Title of Invention

A SYNTHETIC PEPTIDE

Abstract The present invention comprises novel and modified peptides capable of inducing an HIV-1 specific immune response without antagonizing the cytotoxic T-cell activity in order to achieve an effective prophylactic and therapeutic vaccine against HIV. The peptides are based on conserved regions of HIV gag p24 proteins. Antigens in free- or carrier-bound form comprising at least one of the said peptides, vaccine compositions containing at least one of the antigens, immunoassay kits and a method of detecting antibodies induced by HIV or HIV specific peptides using such antigens, are described.
Full Text FORM 2
THE PATENTS ACT 1970
[39 OF 1970]
&
THE PATENTS RULES, 2006
COMPLETE SPECIFICATION
[See Section 10; rule 13]
"HIV PEPTIDES OF SEQ ID NO 9, ANTIGENS, VACCINE COMPOSITIONS, IMMUNOASSAY KIT AND A METHOD OF DETECTING
ANTIBODIES INDUCED BY HIV
BIONOR IMMUNO AS, a Norwegian company of P.O. Box 1893 Gulset, N-3703 skien, Norway,
The following specification particularly describes the invention and the manner in which it is to be performed:

The present invention relates to novel peptides based on conserved regions of HIV gag p24, antigens in free or carrier-bound form comprising at least one of the said peptides, vaccine compositions containing at least one of the antigens, immunoassay kits and a method of detecting antibodies, induced by human immunodeficiency virus (HIV) or HIV-specific peptides, using such antigens.

BACKGROUND
There is an urgent heed to control the global epidemic of HIV infection and the development of a vaccine against HIV is one of the major objectives in AIDS research. In general vaccines should activate antigen presenting cells, overcome genetic restriction in T-cell responses and generate T- and B-memory cells. The variability of the viral population poses a further difficulty in obtaining an effective HIV vaccine. A break through in the ongoing attempts to develop a vaccine against AIDS has so far not been reported. It is now generally accepted that an induction of antigen-specific humoral and cell-mediated immunity is crucial for a development of an effective prophylactic and therapeutic vaccine. All three arms of the immune system including neutralizing antibodies; CD8+CTL and T-helper-1 (TH1) cells might be required for protective immunity to HIV. It is known that CTL can clear other viral infections (Ada, Immunol. Cell Biol., 72:447-454, 1994) and that CTL can lyse infected targets early in infection before viral progeny can be produced and released by cell lysis, Ada et a!., supra. The focus has been on selection of antigens as well as on design and evaluation of different adjuvances. The antigens used in different in vitro and in vivo studies have been all from crude proteins to various synthetic peptides mainly from gp160 and to some extent from p24. A large number of studies have been done on the V3 loop of gp120. induction of both B- and T-cell responses have been observed, however, it has been reported from an in vitro study that a peptide from the conserved region of gp41 have indicated infection enhancement (Bell S.J., et al., Clin. Exp. Immunol., 87 (1): 37-45, (January 1992).

WO 00/52040 PCT/NO00/00075

Naturally occurring HIV sequences in vaccine candidates are not capable of stimulating a stable immune response due to the viruses inherent ability to hide by changing the appearance of the epitopes presented on the cell surface of infected ceils. The immune system is fooled to believe that a particular amino acid sequence is relevant when in fact the amino acids of importance is hidden.
A resent study of titers of antibodies against the gag p24 protein, has shown that slow progression towards development of AIDS Is associated with high titers, while fast progression towards development of AIDS is associated with low titers. It is shown that persons with low p24 antibody titer develop significantly faster AIDS than persons with high P24 antibody indicating that p24 can play a key role to control the development of AIDS.
New HIV p24 peptides are described in WO91/13360, wherein the peptides are used in
a method of discriminating between a false and true diagnosed HIV-positive serum
sample.
Johnson R.P., et al., The Journal of Immunology, Vol.147, p.1512-1521, No:5,
September 1, 1991 describe an analysis of the fine specificity of gag-specific CTL-
responses in three HlV-1 seropositive individuals, the gag-specific CTL-responses were
found to be mediated By CD3+CD8+ lymphocytes which are HLA class I restricted.
EP-A-0 356 007 discloses antigenic determinants, in particular it relates to synthetic polypeptide sequences which are related to proteins present in the HIV-1 and which can be used as a basis for a potential vaccine against AIDS.
Rosenberg E.S. et al., Science, Vol.278, 21 November 1997, p. 1447-1450 describe that virus specific CD4+ T helper lymphocytes are critical to the maintenance of effective immunity in a number of chronic viral infections, but are characteristically undetectable in chronic human immunodeficiency virus-type 1 (HIV-1) infection. HIV-1-specific proliferative responses to p24 were inversely related to viral load. They

WO 00/52040 PCT/NO00/00075
conclude that the HIV-1-specific helper cells are likely to be important in immunotherapeutic interventions and vaccine development.
EP 0 230 222, EP 0 270 114, DE 37 11 016 and GB 2 188 639 all in the name of F. Hoffmann-La Roche & Co. Aktiengesellschaft concern recombinant expression and purification of an HTLVIII Gag/Env gene protein or fusionproteins. The proteins consisting of native sequences can be purified to homogeneity and used as a basis for diagnostic tests for detection of antibodies against viruses associated with AIDS. The gag/env protein may also be formulated for use as a vaccine for protection against AIDS through prophylactic immunization.
From a diagnostic and therapeutic point of view, the major problems with using p24 as part of an assay or therapy is associated with the high number of epitopes on p24 which stimulates production of a large number of antibodies with poor specificity, which through repeated boostering on potential mutated sequences can create autoantibodies (Autoantibodies to the alfa/beta T-cell receptors in HIV infection; dysregulation and mimicry. Lake D.F., et aj., Proc. Natl. Acad. Sci. USA, (23): 10849-53, Nov. 8 1994). Further, it is reported that the p24 antibody titer does not reach the same high levels as for the envelope proteins (gp120 and gp41). Normally antibodies to p24 are developed in the very early phase of the infection, but the titer is fairly quickly stabilized after the initial infection period. Later the p24 titer is gradually decreasing while the opposite happens with gp160. These findings can also be seen in relation to recent reports stating that cytotoxic T-cell activity is antagonized by naturally occurring HIV-1 gag variants (Klenerman P., et al., Nature, 2:369 (6479), p. 355, 2 June 1994). This can be one of the reasons why a rapid stabilization of the p24 titer is seen and why it later starts to decrease.
Based on the above background data, we decided to investigate the possibility of designing novel synthetic peptides which can mimic the p24 epitope without antagonizing the cytotoxic T-cell activity, in order to meet the need for an effective prophylactic and therapeutic vaccine.


NO 000000075
Amended page

The intital work was based on one epitope which was published by Korber B., et al., Human Retroviruses and AIBB 1997 Eds-Theoretical Biology and Biophysics: Group; Los Alamos National Laboratory. Los Alamos. NM. The amino acid sequence of this epitope (203-222) was:
KAL GPGATLEEMMTA CQGVG
RRM R T K SI KD L S S S R R
G V R V
S A A
S E
Q Q
The one letter as well as the three letter codes defining the amino adds in the. sequences given throughout this specification are in accordance with International standards and given in textbooks, for instance Lehninger A.L, Princlples of Biochemistry». Worth Publishers Inc.. New York. 1982. The aminoadds given below the head sequence represent the natural variation of the sequence. An initial study of a sequence containing this modified epitope was conducted on the sequence:
ANPDCKQILKSLGPGATLEEXXTACQGVG-
NH2

wherein X indicates 2-amiriohexanoic add and the cysteine residues are in an oxidized state, i.e. are forming an intrachain disulphide bridge. The results (unpublished) from studies using this peptide as part of a diagnostic kit showed that the specificity became 87% (n=279) on a preselected panel of African sera. The sensitivity was surprisingly 100% on a panel of HIV positive sera including HIV-1 • subtype O sera, which is quite different from the other subtypes.
In order to improve specificity, i.e. define the amino acids which contribute to a pure non-crossreacting antibody response, a similar study was applied to a significantly shorter and further modified peptide:
LIWGATCQEHXTA CQGVG-NH2


WO 00/52040 PCT/NO00/00075

wherein X has the above mentioned meaning and the cysteine residues are forming an intrachain disulphide bridge.
The results from this study showed that the specificity of the assay increased to 96%, and (n=293) which is similar to the specificity obtained in the assay without using the p24 peptide. With a specificity of 87% to the assay where the first peptide was included, it would be likely that the peptide would induce immune response to more than one' epitope since it was recognized by unspecific antibodies, if it was used as a vaccine candidate. The latter, however, show that the peptide sequence is picking up an immune response which is unique to HIV-1. Consequently, if a sequence based on this is used as an antigen in a vaccine candidate, it would most likely boost an unique immune response to HlV-li
To further increase the number of T-cell epitopes and reduce the probability for development of escape mutants three additional peptide sequences were based on the
following three sequences from residues 264-284, 253-271 and 166-186, respectively published in Human Retroviruses and AIDS 1997; A Compilation and Analysis of Nucleic Acid and Amino Acid Sequences. Eds. Theoretical Biology and Biophysics Group, Los Alamos National Laboratory., Los Alamos :
RWI I L GLNKI VRMYSPT S I L D
K G V V M M K C V G E
D M V V Q I G
S
A
NNPPIPVGEIYKRWIILGL
S Q A V K D M L R K G M V-M'"
G G S N, K V D V V
H GT
A
P

WO 00/52040 PCT/NO00/00075

and
PEV PMFSALSEGATPQDLN T
R I TT T LTE AD ISYNIYM
L N AL V H V I
M LA V
Several modified peptides have been synthesized in order to determine unique sequences which are both specific and sensitive towards HIV-1.
DESCRIPTION OF THE INVENTION
The peptides according to the invention are originating from the four different conservec areas of the HIV-1 core protein p24 which are described above, having the properties ol maintaining the uniqueness (sensitivity and specificity) of the HIV-1 -epitope. Further the new peptides according to the invention possess no recognized cytotoxic T lymphocyte (CTL) antagonistic effect and shall have at least one potential CTL epitope.
The peptides, according to the invention, which have met the above criteria are
selected from the following groups ;
Xaa, Xaa2Xaa3Xaa4 Xaa5 Xaa6 Ala Xaa8 Xaa9 Gin Thr Pro Trp Xaa14Xaa15 Xaa16 Xaa17 Xaa18Val Xaa20 (SEQ ID NO : 1)
wherein the amino acids of the chain could have the following meanings ;
Xaa in position 1 of the peptide derivate is Lys or Arg,
Xaa in position 2 is Ala, Gly, Ser or Arg,
Xaa in position 3 is Leu or Met,
Xaa in position 4 is Gly or Arg,
Xaa in position 5 is Pro, Thr, Val, Ser, Gin or Ala,
Xaa in position 6 is Gly, Ala, Lys, Arg, Gin or Glu,
Xaa in position 8 is Thr or Ser,

WO 00/52040 PCT/NO00/00075

Xaa in position 9 is Leu or lie ,
Xaa in position 14 is Thr, Ser or Val, Xaa in position 15 is Ala or Ser,
Xaa in position 16 is Cys or Ser,
Xaa in position 17 is Gin or Leu
Xaa in position 18 is Gly, Glu or Arg, Xaa in position 20 is Gly or Arg,
the peptide comprises at least nine consecutive amino acids of the sequence of SEQ ID NO: 1,
Xaa,Xaa2 Xaa3 Xaa4 Xaa5 Gly Leu Asn Pro Leu Val [Gly]nXaa12Xaa13 Tyr Xaa15 Pro Xaa17Xaa18 lie Leu Xaa21 Xaa22 (SEQ ID NO : 4)
wherein the amino acids of the chain have the following meaning;
Xaa in position 1 is Arg, Lys, Asp or none
Xaa in position 2 is Trp, Gly, Lys or Arg,
Xaa in position 3 is lie, Leu, Val or Met
Xaa in position 4 is He, Val or Leu
Xaa in position 5 Leu, Met, Val or Pro
Xaa in position 12 is Arg, Lys
Xaa in position 13 is Met or Leu,
Xaa in position 15 is Ser, Cys or Gin,
Xaa in position 17 is Thr, Val, lie, Ser or Ala,
Xaa in position 18 is Ser, Gly or Thr,
Xaa in position 21 is Asp, Glu, Cys or Gly,
Xaa in position 22 is Gly or none
wherein the sequence of SEQ ID NO : 4 comprises at least six consecutive amino acids
and n = 0,1,2 or 3,
Xaa, Xaa2Xaa3 Pro lie Pro Xaa7 Xaa„ Xaa9 Xaa10Xaa11 Xaa12 [Gly]n Xaa13 Xaa14Xaa15 Xaa18 Xaa17 Xaa18 Xaa19 Xaa20 Xaa21 Xaa^ Xaa23 Xaa24 (SEQ ID NO : 9)

WO 00/52040 PCT/NO00/00075

wherein Xaa in position 1 is Asn, Ser, Gly, His, Ala, Pro, Arg or none
Xaa in position 2 is Asn, Ala or Lys
Xaa in position 3 is Pro, Gin, Gly, He or Leu
Xaa in position 7 is Val or Ala
Xaa in position 8 is Gly or Lys
Xaa in position 9 is Glu, Asp, Lys, Phe or Thr
Xaa in position 10 is lie, Met, Val or Leu
Xaa in position 11 is Tyr, Leu or none
Xaa in position 12 is Ser or none
Xaa in position 13 is Arg or none
Xaa in position 14 is Asp, Arg, Trp, Ala or none
Xaa in position 15 is He or none
Xaa in position 16 is Tyr or none
Xaa in position 17 is Lys or Arg
Xaa in position 18 is Arg, Lys or Asp
Xaa in position 19 is Trp or Gly
Xaa in position 20 is lie, Met, Val, Gin or Ala
Xaa in position 21 is lie, Val or Ala
Xaa in position 22 is Leu, Met or Val
Xaa in position 23 is Gly or Cys
Xaa in position 24 is Leu or none
wherein the sequence of SEQ ID NO : 9 consists of at least six consecutive amino acids and n = 1,2 or 3,
Xaa, Xaa2 IIe IIe Xaas Xaa6 Xaa7Xaa8 Xaa9 Leu Xaa„ [Gly]n[Arg]m Xaa12 Xaa13 Xaa14 Xaa15Xaa16 Xaa17 Xaa18 Xaa19Xaa20 Xaa21Xaa22Xaa23 Xaa24 Xaa2S(SEQ ID NO: 15)
wherein the Xaa in position 1 is Pro, Lys, Arg or none
Xaa in position 2 is Glu, Arg', Phe or Lys
Xaa in position 5 is Pro or Thr
Xaa in position 6 is Met, Thr or Nleu
Xaa in position 7 is Phe or Leu

WO 00/52040 PCT/NO00/00075

Xaa in position 8 is Ser, Thr, Ala or Met
Xaa in position 9 is Ala, Glu or Leu
Xaa in position 11 is Ser or none
Xaa in position 12 is Ala, Arg or none
Xaa in position 13 is He, Leu or none
Xaa in position 14 is Ser, Ala, Leu or none
Xaa in position 15 is Tyr, Glu or Asp
Xaa in position 16 is Gly or Asp
Xaa in position 17 is Ala or Leu
Xaa in position 18 is Thr, He, Val, Leu or Asn,
Xaa in position 19 is Pro, Thr or Ser
Xaa in position 20 is Tyr, Phe, Nleu, His or Gin
Xaa in position 21 is Asp, Asn, Leu or Ala
Xaa in position 22 is Leu, lie, Val or Asn
Xaa in position 23 is Asn, Tyr, Cys or Gly
Xaa in position 24 is Thr, Met, lie, Ala, Val or none
Xaa in position 25 is Gly or none
wherein the sequence of SEQ ID NO : 15 consists of at least six consecutive amino
acids, n = 1,2 or 3 and m=0,1,2 or 3,
the terminal ends of the sequences may be free carboxyl- or amino groups, amides, acyls, acetyls or salts thereof,
two or more of the Cys residues may form part of an intrachain- or interchain disulphide binding, a -S-(CH2)P-S- or a - (CH2)p-bridge wherein p = 1-8, optionally intervened by one or more heteroatoms such as O, N or S and/or the said peptide sequences are immobilized to a solid support.
The new peptide sequences have the potential to serve as a good antigen wherein the antigen comprises at least one peptide selected form the group of sequences
ID NO: 1 SEQ ID NO: 4 SEQ ID NO: 9 or SEQ ID NO; 15 The antigenicity, may be
adapted through adjusting the ratio or concentration of different peptides or size of the peptides by for instance dimerisation or polymerisation and/or immobilisation to a solid phase. The antigen comprises two or more polypeptide sequences, according to the

WO 00/52040 PCT/NO00/00075

invention, which are either linked by a bridge for instance a disulphide bridge between the Cys residues of the chains or bridges like C1-C8 alkylen possibly intervened by one or more heteroatoms like O, S, or N or preferably they are unlinked. The chains may be immobilized to a solid phase in monomeric, dimeric or oligomeric forms. Further amino acids may be added to the ends in order to achieve an «arm» to facilitate immobilization.
All amino acids in the peptides of the invention can be in both D- or L-form, although the naturally occurring L- form is preferred.
. The C- and N-terminal ends of the peptide sequences could deviate from the natural
sequences by modification of the terminal NH2- group and/or COOH-group, they may for instance be acylated, acetylated, amidated or modified to provide a binding site for a carrier or another molecule.
The peptides according to the invention are consisting of 6 to 50 amino acids, preferably between 10 and 30 amino acids. They are covering all natural variation of amino acids in the identified positions.
The polypeptide antigen according to the invention is either in a free or in a carrier-bound form. The carrier or solid phase to which the peptide is optionally bound can be selected from a vide variety of known earners. It should be selected with regard to the intended use of the immobilized polypeptide as a diagnostic antigen or as an immunizing component in a vaccine.
Examples of carriers that can be used for e.g. diagnostic purposes are magnetic beads or latex of co-polymers such as styrene-divinyl benzene, hydroxylated styrene-divinyl benzene, polystyrene, carboxylated polystyrene, beads of carbon black, non-activated or polystyrene or polyvinyl chloride activated glass, epoxy-activated porous magnetic glass gelatine or polysaccharide particles or other protein particles, red blood cells, mono- or polyclonal antibodies or fab fragments of such antibodies.

WO 00/52040 PCT/NO00/00075
According to a further embodiment of the present invention, the antigens may form part of a vaccine possibly combined with carriers, adjuvants or combined with other immunostimulating elements such as canarypox virus carrying the env gene. Examples of earners and/or adjuvants for vaccine purposes are other proteins such as human or bovine serum albumin and keyhole limpet haemocyanin. Immunostimulatory materials may be divided into three groups; adjuvants, carriers for antigens and vehicles. Examples of adjuvants include aluminum hydroxyd, aluminum salts, saponin, muramyl di- and tri-peptides, monophosphpryl lipid A, B.pertussis and various cytokines including the Th1 cytokine IL-12 and IL-1. A number of protein toxins can be used to carry passenger proteins across cellular membranes into the cytosol, which are useful in developing CTL vaccines Carriers include bacterial toxoids such as inactivated inactivated tetanus arid cholera toxins, genetically detoxified bacterial toxins such as heat labile enterotoxin from E.coli, fatty acids, live vectors Such as polio chimeras and hybrid proteins that form particulates for example yeast retrotransposon hybrid TY particles and HBcAg particles. Vehicles which are frequently occurring components in modem vaccines are consisting of mineral oil emulsion, Freunds complete and incomplete adjuvant, vegetable oil emulsions, nonionic block co-polymer surfactants, squalene or squalane, liposomes and biodegradable microspheres. Two novel adjuvants which possess significant potential for the development of new vaccines include an oil-in-water microemulsion (MF59) and polymeric microparticles. Any substance that can enhance the immunogenicity of the antigen may be used and several further alternatives of carriers or adjuvants are given in the US or European Pharmacopoeia.
A suitable formulation of the antigen for immunostimulatory uses may also comprise interferons such as INF-y, antiviral chemokines or haematopoietic growth factors such as granulocyte macrophage growth factor,
Another approach in order to enhance the stimulation and absorption in for instance the
intestine is to administer the peptides of the invention, with small peptides such as di-
tri- or tetra peptides. These peptides can be administered in addition to or in combination with the peptides of the invention Preferably the peptides are administered
together with the tripeptide YGG, consisting of amino acids in the D- or L-forms; preferably in the D-form.

WO 00/52040 PCT/NO00/00075

Recent approaches to non-parenteral delivery of vaccines, for instance via mucosa include; gene fusion technology to create non-toxic derivatives of mucosal adjuvants, genetically inactivated antigens with a deletion in an essential gene, coexpression of an antigen and a specific cytokine that is important in the modulation and control of a mucosal immune response, and genetic material itself that would allow DNA or RNA uptake and its endogenous expression in the host's cells.
One approach for developing durable responses where cell-mediated immunity is ■ required, is to vaccinate with plasmid DNA encoding one or more specific antigen(s).
In order to protect against HIV infection, vaccines should induce both mucosal and systemic immune responses and could be administered by any convenient route, parenterally or non-parenterally, such as subcutanously, intracutanously, intravenously, intramuscularly, perorally, mucosally or intranasally for example.
In a preferred embodiment of the vaccine according to the present invention it
comprises antigens containing the peptides of the SEQ ID NO : 1, 4, 9 and 15, more preferred the peptides occur in the ratio 1:1:1:1.
In a further preferred embodiment the vaccine composition contains the antigens ;
RAL GPAATLQTPWTASLGVG-NH2(SEQ ID NO : 3)
RWLLLGLNPLVGGGRLYSPTSILG-NH2 (SEQ ID NO : 6)
RAIPIPAGTLLSGGGRAIYKRTAILG- NH2(SEQ ID NO : 11)
and
RFMPNIFTALSGGRRALLYGATPYAIG-NH2 (SEQ ID NO : 18).
One of the sequences contains a B-cell epitope and will activate the humoral immune system, whereas the other sequences contribute with CTL-epitopes and the amino acid changes implemented within the frame of the CTL-epitope are designed to achieve enhanced binding. Other amino acid changes have been conducted in order to facilitate the synthesis of the peptide and/or increase the solubility of the peptide.

WO 00/52040 PCT/NO00/00075

A method for detecting antibodies, induced by HIV-1 or HIV-1 specific peptides or proteins, in a sample of body fluid using the present antigens is a further embodiment of the invention. Also immunoassay kit designed for this detection and antibodies capable of selectively reacting with the said antigens are encompassed by the present invention.
DESCRIPTION OF THE PREPARATION OF THE PEPTIDES
The peptides of the invention can be produced by any known method of producing a linear amino acid sequence, such as recombinant DNA techniques. A nucleic acid sequence which encodes a peptide of the invention or a multimer of the said peptides, is introduced into an expression vector. Suitable expression vectors are for instance plasmids, cosmids, viruses and YAC (yeast artifical chromosome) which comprise necessary control regions for replication and expression. The expression vector may be stimulated to expression in a host cell. Suitable host cells are for example bacteria, yeast cells and mammal cells. Such techniques are well known in the art and described for instance by Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, 1989. Other well-known techniques are degradation or synthesis by coupling of one amino acid residue to the next one in liquid phase or preferably on a solid phase (resin) for instance by the so-called Merrifield synthesis. See for instance Barany and Merrifield in the Peptides, Analysis, Synthesis, Biology, Vol.2, E. Gross and Meinhofer, Ed. (Acad.Press, N.Y., 1980), Kneib-Coronier and Mullen Int. J. Peptide Protein Res.,30, p.705-739 (1987) and Fields and Noble IntJ.Peptide Protein Res., 35, p.161-214 (1990).
In case a linked or cyclic peptide is desired, the amino acid sequence is subjected to a chemical oxidation step in order to cyclize or link the two cysteine residues within one or between two peptide sequences, when the appropriate linear amino acid sequences are synthesized, see Akaji et al., Tetrahedron Letter, 33, 8, p. 1073-1076,1992.

WO 00/52040 PCT/NO00/00075

GENERAL DESCRIPTION OF SYNTHESIS
All peptide derivatives prepared in the Examples given below were synthesized on a Milligen 9050 Peptide Synthesizer using a standard program. The resin used was Tenta Gel P RAM with a theoretical loading of 0,20 meq/g (RAPP POLYMERE GmbH,
Tubingen). The final product of the synthesis was dried in vacuo overnight. The peptide was then cleaved from the resin by treatment with 90% trifluoroacetic acid in the presence of ethandithiol (5%) and water (5%) as scavengers (1,5 hours at RT). Then the resin was filtered and washed on filter with additional trifluoroacetic acid (100%) (2 x 20 ml). The combined filtrates were evaporated in vacuo (water bath at RT) and the residue was triturated with ethyl ether (200 ml) and the precipitated product filtered off. The solid was promptly dissolved on filter with glacial acetic acid (100 ml) and added to 1,5 I of 20% acetic acid in methanol and treated with 0,1 M solution of iodine in methanol until a faint brown colour remained. Then Dowex 1 x 8 ion exchange in acetate form (15g) (Bio-Rad, Richmond, CA) was added and the mixture filtered. The filtrate was evaporated and the residue freeze-dried from acetic acid. The product was then purified by reversed phase liquid chromatography on a column filled with Kromasil® 100 - 5 C8 (EKA Nobel, Suite, Sweden) in a suitable system containing acetonitrile in 0,1 % trifluoroacetic acid water solution. The samples collected from the column were analyzed by analytical high performance liquid chromatography (HPLC) (Beckman System Gold, USA) equipped with a Kromasil® 100 - 5 C8 Column (EKA Nobel, Surte, Sweden). Fractions containing pure substance were pooled, the solvent was evaporated and the product freeze-dried from acetic acid. The final HPLC analysis was performed on final product, and the structure of the peptide was confirmed by amino acid analysis and mass spectrometry (LDI-MS).
All amino acids used during the synthesis were L-amino acids and they were protected with a fluorenylmethoxy-carbonyl group at the a-amino function. The side chains were protected as follows:
Cys (Trt), Gln(Trt), Glu(OtBu), Thr(tBu). The abbreviations, within the brackets are :

WO 00/52040 PCT/NO00/00075

Trt = triphenylmethyl
t-Bu = tert. Butyl
OtBu = tert. Butylester
The amino acid derivatives was supplied by Bachem AG, Switzerland.

EXAMPLE 1
Preparation of K A L G P G AT L Q T P WT A C Q G VG - NH2(SEQ ID NO : 2).' The peptide was synthesized in amide form, from corresponding starting materials according to the general description of synthesis. The purity was determined by HPLC analysis and the structure was confirmed by amino acid analysis and mass spectrometry (LDI-MS).
Purity (HPLC): 87%
EXAMPLE 2
Preparation of RALGPAATLQTPWTASLGVG (SEQ ID NO : 3).
The peptide was synthesized in amide form, from corresponding starting materials
according to the general description of synthesis. The purity was determined by HPLC
analysis and the structure was confirmed by amino acid analysis and mass
spectrometry (LDI-MS).
Purity (HPLC): more than 95%
Molecular weight (free base): 1966
Molecular formula : C88H144O25N26
EXAMPLE 3
Preparation of WMPGLNPLVGGGKLYSPTSILCG-NH2 (SEQ ID NO : 5).
The peptide was synthesized in amide form, from the corresponding starting materials
according to the general description of synthesis. The purity was determined by HPLC
analysis and the structure was confirmed by amino acid analysis and mass
spectrometry (LDI-MS).
Purity (HPLC): 95%
Mass spectral analysis : Theoretical molecular weight: 2454.9
Experimental molecular weight: 2454.8 ES+

WO 00/52040 PCT/NO00/00075

EXAMPLE 4
Preparation ofRWLLLGLNPLVGGGRLYSPTSILG (SEQ ID NO : 6). The peptide was synthesized in amide form, from the corresponding starting materials according to the general description of synthesis. The purity was determined by HPLC analysis and the structure was confirmed by amino acid analysis and mass spectrometry (LDI-MS).
Purity (HPLC): more than 95 %
Molecular weight (free base): 2552
Molecular formula: C119H195O29N33 '
EXAMPLE 5
Preparation of K I L L G L N P L V G G G R L Y S P T S I L G (SEQ ID NO : 7). R L L
LGLNPLVGGG R (SEQ l.D NO : 8) and N I P I P V G D I Y G G
G D I Y K R W,Q A L C L (SEQ ID NO : 24). The peptides are synthesized in amide
form, from the corresponding starting materials according to the general description of
synthesis. The purity are determined HPLC analysis and the structures are
confirmed by amino acid analysis arid mass spectrometry (LDI-MS).
EXAMPLE 6
Preparation of RNIPIPVGDIYGG G D I Y K R W Q A L C L (SEQ ID NO: 10); The peptide was synthesized in amide form, from the corresponding starting materials according to the general description of synthesis. The purity was determined by HPLC analysis and the structure was confirmed by amino acid analysis and mass spectrometry (LDI-MS). Purity (HPLC): 85%
Mass spectral analysis : theoretical molecular weight: 2817.3 Experimental molecular weight: 2813.7 ES+
EXAMPLE 7
Preparation of R A I P I P A G T L L S G G G R A I Y K R W A I L G (SEQ ID NO ; 11):_ The peptide was synthesized iri amide form, from the corresponding starting materials according to the general description of synthesis. The purity was determined by HPLC

WO 00/52040 PCT/NO00/00075

analysis and the structure was confirmed by amino acid analysis and mass
spectrometry (LDI-MS).
Purity (HPLC): more than 95 %
Molecular weight (free base): 2707
Molecular formula: C125H208O29N38
EXAMPLE 8
Preparation of ALPIPAGFIYGGGRIYKRWQALG (SEQ ID NO : 12), K I P IPVGFIGGGWIYKRWAILG (SEQ ID NO : 13) and K I P I P V G T L L S G G GRIYKRWA|LG( SEQ ID NO : 14). The peptides are synthesized in amide form, from the corresponding starting materials ^according to the general description of synthesis. The purity are determined by HPLC analysis and the structures are confirmed by amino acid analysis and mass spectrometry (LDI-MS).
EXAMPLE 9
Preparation of KFIIPNIFSALGGAISYDLNTNILNCI (SEQ ID NO : 10). The peptide was synthesized in amide form, from the corresponding starting materials according to the general description of synthesis. Nl in the sequence is Norleucine. The purity was determined by HPLC analysis and the structure was confirmed by amino acid analysis and mass spectrometry (LDI-MS). Purity (HPLC): more than 80 %
Mass spectral analysis : Theoretical molecular weight: 2783.3 Experimental molecular weight: 2783.3 ES+
EXAMPLE 10
Preparation ofKFIIPNIFSALSGGGAISYDLNTFLNCIG (SEQ ID NO :
17).
The peptide was synthesized in amide form, from the corresponding starting materials
according to the general description of synthesis. Nl in the sequence is Norleucine. The
purity was determined by HPLC analysis and the structure was confirmed by amino
acid analysis arid mass spectrometry (LDI-MS).
Purity (HPLC): more than 80 %
Mass spectral analysis : Theoretical molecular weight: 2932.4

WO 00/52040 PCT/NO00/00075

Experimental molecular weight: 2931.8 ES+
EXAMPLE 11
Preparation of RFIIPNIFTALSGGRRALLYGATPYAIG (SEQ ID NO :
18).
The peptide was synthesized in amide form, from the corresponding starting materials
according to the general description of synthesis. Nl in the sequence is Norleucine. The
purity was determined by HPLC analysis and the structure was confirmed by amino
acid analysis and mass spectrometry (LDI-MS).
Purity (HPLC): more than 95 %
Molecular weight (free base): 2894
Molecular formula: C137H217032N37
EXAMPLE 12
Preparation of KM PNIFSALGGGRLLYGATPYAIG (SEQ ID NO : 19), R I IPNIFTALSGGGRLLYGATPYAIG (SEQ ID NO : 20) and W11 P Nl F S A LGGAISYDLNTNILNCI (SEQ ID NO : 25). The peptides are synthesized in amide form, from the corresponding starting materials according to the general description of synthesis. The purity are determined by HPLC analysis and the structures are confirmed by amino acid analysis and mass spectrometry (LDI-MS).
EXAMPLE 13
Dimerisation via disulphide bridge.
The peptide sequences of the Examples 1 and 3 were linked via an oxidation step to form a dipeptide wherein the cysteine residues formed a disulphide bridge. The bridge was formed in either ways;
A) Oxidation with l2 Equal amounts of the peptides were dissolved in acetic acid/methanol (1:4) and 0.1 M l2 in methanol was added yielding a mixture of the dimer. or
B) Oxidation via [Cys(Spy)16]-SEQ ID NO : 2.2,3mM of the peptide of SEQ ID NO : 2 dissolved in 2 M AcOH (aq) and 2-propanol (1:1) was treated with 2,2 dithiodipyridin (3 eqv) to yield [Cys(Spy)16]-SEQ ID NO : 2. Equal ambunts of [Cys(Spy)16]-SEQ ID NO : 2

WO 00/52040 PCT/NO00/00075

and peptide of SEQ ID NO : 5 were dissolved in 10 mM NH4Oac (aq pH=6, 5) and methanol (5:2) to yield the dimer of SEQ ID NO : 21.
The purity of the peptide was determined by HPLC analysis and the peptide structure was confirmed by amino acid analysis. The peptide content (aminoacid free base) was 80%, Purity (HPLC): 92%.
EXAMPLE 14
A vaccine comprising the peptides of the SEQ ID NO : 3, 6,11 and 18 was prepared. The freeze-dried peptides were dissolved in sterile water at a final concentration of 4 mg/ml. The final salt concentration was 0,9 %. A preparation of a granulocyte-macrophage-colony stimulating factor (GM-CSF) was also prepared, according to the manufacturers directions for use, to a final concentration of 0.3 mg/ml. The two solutions are administered intracutaneously. A typical injection dose is 100 ml.
EXAMPLE 15
An antigen solution or suspension is mixed with equal parts of Freund's adjuvant of Behring, complete or incomplete, and is then finely emulsified by being drawn up into, and vigurously pressed out of, an injection syringe, or with a homogenator. The emulsion should remain stable for at least 30 minutes. The antigen-adjuvant emulsions is best injected subcutaneously as a depot.
EXAMPLE 16 Toxicity data.
The dipeptide of Example 13 was diluted in 0,9% NaCI to a test solution concentration of 4 mg/ml. The peptide was administered by injection to NMFI female mice in a dose of 100 ng per kg bodyweight. No toxicological effects were observed and the peptide was deemed not toxic.
Toxicity studies were performed in mice and rats on the peptide composition of the vaccine in Example 14. The mouse was selected for the study to provide comparative data from a second commonly used rodent species. The test substance was a mixture

WO 00/52040 PCT/NO00/00075

of four peptides supplied as one vial containing lyophilised material for reconstitution with physiological saline, and dose levels were expressed in terms of total peptide load. The individual peptides was present in ratio 1:1:1:1 giving dose levels of each peptide of 0.0075 mg/kg body weight, 0.075 mg/kg body weight and 0.75 mg/kg body weight, which are up to 500 fold the intended human dose. The test animals were divided into four groups often animals each (five males and five females); a saline control group and groups for low, intermediate and high doses. The test composition was administered once, by intravenous infusion into a tail vein at a dose rate of 3 ml/minute. The animals were killed at day 15 and 16 by intraperitoneal injection of sodium pentobarbitone.
The results of these studies indicated that the dose levels administered to the mice and rats elicited no adverse reactions and that the no effect level was in excess of 3 mg/kg.
EXAMPLE 17
Immunoassay for detection of antibodies induced by HIV-1.
The magnetic particle reagents are to be prepared according to the manufacturers recommended protocol. Dynal AS, is the manufacturer of the Dynabeads, which are employed. The magnetic particles coated with ligand are called Reagent 1. A peptide according to the invention is covalently coupled to the pre-activated surface of the magnetic particles. It is also possible to physically absorb the peptide to the surface of the magnetic particles. The concentration of particles in Reagent 1 is within the range from 1 mg/ml to 15 mg/ml. The particle size varies between 0,2 mm to 15 urn. The concentration of peptides is within the range from 0,01 mg/mg particle to 1 mg/mg particle.
The anti human Ig Alkaline Phosphatase (AP) conjugated antibody reagent is prepared according to the recommended protocol of Dako AS. This protocol is a standard procedure in this field. This reagent is called Reagent 2.
The substrate solution phenolphtalein-monophosphate is to be prepared according to the recommended protocol of Fluka AG. This protocol is a standard procedure in this field. The substrate solution is called Reagent 3.

WO 00/52040 PCT/NO00/00075

The washing and incubation buffer which is used is standard 0.05M tris-base buffer with the following additional compounds; Tween 20 (0,01% to 0,1%), glycerol (0,1% to 10%) and sodium chloride (0,2% to 0,1%).
The assay procedure comprises an incubation step wherein 1 drop of Reagent 1 is mixed with 2 drops of washing buffer in each well. After mixing, 30 ml of sample is added and the solution is incubated for 5 minutes. The magnetic particles can be trapped by a magnet and the liquid removed, before the magnet is separated. Then the wells are washed twice in 4 drops of washing solution, before incubation with Reagent 2. 1 drop of Reagent 2 is added with 2 drops of washing buffer and the solution is incubated for 5 minutes. The magnetic particles can be trapped by a magnet and the liquid removed, before the magnet is separated. Then the washing step is repeated before incubation with Reagent 3. 2 drops of Reagent 3 is added to each well and the solution is incubated for 3 minutes. The results can be read against a white background. Positive results are red (3+ = strong red) whereas negative results are clearly light yellow/brown solutions as obtained in the negative control.
The immunoassay kit could be used in detection of antibodies, induced either by HIV virus or HIV-specific peptides or proteins, for instance the peptides of the present invention.
The above Examples are only meant as illustrating the invention. It must be understood that a person skilled in the art can modify the peptides, antigens and vaccines herein described without deviating from the concept and scope of this invention as set forth in the claims.
The polypeptides of the invention can be used in a combination of at least one peptide selected from each group of sequences, SEQ ID NO : 1, SEQ ID NO : 4, SEQ ID NO : 9 and SEQ ID NO : 15 to form antigens and the the active principle of a prophylactic or therapeutic vaccine intended to provide protection against the human immunodeficiency virus type 1 (HIV-1). The vaccine may include compounds having beneficial effects in protecting or stimulating the host's immune system (human being or vertebrate animal) for instance interleukins, interferons, granulocyte macrophage growth factors, haematopoietic growth factors or similar. Preferably the vaccine

WO 00/52040 PCT/NO00/00075

composition further contain an adjuvant vehicle more preferably the adjuvant or
vehicle is Monophosphoryl Lipid A (MPL ®) possibly with alum, Freund*s adjuvant
(complete or incomplete) or aluminum hydroxyd. The optimal amount of
adjuvant/vehicle will depend 6n the type(s) which is chosen.
The peptide or vaccine formulation can be freeze-dried prior to storage. The vaccine
may be stored preferably at low temperature, in ampoules containing one or more
dosage units, ready for use. A typical dosage unit of the peptide according to the
invention is within the concentration range : 1 ng-1 mg per kg bodyweight, preferably
within 2 mg-0.15 mg per kg body Weight Persons skilled in the art will appreciate that a suitable dose will depend on the body weight of the pasient the type of disease,
seventy of condition, administration route and several other factors. The vaccine might be administered up to twelve times and through injection, typically it will be
administered about three times. In preparation of an injection solution the peptides are
dissolved in sterile sodium chloride solution at a final concentration of 1 mg/ml per
peptide and 0,9% sodium chloride. Typically an injection volume is 100ml to 200ml (2 x 100ml). the peptide is preferably co-administered with a suitable adjuvant and/or a
granulocyte-macrophage growth factor for instance Leucomax® «Shering Plough».
Suitable administration may be intracutane, subcutane, intravenous, peroral,
intramuscular, intranasal, mucosal or any other suitable route. Booster administrations
may be required in order to maintain protection. For persons skilled in the, art it will be
understood that the vaccine compositions according to the invention are useful hot only
in prevention of infection, but also in treatment of infection.

WE CLAIM;
1. Peptide derived from HIV gag p24 protein that comprises a modified amino acid sequence containing modifications compared to the native sequence and comprising the amino acid sequence:
Xaa1 Xaa2 Xaa3 Pro lie Pro Xaa7 Xaa8 Xaa9 Xaa10 Xaa11 Xaa12 [Gly]n Xaa13 Xaa14 Xaa15 Xaa16 Xaa17 Xaa18 Xaa19 Xaa20 Xaa21 Xaa22 Xaa23 Xaa24 (SEQ ID NO: 9)
wherein Xaa in position 1 is Asn, Ser, Gly, His, Ala, Pro, Arg or none,
Xaa in position 2 is Asn, Ala or Lys,
Xaa in position 3 is Pro, Gin, Gly, He or Leu,
Xaa in position 7 is Val or Ala,
Xaa in position 8 is Gly or Lys,
Xaa in position 9 is Glu, Asp, Lye, Phe or Thr,
Xaa in position 10 is He, Met, Val or Leu,
Xaa in position 11 is Tyr, Leu or none,
Xaa in position 12 is Ser or none,
Xaa in position 13 is Arg or none,
Xaa in position 14 is Asp, Arg, Trp, Ala or none,
Xaa in position 15 is lie or none,
Xaa in position 16 is Tyr or none,
Xaa in position 17 is Lys or Arg,
Xaa in position 18 is Arg, Lys or Asp,
Xaa in position 19 is Trp or Gly,
Xaa in position 20 is He, Met, Val, Gin or Ala,
Xaa in position 21 is He, Val or Ala,
Xaa in position 22 is Leu, Met or Val,
Xaa in position 23 is Gly or Cys,
Xaa in position 24 is Leu or none,


wherein the sequence of SEQ ID NO: 9 consists of at least six consecutive amino acids and n = 1, 2 or, and
the terminal ends of the sequences may be free carboxyl- or amino groups, amides, acyls, acetyls or salts thereof,
two or more of the Cys residues may form part of an intrachain- or interchain disulphide binding, a -S-(CH2)p-S- or a - (CH2) p-bridge wherein p = 1-8 optionally intervened by one or more heteroatoms such as O, N and S and/or the said peptide sequences are immobilized to a solid support.
2. Peptide as claimed in claim 1, wherein the amino acid sequence
of SEQ ID NO: 9 is selected from the groups of SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 and SEQ ID NO:
14.
3. Antigen, that comprises at least one peptide as claimed in claim 1.
4. Antigen as claimed in claim 3 that comprises at least one peptide selected from SEQ ID NO: 9.
5. Vaccine composition, that comprises an antigen as claimed in claim 3 with a pharmaceutically acceptable diluent and optionally an adjuvant, carrier and/or vehicle and optionally additional immunostimulatory compound(s).
6. Vaccine composition as claimed in claim 5 that comprises at least four peptides selected from each of the peptides of claim 1.
7. Vaccine composition as claimed in claim 6, that comprises the peptides of SEQ ID NO: 11.


8. Vaccine composition as claimed in one of claims 5-7 wherein the peptides are dissolved in a saline water solution and the optional immunostimulatory compound is a granulocyte macrophage growth factor.
9. Vaccine composition as claimed in one of claims 5-8 wherein the composition comprises an adjuvant selected from the group Monophosphoryl Lipid A (MPL®), Freund's complete or incomplete adjuvant or aluminium hydroxyd.
10. A method of detecting antibodies, induced by a HIV or HIV-specific peptides or proteins, in a sample of body fluid comprising subjecting the said sample to an immunoassay, wherein the antigen(s) is/ are selected from the peptides of claims 1 or 2.
11. An immunoassay kit for the detection of antibodies, induced by a HIV or HIV-specific peptides or proteins, in a sample of body fluid wherein the diagnostic antigen is a peptide of any one of claims 1 to 2.
12. Antibody that is capable of selectively reacting with the antigen of claims 3 or 4.

ABSTRACT
"HIV PEPTIDES OF SEQ ID NO 9, ANTIGENS, VACCINE COMPOSITIONS, IMMUNOASSAY KIT AND A METHOD OF DETECTING ANTIBODIES
INDUCED BY HIV"
Peptide derived from HIV gag p24 protein that comprises a modified amino
acid sequence containing modifications compared to the native sequence
and comprising the amino acid sequence:
Xaa1 Xaa2 Xaa3 Pro He Pro Xaa7 Xaa8 Xaa9 Xaa10 Xaa11 Xaa12 [Gly]n Xaa13
Xaa14 Xaa15 Xaa16 Xaa17 Xaa18 Xaa19 Xaa20 Xaa21 Xaa22 Xaa23 Xaa24 (SEQ ID
NO: 9)
wherein Xaa in position 1 is Asn, Ser, Gly, His, Ala, Pro, Arg or none,
Xaa in position 2 is Asn, Ala or Lys,
Xaa in position 3 is Pro, Gin, Gly, He or Leu,
Xaa in position 7 is Val or Ala,
Xaa in position 8 is Gly or Lys,
Xaa in position 9 is Glu, Asp, Lye, Phe or Thr,
Xaa in position 10 is He, Met, Val or Leu,
Xaa in position 11 is Tyr, Leu or none,
Xaa in position 12 is Ser or none,
Xaa in position 13 is Arg or none,
Xaa in position 14 is Asp, Arg, Trp, Ala or none,
Xaa in position 15 is lie or none,
Xaa in position 16 is Tyr or none,
Xaa in position 17 is Lys or Arg,
Xaa in position 18 is Arg, Lys or Asp,
Xaa in position 19 is Trp or Gly,
Xaa in position 20 is lie, Met, Val, Gin or Ala,
Xaa in position 21 is lie, Val or Ala,
Xaa in position 22 is Leu, Met or Val,
Xaa in position 23 is Gly or Cys,
Xaa in position 24 is Leu or none,
wherein the sequence of SEQ ID NO: 9 consists of at least six consecutive
amino acids and n = 1, 2 or, and
antigens, vaccine compositions, immunoassay kit and a method of detecting
antibodies induced by HIV.

Documents:

1391-MUMNP-2007-ABSTRACT(1-12-2011).pdf

1391-mumnp-2007-abstract(11-9-2007).pdf

1391-MUMNP-2007-ABSTRACT(21-5-2012).pdf

1391-MUMNP-2007-ABSTRACT(GRANTED)-(28-6-2012).pdf

1391-mumnp-2007-abstract.doc

1391-mumnp-2007-abstract.pdf

1391-MUMNP-2007-CANCELLED PAGES(21-5-2012).pdf

1391-MUMNP-2007-CLAIMS(AMENDED)-(1-12-2011).pdf

1391-MUMNP-2007-CLAIMS(AMENDED)-(21-5-2012).pdf

1391-mumnp-2007-claims(complete)-(11-9-2007).pdf

1391-MUMNP-2007-CLAIMS(GRANTED)-(28-6-2012).pdf

1391-mumnp-2007-claims.doc

1391-mumnp-2007-claims.pdf

1391-MUMNP-2007-CORRESPONDENCE(1-4-2010).pdf

1391-MUMNP-2007-CORRESPONDENCE(12-4-2012).pdf

1391-MUMNP-2007-CORRESPONDENCE(14-5-2009).pdf

1391-MUMNP-2007-CORRESPONDENCE(20-12-2011).pdf

1391-MUMNP-2007-CORRESPONDENCE(20-2-2009).pdf

1391-MUMNP-2007-CORRESPONDENCE(3-11-2010).pdf

1391-MUMNP-2007-CORRESPONDENCE(31-1-2012).pdf

1391-MUMNP-2007-CORRESPONDENCE(9-5-2012).pdf

1391-MUMNP-2007-CORRESPONDENCE(IPO)-(28-6-2012).pdf

1391-mumnp-2007-correspondence-others.pdf

1391-mumnp-2007-correspondence-received.pdf

1391-mumnp-2007-description (complete).pdf

1391-mumnp-2007-description(complete)-(11-9-2007).pdf

1391-MUMNP-2007-DESCRIPTION(GRANTED)-(28-6-2012).pdf

1391-MUMNP-2007-FORM 1(1-12-2011).pdf

1391-MUMNP-2007-FORM 1(20-2-2009).pdf

1391-mumnp-2007-form 13(14-5-2009).pdf

1391-mumnp-2007-form 13(20-2-2009).pdf

1391-MUMNP-2007-FORM 13(9-5-2012).pdf

1391-mumnp-2007-form 18(20-2-2008).pdf

1391-mumnp-2007-form 2(complete)-(11-9-2007).pdf

1391-MUMNP-2007-FORM 2(GRANTED)-(28-6-2012).pdf

1391-MUMNP-2007-FORM 2(TITLE PAGE)-(1-12-2011).pdf

1391-mumnp-2007-form 2(title page)-(complete)-(11-9-2007).pdf

1391-MUMNP-2007-FORM 2(TITLE PAGE)-(GRANTED)-(28-6-2012).pdf

1391-MUMNP-2007-FORM 3(1-4-2010).pdf

1391-MUMNP-2007-FORM 3(20-12-2011).pdf

1391-MUMNP-2007-FORM 3(3-11-2010).pdf

1391-mumnp-2007-form-1.pdf

1391-mumnp-2007-form-2.doc

1391-mumnp-2007-form-2.pdf

1391-mumnp-2007-form-3.pdf

1391-mumnp-2007-form-5.pdf

1391-MUMNP-2007-OTHER DOCUMENT(1-12-2011).pdf

1391-MUMNP-2007-OTHER DOCUMENT(1-4-2010).pdf

1391-MUMNP-2007-POWER OF ATTORNEY(1-12-2011).pdf

1391-mumnp-2007-power of authority(17-1-2008).pdf

1391-MUMNP-2007-REPLY TO EXAMINATION REPORT(1-12-2011).pdf

1391-MUMNP-2007-REPLY TO HEARING(21-5-2012).pdf

1391-mumnp-2007-sequence listing(30-1-2008).pdf


Patent Number 253156
Indian Patent Application Number 1391/MUMNP/2007
PG Journal Number 27/2012
Publication Date 06-Jul-2012
Grant Date 28-Jun-2012
Date of Filing 11-Sep-2007
Name of Patentee BIONOR IMMUNO AS
Applicant Address P.O. BOX 1893 GULSET, N-3703 SKIEN
Inventors:
# Inventor's Name Inventor's Address
1 BIRGER SORENSEN MEIERLIA 3, N-3727 SKIEN
PCT International Classification Number C07K14/16
PCT International Application Number PCT/NO00/00075
PCT International Filing date 2000-03-02
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 19991078 1999-03-04 Norway