of claim 1 , wherein the amino acid sequences used in step a) are available from a genomic database.

3. The method of claim 1 , wherein the host organism is a vertebrate.

4. The method of claim 3, wherein the host organism is a human.

5. A polypeptide comprising an amino acid sequence obtainable by the method of any one of claims 1 to 4.

6. A polypeptide comprising: (i) any one of the amino acid sequences listed in Table 3; (ii) an amino acid sequence having at least 85% sequence identity to an amino acid sequence from Table 3 and/or comprising an amino acid sequence consisting of a fragment of at least 5 contiguous amino acids from an amino acid sequence from Table 3; or (iii) an amino acid sequence listed in Table 3, except that the amino acid sequence contains one or more variations.

7. The polypeptide of claim 5 or claim 6, being at least 8 amino acids long.

8. The polypeptide of claim 5, claim 6 or claim 7, being fewer than 100 amino acids long.

9. A polymer comprising any one of the sequences listed in Table 3, wherein (a) the polymer comprises monomers selected from the group consisting of: L-amino acids; D-amino acids; and amino acid mimetics, and/or (b) the bonds between monomers are not peptide bonds, wherein said polymer does not consist of: a chain of L-amino acids joined by peptide bonds to form a linear unbranched polypeptide chain.

10. Nucleic acid comprising a nucleotide sequence which encodes a polypeptide of any one of claims 5 to 8.

11. A composition comprising a polypeptide or polymer of claims 5 to 9, and a pharmaceutically acceptable carrier.

12. The composition of claim 11 which further comprises at least one of a tumor antigen, a bacterial antigen or a viral antigen.

13. A method of raising an immune response in a patient, comprising administering to a patient the composition of claim 11. or 12.

14. The nucleic acid of claim 10 which further encodes at least one of a tumor antigen, bacterial antigen or viral antigen.

15. A method of raising an immune response in a patient, comprising administering to a patient the composition of claim 10. or 14.

16. A method of raising an immune response in a patient comprising administering to the patient the composition of claim 10 or 14.

Description:

PAMPS, Pathogen Associated Molecular Patterns

All documents cited herein are incorporated by reference in their entirety.

TECHNICAL FIELD

This invention is in the field of vaccine adjuvants. BACKGROUND ART

Sub-unit vaccines often require the aid of an adjuvant to help boost immune activity. Chemical adjuvants such as aluminium salts and MF59™ have been approved for human use. However, aluminium salts are subject to safety concerns and are incompatible with some antigens. Furthermore, it produces a Th2 type of helper T cell response, which is often inappropriate or insufficient for protective immunity.

The best known adjuvant in laboratory use is Complete Freund's Adjuvant, which consists of killed Mycobacterium tuberculosis suspended in oil. Although this adjuvant is not suitable for human use due to its toxicity, safer adjuvants have been derived from other pathogenic organisms.

The immunostimulatory activity of materials derived from pathogens is believed to reflect the natural host-pathogen interaction. When the antigen-specific immune response evolved, it would have done so in an environment containing adjuvant-active bacterial components. The response to a pure bacterial antigen, injected without adjuvant-active bacterial components, is therefore an artificial situation to which the host would not be adapted to respond.

Components of pathogens are therefore believed to act as "danger signals", which put the immune system on alert. Examples of adjuvants in this category are components of bacterial capsules, LPS (lipopolysaccharides) from Gram negative bacteria, the glycolipids and arabinogalactans in mycobacteria and the peptidoglycans of spirochaetes. Other known adjuvants include DNA comprising unmethylated CpG dinucleotide motifs, which are relatively rare in vertebrate DNA compared to bacterial DNA, and double-stranded RNA, which mimics the presence of an invading virus.

Polypeptides from pathogens have not received much attention as potential adjuvants. One means to identify adjuvant-active polypeptide sequences from pathogens would be by

high-throughput screening, but this approach is essentially random and undirected, such that effort will be wasted on screening polypeptides which are unlikely to function as adjuvants.

Many of the most widely used vaccines consist of whole organisms. These include live organisms that have been rendered safe by attenuating mutations (e.g. tuberculosis and rubella) and organisms killed or inactivated by chemical treatment (e.g. influenza and hepatitis A virus). That these types of vaccines are based on whole organisms presents both advantages and disadvantages. While having all of the components of the pathogen contained within the vaccine is useful for eliciting immune responses against multiple antigens that are structurally similar to those found on the infecting pathogen, some components of whole organism vaccines can cause undesirable side effects. Furthermore, live organism vaccines, although attenuated, can sometimes cause problems in immunosuppressed individuals and have the potential to revert to a virulent state. These disadvantages spurred a movement towards potentially safer, more defined vaccines consisting of partially purified subunits known to be targets for protective immune responses (e.g. tetanus toxoid and influenza haemagglutinin). With the advent of recombinant DNA technology came the ability to produce protein antigens in heterologous expression systems (e.g. hepatitis B surface antigen). In this way, high levels of protein can be manufactured, while eliminating contamination by toxic components of the pathogen. The progression from whole organisms to subunit vaccines has highlighted a need to augment these more purified vaccine components with adjuvants, as vaccines based on live attenuated organisms contain built-in adjuvants in the form of PAMPs. In contrast, subunit vaccines often lack these elements, thus requiring that they be added back.

There is thus a need for new adjuvants, particularly for human vaccines, and for methods for identifying them. It is an object of the invention to provide further and improved adjuvants for use in vaccines and also a directed method for identifying such adjuvants.

DISCLOSURE OF THE INVENTION

The invention is based on the identification of various pathogen-associated molecular patterns (PAMPs [refs. 1-6]) and the use of these patterns in identifying adjuvant-active polypeptides. Polypeptide PAMPs are motifs present in pathogenic polypeptides but rare or absent in the host organism's own polypeptides. Such motifs are commonly found in microbes but not in vertebrates. Thus, they are recognised as foreign by the host, resulting in the triggering of an appropriate immune response, which makes these polypeptide PAMPs ideal candidates for adjuvants in vaccines. Furthermore, because they are proteinaceous in nature, their amino acid

sequence could be directly incorporated into the amino acid sequence of a protein antigen to increase potency.

Methods for identifying adjuvant-active polypeptides

The invention thus provides a method for identifying a polypeptide which acts as an adjuvant in a host organism comprising the steps of: a) generating protein families by grouping together amino acid sequences from at least a different pathogenic organisms, which sequences share a BLAST alignment with an E- score less than 1 E "05 (i.e. I E "05 , 1 E "07 , 1 E "08 , 1 E "09 , 1 E "10 , 1 E "15 , 1 E "20 or less); b) selecting a protein family of step a) wherein: (i) the family includes sequences from at least b different pathogenic organisms, and

(ii) at least one of the proteins in the family does not share a BLAST alignment with an E-score smaller than 1 E-° 5 (/.e. 1 E "06 , I E "07 , 1 E "08 , 1 Eλ 1 E "10 , 1E~ 15 , 1 E '20 Or smaller) with amino acid sequences from a chosen non-pathogenic organism; c) determining the sequence motifs from the resulting families of step b) that are conserved within the family; and d) selecting polypeptide sequences that comprise the motifs of step c), wherein: a is at least 60 (e.g. 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 99 or more);and b is at least 30 (e.g. 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60 or more), where b<a.

Compared to random high throughput screening methods, the method of the invention is a more directed approach for identifying polypeptide adjuvants, as it takes into account that a host immune response is invoked against compounds considered to be foreign rather than compounds considered to be self. The method therefore selects polypeptide motifs that are present in pathogens but not in the host organism.

By "BLAST E score" we mean the E score achieved when amino acid sequences are aligned using BLAST P 2.2.1 from the Wisconsin GCG package version 10.3 [7].

II"'" 11.,,It I! ,/ 1 I'..!' " ""'" " » •" •' " ' " ' '" "

"Pathogenic" or "pathogen" in the context of this invention refers to an organism that is capable of causing disease, for example, viruses and bacteria. Preferably, the pathogenic organism is a bacterium as most known pathogen -derived adjuvants are from bacteria.

5 "Non-pathogenic" in the context of this invention refers to an organism that is not capable of directly causing disease in a host organism. For example, a rat is considered to be nonpathogenic to a human even though the rat may be harbouring fleas which in turn carry the pathogen Yersinia pestis which can give rise to bubonic plague in a human host.

10 A "host organism" in the context of this invention refers to the organism being targeted by the pathogen. Preferably, the host organism is a human.

Step a) - generating protein families

Step a) provides protein families from which the polypeptide adjuvant is derived. By starting

15 with amino acid sequences derived from pathogenic organisms, however, rather than just random sequences, the method minimises the total number of sequences needed to be screened compared to random methods such as high throughput screening. Some of the protein families identified in this step will not be exclusive to pathogen genomes and will be excluded later on in the method. Preferably, the amino acid sequences used to generate the

20 protein families of step a) are available from a genomic database, more preferably, a cDNA or expressed sequence database.

The most desirable immune response to be generated by an adjuvant is the innate host response, a response which is based on 'broad spectrum' mechanisms. Such 'broad spectrum'

25 mechanisms, including but not limited to activation of complement via the alternative pathway, are known to be triggered by common microbial (i.e. pathogenic) components. Such common microbial components have common amino acid sequences. Step a) therefore specifies that the organisms used to generate the protein families are pathogenic. At least a different pathogenic organisms are used for generating the families, and higher values of a give a higher

30 probability that the protein families encode a common microbial component present in a large number of pathogens.

Step b) - selecting a protein family

Having created a number of protein families, the method selects only those families which include sequences from at least b different pathogenic organisms, where b is equal to or less than a. This step excludes any protein family that does not encompass sequences from a large number of pathogenic organisms, as the objective of the method of this invention is to identify polypeptides that encode a common or conserved microbial component present in several pathogens. The closer the value of b is to a, the higher is the stringency of this step.

The second part of step b) is a filter to exclude those sequence motifs common to pathogenic and non-pathogenic organisms. Thus, only those families containing at least one sequence having no BLAST alignment over their full length with an E-score exceeding 1 E "05 (i.e. 1 E "05 , 1 E " ° 6 , 1 E "07 , I E "08 , I E "09 , 1 E "10 , 1 E ~ 15 , 1 E '20 or smaller) with a chosen non-pathogenic organism are selected for the next step. In most cases, this guarantees that all members of the selected family share a domain that is not present in the chosen non-pathogenic organism. By excluding protein families that include sequences common to both pathogens and non-pathogens, the probability that the selected protein family contains a PAMP is increased and therefore the probability of identifying a polypeptide which acts as an adjuvant is increased.

The non-pathogenic organism used in step b) may be the host organism (e.g. a human). However, it is preferably not a human, particularly when the method comprises a further step of selecting polypeptide sequences not present in the human genome (see below). A preferred non-pathogenic genome is a fly genome such as that of Drosophila melanogaster.

Step c) - selecting a polypeptide sequence from the selected protein family

Having selected a protein family that includes sequences common to a number of pathogenic organisms but not to a non-pathogenic organism, step c) identifies conserved sub-sequences or motifs within the selected protein family. These sub-sequences are tested for by means of their statistical relevance.

Preferably, a computer program is used for step c). Conserved patterns in protein sequences can be conveniently represented as a set of regular expressions, i.e. strings of symbols that, for each sequence position, specify the amino acid or list of amino acids that can occur in such a position. Efficient algorithms exist, both for the extraction of conserved motifs in the form of regular expressions from a set of related sequences, and for testing the occurrence of a given pattern in a set of sequences and will be well-known to a skilled artisan. More preferably, the

PRATT program is used for step c) [8]. The PRATT program is able to discover patterns conserved in sets of unaligned protein sequences and can be found on-line at http://us.expasy.org/tools/pratt/.

Further embodiments of the present method

Preferably, the sequences of step a) are derived from expressed or open reading frame (ORF) sequences. This is because the adjuvants of the present invention are based on expressed peptide sequences of pathogens.

Preferably, the host organism is a vertebrate, more preferably a mammal, most preferably a human.

In addition to the steps described above, the method may comprise the step of selecting those polypeptide sequences that are found, or predicted to be found, on the surface of, or are secreted by, a pathogenic cell. This is because in nature, the host immune system is more likely to initially encounter a secreted protein or surface protein of the pathogen rather than any intracellular component. This step therefore mimics the natural environment in this respect.

In a preferred embodiment, this step is carried out between steps b) and c) of the method. More preferably, only a protein family that included at least one amino acid sequence that is predicted, or has been annotated, as a surface or secreted protein is selected for use in step c).

Preferably, this step comprises inferring whether the polypeptide sequence is expressed on the surface or secreted by a pathogenic cell by making use of published annotation data. Such annotation data are generally available on public databases known to a skilled person including the NCBI databases available at http://www.ncbi.nlm.nih.gov.

Alternatively, this step may comprise predicting whether the polypeptide sequence is expressed on the surface or secreted by a pathogenic cell by using an algorithm or program that provides accurate predictions of function. Such programs may make use of certain characteristics that are known to be shared by surface or secreted peptides such as a localisation signal. Usually, the localisation signal takes the form of a short peptide sequence. Often, but not always, this

constitutes a signal sequence (or leader sequence). In the case of bacterial secreted proteins, most comprise leader sequences that are often of 15-40 amino acids in length (most commonly about 20), have a charged segment at the amino terminus, with one or two basic amino acids (e.g. lysine) followed by a stretch of hydrophobic amino acids, which usually includes two glycines or pralines, have a hydrophobic sequence followed by a stretch of about six amino acids that is thought to make a reverse turn in the chain.

An example of such a program is SignalP, a powerful tool for the detection of signal peptides and their cleavage sites [9] and available on-line at http://www.cbs.dtu.dk/services/SignalP/. Another example is PSORT, which annotates sequences as surface exposed or as surface, membrane or periplasm related and available at http://psort.nibb.ac.jp/. Preferably, the PSORT program is used to predict whether the polypeptide sequence is expressed on the surface or secreted by a pathogenic cell.

The method may additionally comprise the step of selecting a polypeptide sequence from step b) or step c) that is not identical to an endogenous human amino acid sequence. Like step b) ii), this provides a filtering step to exclude those protein families or amino acid sequences within a protein family that are not exclusive to pathogenic organisms. This step therefore preferably excludes all patterns that encode at least one amino acid sequence present in the human genome.

In addition to those steps described above, the method may additionally comprise the further step of producing a polypeptide comprising or consisting of the sequence identified by the method of the present invention. Methods for producing such polypeptides are well known to a skilled person and are described in more detail below.

Adjuvant polypeptides

The method of the invention reveals amino acid sequences suitable for use/testing as adjuvants. Therefore the invention provides a polypeptide comprising an amino acid sequence obtainable by the method described above. Because of their pathogen-specific nature, such polypeptides are ideal candidates for vaccine adjuvants. The immune stimulating properties of polypeptides are well known [e.g. see ref. 10]. Many known polypeptide adjuvants are modified with lipids or glycans (especially from bacteria, e.g. MDP), but unmodified peptide adjuvants have also been disclosed [17].

The invention provides polypeptides identified by methods of the invention. For instance, the invention provides a polypeptide comprising any one of the amino acid sequences listed in Table 3. The amino acid sequences in Table 3 are represented in PROSITE notation, with the amino acids being represented by their one-letter codes [11]. Briefly, a peptide comprising the following formula:

A(1)-x(i1,j1)-A(2)-x(i2 I j2)-....A{p-1}-x(i{p-1},j{p-1})-Ap is to be interpreted in the following manner: A(k) is a component, either specifying one amino acid, e.g. C, or a set of amino acids, e.g. [ILVF]. A component A(k) is an identity component if it specifies exactly one amino acid (for instance C or L) or an ambiguous component if it specifies more than one (for instance [ILVF] or [FWY]). i(k), j(k) are integers so that i(k)<=j(k) for all k. The part x(ikjk) specifies a wildcard region of the pattern matching between ik and jk arbitrary amino acids. A wildcard region x(ikjk) is "flexible" if jk is bigger than ik (for example x(2,3)). The flexibility of such a region is jk-ik. For example the flexibility of x(2,3) is 1. The wildcard region is fixed if j(k) is equal to i(k), e.g., x(2,2) which can be written as x(2). The product of flexibility for a pattern is the product of the flexibilities of the flexible wildcard regions in the pattern, if any, otherwise it is defined to be one.

For example, C-x(2)-H is a pattern with two components (C and H) and one fixed wildcard region. It matches any sequence containing a C followed by any two arbitrary amino acids followed by an H. Amino acid sequences ChgHyw (SEQ ID NO: 1) and liChgHlyw (SEQ ID NO: 2) would be included in the formula. C-x(2,3)-H is a pattern with two components (C and H) and one flexible wildcard region. It matches any sequence containing a C followed by any two or three arbitrary amino acids followed by an H such as aaChgHywk (SEQ ID NO: 3) and liChgaHlyw (SEQ ID NO: 4). C-x(2,3)-[ILV] is a pattern with two components (C and [ILV]) and one flexible wildcard region. It matches any sequence containing a C followed by any two or three arbitrary amino acids followed by an I, L or V.

The invention also provides a polypeptide comprising an amino acid sequence having at least c% sequence identity to the amino acid sequences in Table 3 and/or comprising an amino acid sequence consisting of a fragment of at least x contiguous amino acids from an amino acid sequence of Table 3. Preferably the polypeptide has adjuvant activity e.g. in humans. The value of c is at least 85 e.g. 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 99.5, or more. The value of x is at least 5 e.g. 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50. The fragment preferably retains adjuvant activity. Adjuvant activity can be assessed by measuring the immune response induced following co

administration of antigen in the presence or absence of the test adjuvant. Adjuvants enhance the immune response against a co-administered antigen. Examples of such methods are described in references 12 and 13.

The invention also provides a polypeptide comprising an amino acid sequence listed in Table 3, except that the amino acid sequence contains one or more variations. The mutations may each independently be a substitution, an insertion, or a deletion. Preferably, the amino acid sequences contains fewer than twenty mutations (e.g. 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1). Each variation preferably involves a single amino acid. It is preferred that substitutions are conservative i.e. replacement of one amino acid with another which has a related side chain. Genetically-encoded amino acids are generally divided into four families: (1) acidic i.e. aspartate, glutamate; (2) basic i.e. lysine, arginine, histidine; (3) non-polar i.e. alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar i.e. glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. In general, substitution of single amino acids within these families does not have a major effect on the biological activity.

Polypeptides of the invention will be at least 3 (e.g. 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 or more) amino acids in length. Polypeptides as short as 3 amino acids have been shown in the prior art to stimulate an immune response [14, 15, 16]. A 5-mer peptide (ALTTE) (SEQ ID NO: 5) from a bacterial fimbriae protein has also been shown to induce cytokine production from cells in vitro [17]. Although the over-riding factor that determines the length of the polypeptide is that it has to possess adjuvant activity, other factors may also contribute to the determination of the final length of the polypeptide. Such factors may include the expense involved in manufacturing said polypeptide, with shorter polypeptides being generally cheaper to synthesise.

Polypeptides of the invention may comprise fewer than 100 (e.g. 90, 80, 70, 60, 50, 45, 40, 35, 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11 , 10, 9, 8, 7, 6, 5, 4) amino acids.

Also provided in this invention is a polypeptide comprising the formula NH 2 -A-(B-C) n -D-COOH, wherein: A is an optional N-terminal amino acid sequence; B is an adjuvant polypeptide

sequence obtainable by the method of the invention (e.g. a sequence in Table 3); C is an optional amino acid linker sequence; D is an optional C-terminal amino acid sequence; and n>1. Where A7>1 , each B and/or C may be the same or different in each of the n repetitions of B-C. Thus the polypeptide sequence A-(B 1 -C 1 )-(B 2 -C 2 )-D, where B 1 ^B 2 and C^C 2 , still satisfies the formula A-(B-C) n -D.

The/each sequence -B- may consist of fewer than 100 (e.g. 90, 80, 70, 60, 50, 45, 40, 35, 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4) amino acids.

The sequences -A-, -C- and -D- may comprise a tag sequence to aid in purification, a sequence that confers higher protein stability, etc. The sequence(s) -C- may comprise a linker sequence (e.g. a poly-glycine linker). The optional N-terminal -A- may contain a secretory or leader sequence for directing protein trafficking. The/each sequence -A-, -C- and/or -D- may consist of fewer than 100 (e.g. 90, 80, 70, 60, 50, 45, 40, 35, 30, 25, 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4) amino acids.

When A and/or D sequences are present, these may be short (e.g. each <30 amino acids) or long (e.g. each longer than 30 amino acids). In the long form, A and/or D preferably comprises an immunogenic polypeptide sequence, thereby giving a fusion polypeptide including an antigen that makes use of the adjuvant sequence B. Having an antigen and adjuvant in the same polypeptide molecule simplifies production and purification, and enhancement of immunogenicity in this way has been shown using adjuvant portions of HSP [18].

Polypeptides of the invention can be prepared in any suitable manner e.g. by chemical synthesis (at least in part), by digesting longer polypeptides using proteases, by translation from

RNA, by purification from cell culture (e.g. from recombinant expression), etc. The choice of how to prepare the polypeptide will depend on various factors. For short polypeptides, in vitro chemical synthesis [19, 20] will usually be the choice. Solid-phase peptide synthesis is particularly preferred, such as methods based on t-Boc or Fmoc [21] chemistry. Enzymatic synthesis [22] may also be used in part or in full.

For longer polypeptides, particularly those which include antigen and adjuvant within a single fusion polypeptide chain, biological synthesis will generally be the choice e.g. the polypeptides may be produced by translation. Translation may be carried out in vitro or in vivo.

In addition to their essential nature as polymers of amino acids, polypeptides of the invention may include modifications at various positions, including the peptide backbone, the amino acid side-chains and at the amino or carboxyl termini. Blockage of the amino and/or carboxyl terminus of a polypeptide by a covalent modification is common in naturally-occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention. Similarly, modified amino acids (e.g. hydroxyproline, γ-carboxyglutamate, O-phosphoserine, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium, /V-formyl-methionine) may be present.

Polypeptides of the invention are generally provided in substantially pure form e.g. such that less than 50%, usually less than 60% and more usually less than 90% of the composition is made up of other polypeptide(s).

Polypeptides of the invention can be prepared in various forms (e.g. native, fusions, glycosylated, non-glycosylated, etc.). Polypeptides of the invention may be attached to a solid support. Polypeptides of the invention may comprise a detectable label (e.g. a radioactive or fluorescent label, or a biotin label).

The polypeptide of the invention is preferably not a full-length wild type polypeptide. It is preferably not an unmodified NH 2 -ALTTE-COOH pentapeptide (SEQ ID NO: 5).

Peptidomimetics

Polypeptides of the invention are useful adjuvants in their own right. However, they may be refined to improve adjuvant activity (either general or specific) or to improve pharmacologically important features such as bio-availability, toxicology, metabolism, pharmacokinetics, etc. The polypeptides may therefore be used as lead compounds for further research and refinement.

II'"" 11,1.1' Il / 1 I"'! 1 ''"'I' "'"" " "" '' "'"" •■ '' "" " ''"'" ' "

Polypeptides of the invention can be used for designing peptidomimetic molecules [e.g. refs. 23 to 28] with adjuvant activity. These will typically be isosteric with respect to the polypeptides of the invention but will lack one or more of their peptide bonds. For example, the peptide backbone may be replaced by a non-peptide backbone while retaining important amino acid 5 side chains.

The invention therefore provides a polymer comprising any one of the sequences listed in Table 3, wherein (a) the polymer comprises monomers selected from the group consisting of (but not limited to): L-amino acids; D-amino acids; and amino acid mimetics (such as those discussed in 10 reference 29), and/or (b) the bonds between monomers are not peptide bonds. This polymer will not consist of a chain of L-amino acids joined by peptide bonds to form a linear unbranched polypeptide chain.

Different types of monomers (e.g. L- and D-amino acids) may be included in the same polymer, 15 or the polymer may include a single type of monomer (e.g. all D-amino acids).

The polymer may include "peptoid" residues may be used. "Peptoids" result from the oligomeric assembly of N-substituted glycines [30]. Peptidomimetic compounds are advantageous because they omit classical peptide characteristics such as enzymatically scissille peptidic 20 bonds. The polymer may comprise sugar amino acids [31].

Peptidomimetic compounds of the invention will generally be prepared by chemical synthetic routes, as biological methods are in general restricted to the production of polypeptides based on L-amino acids. However, manipulation of translation machinery (e.g. of aminoacyl-tRNA 25 molecules) can be used to allow the introduction of D-amino acids (or of other non-natural amino acids, such as iodotyrosine or methylphenylalanine, azidohomoalanine, efc.) [32].

Nucleic acid molecules encoding the adjuvant polypeptide sequences and related products

30 The invention provides nucleic acid comprising a nucleotide sequence which encodes a polypeptide of the invention.

i 1 ' IUI' '...'I' 'UH 1 ImI 1 I'' "mi' - 1 ' 1 H 'ImI' Il

The invention also provides nucleic acid which hybridises under high stringency conditions to a nucleic acid which encodes a polypeptide of the invention.

The invention also provides nucleic acid comprising a nucleotide sequence which has at least 75% identity {e.g. 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 100% identity) to a nucleotide sequence which encodes a polypeptide of the invention.

Further, the invention provides a vector, such as an expression vector, including such nucleic acids as plasmid DNA and recombinant viral and bacterial sequences.

The invention further provides a host cell transformed with a vector of the invention.

Medicaments and immunogenic compositions The invention provides a composition comprising a polypeptide or polymer of the invention, and a pharmaceutically acceptable carrier.

The term "pharmaceutically acceptable carrier" refers to a carrier for administration of a therapeutic agent, such as antibodies or a polypeptide, genes, and other therapeutic agents. The term refers to any pharmaceutical carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition, and which can be administered without undue toxicity. Suitable carriers can be large, slowly metabolised macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. Pharmaceutically acceptable carriers in therapeutic compositions can include liquids such as water, saline, glycerol and ethanol. Auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, can also be present in such vehicles. Typically, the therapeutic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared. Pharmaceutically acceptable salts can also be present in the pharmaceutical composition, e.g. mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of pharmaceutically acceptable

iι,,,ιι Ii .•' 1 U 1 'imi' i' 'i.'.ii . ■■ ii,,,,, ,ιi ir n;;;ii "T 1 excipients is available in reference 33. Preferred medicaments are aqueous, buffered at pH 7.0+0.5, pyrogen-free and sterile.

In one embodiment, the invention provides a composition comprising microparticles and/or 5 microemulsions and a polypeptide or polymer of the invention. Such microparticles and emulsions have been shown to potentiate the adjuvant activity of other known adjuvants [34- 36].

Compositions of the invention are preferably immunogenic e.g. vaccines. Vaccines according to 10 the invention may either be prophylactic (i.e. to prevent infection) or therapeutic (i.e. to treat disease after onset), but will typically be prophylactic. Therapeutic vaccines can also be used to treat non-infectious diseases such as cancer, allergy and asthma.

Immunogenic compositions used as vaccines comprise an immunologically effective 15 amount of an antigen, as well as any other of the above-mentioned components, as needed. By 'immunologically effective amount', it is meant that the administration of that amount to an individual, either in a single dose or as part of a series, is effective for treatment or prevention. This amount varies depending upon the health and physical condition of the individual to be treated, age, the taxonomic group of individual to be treated 20 (e.g. non-human primate, primate, etc.), the capacity of the individual's immune system to synthesise antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.

25

Compositions of the invention may be administered in conjunction with one or more antigens for use in therapeutic, prophylactic, or diagnostic methods of the present invention. Preferred antigens include those listed below. Additionally, the compositions of the present invention may be used to treat or prevent infections caused by any of the below-listed 30 microbes.

In addition to combination with the antigens described below, the compositions of the invention may also be combined with an adjuvant as described herein. Antigens for use with

the invention include, but are not limited to, one or more of the following antigens set forth below, or antigens derived from one or more of the pathogens set forth below:

A. Bacterial Antigens Bacterial antigens suitable for use in the invention include proteins, polysaccharides, lipopolysaccharides, and outer membrane vesicles which may be isolated, purified or derived from a bacteria. In addition, bacterial antigens may include bacterial lysates and inactivated bacteria formulations. Bacteria antigens may be produced by recombinant expression. Bacterial antigens preferably include epitopes which are exposed on the surface of the bacteria during at least one stage of its life cycle. Bacterial antigens are preferably conserved across multiple serotypes. Bacterial antigens include antigens derived from one or more of the bacteria set forth below as well as the specific antigens examples identified below.

Neisseria meningitides: Meningitides antigens may include proteins (such as those identified in References A-G), saccharides (including a polysaccharide, oligosaccharide or lipopolysaccharide), or outer-membrane vesicles (References H, I, J, K) purified or derived from N. meningitides serogroup A, C, W135, Y, and/or B. Meningitides protein antigens may be selected from adhesions, autotransporters, toxins, Fe acquisition proteins, and membrane associated proteins (preferably integral outer membrane protein).

Streptococcus pneumoniae: Streptococcus pneumoniae antigens may include a saccharide (including a polysaccharide or an oligosaccharide) or protein from Streptococcus pneumoniae. Saccharide antigens may be selected from serotypes 1 , 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11 A, 12F, 14, 15B, 17F 1 18C, 19A, 19F, 20, 22F, 23F, and 33F. Protein antigens may be selected from a protein identified in WO 98/18931 , WO 98/18930, US Patent No. 6,699,703, US Patent No. 6,800,744, WO 97/43303, and WO 97/37026. Streptococcus pneumoniae proteins may be selected from the Poly Histidine Triad family (PhtX), the Choline Binding Protein family (CbpX), CbpX truncates, LytX family, LytX truncates, CbpX truncate-LytX truncate chimeric proteins, pneumolysin (Ply), PspA, PsaA, Sp128, Sp101 , Sp130, Sp125 or Sp133.

Streptococcus pyogenes (Group A Streptococcus): Group A Streptococcus antigens may include a protein identified in WO 02/34771 or WO 2005/032582 (including GAS 40), fusions of fragments of GAS M proteins (including those described in WO 02/094851, and Dale, Vaccine

(1999) 17:193-200, and Dale, Vaccine 14(10): 944-948), fibronectin binding protein (Sf b1), Streptococcal heme-associated protein (Shp), and Streptolysin S (SagA).

Moraxella catarrhalis: Moraxella antigens include antigens identified in WO 02/18595 and WO 99/58562, outer membrane protein antigens (HMW-OMP), C-antigen, and/or LPS.

Bordetella pertussis: Pertussis antigens include petussis holotoxin (PT) and filamentous haemagglutinin (FHA) from β. pertussis, optionally also combination with pertactin and/or agglutinogens 2 and 3 antigen.

Staphylococcus aureus: Staph aureus antigens include S. aureus type 5 and 8 capsular polysaccharides optionally conjugated to nontoxic recombinant Pseudomonas aeruginosa exotoxin A, such as StaphVAX™, or antigens derived from surface proteins, invasins (leukocidin, kinases, hyaluronidase), surface factors that inhibit phagocytic engulfment (capsule, Protein A), carotenoids, catalase production, Protein A, coagulase, clotting factor, and/or membrane-damaging toxins (optionally detoxified) that lyse eukaryotic cell membranes (hemolysins, leukotoxin, leukocidin).

Staphylococcus epidermis: S. epidermidis antigens include slime-associated antigen (SAA).

Tetanus: Tetanus antigens include tetanus toxoid (TT), preferably used as a carrier protein in conjunction/conjugated with the compositions of the present invention.

Diphtheria: Diphtheria antigens include diphtheria toxin, preferably detoxified, such as CRM 197 , additionally antigens capable of modulating, inhibiting or associated with ADP ribosylation are contemplated for combination/co-administration/conjugation with the compositions of the present invention, the diphtheria toxoids are preferably used as carrier proteins.

Haemophilus influenzae B (Hib): Hib antigens include a Hib saccharide antigen.

Pseudomonas aeruginosa: Pseudomonas antigens include endotoxin A, Wzz protein, P. aeruginosa LPS, more particularly LPS isolated from PAO1 (05 serotype), and/or Outer Membrane Proteins, including Outer Membrane Proteins F (OprF) (Infect Immun. 2001 May; 69(5): 3510-3515).

Legionella pneumophila (Legionnairs' Disease): L. pneumophila antigens may optionally derived from cell lines with disrupted asd genes (Infect Immun. 1998 May; 66(5): 1898).

Streptococcus agalactiae (Group B Streptococcus): Group B Streptococcus antigens include a protein or saccharide antigen identified in WO 02/34771 , WO 03/093306, WO 04/041157, or WO 2005/002619 (including proteins GBS 80, GBS 104, GBS 276 and GBS 322, and including saccharide antigens derived from serotypes Ia, Ib, la/c, II, III, IV, V, Vl, VII and VIII).

Neiserria gonorrhoeae: Gonorrhoeae antigens include Por (or porin) protein, such as PorB (see Zhu et a/., Vaccine (2004) 22:660 - 669), a transferring binding protein, such as TbpA and TbpB (See Price et a/., Infection and Immunity (2004) 71(1):277 - 283), a opacity protein (such as Opa), a reduction-modifiable protein (Rmp), and outer membrane vesicle (OMV) preparations (see Plante et al., J Infectious Disease (2000) 182:848 - 855), also see e.g. WO99/24578, WO99/36544, WO99/57280, WO02/079243).

Chlamydia trachomatis: Chlamydia trachomatis antigens include antigens derived from serotypes A, B, Ba and C are (agents of trachoma, a cause of blindness), serotypes L 1 , L 2 & L 3 (associated with Lymphogranuloma venereum), and serotypes, D-K. Chlamydia trachomas antigens may also include an antigen identified in WO 00/37494, WO 03/049762, WO 03/068811 , or WO 05/002619.

Treponema pallidum (Syphilis): Syphilis antigens include TmpA antigen.

Haemophilus ducreyi (causing chancroid): Ducreyi antigens include outer membrane protein (DsrA).

Enterococcus faecalis or Enterococcus faecium: Antigens include a trisaccharide repeat or other Enterococcus derived antigens provided in US Patent No. 6,756,361.

Helicobacter pylori: H pylori antigens include Cag, Vac, Nap, HopX, HopY and/or urease antigen.

Staphylococcus saprophytics: Antigens include the 160 kDa hemagglutinin of S. saprophyticus antigen.

Yersinia enterocolitica Antigens include LPS (Infect Immun. 2002 August; 70(8): 4414).

E. coli: E. coli antigens may be derived from enterotoxigenic E. coli (ETEC), enteroaggregative E. coli (EAggEC), diffusely adhering E. coli (DAEC), enteropathogenic E. coli (EPEC), and/or enterohemorrhagic E. coli (EHEC).

Bacillus anthracis (anthrax); β. anthracis antigens are optionally detoxified and may be selected from A-components (lethal factor (LF) and edema factor (EF)), both of which can share a common B-component known as protective antigen (PA).

Yersinia pestis (plague): Plague antigens include F1 capsular antigen (Infect Immun. 2003 Jan; 71 (1 )): 374-383, LPS (Infect Immun. 1999 Oct; 67(10): 5395), Yersinia pestis V antigen (Infect Immun. 1997 Nov; 65(11 ): 4476-4482).

Mycobacterium tuberculosis: Tuberculosis antigens include lipoproteins, LPS, BCG antigens, a fusion protein of antigen 85B (Ag85B) and/or ESAT-6 optionally formulated in cationic lipid vesicles (Infect Immun. 2004 October; 72(10): 6148), Mycobacterium tuberculosis (Mtb) isocitrate dehydrogenase associated antigens (Proc Natl Acad Sci U S A. 2004 Aug 24; 101(34): 12652), and/or MPT51 antigens {Infect Immun. 2004 July; 72(7): 3829).

Rickettsia: Antigens include outer membrane proteins, including the outer membrane protein A and/or B (OmpB) (Biochim Biophys Acta. 2004 Nov 1 ;1702(2):145), LPS, and surface protein antigen (SPA) (J Autoimmun. 1989 Jun;2 Suppl:81).

Listeria monocytogenes : Antigens derived from L. monocytogenes are preferably used as carriers/vectors for intracytoplasmic delivery of conjugates/associated compositions of the present invention.

Chlamydia pneumoniae: Antigens include those identified in WO 02/02606.

Vibrio cholerae: Antigens include proteinase antigens, LPS, particularly lipopolysaccharides of Vibrio cholerae II, 01 lnaba O-specific polysaccharides, V. cholera 0139, antigens of IEM108 vaccine (Infect Immun. 2003 Oct;71(10):5498-504), and/or Zonula occludens toxin (Zot).

Salmonella typhi (typhoid fever): Antigens include capsular polysaccharides preferably conjugates (Vi, i.e. vax-TyVi).

Borrelia burgdorferi (Lyme disease): Antigens include lipoproteins (such as OspA, OspB, Osp C and Osp D), other surface proteins such as OspE-related proteins (Erps), decorin-binding proteins (such as DbpA), and antigenically variable Vl proteins. , such as antigens associated with P39 and P13 (an integral membrane protein, Infect Immun. 2001 May; 69(5): 3323-3334), VIsE Antigenic Variation Protein (J CHn Microbiol. 1999 Dec; 37(12): 3997).

Porphyromonas gingivalis: Antigens include P. gingivalis outer membrane protein (OMP).

Klebsiella: Antigens include an OMP, including OMP A, or a polysaccharide optionally conjugated to tetanus toxoid.

Where not specifically referenced, further bacterial antigens of the invention may be capsular antigens, polysaccharide antigens or protein antigens of any of the above. Further bacterial antigens may also include an outer membrane vesicle (OMV) preparation. Additionally, antigens

include live, attenuated, split, and/or purified versions of any of the aforementioned bacteria. The bacterial or microbial derived antigens of the present invention may be gram-negative or gram-positive and aerobic or anaerobic.

Additionally, any of the above bacterial-derived saccharides (polysaccharides, LPS, LOS or oligosaccharides) can be conjugated to another agent or antigen, such as a carrier protein (for example CRM 197 ). Such conjugation may be direct conjugation effected by reductive amination of carbonyl moieties on the saccharide to amino groups on the protein, as provided in US Patent No. 5,360,897 and Can J Biochem Cell Biol. 1984 May;62(5):270-5. Alternatively, the saccharides can be conjugated through a linker, such as, with succinamide or other linkages provided in Bioconjugate Techniques, 1996 and CRC, Chemistry of Protein Conjugation and Cross-Linking, 1993.

B. Viral Antigens Viral antigens suitable for use in the invention include inactivated (or killed) virus, attenuated virus, split virus formulations, purified subunit formulations, viral proteins which may be isolated, purified or derived from a virus, and Virus Like Particles (VLPs). Viral antigens may be derived from viruses propagated on cell culture or expressed recombinantly. Viral antigens preferably include epitopes which are exposed on the surface of the virus during at least one stage of its life cycle. Viral antigens are preferably conserved across multiple serotypes. Viral antigens include antigens derived from one or more of the viruses set forth below as well as the specific antigens examples identified below.

Orthomyxovirus: Viral antigens may be derived from an Orthomyxovirus, such as Influenza A, B and C. Orthomyxovirus antigens may be selected from one or more of the viral proteins, including hemagglutinin (HA), neuraminidase (NA), nucleoprotein (NP), matrix protein (M1 ), membrane protein (M2), one or more of the transcriptase components (PB1, PB2 and PA). Preferred antigens include HA and NA.

Influenza antigens may be derived from interpandemic (annual) flu strains. Alternatively influenza antigens may be derived from strains with the potential to cause pandemic a pandemic outbreak (i.e., influenza strains with new haemagglutinin compared to the haemagglutinin in currently circulating strains, or influenza strains which are pathogenic in avian

If 1 "" Il It ,.'' ifiul 1 '....Ii limn Ui .•' «.,.,. J 1 Ii limn ii subjects and have the potential to be transmitted horizontally in the human population, or influenza strains which are pathogenic to humans).

Paramyxoviridae viruses: Viral antigens may be derived from Paramyxoviridae viruses, such as 5 Pneumoviruses (RSV), Paramyxoviruses (PIV) and Morbilliviruses (Measles).

Pneumovirus: Viral antigens may be derived from a Pneumovirus, such as Respiratory syncytial virus (RSV), Bovine respiratory syncytial virus, Pneumonia virus of mice, and Turkey rhinotracheitis virus. Preferably, the Pneumovirus is RSV. Pneumovirus antigens may be

10 selected from one or more of the following proteins, including surface proteins Fusion (F), Glycoprotein (G) and Small Hydrophobic protein (SH), matrix proteins M and M2, nucleocapsid proteins N, P and L and nonstructural proteins NS1 and NS2. Preferred Pneumovirus antigens include F, G and M. See e.g., J Gen Virol. 2004 Nov; 85(Pt 11):3229). Pneumovirus antigens may also be formulated in or derived from chimeric viruses. For example, chimeric RSV/PIV

15 viruses may comprise components of both RSV and PIV.

Paramyxovirus: Viral antigens may be derived from a Paramyxovirus, such as Parainfluenza virus types 1 - 4 (PIV), Mumps, Sendai viruses, Simian virus 5, Bovine parainfluenza virus and Newcastle disease virus. Preferably, the Paramyxovirus is PIV or Mumps. Paramyxovirus

20 antigens may be selected from one or more of the following proteins: Hemagglutinin - Neuraminidase (HN), Fusion proteins F1 and F2, Nucleoprotein (NP), Phosphoprotein (P), Large protein (L), and Matrix protein (M). Preferred Paramyxovirus proteins include HN, F1 and F2. Paramyxovirus antigens may also be formulated in or derived from chimeric viruses. For example, chimeric RSV/PIV viruses may comprise components of both RSV and PIV.

25 Commercially available mumps vaccines include live attenuated mumps virus, in either a monovalent form or in combination with measles and rubella vaccines (MMR).

Morbillivirus: Viral antigens may be derived from a Morbillivirus, such as Measles. Morbillivirus antigens may be selected from one or more of the following proteins: hemagglutinin (H), 30 Glycoprotein (G), Fusion factor (F), Large protein (L), Nucleoprotein (NP), Polymerase phosphoprotein (P), and Matrix (M). Commercially available measles vaccines include live attenuated measles virus, typically in combination with mumps and rubella (MMR).

Picornavirus: Viral antigens may be derived from Picornaviruses, such as Enteroviruses, Rhinoviruses, Heparnavirus, Cardioviruses and Aphthoviruses. Antigens derived from Enteroviruses, such as Poliovirus are preferred.

Enterovirus: Viral antigens may be derived from an Enterovirus, such as Poliovirus types 1 , 2 or 3, Coxsackie A virus types 1 to 22 and 24, Coxsackie B virus types 1 to 6, Echovirus (ECHO) virus) types 1 to 9, 11 to 27 and 29 to 34 and Enterovirus 68 to 71. Preferably, the Enterovirus is poliovirus. Enterovirus antigens are preferably selected from one or more of the following Capsid proteins VP1 , VP2, VP3 and VP4. Commercially available polio vaccines include Inactivated Polio Vaccine (IPV) and Oral poliovirus vaccine (OPV).

Heparnavirus: Viral antigens may be derived from an Heparnavirus, such as Hepatitis A virus (HAV). Commercially available HAV vaccines include inactivated HAV vaccine.

Togavirus: Viral antigens may be derived from a Togavirus, such as a Rubivirus, an Alphavirus, or an Arterivirus. Antigens derived from Rubivirus, such as Rubella virus, are preferred. Togavirus antigens may be selected from E1, E2, E3, C, NSP-1 , NSPO-2, NSP-3 or NSP-4. Togavirus antigens are preferably selected from E1, E2 or E3. Commercially available Rubella vaccines include a live cold-adapted virus, typically in combination with mumps and measles vaccines (MMR).

Flavivirus: Viral antigens may be derived from a Flavivirus, such as Tick-borne encephalitis (TBE), Dengue (types 1 , 2, 3 or 4), Yellow Fever, Japanese encephalitis, West Nile encephalitis, St. Louis encephalitis, Russian spring-summer encephalitis, Powassan encephalitis. Flavivirus antigens may be selected from PrM, M, C, E, NS-1 , NS-2a, NS2b, NS3, NS4a, NS4b, and NS5. Flavivirus antigens are preferably selected from PrM, M and E. Commercially available TBE vaccine include inactivated virus vaccines.

Pestivirus: Viral antigens may be derived from a Pestivirus, such as Bovine viral diarrhea (BVDV), Classical swine fever (CSFV) or Border disease (BDV).

Hepadnavirus: Viral antigens may be derived from a Hepadnavirus, such as Hepatitis B virus.

Hepadnavirus antigens may be selected from surface antigens (L, M and S), core antigens

II ' " IUi' Il • ' 'I '"' """' """' " "'"" -" ' " "

(HBc, HBe). Commercially available HBV vaccines include subunit vaccines comprising the surface antigen S protein.

Hepatitis C virus: Viral antigens may be derived from a Hepatitis C virus (HCV). HCV antigens 5 may be selected from one or more of E1 , E2, E1/E2, NS345 polyprotein, NS 345-core polyprotein, core, and/or peptides from the nonstructural regions (Houghton et al., Hepatology (1991 ) 14:381 ; See also WO92/08734).

Rhabdovirus: Viral antigens may be derived from a Rhabdovirus, such as a Lyssavirus (Rabies 10 virus) and Vesiculovirus (VSV). Rhabdovirus antigens may be selected from glycoprotein (G), nucleoprotein (N), large protein (L), nonstructural proteins (NS). Commercially available Rabies virus vaccine comprise killed virus grown on human diploid cells or fetal rhesus lung cells.

Caliciviridae; Viral antigens may be derived from Calciviridae, such as Norwalk virus.

15

Coronavirus: Viral antigens may be derived from a Coronavirus, SARS, Human respiratory coronavirus, Avian infectious bronchitis (IBV), Mouse hepatitis virus (MHV), and Porcine transmissible gastroenteritis virus (TGEV). Coronavirus antigens may be selected from spike (S), envelope (E) 1 matrix (M), nucleocapsid (N), and Hemagglutinin-esterase glycoprotein (HE). 20 Preferably, the Coronavirus antigen is derived from a SARS virus. SARS viral antigens are described in WO 04/92360;

Retrovirus: Viral antigens may be derived from a Retrovirus, such as an Oncovirus, a Lentivirus or a Spumavirus. Oncovirus antigens may be derived from HTLV-1 , HTLV-2 or HTLV-5. 25 Lentivirus antigens may be derived from HIV-1 or HIV-2. Retrovirus antigens may be selected from gag, pol, env, tax, tat, rex, rev, nef, vif, vpu, and vpr. HIV antigens may be selected from gag (p24gag and p55gag), env (gp160 and gp41 ), pol, tat, nef, rev vpu, miniproteins, (preferably p55 gag and gp140v delete). HIV antigens may be derived from one or more of the following strains: HIV, llb , HIV SF2 , HIVLAV, HIVLA,, HIV MN , HIV-1 C M 235 , HIV-1 US 4.

30

Reovirus: Viral antigens may be derived from a Reovirus, such as an Orthoreovirus , a Rotavirus, an Orbivirus, or a Coltivirus. Reovirus antigens may be selected from structural proteins λ1 , λ2, A3, μ1 , μ2, σ1 , σ2, or σ3, or nonstructural proteins σNS, μNS, or σ1s. Preferred

Reovirus antigens may be derived from a Rotavirus. Rotavirus antigens may be selected from VP1 , VP2, VP3, VP4 (or the cleaved product VP5 and VP8), NSP 1 , VP6, NSP3, NSP2, VP7, NSP4, or NSP5. Preferred Rotavirus antigens include VP4 (or the cleaved product VP5 and VP8), and VP7.

Parvovirus: Viral antigens may be derived from a Parvovirus, such as Parvovirus B19. Parvovirus antigens may be selected from VP-1 , VP-2, VP-3, NS-1 and NS-2. Preferably, the Parvovirus antigen is capsid protein VP-2.

Delta hepatitis virus (HDV): Viral antigens may be derived HDV, particularly δ-antigen from HDV (see, e.g., U.S. Patent No. 5,378,814).

Hepatitis E virus (HEV): Viral antigens may be derived from HEV.

Hepatitis G virus (HGV): Viral antigens may be derived from HGV.

Human Herpesvirus: Viral antigens may be derived from a Human Herpesvirus, such as Herpes Simplex Viruses (HSV), Varicella-zoster virus (VZV), Epstein-Barr virus (EBV), Cytomegalovirus (CMV), Human Herpesvirus 6 (HHV6), Human Herpesvirus 7 (HHV7), and Human Herpesvirus 8 (HHV8). Human Herpesvirus antigens may be selected from immediate early proteins (α), early proteins (β), and late proteins (Y). HSV antigens may be derived from HSV-1 or HSV-2 strains. HSV antigens may be selected from glycoproteins gB, gC, gD and gH, fusion protein (gB), or immune escape proteins (gC, gE, or gl). VZV antigens may be selected from core, nucleocapsid, tegument, or envelope proteins. A live attenuated VZV vaccine is commercially available. EBV antigens may be selected from early antigen (EA) proteins, viral capsid antigen (VCA), and glycoproteins of the membrane antigen (MA). CMV antigens may be selected from capsid proteins, envelope glycoproteins (such as gB and gH), and tegument proteins.

Papovaviruses: Antigens may be derived from Papovaviruses, such as Papillomaviruses and Polyomaviruses. Papillomaviruses include HPV serotypes 1 , 2, 4, 5, 6, 8, 11 , 13, 16, 18, 31 , 33, 35, 39, 41 , 42, 47, 51 , 57, 58, 63 and 65. Preferably, HPV antigens are derived from serotypes 6, 11 , 16 or 18. HPV antigens may be selected from capsid proteins (L1) and (L2), or

E1 - E7, or fusions thereof. HPV antigens are preferably formulated into virus-like particles (VLPs). Polyomyavirus viruses include BK virus and JK virus. Polyomavirus antigens may be selected from VP1 , VP2 or VP3.

Further provided are antigens, compostions, methods, and microbes included in Vaccines, 4 th Edition (Plotkin and Orenstein ed. 2004); Medical Microbiology 4 th Edition (Murray et al. ed. 2002); Virology, 3rd Edition (W.K. Joklik ed. 1988); Fundamental Virology, 2nd Edition (B.N. Fields and D. M. Knipe, eds. 1991), which are contemplated in conjunction with the compositions of the present invention.

Fungal Antigens

In some embodiments compositions of the present invention are combined with fungal antigens for use in methods of the present invention, including treatment or prevention of mycoses. Fungal antigens for use herein, associated with vaccines include those described in: U.S. Pat. Nos. 4,229,434 and 4,368,191 for prophylaxis and treatment of trichopytosis caused by Trichophyton mentagrophytes; U.S. Pat. Nos. 5,277,904 and 5,284,652 for a broad spectrum dermatophyte vaccine for the prophylaxis of dermatophyte infection in animals, such as guinea pigs, cats, rabbits, horses and lambs, these antigens comprises a suspension of killed T. equinum, T. mentagrophytes (var. granulare), M. canis and/or M. gypseum in an effective amount optionally combined with an adjuvant; U.S. Pat. Nos. 5,453,273 and 6,132,733 for a ringworm vaccine comprising an effective amount of a homogenized, formaldehyde-killed fungi, i.e., Microsporum canis culture in a carrier; U.S. Pat. No. 5,948,413 involving extracellular and intracellular proteins for pythiosis. Additional antigens identified within antifungal vaccines include Ringvac bovis LTF-130 and Bioveta.

Further, fungal antigens for use herein may be derived from Dermatophytres, including: Epidermophyton floccusum, Microsporum audouini, Microsporum canis, Microsporum distortum, Microsporum equinum, Microsporum gypsum, Microsporum nanum, Trichophyton concentricum, Trichophyton equinum, Trichophyton gallinae, Trichophyton gypseum, Trichophyton megnini, Trichophyton mentagrophytes, Trichophyton quinckeanum, Trichophyton rubrum, Trichophyton schoenleini, Trichophyton tonsurans, Trichophyton verrucosum, T. verrucosum var. album, var. discoides, var. ochraceum, Trichophyton violaceum, and/or Trichophyton faviforme.

Fungal pathogens for use as antigens or in derivation of antigens in conjunction with the compositions of the present invention comprise Aspergillus fumigatus, Aspergillus flavus, Aspergillus niger, Aspergillus nidulans, Aspergillus terreus, Aspergillus sydowi, Aspergillus flavatus, Aspergillus glaucus, Blastoschizomyces capitatus, Candida albicans, Candida enolase, Candida tropicalis, Candida glabrata, Candida krusei, Candida parapsilosis, Candida stellatoidea, Candida kusei, Candida parakwsei, Candida lusitaniae, Candida pseudotropicalis, Candida guilliermondi, Cladosporium carrionii, Coccidioides immitis, Blastomyces dermatidis, Cryptococcus neoformans, Geotrichum clavatum, Histoplasma capsulatum, Klebsiella pneumoniae, Paracoccidioides brasiliensis, Pneumocystis carinii, Pythiumn insidiosum, Pityrosporum ovale, Sacharomyces cerevisae, Saccharomyces boulardii, Saccharomyces pombe, Scedosporium apiosperum, Sporothrix schenckii, Trichosporon beigelii, Toxoplasma gondii, Penicillium marneffei, Malassezia spp., Fonsecaea spp., Wangiella spp., Sporothrix spp., Basidiobolus spp., Conidiobolus spp., Rhizopus spp, Mucor spp, Absidia spp, Mortierella spp, Cunninghamella spp, and Saksenaea spp.

Other fungi from which antigens are derived include Altemaria spp, Curvularia spp, Helminthosporium spp, Fusarium spp, Aspergillus spp, Penicillium spp, Monolinia spp, Rhizoctonia spp, Paecilomyces spp, Pithomyces spp, and Cladosporium spp.

Processes for producing a fungal antigens are well known in the art (see US Patent No. 6,333,164). In a preferred method a solubilized fraction extracted and separated from an insoluble fraction obtainable from fungal cells of which cell wall has been substantially removed or at least partially removed, characterized in that the process comprises the steps of: obtaining living fungal cells; obtaining fungal cells of which cell wall has been substantially removed or at least partially removed; bursting the fungal cells of which cell wall has been substantially removed or at least partially removed; obtaining an insoluble fraction; and extracting and separating a solubilized fraction from the insoluble fraction.

STD Antigens Embodiments of the invention include compositions and methods related to a prophylactic and therapeutic treatments for microbes that can be neutralized prior to infection of a cell. In particular embodiments, microbes (bacteria, viruses and/or fungi) against which the present compositions and methods can be implement include those that cause sexually transmitted diseases (STDs) and/or those that display on their surface an antigen that can be the target or

antigen composition of the invention. In a preferred embodiment of the invention, compositions are combined with antigens derived from a viral or bacterial STD. Antigens derived from bacteria or viruses can be administered in conjunction with the compositions of the present invention to provide protection against at least one of the following STDs, among others: chlamydia, genital herpes, hepatitis (particularly HCV), genital warts, gonorrhoea, syphilis and/or chancroid (See, WO00/15255).

In another embodiment the compositions of the present invention are co-administered with an antigen for the prevention or treatment of an STD.

Antigens derived from the following viruses associated with STDs, which are described in greater detail above, are preferred for co-administration with the compositions of the present invention: hepatitis (particularly HCV), HPV, HIV, or HSV.

Additionally, antigens derived from the following bacteria associated with STDs, which are described in greater detail above, are preferred for co-administration with the compositions of the present invention: Neiserria gonorrhoeae, Chlamydia pneumoniae, Chlamydia trachomatis, Treponema pallidum, or Haemophilus ducreyi.

Respiratory Antigens

The invention provides methods of preventing and/or treating infection by a respiratory pathogen, including a virus, bacteria, or fungi such as respiratory syncytial virus (RSV), PIV, SARS virus, influenza, Bacillus anthracis, particularly by reducing or preventing infection and/or one or more symptoms of respiratory virus infection. A composition comprising an antigen described herein, such as one derived from a respiratory virus, bacteria or fungus is administered in conjunction with the compositions of the present invention to an individual which is at risk of being exposed to that particular respiratory microbe, has been exposed to a respiratory microbe or is infected with a respiratory virus, bacteria or fungus. The com position (s) of the present invention is/are preferably co-administered at the same time or in the same formulation with an antigen of the respiratory pathogen. Administration of the composition results in reduced incidence and/or severity of one or more symptoms of respiratory infection.

Tumor Antigens

One embodiment of the present involves a tumor antigen or cancer antigen in conjunction with the compositions of the present invention. Tumor antigens can be, for example, peptide- containing tumor antigens, such as a polypeptide tumor antigen or glycoprotein tumor antigens. A tumor antigen can also be, for example, a saccharide-containing tumor antigen, such as a glycolipid tumor antigen or a ganglioside tumor antigen. The tumor antigen can further be, for example, a polynucleotide-containing tumor antigen that expresses a polypeptide-containing tumor antigen, for instance, an RNA vector construct or a DNA vector construct, such as plasm id DNA.

Tumor antigens appropriate for the practice of the present invention encompass a wide variety of molecules, such as (a) polypeptide-containing tumor antigens, including polypeptides (which can range, for example, from 8-20 amino acids in length, although lengths outside this range are also common), lipopolypeptides and glycoproteins, (b) saccharide-containing tumor antigens, including poly-saccharides, mucins, gangliosides, glycolipids and glycoproteins, and (c) polynucleotides that express antigenic polypeptides.

The tumor antigens can be, for example, (a) full length molecules associated with cancer cells, (b) homologs and modified forms of the same, including molecules with deleted, added and/or substituted portions, and (c) fragments of the same. Tumor antigens can be provided in recombinant form. Tumor antigens include, for example, class l-restricted antigens recognized by CD8+ lymphocytes or class ll-restricted antigens recognized by CD4+ lymphocytes.

Numerous tumor antigens are known in the art, including: (a) cancer-testis antigens such as NY-ESO-1 , SSX2, SCP1 as well as RAGE, BAGE, GAGE and MAGE family polypeptides, for example, GAGE-1 , GAGE-2, MAGE-1 , MAGE-2, MAGE-3, MAGE-4, MAGE-5, MAGE-6, and MAGE-12 (which can be used, for example, to address melanoma, lung, head and neck, NSCLC, breast, gastrointestinal, and bladder tumors), (b) mutated antigens, for example, p53 (associated with various solid tumors, e.g., colorectal, lung, head and neck cancer), p21/Ras (associated with, e.g., melanoma, pancreatic cancer and colorectal cancer), CDK4 (associated with, e.g., melanoma), MUM1 (associated with, e.g., melanoma), caspase-8 (associated with, e.g., head and neck cancer), CIA 0205 (associated with, e.g., bladder cancer), HLA-A2-R1701 , beta catenin (associated with, e.g., melanoma), TCR (associated with, e.g., T-cell non-Hodgkins

lymphoma), BCR-abl (associated with, e.g., chronic myelogenous leukemia), triosephosphate isomerase, KIA 0205, CDC-27, and LDLR-FUT, (c) over-expressed antigens, for example, Galectin 4 (associated with, e.g., colorectal cancer), Galectin 9 (associated with, e.g., Hodgkin's disease), proteinase 3 (associated with, e.g., chronic myelogenous leukemia), WT 1 (associated with, e.g., various leukemias), carbonic anhydrase (associated with, e.g., renal cancer), aldolase A (associated with, e.g., lung cancer), PRAME (associated with, e.g., melanoma), HER-2/neu (associated with, e.g., breast, colon, lung and ovarian cancer), alpha-fetoprotein (associated with, e.g., hepatoma), KSA (associated with, e.g., colorectal cancer), gastrin (associated with, e.g., pancreatic and gastric cancer), telomerase catalytic protein, MUC-1 (associated with, e.g., breast and ovarian cancer), G-250 (associated with, e.g., renal cell carcinoma), p53 (associated with, e.g., breast, colon cancer), and carcinoembryonic antigen (associated with, e.g., breast cancer, lung cancer, and cancers of the gastrointestinal tract such as colorectal cancer), (d) shared antigens, for example, melanoma-melanocyte differentiation antigens such as MART-1/Melan A, gplOO, MC1 R, melanocyte-stimulating hormone receptor, tyrosinase, tyrosinase related protein-1/TRP1 and tyrosinase related protein-2/TRP2 (associated with, e.g., melanoma), (e) prostate associated antigens such as PAP, PSA, PSMA, PSH-P1 , PSM-P1 , PSM-P2, associated with e.g., prostate cancer, [T) immunoglobulin idiotypes (associated with myeloma and B cell lymphomas, for example), and (g) other tumor antigens, such as polypeptide- and saccharide-containing antigens including (i) glycoproteins such as sialyl Tn and sialyl Le x (associated with, e.g., breast and colorectal cancer) as well as various mucins; glycoproteins may be coupled to a carrier protein (e.g., MUC-1 may be coupled to KLH); (ii) lipopolypeptides (e.g., MUC-1 linked to a lipid moiety); (iii) polysaccharides (e.g., Globo H synthetic hexasaccharide), which may be coupled to a carrier proteins (e.g., to KLH), (iv) gangliosides such as GM2, GM12, GD2, GD3 (associated with, e.g., brain, lung cancer, melanoma), which also may be coupled to carrier proteins (e.g., KLH).

Additional tumor antigens which are known in the art include p15, Hom/Mel-40, H-Ras, E2A- PRL, H4-RET, IGH-IGK, MYL-RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens, including E6 and E7, hepatitis B and C virus antigens, human T-cell lymphotropic virus antigens, TSP-180, p185erbB2, p180erbB-3, c-met, mn-23H1 , TAG-72-4, CA 19-9, CA 72-4, CAM 17.1 , NuMa, K-ras, p16, TAGE, PSCA, CT7, 43-9F, 5T4, 791 Tgp72, beta- HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29\BCAA), CA 195, CA 242, CA-50, CAM43, CD68\KP1 , CO-029, FGF-5, Ga733 (EpCAM), HTgp-175, M344, MA-50, MG7-Ag, MOV18, NB/70K, NY-CO-1 , RCAS1 , SDCCAG16, TA-90 (Mac-2 binding protein\cyclophilin C-associated protein), TAAL6, TAG72, TLP, TPS, and the like. These as well as other cellular components are described for example in United States Patent Application 20020007173 and references cited therein.

Polynucleotide-containing antigens in accordance with the present invention typically comprise polynucleotides that encode polypeptide cancer antigens such as those listed above. Preferred polynucleotide-containing antigens include DNA or RNA vector constructs, such as plasmid vectors (e.g., pCMV), which are capable of expressing polypeptide cancer antigens in vivo.

Tumor antigens may be derived, for example, from mutated or altered cellular components. After alteration, the cellular components no longer perform their regulatory functions, and hence the cell may experience uncontrolled growth. Representative examples of altered cellular components include ras, p53, Rb, altered protein encoded by the Wilms' tumor gene, ubiquitin, mucin, protein encoded by the DCC, APC, and MCC genes, as well as receptors or receptor- like structures such as neu, thyroid hormone receptor, platelet derived growth factor (PDGF) receptor, insulin receptor, epidermal growth factor (EGF) receptor, and the colony stimulating factor (CSF) receptor. These as well as other cellular components are described for example in U.S. Patent No. 5,693,522 and references cited therein.

Additionally, bacterial and viral antigens, may be used in conjunction with the compositions of the present invention for the treatment of cancer. In particular, carrier proteins, such as CRM 197 , tetanus toxoid, or Salmonella typhimurium antigen can be used in conjunction/conjugation with compounds of the present invention for treatment of cancer. The cancer antigen combination therapies will show increased efficacy and bioavailability as compared with existing therapies.

Additional information on cancer or tumor antigens can be found, for example, in Moingeon P, "Cancer vaccines," Vaccine, 2001, 19:1305-1326; Rosenberg SA, "Progress in human tumor immunology and immunotherapy," Nature, 2001 , 411 :380-384; Dermine, S. et al, "Cancer Vaccines and Immunotherapy," British Medical Bulletin, 2002, 62, 149-162; Espinoza-Delgado I., "Cancer Vaccines," The Oncologist, 2002, 7(suppI3):20-33; Davis, I. D. et al., "Rational approaches to human cancer immunotherapy," Journal of Leukocyte Biology, 2003, 23: 3-29; Van den Eynde B, et al., "New tumor antigens recognized by T cells," Curr. Opin. Immunol., 1995, 7:674-81 ; Rosenberg SA, "Cancer vaccines based on the identification of genes encoding cancer regression antigens, Immunol. Today, 1997, 18:175-82; Offringa R et al., "Design and evaluation of antigen-specific vaccination strategies against cancer," Current Opin. Immunol., 2000, 2:576-582; Rosenberg SA, "A new era for cancer immunotherapy based on the genes that encode cancer antigens," Immunity, 1999, 10:281 -7; Sahin U et al., "Serological identification of human tumor antigens," Curr. Opin. Immunol., 1997, 9:709-16; Old LJ et al.,

"New paths in human cancer serology," J. Exp. Med., 1998, 187:1163-7; Chaux P, et al., "Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes," J. Exp. Med., 1999, 189:767-78; Gold P, et al., "Specific carcinoembryonic antigens of the human digestive system," J. Exp. Med., 1965, 122:467-8; Livingston PO, et al., Carbohydrate vaccines that induce antibodies against cancer: Rationale," Cancer Immunol. Immunother., 1997, 45:1-6; Livingston PO, et al., Carbohydrate vaccines that induce antibodies against cancer: Previous experience and future plans," Cancer Immunol. Immunother., 1997, 45:10-9; Taylor-Papadimitriou J, "Biology, biochemistry and immunology of carcinoma-associated mucins," Immunol. Today, 1997, 18:105-7; Zhao X-J et al., "GD2 oligosaccharide: target for cytotoxic T lymphocytes," J. Exp. Med., 1995, 182:67-74; Theobald M, et al., "Targeting p53 as a general tumor antigen," Proc. Natl. Acad. Sci. USA, 1995, 92:11993-7; Gaudernack G, "T cell responses against mutant ras: a basis for novel cancer vaccines," Immunotechnology, 1996, 2:3-9; WO 91/02062; U.S. Patent No. 6,015,567; WO 01/08636; WO 96/30514; U.S. Patent No. 5,846,538; and U.S. Patent No. 5,869,445.

Pediatric/Geriatric Antigens

In one embodiment the compositions of the present invention are used in conjunction with an antigen for treatment of a pediatric population, as in a pediatric antigen. In a more particular embodiment the pediatric population is less than about 3 years old, or less than about 2 years, or less than about 1 years old. In another embodiment the pediatric antigen (in conjunction with the composition of the present invention) is administered multiple times over at least 1 , 2, or 3 years.

In another embodiment the compositions of the present invention are used in conjunction with an antigen for treatment of a geriatric population, as in a geriatric antigen.

Other Antigens

Other antigens for use in conjunction with the compositions of the present include hospital acquired (nosocomial) associated antigens.

In another embodiment, parasitic antigens are contemplated in conjunction with the compositions of the present invention. Examples of parasitic antigens include those derived from organisms causing malaria and/or Lyme disease.

In another embodiment, the antigens in conjunction with the compositions of the present invention are associated with . or effective against a mosquito born illness. In another embodiment, the antigens in conjunction with the compositions of the present invention are associated with or effective against encephalitis. In another embodiment the antigens in conjunction with the compositions of the present invention are associated with or effective against an infection of the nervous system.

In another embodiment, the antigens in conjunction with the compositions of the present invention are antigens transmissible through blood or body fluids.

Antigen Formulations

In other aspects of the invention, methods of producing microparticles having adsorbed antigens are provided. The methods comprise: (a) providing an emulsion by dispersing a mixture comprising (i) water, (ii) a detergent, (iii) an organic solvent, and (iv) a biodegradable polymer selected from the group consisting of a poly(α-hydroxy acid), a polyhydroxy butyric acid, a polycaprolactone, a polyorthoester, a polyanhydride, and a polycyanoacrylate. The polymer is typically present in the mixture at a concentration of about 1% to about 30% relative to the organic solvent, while the detergent is typically present in the mixture at a weight-to- weight detergent-to-polymer ratio of from about 0.00001 :1 to about 0.1 :1 (more typically about 0.0001 :1 to about 0.1 :1 , about 0.001 :1 to about 0.1 :1, or about 0.005:1 to about 0.1 :1); (b) removing the organic solvent from the emulsion; and (c) adsorbing an antigen on the surface of the microparticles. In certain embodiments, the biodegradable polymer is present at a concentration of about 3% to about 10% relative to the organic solvent.

Microparticles for use herein will be formed from materials that are sterilizable, non-toxic and biodegradable. Such materials include, without limitation, poly(α-hydroxy acid), polyhydroxybutyric acid, polycaprolactone, polyorthoester, polyanhydride, PACA, and polycyanoacrylate. Preferably, microparticles for use with the present invention are derived from a poly(α-hydroxy acid), in particular, from a poly(lactide) ("PLA") or a copolymer of D 1 L- lactide and glycolide or glycolic acid, such as a poly(D,L-Iactide-co-glycolide) ("PLG" or "PLGA"), or a copolymer of D,L-lactide and caprolactone. The microparticles may be derived from any of various polymeric starting materials which have a variety of molecular weights and, in the case of the copolymers such as PLG, a variety of lactide:glycolide ratios, the selection of

which will be largely a matter of choice, depending in part on the coadministered macromolecule. These parameters are discussed more fully below.

Further antigens may also include an outer membrane vesicle (OMV) preparation.

Additional formulation methods and antigens (especially tumor antigens) are provided in U.S. Patent Serial No. 09/581 ,772.

Antigen References The following references include antigens useful in conjunction with the compositions of the present invention:

A. International patent application WO99/24578

B. International patent application WO99/36544. C. International patent application WO99/57280.

D. International patent application WO00/22430.

E. Tettelin et al. (2000) Science 287:1809-1815.

F. International patent application WO96/29412.

G. Pizza et al. (2000) Science 287:1816-1820. H. PCT WO 01/52885.

I. Bjune et al. (1991) Lancet 338(8775). J. Fuskasawa et al. (1999) Vaccine 17:2951-2958. K. Rosenqist et al. (1998) Dev. Biol. Strand 92:323-333. Constantino et al. (1992) Vaccine 10:691-698. Constantino et al. (1999) Vaccine 17:1251-1263.

Watson (2000) Pediatr Infect Dis J 19:331-332. Rubin (20000) Pediatr Clin North Am 47:269-285,v.

If" IUir H ,' 'I...I' '»iil' 'W II."!' i' 1 ,|l' f '" 1 II" ICiIIi Il

Jedrzejas (2001 ) Microbiol MoI Biol Rev 65:187-207.

International patent application filed on 3 rd July 2001 claiming priority from GB- 0016363.4;WO 02/02606; PCT IB/01 /00166.

Kalman et al. (1999) Nature Genetics 21 :385-389. 5 Read et al. (2000) Nucleic Acids Res 28:1397-406.

Shirai et al. (2000) J. Infect. Dis 181(Suppl 3):S524-S527.

International patent application WO99/27105.

International patent application WO00/27994.

International patent application WO00/37494. 10 International patent application WO99/28475.

Beil (2000) Pediatr Infect Dis J 19:1187-1188.

Iwarson (1995) APMIS 103:321-326.

Gerlich et al. (1990) Vaccine 8 Suppl:S63-68 & 79-80.

Hsu et al. (1999) Clin Liver Dis 3:901-915. 15 Gastofsson et al. (1996) N. Engl. J. Med. 334-:349-355.

Rappuoli et al. (1991 ) TIBTECH 9:232-238.

Vaccines (1988) eds. PIotkin & Mortimer. ISBN 0-7216-1946-0.

Del Guidice et al. (1998) Molecular Aspects of Medicine 19:1-70.

International patent application WO93/018150. 20 International patent application WO99/53310.

International patent application WO98/04702.

Ross et al. (2001) Vaccine 19:135-142.

Sutter et al. (2000) Pediatr Clin North Am 47:287-308.

Zimmerman & Spann (1999) Am Fan Physician 59:113-118, 125-126. 25 Dreensen (1997) Vaccine 15 SuppI"S2-6.

MMWR Morb Mortal WkIy rep 1998 Jan 16:47(1 ):12, 9.

McMichael (2000) Vaccine19 Suppl 1 :S101-107.

Schuchat (1999) Lancer 353(9146) :51 -6.

GB patent applications 0026333.5, 0028727.6 & 0105640.7.

Dale (1999) Infect Disclin North Am 13:227-43, viii.

Ferretti et al. (2001) PNAS USA 98: 4658-4663. Kuroda et al. (2001 ) Lancet 357(9264):1225-1240; see also pages 1218-1219.

Ramsay et al. (2001) Lancet 357(9251 ):195-196.

Lindberg (1999) Vaccine 17 Suppl 2:S28-36.

Buttery & Moxon (2000) J R Coil Physicians Long 34:163-168.

Ahmad & Chapnick (1999) Infect Dis Clin North Am 13:113-133, vii. Goldblatt (1998) J. Med. Microbiol. 47:663-567.

European patent 0 477 508.

U.S. Patent No. 5,306,492.

International patent application WO98/42721.

Conjugate Vaccines (eds. Cruse et al.) ISBN 3805549326, particularly vol. 10:48-114. Hermanson (1996) Bioconjugate Techniques ISBN: 012323368 & 012342335X.

European patent application 0372501.

European patent application 0378881.

European patent application 0427347.

International patent application WO93/17712. International patent application WO98/58668.

European patent application 0471177.

International patent application WO00/56360.

International patent application WO00/67161.

Compositions of the invention may be formulated for administration by mucosal or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway

(aerosol), oral, intranasal, rectal, vaginal and topical (including buccal and sublingual) administration. Thus the compositions may be prepared in injectable form.

Methods of treatment

The invention provides a method of raising an immune response in a patient, comprising administering a patient with a composition of the invention. The composition may be administered intravenously, intramuscularly, intraperitoneally, subcutaneously, transdermally, orally, intranasally, rectally, vaginally or topically. The immune response is preferably protective.

The invention also provides the use of a polypeptide or polymer of the invention in the manufacture of a medicament for immunising a patient. The invention also provides a polypeptide or polymer of the invention for use in medicine.

Medicaments may be administered by mucosal or parenteral routes, including intravenous, intramuscular, intraperitoneal, subcutaneous, transdermal, airway (aerosol), oral, intranasal, rectal, vaginal and topical (including buccal and sublingual) administration. Intramuscular administration to the thigh or the upper arm is preferred. Injection may be via a needle (e.g. a hypodermic needle), but needle-free injection may alternatively be used.

Administration may be a single dose schedule or a multiple dose schedule. A primary dose schedule may be followed by a booster dose schedule. Suitable timing between priming and boosting can be routinely determined.

Administration will generally be to an animal and, in particular, human subjects can be treated. The compositions are particularly useful for vaccinating children and teenagers.

Certain bacterial proteins (e.g., flagellin, fimbriae, HSP) are known, or thought, to signal to the innate immune system through toll-like receptors (TLRs) [37,38], and this may be mediated through linear peptide sequences (e.g., ALTTE in fimbriae). Thus, another aspect of the present invention is the use of a polypeptide of the invention as an agonist or antagonist of a toll-like receptor or a related receptor of the innate immune system. The term "agonist" refers to a substance that has affinity for and stimulates physiological activity at a cell receptor normally

stimulated by naturally occurring substances, thus triggering a biochemical response. Assays for TLR agonism are known [e.g. 39]. One indicator of activity of a TLR agonist is the production of cytokines from one or more cells of the immune system, such as antigen presenting cells, lymphocytes or macrophages. The term "antagonist" refers to a substance that nullifies the action of another, such as a drug binding to a cell receptor without eliciting a biological response. Antagonists are sometimes referred to as inhibitors. Assays for TLR agonism are known [e.g. 40].

Definitions

The term "comprising" can mean "including" as well as "consisting" e.g. a composition "comprising" X may consist exclusively of X or may include something additional e.g. X + Y.

As well as being adjuvants, it would be understood by a skilled person that the polypeptides of the present invention may also be useful in immune-based therapies for cancer or infectious diseases, and immunomodulatory agents for autoimmune diseases such as allergies and asthma.

References to a percentage sequence identity between two amino acid sequences means that, when aligned, that percentage of amino acids are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of reference 41. A preferred alignment is determined by the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, BLOSUM matrix of 62. The Smith-Waterman homology search algorithm is taught in reference 42.

References to a percentage sequence identity between two nucleic acid sequences mean that, when aligned, that percentage of bases are the same in comparing the two sequences. This alignment and the percent homology or sequence identity can be determined using software programs known in the art, for example those described in section 7.7.18 of reference 41. A preferred alignment program is GCG Gap (Genetics Computer Group, Wisconsin, Suite Version 10.1), preferably using default parameters, which are as follows: open gap = 3; extend gap = 1.

MODES FOR CARRYING OUT THE INVENTION

1. Generation of protein families

A database consisting of 60 complete bacterial genomes (see Table 1 below) was found to comprise 148251 open reading frames (ORFs). The polypeptides encoded by these 148251 ORFs were grouped together into protein families according to their sequence identity. The BLAST program from the GCG program suite was used, using the BLOSUM62 substitution matrix and default parameters to identify all pairs of proteins sharing an alignment with an E- score smaller than 1.0E "05 . The BLAST E-score is the probability that an alignment of better quality, as measured by the alignment score, is found by chance in the same data set. The protein dataset was then partitioned into non-overlapping families, which include only proteins that share an alignment with an E-score smaller than 1.0E "05 with all other members of the same family.

2. Selecting a protein family that comprises a PAMP

In order to identify only those protein families containing PAMPs, which by definition are motifs that are exclusive to pathogens (and not present in the host human), only those protein families that did not contain proteins from the Drosophila genome were chosen. This means that at least one of the proteins in the family had no BLAST alignment with any protein in the Drosophila genome with an E-score smaller than the threshold of 1.0E "05 . Although the Drosophila is not the host organism for the present example, a pathogenic protein family sequence that shares a high identity with a Drosophila (i.e. non-pathogenic) protein sequence has an increased probability of sharing a high identity with a human sequence.

3. Predicting the cellular localisation of the protein families

Protein families that encode a secreted protein or surface protein of a pathogen are preferred. Only those protein families that contained at least one amino acid sequence which was either already annotated as encoding a surface related protein or was predicted as being a protein that is localised on the cell surface by PSORT were kept. 251 protein families were identified at this stage.

4. Identifying a conserved polypeptide sequence from within the protein family

Of the families that passed the previous selections, a list of conserved polypeptide sequences from within the protein family sequences were identified. The list was compiled with the aid of the PRATT algorithm. The following default parameters were used, i.e. i) pattern conserved in

100% of the sequences, ii) max length :50, iii) max number of consecutive x's is 5, iv) max number of flexible spacers is 2, v) max flexibility is 2 and vi) mac flexible product is 10. The patterns were ranked using PRATT as specified in [8]. For each protein family, only the top 10 scoring patterns were retained. PRATT analysis of the 251 families yielded a list of over 2500 polypeptide sequences.

5. Excluding pathogenic polypeptide sequences that are also present in the human genome

Using the list of over 2500 polypeptide sequences in the EMBOSS [43] software suite, a pattern search was carried out on the published human genome sequence. EMBOSS was able to test whether any of the over 2500 polypeptide sequences shared a high identity with any human polypeptide sequences. Using fuzzpro, which is part of the EMBOSS suite, a search was run for each pattern against the complete set of human proteins. Those patterns having at least one hit in the human genome were discarded. This last selection step yielded a list of 312 polypeptide sequences, which are listed in Table 3.

6. Confirmation of adjuvant activity

To evaluate adjuvant activity of the putative peptide PAMPs listed in Table 2, a set of peptides relating to the bacterial signature sequence PDCG-[LM]-[KR] were synthesised, purified and demonstrated to be free of endotoxin. The possible combinations of sequences included PDCGLR, PDCGLK, PDCGMR, and PDCGMK. The human monocytic cell line THP-1 was incubated with each of the peptides and adjuvant activity was observed for the peptide PDCGLR, as measured by the specific production of cytokines (IL1-β, IL-8 and TNFα). This peptide was further evaluated on primary human peripheral blood mononuclear cells (hPBMC), where cytokine production (IL-6 and TNFα) was also demonstrated. Therefore, the specific peptide sequence PDCGLR, which was identified as a bacterial signature found commonly in bacterial proteins but not in Drosophila, is recognised by the human immune system as a PAMP.

For the assay, the human cells used were cultured at 1 million cells/ml in 96-well plates with the peptides in complete RPMI medium with 5% FBS. After culture for 18 hours at 37°C and 5% CO 2 , the culture supernatants were measured for the cytokines generated by the cells with an Upstate multiplex cytokine kit. The peptide solutions made in PBS buffer were found to have

<0.01 U/ml endotoxin, and this level of endotoxin can not stimulate the immune cells used to secrete detectable cytokines.

It will be understood that the invention has been described by way of example only and modifications may be made whilst remaining within the scope and spirit of the invention.

TABLE 1

TABLE 2: Cytokine production (pg/ml) by human cells after peptide stimulation

Note: ND, not determined.

TABLE 3

312 pathogenic adjuvant polypeptide sequences identified using the method of the invention

G-[FY]-x(2)-R-x(2)-[ST]-x(2)-G-[QR]-x-[ILV]-[ILV]-x(2)-R- R-x-[HKR]-[GNQ]-R-x(2)-L (SEQ ID NO: 10)

H-E-x-[AG]-H-x(3)-[AG]-x(4)-[l MV]-x(4)-[FY]-x-[l LV]-G-[FI M]-G-x(2)-[FIL] (SEQ ID NO: 11) H-E-x(2)-H-x(3)-[AG]-x(4)-[IMV]-x(4)-[FY]-x-[ILV]-G-[FIM]-G- x(2)-[FIL] (SEQ ID NO: 12)

G-[ILMV]-X-L-X-G-X-E-[IV]-X-[AS]-[ILMV] (SEQ ID NO: 13) D-[DEGNQS]-X-[DEG]-X-D-X-K-[ILV]-[ILV]-[ACG]-[LV]-X(S)-[DHK] (SEQ ID NO: 14)

G-G-x-S-x-E-[HR]-x-[IV]-S-x(2)-[ST]-[AGS] (SEQ ID NO: 15) G-x-S-x-E-[HR]-x-[IV]-S-x(2)-[Sη-[AGS] (SEQ ID NO: 16) H-G-x(2)-G-x(0,2)-E-[DT]-G-x-[ILMV]-x-[AGS] (SEQ ID NO: 17) V-x(3)-[ASV]-x-K-x(2)-[AILV]-x(2)-A-[IMV]-x(2)-[AILV]-F-x(1, 2)-V-x(1,2)-V (SEQ ID NO: 18) K-x(2)-[AILV]-x(2)-A-[IMV]-x(2)-[AILV]-F-x(1,2)-V-x(1,2)-V(S EQ ID NO: 19) A-x(3)-G-x(2)-E-x-[DNP]-x(3)-D-[ILV]-x-[AG]-x-D-x(3)-K-x(2)- [ILV] (SEQ ID NO: 20) A-x(3)-G-x(2)-E-x(5)-D-[ILV]-x-[AG]-x-D-x(3)-K-x(2)-[ILV] (SEQ ID NO: 21) G-x(2)-E-x-[DNP]-x(3)-D-[ILV]-x-[AG]-x-D-x(3)-K-x(2)-[ILV] (SEQ ID NO: 22) G-x(2)-E-x(5)-D-[ILV]-x-[AG]-x-D-x(3)-K-x(2)-[ILV] (SEQ ID NO: 23) E-x-[DNP]-x(3)-D-[ILV]-x-[AG]-x-D-x(3)-K-x(2)-[ILV] (SEQ ID NO: 24) Q-[FILM]-[IV]-x-H-x(2)-[FIV]-x-[ILV]-[ADGN]-[DGN]-x(2)-[AILV ]-x(3)-[GS] (SEQ ID NO: 25) D-x-[AIV]-[FILV]-[AS]-G-[IV]-N-x(1,2)-G-x(0,1)-N (SEQ ID NO: 26) N-D-D-G-x(2)-[AS]-x-[GS]-[ILMV]-x(2)-[AL] (SEQ ID NO: 27) D-[FILMV]-X-[FILV]-S-G-X(O 1 I)-N-X-[AGT]-X-[NT] (SEQ ID NO: 28) K-x-[AS]-R-x-[ILMV]-[AGS]-L-[ILMV]-P-[FY] (SEQ ID NO: 29) G-W-x(5)-G-R-x-P-[WY] (SEQ ID NO: 30) H-[AGS]-x(2)-H-[ILM]-x-G-x-[DK]-[DH] (SEQ ID NO: 31) G-x-G-N-x(0,1 )-Y-x(2,3)-E-x(2)-[FHW]-x(3)-[FILV]-x-[GP] (SEQ ID NO: 32) G-N-x(0,1)-Y-x(2,3)-E-x(2)-[FHW]-x(3)-[FILV]-x-[GP] (SEQ ID NO: 33) R-x(2)-L-N-x-G-H-[ST]-[FILV]-[AG]-x-[AGLV]-[ILV]-E (SEQ ID NO: 34)

R-x(1,2)-L-x(0,1)-N-x-G-H-[ST]-[FILV]-[AG]-x-[AGLV]-[ILV] -E (SEQ ID NO: 35)

G-G-G-[ATV]-x(2)-D-x-[AGTV]-G-x(1,2)-A-x(4,5)-R-G (SEQ ID NO: 36) L-N-x-G-H-[ST]-[FILV]-[AG]-x-[AGLV]-tlLV]-E (SEQ ID NO: 37) L-x(0,1)-N-x-G-H-[ST]-[FILV]-[AG]-x-[AGLV]-[ILV]-E (SEQ ID NO: 38) D-[AST]-[AS]-[AIV]-G-G-K-[NTV]-[AG]-[ILV] (SEQ ID NO: 39) R-x(2,3)-L-x(2,3)-G-H-[Sη-[FILV]-[AG]-x-[AGLV]-[ILV]-E (SEQ ID NO: 40) G-G-[ATV]-x(2)-D-x-[AGTV]-G-x(1,2)-A-x(4,5)-R-G (SEQ ID NO: 41) G-[ATV]-x(2)-D-x-[AGTV]-G-x(1,2)-A-x(4,5)-R-G (SEQ ID NO: 42) D-E-P-X-[AG]-[AENS]-L-D (SEQ ID NO: 43)

G-S-S-R-E-x-A-[APV]-x(2)-[ILP]-x(4)-[FIV]-x(2)-[ILMV]-[IL V]-[AGS]-x(2)-[FY]-[AGS]-x-l-[FHY]-x(2)-N (SEQ ID NO: 44) S-S-R-E-x-A-[APV]-x(2)-[ILP]-x(4)-[FIV]-x(2)-[ILMV]-[ILV]-[A GS]-x(2)-[FY]-[AGS]-x-l-[FHη-x(2)-N (SEQ ID NO: 45) S-R-E-x-A-[APV]-x(2)-[ILP]-x(4)-[FIV]-x(2)-[ILMV]-[ILV]-[AGS ]-x(2)-[Fη-[AGS]-x-l-[FHY]-x(2)-N (SEQ ID NO: 46) R-E-x-A-[APV]-x(2)-[ILP]-x(4)-[FIV]-x(2)-[ILMV]-[ILV]-[AGS]- x(2)-[FY]-[AGS]-x-l-[FHY]-x(2)-N (SEQ ID NO: 47)

S-R-x(3,5)-W-x-K-G-[ADES]-x-S-G-x(4)-[ILV]-x(2HFILMV]-x(2 )-D-C-D-x-D-[ATV]-[ILMV]-x(3)-[AIV]-x(3)-G-x(2)-[ACG] (SEQ ID NO: 48)

S-x(2,4)-R-x(13)-W-x-K-G-[ADES]-x-S-G-x(4)-[ILV]-x(2)-[FI LMV]-x(2)-D-C-D-x-D-[ATV]-[ILMV]-x(3)-[AIV]-x(3)-G-x(2)- [ACG] (SEQ ID NO: 49)

R-x(3,5)-W-x-K-G-[ADES]-x-S-G-x(4)-[ILV]-x(2)-[FILMV]-x(2 )-D-C-D-x-D-[ATV]-[ILMV]-x(3)-[AIV]-x(3)-G-x(2HACG] (SEQ ID NO: 50)

W-x-K-G-[ADES]-x-S-G-x(4)-[ILV]-x(2)-[FILMV]-x(2)-D-C-D-x -D-[ATV]-[ILMV]-x(3)-[AIV]-x(3)-G-x(2)-[ACG] (SEQ ID NO: 51)

K-G-[ADES]-x-S-G-x(4)-[ILV]-x(2)-[FILMV]-x(2)-D-C-D-x-D-[ ATV]-[ILMV]-x(3)-[AIV]-x(3)-G-x(2)-[ACG] (SEQ ID NO: 52) K-G-x(2)-S-G-x(4)-[ILV]-x(2)-[FILMV]-x(2)-D-C-D-x-D-[ATV]-[I LMV]-x(3)-[AIV]-x(3)-G-x(2)-[ACG] (SEQ ID NO: 53) G-x(2,4)-C-H-x-G-x(2)-[AST]-C-[FW] (SEQ ID NO: 54) C-H-X-G-X(^-[AST]-C-[FW] (SEQ ID NO: 55) l-x(3,5)-D-x-V-x-[ILV]-[AE]-[FILM]-[ST]-[APη-[HY]-[DS]-χ(3 )-[AG]-R-[IV]-x(2)-R (SEQ ID NO: 56) D-x-V-x(2)-E-x(2)-[APT]-x-[DST]-x(3)-[AGV]-[QR]-[ILV3 (SEQ ID NO: 57)

Y-[IV]-G-K-[AS]-x(2)-[IL]-x(2)-R-x(3)-[HY] (SEQ ID NO: 58)

P-[FL]-[ADEG]-x-G-x(2,3)-T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(4)-[C GNS]-x(2)-[AGS]-x-[ASV] (SEQ ID NO: 59) P-x(3)-G-x-[AGV]-x-T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(1,2)-L-x-[A CGST] (SEQ ID NO: 60) P-x(2,3)-G-x-[AGV]-x-T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(1 l 2)-L-x-[ACGSη (SEQ ID NO: 61)

P-x(3)-G-x(3)-T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(1,2)-L-x-[ACG Sη (SEQ ID NO: 62) P-x(2,3)-G-x(3)-T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(1,2)-L-x-[ACGS η (SEQ ID NO: 63) G-x-[AGV]-x-T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(1,2)-L-x-[ACGSη (SEQ ID NO: 64) T-[ILV]-[AG]-x(2)-[ILM]-R-R-x(1,2)-L-x-[ACGSη (SEQ ID NO: 65)

P-x(1,2)-L-x(2,3)-G-[ACGSTV]-x-G-l-[AS]-[ASV]-G-x-[AST]-[ Aη-x-[IMV]-x-[PSη-H-x(3HDE]-x(3)-[AGS] (SEQ ID NO: 66)

P-x-[ILV]-[FIL]-x(2)-G-x(1 ,2)-G-x(0,1)-l-[AS]-[ASV]-G-x-[ASη-[AT]-x-[IMV]-x-[PSη-H-x (3)-[DE]-x(3)-[AGS] (SEQ ID NO: 67)

L-x(2,3)-G-[ACGSTV]-x-G-l-[AS]-[ASV]-G-x-[ASη-[AT]-x-[IM V]-x-[PSη-H-x(3)-[DE]-x(3)-[AGS] (SEQ ID NO: 68) L-x(1 ,3)-G-[ACGSTV]-x-G-l-[AS]-[ASV]-G-x-[ASη-[AT]-x-tlMV]-x-[PS η-H-x(3)-[DE]-x(3)-[AGS] (SEQ ID NO: 69) G-x(2)-G-l-[AS]-[ASV]-G-x-[ASη-[AT]-x-[IMV]-x-[PSη-H-x(3)- [DE]-x(3),[AGS] (SEQ ID NO: 70) D-G-X-K-P-[SV]-X-R-R-[AILV]-[ILMV]-[FHWY]-[AGST] (SEQ ID NO: 71) G-x-K-P-[SV]-x-R-R-[AILV]-[ILMV]-[FHWY]-[AGSη (SEQ ID NO: 72) A-x-[AIPV]-x(3)-D-x(0,1)-G-x(2)-[LP]-[SV]-x-R-x-[AILV]-x-[FH WY]-[AGSη (SEQ ID NO: 73) K-P-[SV]-x-R-R-[AILV]-[ILMV]-[FHWY]-[AGSη (SEQ ID NO: 74) K-P-x(2)-R-R-[AILV]-[ILMV]-[FHWYHAGST] (SEQ ID NO: 75) R-x(3)-R-x(5)-G-x-[ILV]-P-G-x(2)-K-[AS]-S-W (SEQ ID NO: 76) R-x(2)-[FILV]-R-x(5)-G-x-[ILV]-P-G-x(1,2)-K-x(1,2)-S-W(SEQ ID NO: 77) R-x(5)-G-x-[ILV]-P-G-x(1,2)-K-x(1,2)-S-W(SEQ ID NO: 78) G-x(2)-P-G-x(2)-K-[AS]-S-W(SEQ ID NO: 79) G-x-[ILV]-P-G-x(1 ,2)-K-x(1,2)-S-W (SEQ ID NO: 80)

G-x(4)-F-x-R-x-[AGNQST]-x-C-x(3)-[CS]-x(3)-[ADNQ] (SEQ ID NO: 81) G-x(3,4)-F-x-R-x-[AGNQSη-x-C-x(3)-[CS]-x(3)-[ADNQ] (SEQ ID NO: 82)

F-N-[FILV]-A-D-x(2)-[ILV]-x(2)-[CG] (SEQ ID NO: 83) F-N-x-A-D-x(2)-[ILV]-x(2)-[CG] (SEQ ID NO: 84) G-x-[PS]-x-[ILV]-[ACNS]-D-P-G-x(2)-[ILM]-[AIV] (SEQ ID NO: 85) G-[ASη-[ANPSη-[APS]-G-Y-[IV]-G-x(3)-[AGNS]-[GNSη-x(3)-[DE NQT] (SEQ ID NO: 86) E-[FL]-[DPSη-[DE]-x-Q-G-x(2)-G-x(0,2)-Y-x(4)-[ADEGN]-x(4)-[ AIV]-[ACSη-x(4)-[DEGQ]-x-[Hη (SEQ ID NO: 87) P-x(3,4)-D-P-[FY]-[AG]-L-x-[RS]-x-[ASTV]-x-[AEGT]-[AILV] (SEQ ID NO: 88) Q-G-x(2)-G-x(0,2)-Y-x(4)-[ADEGN]-x(4)-[AIV]-[ACSη-x(4)-[DEG Q]-x-[Hη (SEQ ID NO: 89) T-x-[ANPSη-H-x-[FILMV]-x(3)-[FILMV]-[ILMV]-x-E-[AQS]-[ILV]- [FY]-R-[AG]-x(2)-[ILV] (SEQ ID NO: 90) L-[DE]-[IV]-[AST]-S-x-G-x(2)-R-x-[ILM] (SEQ ID NO: 91)

R-x(0,2)-L-x(0,2)-D-x(2)-[ASη-x-G-[ALV]-[ILMV]-[FILV]-[F ILMV]-[Sη-[DNQη (SEQ ID NO: 92) L-D-x(2)-[ASη-x-G-x(0,1)-L-[ILMV]-[FILV]-x-[DNQST]-[DNQSη (SEQ ID NO: 93) M-[KR]-[IV]-[NQR]-x-S-V-K-x(2)-C-x(2)-C-[KR]-x-[IV]-[KR]-R (SEQ ID NO: 94) S-V-K-x(2)-C-x(2)-C-[KR]-x-[IV]-[KR]-R (SEQ ID NO: 95) V-K-x(2)-C-x(2)-C-[KR]-x-[IV]-[KR]-R (SEQ ID NO: 96) K-x(2)-C-x(2)-C-[KR]-x-[IV]-[KR]-R (SEQ ID NO: 97) l-[GV]-x(2)-[EG]-x-N-x(2)-L-x-[ANST]-x-[ILV]-x-[DEGNQS]-x(3) -[DENT] (SEQ ID NO: 98) Q-F-H-[Pη-E-[KR]-S-x(3)-G-x(2)-[FILMV] (SEQ ID NO: 99) G-[IV]-C-[LV]-G-x-Q-x-[FLM]-x(3)-[GS]-x-E (SEQ ID NO: 100) Q-F-H-[PT]-E-[KR]-S-x(2,3)-G-x(2)-[FILMV] (SEQ ID NO: 101) F-H-[Pη-E-[KR]-S-x(3)-G-x(2)-[FILMV] (SEQ ID NO: 102) F-H-x-E-[KR]-S-x(3)-G-x(2)-[FILMV] (SEQ ID NO: 103) C-[LV]-G-x-Q-x-[FLM]-x(3)-[GS]-x-E (SEQ ID NO: 104)

H-[PT]-E-[KR]-S-x(3)-G-x(2)-[FILMV] (SEQ ID NO: 105)

H-P-x-W-x-M-G-x(3)-[ST]-[ILV]-[DN]-S-[AS]-[ST]-[LM]-x-N-K -[AGL]-[FL]-E-x-[ILM]-E-[ACT]-x(2)-[FL]-[FY] (SEQ ID NO: 106)

P-x-W-x-M-G-x(3)-[Sη-[ILV]-[DN]-S-[AS]-[Sη-[LM]-x-N-K-[ AGL]-[FL]-E-x-[ILM]-E-[ACT]-x(2)-[FL]-[FY] (SEQ ID NO: 107)

S-[AS]-[ST]-[LM]-x-N-K-[AGL]-[FL]-E-x-[ILM]-E-[ACT]-x(2)- [FL]-[FY] (SEQ ID NO: 108) N-K-[AGL]-[FL]-E-x-[ILM]-E-[ACη-x(2)-[FL]-[FY] (SEQ ID NO: 109)

P-[AIV]-D-S-E-H-x-[AG]-[ILV]-x(3)-[ILM] (SEQ ID NO: 110) D-S-E-H-x-[AG]-[!LV]-x(3)-[ILM] (SEQ ID NO: 111)

G-x(3)-E-x-A-x(2)-E-x(3,5)-E-x(3)-[ILV]-x-[DNR]-x-[FILMV] -x(4)-[FILMV] (SEQ ID NO: 112) E-[CGTV]-x(2)-D-L-x(1,3)-G-x(4,5)-L-x-G (SEQ ID NO: 113) E-x-A-x(2)-E-x(3,5)-E-x(3)-[ILV]-x-[DNR]-x-[FILMV]-x(4)-[FIL MV] (SEQ ID NO: 114) F-x(2,4)-E-x(3,5)-D-L-[FIM]-x-E-x-[AGTV] (SEQ ID NO: 115)

L-x(2,4)-L-x(1,3)-R-x(3)-[ESη-[IL]-S-[GP]-G-E-x(2)-R-[AI LV]-x-[ILM]-x(3)-[ILV] (SEQ ID NO: 116) V-x(2,3)-G-[ILPV]-[AS]-G-[AS]-G-K-[ST]-[STV]-L-[AIL] (SEQ ID NO: 117) R-x(0,1 )-S-[ILM]-N-[ILV]-[AS]-x-[ACST]-[AV]-[ACGSV]-x(4)€-x(2)-[K R]-Q (SEQ ID NO: 118) L-x(2,3)-P-x(4)-N-[TV]-G-[ANST]-x(2)-R-x-[ACη (SEQ ID NO: 119) P-x(4)-N-[TV]-G-[ANST]-x(2)-R-x-[ACT] (SEQ ID NO: 120) D-[AP]-G-H-G-[DG]-X-[DEN]-X-G (SEQ ID NO: 121) L-x(2)-W-A-x-[EQR]-G-x(1,2)-L-x(1,2)-N-x(3)-[ST]-[TV] (SEQ ID NO: 122) A-x-[ENQR]-G-[FIMV]-L-[GL]-x-N-x(3)-[ST]-[ATV] (SEQ ID NO: 123) W-A-x-[EQR]-G-x(1 ,2)-L-x(1,2)-N-x(3)-[ST]-[TV] (SEQ ID NO: 124) l-x(1,2)-G-Q-D-P-Y-x-[GNQST] (SEQ ID NO: 125) L-x(1 ,3)-A-x-[EQR]-G-x(1,2)-L-[GIL]-x-[Nη-x(3)-[STV] (SEQ ID NO: 126) W-A-x(2)-G-x(1,2)-L-x(1,2)-N-x(3)-[Sη-[TV] (SEQ ID NO: 127) G-Q-D-P-Y-x-[GNQSη (SEQ ID NO: 128)

E-x(4)-R-x-Q-x-[FY]-x-G-x-[CV]-[ILM]-x(3)-[GN]-x-G (SEQ ID NO: 129)

V-X(O 1 I )-R-x(4)-Y-x(1,2)-R-x(3)-G-K (SEQ ID NO: 130)

R-x(4,5)-Y-[ILM]-R-x(3)-G-K (SEQ ID NO: 131)

T-G-x-E-M-E-A-[ILV]-χ4ACGS]-[AW]-[NQSη-x-[ACGST]-x(2)-[ ANTV]-[ILV]-[WY]-D-M-x-K-x(2)-[DEQS]-x-[ADEGS]- x(2)-[GIV] (SEQ ID NO: 132)

G-x-E-M-E-A-[ILV]-x-[ACGS]-[ATV]-[NQST]-x-[ACGST]-x(2)-[A NTV]-[ILV]-[WY]O-M-x-K-x(2)-[DEQS]-x-[ADEGS]-x( [GIV] (SEQ ID NO: 133)

E-M-E-A-[IL\^-x-[ACGS]-[ATV]-[NQSη-x-[ACGSη-x(2)-[ANW]- [IL^-[M]-D-M-x-K-x(2)-[DEQS]-x-[ADEGS]-x(2H^ (SEQ ID NO: 134)

G-x(1,2)-M-E-A-[IL\/]-x-[ACGS]-[ATV]-[NQST]-x-[ACGSη-x(2 )-[ANT\^-[IL^-[M]O-M-x-K-x(2)-[DEQS]-x-[ADEGS]- x(2)-[GIV] (SEQ ID NO: 135)

M-E-A-[ILV]-x-[ACGS]4ATV]-[NQSη-x-[ACGST]-x(2)-[ANTV]4IL V]-[WY]O-M-x-K-x(2)-[DEQS]-x-[ADEGS]-x(2)-[GIV] (SEQ ID NO: 136)

L-x(3,4)-D-M-x-K-x(2)-[DEQS]-x-[ADEGS]-x(2)-[GIV] (SEQ ID NO: 137) G-x(1 ,3)-Y-x(1,3)-G-x-[IV]-x(2)-[FLV]-A-[DEη-x(2)-R-x(2)-[GI] (SEQ ID NO: 138) Y-x(1, 3)-G-x-[IV]-x(2)-[FLV]-A-[DEη-x(2)-R-x(2)-[Gl] (SEQ ID NO: 139) G-x-[IV]-x(2)-[FLV]-A-[DET]-x(2)-R-x(2)-[GI] (SEQ ID NO: 140) C-x-[FHY]-x-[LP]-[ST]-C-S-x-Y-x(4)-[FILV]-x(3)-[GNPS] (SEQ ID NO: 141) P-x(0,2)-G-G-[CD]-x(2)-G-x-R-x(4)-[FH]-x(3)-[FILM]-x(3)-G (SEQ ID NO: 142) G-G-[CD]-x(2)-G-x-R-x(4)-[FH]-x(3)-[FILM]-x(3)-G (SEQ ID NO: 143) P-x(0,2)-G-G-x(3)-G-x-R-x(4)-[FH]-x(3)-[FILM]-x(3)-G (SEQ ID NO: 144) G-G-x(3)-G-x-R-x(4)-[FH]-x(3)-[FILM]-x(3)-G (SEQ ID NO: 145) l-[AGV]-x-A-G-x-[AES]-[AG]-x-L-x(4,5)-A-x(3,5)-P-V-[FI L]-[AG]-V-P (SEQ ID NO: 146) A-G-x-[AES]-[AG]-x-L-x(4,5)-A-x(3,5)-P-V-[FIL]-[AG]-V-P (SEQ ID NO: 147) l-x(2,3)-A-G-x-[AES]-[AG]-x-L-x(4,5)-A-[AGNS]-x(4)-[PV]-[IV] -x-[AGV]-[PV]-[PV] (SEQ ID NO: 148) G-x-[AES]-[AG]-x-L-x(4,5)-A-x(3,5)-P-V-[FIL]-[AG]-V-P (SEQ ID NO: 149)

A-x(4,5)-P-V-[FIL]-[AG]-V-P (SEQ ID NO: 150)

S-X-G-[IM]-X-C-[AGS]-X(^-[DE]-[IL] (SEQ ID NO: 151) G-x-D-x(2)-[AGNS]-G-[AGS]-G-x(2)-[Aη-D-[ILM]-x-[ASTV] (SEQ ID NO: 152) G-x(1,2)-D-x(2)-[AGNS]-G-[AGS]-G-x(2)-[Aη-D-[ILM]-x-[ASTV] (SEQ ID NO: 153)

G-x(1,2)-D-x(3)-G-[AGS]-G-x(2)-[AT]-D-[ILM]-x-[ASTV] (SEQ ID NO: 154) D-x(2)-[AGNS]-G-[AGS]-G-x(2)-[AT]-D-[ILM]-x-[ASTV] (SEQ ID NO: 155) G-x-D-x(2,3)-G-x(0,1)-G-x(3)-D-[lLM]-x-[ASTV] (SEQ ID NO: 156) G-x(0,1)-G-x(0,1)-G-[ST]-G-[ASη-G-[ACGS]-[AST]-P-x-[FILV]-[ ASV] (SEQ ID NO: 157) G-x(0,1)-G-x(0,1)-G-T-G-[AST]-[AG]-x-[ASTV]-[IPV] (SEQ ID NO: 158) H-x-D-N-[AILV]-x-[AP]-x(3)-G-[GN] (SEQ ID NO: 159)

N-L-x-[KR]-G-x(0,1)-A-x(2,3)-A-x(2)-[ACNS]-x-[DEN]-x(3)-[ DGNS] (SEQ ID NO: 160) D-N-L-x(0,2)-G-x(2,4)-A-x(2)-[ACNSV]-x-[CNV]-x(3)-[DEGN] (SEQ ID NO: 161) E-A-D-D-x-[A!LV]-[AG]-x(2)-[ADSTV] (SEQ ID NO: 162)

G-x-[IV]-[ACGTV]-[ILV]-[DN]-G-x-S-L-T-[ILV] (SEQ ID NO: 163) G-X(O 1 I)-S-[FIV]-[ACGSTV]-X-[DN]-G-X-[CS]-L-T-[ILV] (SEQ ID NO: 164) S-[FIV]-[ACGSTV]-X-[DN]-G-X-[CS]-L-T-[ILV] (SEQ ID NO: 165) G-[ADES]-S-[FIV]-[AS]-x-[ADGNQ]-G-x-C-x(1 ,3)-T (SEQ ID NO: 166) G-H-[FILV]-[LI\/lV]-x-G-H-[IV]-x(3)-[AGTV]-x-[FILV] (SEQ ID NO: 167) S-[ANS]-G-x-H-[ASη-N-G-x-[ST]-x-[AIV]-R-x-[AILV3 (SEQ ID NO: 168) S-X(O 1 I)-G-X-H-[AST]-N-G-X-[ST]-X-[AIV]-R-X-[AILV] (SEQ ID NO: 169) G-x-H-[AST]-N-G-x-[ST]-x-[AIV]-R-x-[AILV] (SEQ ID NO: 170) G-x-H-[ASTV]-N-G-x-[ST]-x-[AIV]-R-x-[AILV] (SEQ ID NO: 171) G-G-E-T-A-x(2)-[GP]-[DEGS]-[FILMV] (SEQ ID NO: 172) D-G-[IV]-G-[Sη-K-x(2)-[ILV] (SEQ ID NO: 173)

G-E-T-A-x(2)-[GP]-[DEGS]-[FILMV] (SEQ ID NO: 174)

E-x-[AS]-x-R-[PTV]-H-[DN]-[ST]-G-x(2)-[ST] (SEQ ID NO: 175) E-x-[AS]-x-R-x-H-[DN]-[Sη-G-x(2)-[ST] (SEQ ID NO: 176)

R-[PTV]-H-[DN]-[ST]-G-x(2)-[Sη (SEQ ID NO: 177)

R-G-R-[ST]-[FIL]-x(4)-[ILM\^-[FILV]-[ILV]O-[DE]-[ACSV]-Q- [ENS]-x-[EPQST]-x(3)-[ILIVIV]-x(2)-[FILV]-[ILV]-x-R-χ-G-x(2 )- [AGSTV]-x(2)-[IV]-x(3)-[DN]-x(4)-[DRS] (SEQ ID NO: 178)

G-X-^-G-[CST]-G-K-[ST]-X-[FILM]-[AST]-X-[ACSTV]-X-[AG] (SEQ ID NO: 179) N-x(3,4)-G-[LPV]-x(2)-G-[FWY]-x(3)-[LM]-x-[ILMV]-x(4)-[FY3(S EQ IDNO: 180) L-[HY]- Q -[ILMV]-X-G-R-X-[AGS]-R-X(^-[AGQS](SEQ ID NO: 181) L-[HY]-Q-x(2)-G-R-x-[AGS]-R-x(4)-[AGQS](SEQID NO: 182) L-X-Q-[ILMV]-X-G-R-X-[AGS]-R-X(^-[AGQS] (SEQIDNO: 183) G-x(3)-L-x-[QT]-[ILM]-x-[GN]-R-x(2)-[NR]-x(4)-[AGNQS](SEQ ID NO: 184) D-[ILV]-G-[AST]-G-X-G-X-P-[AGSV]-[ILV] (SEQIDNO: 185) D-[AILV]-G-[AST]-G-x-G-x-P-[AGSV]-[ILV](SEQID NO: 186) R-x(0,1)-R-R-x-C-x(2)-C-x(2)-R-[FY]-[GST]-T-x-E(SEQ ID NO: 187) R-x(0,1)-R-R-x(1,2)-C-x(2)-C-x(2)-R-[FY]-[GST]-T-x-E(SEQ IDNO: 188) R-R-R-x-C-x(2)-C-x(2)-R-x(1,2)-T-x(0,2)-T-x-E(SEQ IDNO: 189) R-R-R-X(1,2)-C-x(2)-C-x(2)-R-x(1,2)-T-x-E (SEQ ID NO: 190) R-R-x-C-x(2)-C-x(2)-R-x(1,2)-T-x(0,2)-T-x-E(SEQIDNO: 191) L-x(1,3)-C-T-H-[FY]-x(2)-[FILMV]-x(3)-[FIL] (SEQ IDNO: 192) C-T-H-[FY]-x(2)-[FILMV]-x(3)-[FIL] (SEQ ID NO: 193)

G-R-D-x-[GT]-x(2)-[ILV]-x(3)-[AS]-x(2)-[KR]-x-[FY]-[ILMV] -x-[AST]-x(4)-R-[AV]-x-R-[KR]-x(2)-[DEQ3(SEQ ID NO: 194) D-G-[PV]-[AGSη-[AGS]-[ASTV]-G-K-[GS]-[Sη-x-[ACGST]-x(2)-[I LMV]-[AGS](SEQ IDNO: 195) D-G-x-[AGSη-[AGS]-[ASTV]-G-K-[GS]-[ST]-x-[ACGST]-x(2)-[ILMV ]-[AGS] (SEQID NO: 196) D-G-x(2)-[AGS]-[ASTV]-G-K-[GS]-[Sη-x-[ACGST]-x(2)-[ILMV]-[A GS](SEQ IDNO: 197) D-G-x(3)-[ASW]-G-K-[GS]-[ST]-x-[ACGST]-x(2)-[ILMV]-[AGS] (SEQ ID NO: 198)

D-G-x(4)-G-K-[GS]-[ST]-x-[ACGSη-x(2)-[ILMV]-[AGS] (SEQ ID NO: 199) D-G-x(3,4)-G-K-[GS]-[ST]-x-[ACGSη-x(2)-[ILMV]-[AGS] (SEQ ID NO: 200) G-[AST]-X-G-K-X(O 1 I)-T-X(O 1 I)-T-[ASTV]-X(^-[ILMV]-X(S)-[FILMV] (SEQ ID NO: 201)

G-[AST]-x-G-K-[AST]-[GST]-[TV]-[AST]-x-[FlLM] (SEQ ID NO: 202) E-[ILMV]-l-[FILV]-A-x-[ADGNPST]-x-[NSη-x-[ADEN]-[GNS]-[DENQ ] (SEQ ID NO: 203)

L-A-A-G-x-[GS]-x-R-[FIM]-x-S-x(3)-K-[TV]-[LM]-x(2)-[ILV]- x-[DGQ]-x(2)-[LM]-[ILV]-x(3)-[FILV] (SEQ ID NO: 204) A-A-G-x-[GS]-x-R-[FIM]-x-S-x(3)-K-[W]-[LM]-x(2)-[ILV]-x-[DGQ ]-x(2)-[LM]-[ILV]-x(3)-[FILV] (SEQ ID NO: 205) A-G-x-[GS]-x-R-[FIM]-x-S-x(3)-K-[TV]-[LM]-x(2)-[ILV]-x-[DGQ] -x(2)-[LM]-[ILV]-x(3)-[FILV] (SEQ ID NO: 206) G-x-[GS]-x-R-[FIM]-x-S-x(3)-K-[TV]-[LM]-x(2)-[ILV]-x-[DGQ]-x (2)-[LM]-[ILV]-x(3)-[FILV] (SEQ ID NO: 207) G-x(1,3)-R-[FIM]-x-S-x(3)-K-[TV]-[LM]-x(2)-[ILV]-x-[DGQ]-x(2 )-[LM]-[ILV]-x(3)-[FILV] (SEQ ID NO: 208) R-[FIM]-x-S-x(3)-K-[TV]-[LM]-x(2)-[ILV]-x-[DGQ]-x(2)-[LM]-[I LV]-x(3)-[FILV] (SEQ ID NO: 209) C-x(4)-T-A-[GNST]-[Sη-x-[AGNQSη-x(3)-[DE]-x-[LM]-G-[FILMV] -x(4)-[ACNSη-[AGS] (SEQ ID NO: 210) G-x(0,1)-S-[ST]-N-x(3)-H-x(2)-A-x-[AS] (SEQ ID NO: 211) C-D-K-x(2)-P-[AGS]-x(2)-[ILM]-[AG] (SEQ ID NO: 212) E-x-H-H-x(2)-K-x-D-x-[ILP]-S-G-[ST]-[AGL] (SEQ ID NO: 213) H-H-x(2)-K-x-D-x-[ILP]-S-G-[ST]-[AGL] (SEQ ID NO: 214) E-x-H-x(3)-K-x-D-x-[ILP]-S-G-[Sη-[AGL] (SEQ ID NO: 215) H-x(2)-K-x-D-x-[ILP]-S-G-[Sη-[AGL] (SEQ ID NO: 216) E-T-G-A-G-x-[HW]-G-x(1 ,3)-A-x(3)-A (SEQ ID NO: 217) E-T-G-A-G-x-[HW]-G-x(1 ,3)-A-x(1,3)-A (SEQ ID NO: 218) T-G-A-G-x-[HW]-G-x(1, 3)-A-x(3)-A (SEQ ID NO: 219) G-x(3)-A-x-E-[PSη-[ACNS]-H-A-[FILV]-x(2)-[ALV] (SEQ ID NO: 220) T-G-A-G-x-[HW]-G-x(1, 3)-A-x(1,3)-A (SEQ ID NO: 221) G-x(3)-A-x(0,1)-E-[PST]-[ACNS]-H-A-[FILV]-x(2)-[ALV] (SEQ ID NO: 222)

G-x(2,3)-A-x(1,2)-E-[PST]-[ACNS]-H-A-[FILV]-x(2)-[ALV] (SEQ ID NO: 223)

G-H-x-P-x(2)-[AP]-x(2)-P-G-V-x-[ILM]-x-E-x(2,4)-A-[AQST] (SEQ ID NO: 224) H-x-P-x(2)-[AP]-x(2)-P-G-V-x-[ILM]-x-E-x(2,4)-A-[AQSη (SEQ ID NO: 225)

H-x-P-x(5)-P-G-V-x-[ILM]-x-E-x(2,4)-A-[AQST] (SEQ ID NO: 226) P-x(2)-[AP]-x(2)-P-G-V-x-[ILM]-x-E-x(2,4)-A-[AQST] (SEQ ID NO: 227)

P-G-V-x-[ILM]-x-E-x(2,4)-A-[AQST] (SEQ ID NO: 228) G-[HR]-[FY]-E-[AG]-X-D-X-R (SEQ ID NO: 229)

D-E-[IMV]-D-x-[GN]-[ILV]-[GS]-G-x(2)-[AGS]-x(2)-[IMV]-[AG S]-x(2)-[ILM]-x(2)-[ILV]-[AGS]-x(3)-Q-[ILV]-[FILMV]- [ACSTV]-[IV]-[Sη-H-x-[APV]-x-[ILV]-x-[ACGS]-x-[AGS] (SEQ ID NO: 230)

G-x-[Sη-G-[ASTV]-G-K-[Sη-[ILMV]-x-[FILV]-x-[AGS]-[ILM]-x(4 )-[GS]-x(3)-[DEGNSη (SEQ ID NO: 231) S-X(O 1 I)-G-X(O 1 I)-E-X-[ANS]-R-[FILMV]-X-L-[AS]-[FILMV] (SEQ ID NO: 232) S-X(I^)-G-X(O 1 I)-E-X-[ANS]-R-[FILMV]-X-L-[AS]-[FILMV] (SEQ ID NO: 233)

K-x-[IL]-P-x(2)-[AGS]-G-x(1,2)-G-G-x(0,1)-S-[ADNST]-[ADN]-x- [AGT]-[AGSTV]-x-[FLMV]-x(2)-[ILMV] (SEQ ID NO: 234) P-x(2)-[AGS]-G-x(1,2)-G-G-x(0,1)-S-[ADNSη-[ADN]-x-[AGT]-[AG STV]-x-[FLMV]-x(2)-[ILMV] (SEQ ID NO: 235) G-[FILMV]-[AG]-G-G-S-x(2,3)-A-[AGSTV]-x-[AILV] (SEQ ID NO: 236) G-x(1,2)-G-G-x(0,1)-S-[ADNST]-[ADN]-x-[AGη-[AGSTV]-x-[FLMV] -x(2)-[ILMV] (SEQ ID NO: 237) G-[ACTV]-[ILV]-[IV]-X-G-[ADEGNQS]-T-X-H-X(Sy-[IV]-[ACSV]-X(S )-[AGNST]-X(^-[ILMV] (SEQ ID NO: 238) D-[IV]-[AILV]-[IV]-[ACSTV]-[DE]-G-[FY]-x-G-N-x(2)-L-K-x(2)-E -x(0 1 2)-G (SEQ ID NO: 239) G-[FY]-x-G-N-x(2)-L-K-x(2)-E-x(0,2)-G (SEQ ID NO: 240) V-[ACSTV]-x-G-x(1 ,3)-N-x(2,3)-L-K-x(2)-[EG]-[AGS] (SEQ ID NO: 241) G-N-x(2)-L-K-x(2)-E-x(0,2)-G (SEQ ID NO: 242)

G-x(0,2)-G-x-P-x(4)-[EGNQ]-x(4)-[ILM]-x(2)-[DG]-[EG]-[FIL ]-[DS]-[AS]-x-[AIV] (SEQ ID NO: 243) C-x-G-V-x(2)-A-[IMV]-x(2)-[AV] (SEQ ID NO: 244)

N-x-D-G-[EGS]-x-[AGPV]-x(2)-C-G-N-[AG]-x-R-[ACTV]-x(4)-[A ILV] (SEQ ID NO: 245) E-R-G-x(3,4)-T-x-[AS]-C-G-[ST]-[AG]-x(2)-[AGS]-[ACSTV] (SEQ ID NO: 246) D-G-[EGS]-x-[AGPV]-x(2)-C-G-N-[AG]-x-R-[ACTV]-x(4)-[AILV] (SEQ ID NO: 247)

R-G-x(3,4)-T-x-[AS]-C-G-[Sη-[AG]-x(2)-[AGS]-[ACSTV] (SEQ ID NO: 248) G-[EGS]-x-[AGPV]-x(2)-C-G-N-[AG]-x-R-[ACTV]-x(4)-[AILV] (SEQ ID NO: 249) G-x(3,4)-T-x-[AS]-C-G-[ST]-[AG]-x(2)-[AGS]-[ACSTV] (SEQ ID NO: 250)

C-G-N-[AG]-x-R-[ACTV]-x(4)-[AILV] (SEQ ID NO: 251)

T-G-[IV]-[NST]-[AD]-x-[DE]-R-x(0,2)-T (SEQ ID NO: 252) R-x-G-x-T-E-x-[AGST]-x-[ADESη-[IL]-x(4)-[GN] (SEQ ID NO: 253) T-x(0,2)-G-[IV]-[NST]-[AD]-x-[DE]-R-x(0,2)-T (SEQ ID NO: 254) K-x(2)-[IL]-T-[AG]-P-x-T-[IM]-x(3)-[STV] (SEQ ID NO: 255) P-D-C-G-[LM]-[KR] (SEQ ID NO: 256)

G-[ST]-F-D-P-x(3)-G-H-x(2)-[ILMV]-[FILV]-x(2)-[AGST] (SEQ ID NO: 257) F-D-P-x(3)-G-H-x(2)-[ILMV]-[FILV]-x(2)-[AGSη (SEQ ID NO: 258) D-P-x(3)-G-H-x(2)-[ILMV]-[FILV]-x(2)-[AGST] (SEQ ID NO: 259)

G-x(3)-H-H-x(2)-E-[AGST]-x(2)-K-[AGSV]-x-[AGS]-x-[ASTV]-[ ILMV]-x(2)-[AC](SEQIDNO:260) H-H-x(2)-E-D-x-[AG]-[IL]-[ACSTV]-[IL]-G-x(2)-[FILV] (SEQ ID NO:261) H-H-x(2)-E-[AGST]-x(2)-K-[AGSV]-x-[AGS]-x-[ASTV]-[ILMV]-x(2) -[AC3(SEQ ID NO: 262) G-x(3)-H-x(2,3)-E-[AGST]-x(2)-K-[AGSV]-x-[AGS]-x-[ASTV]-[ILM V]-x(2)-[AC] (SEQ IDNO:263) G-x(3,4)-H-x(2)-E-[AGST]-x(2)-K-[AGSV]-x-[AGS]-x-[ASTV]-[ILM V]-x(2)-[AC] (SEQ ID NO:264) T-[DGP]-x-[ADGNPS]-F-x-[DENη-H-[LM]-[ILM]-x(2)-[FILMV]-x(2) -[Hη(SEQID NO: 265) H-x(2)-E-D-x-[AG]-[iL]-[ACSW]-[IL]-G-x(2)-[FILV] (SEQIDNO: 266) E-D-x-[AG]-[IL]-[ACSTV]-[IL]-G-x(2)-[FILV](SEQIDNO:267) G-G-Y-x-R-[ILMV]-x-[KR]-x(3)-R-x(2)-[DG]-x-[AGSTV]-x(4)-[lLV ](SEQID NO: 268) G-Y-x-R-[ILMV]-x-[KR]-x(3)-R-x(2)-[DG]-x-[AGSTV]-x(4)-[ILV] (SEQID NO: 269) G-Y-x(0,1)-R-[ILMV]-x-[KR]-x(3)-R-x(2)-[DG]-x-[AGSTV]-x(4)-[ ILV](SEQID NO:270) Y-x-N-[CSη-x(4)-K-[AST]-x-[ACSTV]-[ADG]-x(2)-[CDGV]-[GT]-x- [AGSTV]-x-[AGTV]-x(3)-[AIV] (SEQ ID NO: 271)

G-V-x-F-M-[ACG]-E-x-[ASV]-x(2)-[ILV]-[ACNSη (SEQ IDNO: 272)

V-x-F-M-[ACG]-E-x-[ASV]-x(2)-[ILV]-[ACNSη (SEQ ID NO: 273) S-G-x-A-x-G-[AIV]-D-x(2)-[ACS]-x(3)-[AGSTV]-[ILMV] (SEQ ID NO: 274)

G-x-A-x-G-[AIV]-D-x(2)-[ACS]-x(3)-[ACSTV]-[ILMV] (SEQ ID NO: 275)

E-M-x-T-G-E-G-K-T-[IL]-x(4)-[APV]-x(4)-[AGSV]-[FILM]-x(4) -[CT\/]-x-[ILM\/]-x-T-x-N-[DE]-Y-L-[ASV]-x(2)-[DGQ] (SEQ ID NO: 276)

M-x-T-G-E-G-K-T-[IL]-x(4)-[APV]-x(4)-[AGS\^-[FILM]-x(4)-[ CTV]-x-[ILMV]-x-T-x-N-[DE]-Y-L-[ASV]-x(2)-[DGQ] (SEQ ID NO: 277)

R-x(0,1 )-D-x-Q-[IL]-x-G-R-[ACSη-[AG]-R-x-G-D-x-G-x-[AST]-x-[FI]-x( 2)-S-x-[DEGQ]-D-x-[LV]-[FILMV] (SEQ ID NO: 278)

R-[ILV]-D-x(0,2)-L-x(1 ,3)-G-R-[ACSη-[AG]-R-x-G-D-x-G-x-[ASη-x-[FI]-x(2)-S-x-[DEG Q]-D-x-[LV]-[FILMV] (SEQ ID NO: 279)

Q-[IL]-x-G-R-[ACST]-[AG]-R-x-G-D-x-G-x-[ASη-x-[FI]-x(2)-S-x -[DEGQ]-D-x-[LV]-[FILMV](SEQIDNO:280) D-x(0,2)-L-x(1,3)-G-R-[ACSη-[AG]-R-x-G-D-x-G-x-[AST]-x-[FI] -x(2)-S-x-[DEGQ]-D-x-[LV]-[FILMV] (SEQIDNO: 281) R-x(2,4)-L-x(1,3)-G-R-[ACSη-[AG]-R-x-G-D-x-G-x-[AST]-x-[FI] -x(2)-S-x-[DEGQ]-D-x-[LV]-[FILMV](SEQIDNO:282) L-X(I 1 S)-G-R-[ACSTHAG]-R-X-G-D-X-G-X-[AST]-X-[FI]-^)-S-X-[DEGQ]-D- X-[LV]-[FILMV](SEQ ID NO: 283) T-[NQS]-M-A-G-R-G-[TV]-D-l-x-[ILP]-[DGST]-x-[DEGNS](SEQIDNO: 284) M-A-G-R-G-[TV]-D-l-x-[ILP]-[DGST]-x-[DEGNS] (SEQ ID NO: 285) G-[DES]-S-H-G-x(2)-[lLMV]-[GTV]-x-[ILV]-[ILV]-[DENST]-G(SEQI DNO:286) G-H-[AE]-x-D-x(2)-[IL]-x-D-x-[ASV]-[ASV]-[CD]-x-[RS]-x(2)-T- P-[ST]-x-A-x(3)-[ALV](SEQIDNO: 287) H-[AE]-x-D-x(2)-[IL]-x-D-x-[ASV]-[ASV]-[CD]-x-[RS]-x(2)-T-P- [ST]-x-A-x(3)-[ALV](SEQIDNO:288) D-[lLV]-x-[DEGQSη-[ST]-G-x-T-L-x(4)-L(SEQ ID NO: 289) L-X(I^)-Q-[AILV]-[AGPST]-G-X(O 1 I)-R-X-[GV]-R(SEQIDNO: 290)

H-[IL]-G-[ILV]-T-E-[AS]-G-x(4)-[AGS]-x-[IV]-x-S-[AST]-x-[ AGS]-[FIL]-[AGS]-x-[ILM]-[LM]-x(2)-[GN]-[IL]-G-[ADN]-T- [ILMV]-R-X-S-[ILM]-[AST](SEQID NO: 291)

L-x(0,2)-G-[ILV]-T-E-[AS]-G-x(4)-[AGS]-x-[IV]-x-S-[AST]-x -[AGS]-[FIL]-[AGS]-x-[ILM]-[LM]-x(2)-[GN]-[IL]-G-[ADN]-T- [ILMV]-R-X-S-[ILM]-[AST](SEQID NO: 292)

G-C-x(0,1)-V-x(0,1)-N-[AG]-[ILP]-G-E-x(3)-[ASη-x(2)-[AG] -x-[ASTV] (SEQID NO: 293)

C-x(0,1 )-V-x(0,1)-N-[AG]-[lLP]-G-E-x(3)-[AST]-x(2)-[AG]-x-[ASTV] (SEQ ID NO: 294)

R-[IV]-G-[AV]-N-X-G-S-[IL] (SEQ ID NO: 295) L-x(3,5)-G-[ADN]-T-[ILMV]-R-x-S-[ILM]-[ASη (SEQ ID NO: 296) L-x(3,4)-G-x(1,3)-T-[ILMV]-R-x-S-[ILM]-[ASη (SEQ ID NO: 297)

l-x(0,2)-G-[AV]-N-x-G-S-[IL] (SEQ ID NO: 298)

G-x-[Sη-G-[ACS]-G-K-[ST]-[Sη-x-[IL]-[KR]-x-[FILMV]-[DN] -x-[ILM] (SEQ ID NO: 299) R-x(3)-[GS]-M-[LV]-F-Q-x(3)-[LPV]-[FVyi (SEQ ID NO: 300)

E-x(2)-K-x(2,3)-N-x-[IV]-x-[ANSTV]-x(3)-[CG]-x(4)-[IPV]-x (4)-[AEGN]-x(2)-[ILV] (SEQ ID NO: 301) G-x(2)-[AG]-x-S-D-[AG]-D-[AIV]-[AILV]-x-H-[ASη-[ILV]-x-[DN] -[AS]-x(2HGSHAGV](SEQIDNO: 302) S-D-[AG]-D-[AIV]-[AILV]-x-H-[ASη-[ILV]-x-[DN]-[AS]-x(2)-[GS ]-[AGV](SEQIDNO: 303) G-x(4,5)-D-[AGη-D-[AIPV]-[AILV]-x-H-[ASη-[ILV]-x-[DN]-[AS] -x(2)-[GS]-[AGV](SEQIDNO: 304) D-[AG]-D-[AIV]-[AILV]-x-H-[AST]-[ILV]-x-[DN]-[AS]-x(2)-[GSHA GV](SEQID NO: 305) G-x-G-x-D-x-H-x-[FIL]-x(2)-[DEGNS]-x(4)-[ILP]-[ACGV]-[GIV]-[ ILV](SEQIDNO:306) G-X-G-X(0,1)-D-x-H-x-[FIL]-x(2)-[DEGNS]-x(4)-[ILP]-[ACGV]-[G IV]-[ILV] (SEQIDNO: 307) G-x-D-x-H-x-[FIL]-x(2)-[DEGNS]-x(4)-[ILP]-[ACGV]-[GIV]-[ILV] (SEQIDNO: 308) E-[ASTV]-H-x(0,2)-P-x(2)-A-x-[CS]-D-[AGNS](SEQ ID NO: 309) E-x-H-x(2)-P-x(2)-A-x-[CS]-D-[AGNS] (SEQ ID NO: 310) E-x(0,1)-H-x(2)-P-x(2)-A-x-[CS]-D-[AGNS] (SEQ ID NO: 311) l-x(2)-[LV]-x(2)-H-x(4,5)-K-x(0,1)-D-x(2)-[GSη-x(2)-[AG]-[F L]-x(4)-[AGNS]-x-R-[KR]-x-[LM]-[IL]-x-[FHY] (SEQ ID NO: 312)

H-x(4,5)-K-x(0,1)-D-x(2)-[GST]-x(2)-[AG]-[FL]-x(4)-[AGNS] -x-R-[KR]-x-[LM]-[IL]-x-[FHY] (SEQ IDNO: 313) G-x(2)-[ADEGQS]-[ALV]-Q-x(2,3)-L-x(3,4)-l-x(2)-[LV]-x(2)-H(S EQIDNO:314) G-x-[Sη-E-x-[AIL]-P-[IV]-S-S-x-[AGT]-x(3)-[AILV](SEQIDNO: 315) G-x(1,2)-E-x-[AIL]-P-[IV]-S-S-x-[AGT]-x(3)-[AILV](SEQIDNO: 316) G-x(0,2)-E-x-[AIL]-P-[IV]-S-S-x-[AGη-x(3)-[AILV](SEQID NO: 317)

H-P-x-[FWY]-[Sη-G-x(2)-[KR]-x(4)-[ADEGST]-[GS]-x-[AIV]-[ ADES]-x-F-x(2)-[KR]-[FY] (SEQ ID NO: 318) H-P-x-[FWY]-[AESη-G-x(2)-[KR]-x(4)-[ADEGST]-[GS]-x-[AIV]-[A DES]-x-F-x(2)-[KR]-[FY] (SEQ ID NO: 319) P-x-[FWY]-[ST]-G-x(2)-[KR]-x(4)-[ADEGSη-[GS]-x-[AIV]-[ADES] -x-F-x(2)-[KRHFY] (SEQ ID NO: 320)

REFERENCES (the contents of which are hereby incorporated in full)

[I] O'Hagan et al (2001) BiomolEng. 18:69-85.

[2] Moriwaki et al. (2001) J Biol Chem. 276:23065-76.

[3] Zipfel & Felix (2005) Curr Opin Plant Biol 8:353-60.

[4] Chalifour et α/.(2004) Blood 104:1778-83.

[5] Alvarez (2005) Front Biosci. 10:582-7.

[6] O'Hagan (2001) Curr Drug Targets Infect Disord. 1:273-86.

[7] Altschul et al. (1990) J. MoI. Biol. 215(3):403-10.

[8] Jonassen et al. (1995) Protein Science 4:1587-1595.

[9] Nielsen et al. (1997) Protein Eng. 10:1-6.

[10] Dutta (2002) Immunol Lett. 83:153-161.

[II] Bairoch et al. (1997) Nucl. Acids Res. 25:217-221.

[12] van Dijk et al. (1986) Methods Find. Exp. Clin. Pharmacol. 8(3):189-93.

[13] Romberg et al. (1997) Vaccine 15(17-18):1820-6.

[14] Berthou et al. (1987) FEBS Letters 218:55-58.

[15] Rich et al (1984) J. Med. Chem. 27:417-422.

[16] Audhya et al. (1986) Science 231:997-999.

[17] Ogawa et al. (2002) Eur. J. Immunol. 32:2543-50.

[18] Suzue & Young (1996) J Immunol 156:873-879.

[19] Bodanszky (1993) Principles of Peptide Synthesis (ISBN: 0387564314).

[20] Fields et al (1997) Methods in Enzymology 289: Solid-Phase Peptide Synthesis. ISBN: 0121821900

[21] Chan & White (2000) Fmoc Solid Phase Peptide Synthesis ISBN: 0199637245.

[22] Kullmann (1987) Enzymatic Peptide Synthesis. ISBN: 0849368413.

[23] Kazmierski (1999) Peptidomimetics Protocols. ISBN: 0896035174.

[24] Abell (1999) Advances in Amino Acid Mimetics and Peptidomimetics. ISBN: 0762306149.

[25] US patent 5,331,573 (Balaji).

[26] Goodman et al (2001) Biopolymers 60:229-245.

[27] Hruby & Balse (2000) Curr Med Chem 7:945-970.

[28] Ribka & Rich (1998) Curr Opin Chan Biol 2:441-452.

[29] Barron & Zuckermann (1999) Curr Opin Chem Biol 3:681-687.

[30] WO 91/19735.

[31] Chakraborty et al. (2002) Curr Med Chem 9:421-435.

[32] Ibba (1996) Biotechnol Genet Eng Rev 13:197-216.

[33] Geimaro (2000) Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472

[34] Singh et al (2001) Pharm Res. 18:1476-1479.

[35] Singh & O'Hagan (1999) Nat Biotechnol 17(ll):1075-81 [36] Souberbielle et al. (1996) Gene Titer 3(10):853-8 [37] Asai et al. (2001) Infect Immun. 69:7387-7395. [38] Asea et al. (2002) J Biol Chem. 277:15028-15034. [39] Hirschfeld et al. (2001) Infect Immun 69(3): 1477-82. [40] Mullarkey et al. (2003) J Pharmacol Exp Tlier 304(3): 1093-1102. [41] Current Protocols in Molecular Biology (F.M. Ausubel et al., eds., 1987) Supplement 30. [42] Smith & Waterman (1981) Adv. Appl. Math. 2: 482-489. [43] Rice et al. (2000) Trends in Genetics 16:276-277.