WO2015086738A2 - Hiv vaccine - Google Patents

Hiv vaccine Download PDF

Info

Publication number
WO2015086738A2
WO2015086738A2 PCT/EP2014/077358 EP2014077358W WO2015086738A2 WO 2015086738 A2 WO2015086738 A2 WO 2015086738A2 EP 2014077358 W EP2014077358 W EP 2014077358W WO 2015086738 A2 WO2015086738 A2 WO 2015086738A2
Authority
WO
WIPO (PCT)
Prior art keywords
xaa
peptide
hiv
seq
composition according
Prior art date
Application number
PCT/EP2014/077358
Other languages
French (fr)
Other versions
WO2015086738A3 (en
Inventor
Lars H. HØIE
Anker LUNDEMOSE
Mats ÖKVIST
Arnt Ove HOVDEN
Maja Sommerfelt GRØNVOLD
Original Assignee
Bionor Immuno As
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bionor Immuno As filed Critical Bionor Immuno As
Publication of WO2015086738A2 publication Critical patent/WO2015086738A2/en
Publication of WO2015086738A3 publication Critical patent/WO2015086738A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus

Definitions

  • the present invention relates to novel peptides, compositions and methods for the treatment of HIV infections and AIDS.
  • the present invention relates to novel formulations of HIV-specific vaccine peptides.
  • the novel formulations may further be administered with other therapeutic agents, such as in combination with immunomodulatory compounds and/or reservoir purging agents, such as histone deacetylase (HDAC) inhibitors.
  • HDAC histone deacetylase
  • 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 cells.
  • 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.
  • 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.
  • 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.
  • HIV-l-specific 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-l-specific proliferative responses to p24 were inversely related to viral load. They conclude that the HIV-l-specific helper cells are likely to be important in immunotherapeutic interventions and vaccine development.
  • HIV-l-specific helper cells are likely to be important in immunotherapeutic interventions and vaccine development.
  • 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.
  • the present invention is based on the finding that a particular formulation of HIV- specific vaccine peptides, provides an effective method for treating HIV infections. Such combinations may also provide other advantageous effects particularly in relation to the properties of pharmaceutical compositions when formulated as a combination therapy. It is a further object of embodiments of the invention to enhance the effect of a therapeutic HIV vaccine by the use of immunomodulatory compounds and/or reservoir purging agents, such as histone deacetylase (HDAC) inhibitors in the prevention and/or treatment and/or eradication of HIV infection and AIDS.
  • HDAC histone deacetylase
  • HIV-specific vaccine peptides as a specific salt will provide better and improved vaccines.
  • HIV-specific peptide selected from the group of amino acid sequences:
  • Xaa in position 1 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
  • Xaa in position 9 is Leu or He
  • 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, and
  • Xaa in position 20 is Gly or Arg
  • 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 He, Leu, Val or Met
  • Xaa in position 4 is He, Val or Leu
  • Xaa in position 12 is Arg or 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, He, 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
  • n 0, 1, 2 or 3; Xaai Xaa 2 Xaa 3 Pro He Pro Xaa 7 Xaa 8 Xaa 9 Xaai 0 Xaau Xaa i2 [Gly] n Xaa i3 Xaa i4 Xaa i5 Xaai 6 Xaa i7 Xaa i8 Xaa i9 Xaa 2 o Xaa 2 i Xaa 22 Xaa 23 Xaa 24 (SEQ ID NO : 9) ;
  • 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 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 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 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
  • n 1, 2 or 3; and Xaai Xaa 2 He He Xaa 5 Xaa 6 Xaa 7 Xaa 8 Xaa 9 Leu Xaau [Gly] n [Arg] m Xaa i2 Xaa i3 Xaa ⁇ Xaa ⁇ Xaai 6 Xaa i7 Xaa i8 Xaai 9 Xaa 20 Xaa 2i Xaa 22 Xaa 23 Xaa 24 Xaa 25 (SEQ ID NO : 15) ;
  • 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
  • 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, He, Val or Asn
  • Xaa in position 23 is Asn, Tyr, Cys or Gly,
  • Xaa in position 24 is Thr, Met, He, Ala, Val or none,
  • Xaa in postion 25 is Gly or none
  • n 1, 2 or 3 and m 0, 1, 2 or 3 independent of each other; wherein the terminal ends of each HIV specific peptide may be free carboxyl- or amino- groups, amides, acyls or acetyls; and wherein each peptide is in the form of an acetate salt.
  • a composition comprising at least one HIV-specific peptide selected from the group of amino acid sequences of SEQ ID NOs: 1, 4, 9 and 15, as defined in the first embodiment;wherein the terminal ends of each HIV specific peptide may be free carboxyl- or amino- groups, amides, acyls or acetyls; and wherein each peptide is in the form of an acetate salt.
  • a peptide consisting of the amino acid sequence shown in SEQ ID NO : 18 (Vacc-10) in the form of an acetate salt.
  • a peptide consisting of the amino acid sequence shown in SEQ ID NO : 11 (Vacc-11) in the form of an acetate salt.
  • a peptide consisting of the amino acid sequence shown in SEQ ID NO : 6 (Vacc-12) in the form of an acetate salt.
  • a peptide consisting of the amino acid sequence shown in SEQ ID NO : 3 (Vacc-13) in the form of an acetate salt.
  • composition comprising one, two, three or four peptide acetate salts according to the invention.
  • an immunogenic composition according to the invention optionally comprising a pharmaceutically acceptable diluent, excipient, vehicle and/or one or more immunological adjuvants.
  • an immunoassay kit for the detection of antibodies induced by HIV or an HIV specific peptide or protein in a sample of body fluid wherein the kit comprises a peptide, or a composition according to the invention.
  • a method for reducing and/or delaying pathological effects of human immunodeficiency virus I (HIV) or for reducing the risk of developing acquired immunodeficiency syndrome (AIDS) in a human infected with HIV comprising administering an effective amount of a peptide, or a composition according to the invention.
  • HIV human immunodeficiency virus
  • HIV disease is composed of several stages including the acute HIV infection which often manifests itself as a flu-like infection and the early and medium stage symptomatic disease, which has several non-characteristic symptoms such as skin rashes, fatigue, night sweats, slight weight loss, mouth ulcers, and fungal skin and nail infections. Most HIV infected will experience mild symptoms such as these before developing more serious illnesses. It is generally believed that it takes five to seven years for the first mild symptoms to appear. As HIV disease progresses, some individuals may become quite ill even if they have not yet been diagnosed with AIDS (see below), the late stage of HIV disease.
  • AIDS is the late stage HIV disease and is a condition which progressively reduces the effectiveness of the immune system and leaves individuals susceptible to opportunistic infections and tumors.
  • Reducing and/or delaying pathological effect of HIV is in the present context meant to denote that use of the methods of the invention provides for a statistically significant reduction and/or delay in morbidity seen in individual infected with HIV which are treated according to the present invention. That is, the time of onset of manifest disease symptoms characterizing AIDS is later compared to non-treated controls and/or the number of pathological manifestations is reduced to controls not receiving the treatment of the present invention.
  • peptide is in the present context intended to mean both short peptides of from 2 to 10 amino acid residues, oligopeptides of from 11 to 100 amino acid residues, and polypeptides of more than 100 amino acid residues.
  • amino acids in peptides it is intended that the amino acids are L-amino acids, unless other information is provided.
  • a “variant” or “analogue” of a peptide refers to a peptide having an amino acid sequence that is substantially identical to a reference peptide, typically a native or “parent” polypeptide.
  • the peptide variant may possess one or more amino acid substitutions, deletions, and/or insertions at certain positions within the native amino acid sequence.
  • Constant amino acid substitutions are those in which an amino acid residue is replaced with an amino acid residue having a side chain with similar physicochemical properties. Families of amino acid residues having similar side chains are known in the art, and include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta- branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine
  • a "retro form" of a peptide is a form of a peptide where the order of the amino acids in N- to C-terminal direction has been inverted.
  • the retro form of ALDFR is the peptide RFDLA.
  • substantially identical in the context of two amino acid sequences means that the sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, at least about 95, at least about 98, or at least about 99 percent sequence identity.
  • residue positions that are not identical differ by conservative amino acid substitutions.
  • Sequence identity is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions.
  • the publicly available GCG software contains programs such as "Gap” and "BestFit” which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild-type protein and a mutein thereof. See, e.g. , GCG Version 6.1. Polypeptide sequences can also be compared using FASTA or ClustalW, applying default or recommended parameters.
  • FASTA e.g., FASTA2 and FASTA3
  • FASTA provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson, Methods Enzymol.
  • a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome-binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • operably linked means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • an "isolated" molecule is a molecule that is the predominant species in the composition wherein it is found with respect to the class of molecules to which it belongs (i.e., it makes up at least about 50% of the type of molecule in the composition and typically will make up at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or more of the species of molecule, e.g., peptide, in the
  • composition Commonly, a composition of an antibody molecule will exhibit 98% - 99% homogeneity for antibody molecules in the context of all present peptide species in the composition or at least with respect to substantially active peptide species in the context of proposed use.
  • treatment refers to preventing, alleviating, managing, curing or reducing one or more symptoms or clinically relevant manifestations of a disease or disorder, unless contradicted by context.
  • treatment of a patient in whom no symptoms or clinically relevant manifestations of a disease or disorder have been identified is preventive or prophylactic therapy, whereas "treatment” of a patient in whom symptoms or clinically relevant manifestations of a disease or disorder have been identified generally does not constitute preventive or prophylactic therapy.
  • antigen denotes a substance of matter which is recognized by the immune system's specifically recognizing components (antibodies, T-cells) .
  • immunogen is in the present context intended to denote a substance of matter, which is capable of inducing an adaptive immune response in an individual, where said adaptive immune response targets the immunogen.
  • an immunogen is an antigen, which is capable of inducing immunity.
  • epitope the region in an antigen or immunogen which is recognized by antibodies (in the case of antibody binding epitopes, also known as "B-cell epitopes") or by T-cell receptors when the epitope is complexed to an MHC molecule (in the case of T-cell receptor binding epitopes, i .e. "T-cell epitopes”) .
  • immunogenically effective amount has its usual meaning in the art, I.e. an amount of an immunogen, which is capable of inducing an immune response, which significantly engages pathogenic agents, which share immunological features with the immunogen.
  • vaccine is used for a composition comprising an immunogen and which is capable of inducing an immune response which is either capable of reducing the risk of developing a pathological condition or capable of inducing a therapeutically effective immune response which may aid in the cure of (or at least alleviate the symptoms of) a pathological condition.
  • pharmaceutically acceptable has its usual meaning in the art, I.e. it is used for a substance that can be accepted as part of a medicament for human use when treating the disease in question and thus the term effectively excludes the use of highly toxic substances that would worsen rather than improve the treated subject's condition.
  • T helper lymphocyte epitope (a T H epitope) is peptide, which binds an MHC Class II molecule and can be presented on the surface of an antigen presenting cell (APC) bound to the MHC Class II molecule.
  • An "immunological carrier” is generally a substance of matter which includes one or many T H epitopes, and which increase the immune response against an antigen to which it is coupled by ensuring that T-helper lymphocytes are activated and proliferate. Examples of known immunological carriers are the tetanus and diphtheria toxoids and keyhole limpet hemocyanin (KLH) .
  • adjuvant has its usual meaning in the art of vaccine technology, i.e. a substance or a composition of matter which is 1) not in itself capable of mounting a specific immune response against the immunogen of the vaccine, but which is 2) nevertheless capable of enhancing the immune response against the immunogen .
  • vaccination with the adjuvant alone does not provide an immune response against the immunogen
  • vaccination with the immunogen may or may not give rise to an immune response against the immunogen, but the combined vaccination with immunogen and adjuvant induces an immune response against the immunogen which is stronger than that induced by the immunogen alone.
  • One aspects of the present invention relates to a composition of at least one HIV- specific peptide as defined above.
  • peptides comprise an N- or C-terminal modification, such as an amidation, acylation, or acetylation.
  • suitable amides included those having the formula -C(0)-NR x R y , wherein R x and R y are independently selected from hydrogen and Ci- 6 alkyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH 3 , -CH 2 CH 3 and -CF 3 , a particular amide group which may be mentioned is -C(0)NH 2 .
  • suitable acetylated N-terminal ends include those of formula -NH-C(0)R z , wherein R z is hydrogen, Ci- 6 alkyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH 3 , -CH 2 CH 3 and -CF 3 , or phenyl .
  • the peptides are contemplated as vaccine agents or diagnostic agents, they are in certain embodiments coupled to a carrier molecule, such as an immunogenic carrier.
  • a carrier molecule such as an immunogenic carrier.
  • the peptides may thus be linked to other molecules either as recombinant fusions (e.g . via CLIP technology) or through chemical linkages in an oriented (e.g . using heterobifunctional cross-linkers) or nonoriented fashion.
  • Linking to carrier molecules such as for example diphtheria toxin, latex beads (convenient in diagnostic and prognostic embodiments), and magnetic beads (also convenient in diagnostic and prognostic embodiments), polylysine constructs etc, are all possible according to the invention.
  • the immunogenic carrier is conveniently selected from carrier proteins such as those conventionally used in the art (e.g . diphtheria or tetanus toxoid, KLH etc.), but it is also possible to use shorter peptides (T-helper epitopes) which can induce T-cell immunity in larger proportions of a population. Details about such T-helper epitopes can e.g . be found in WO 00/20027, which is hereby incorporated by reference herein - all immunolgic carriers and "promiscuous" (i .e. universal) T-helper epitopes discussed therein are useful as immunogenic carriers in the present invention.
  • carrier proteins such as those conventionally used in the art (e.g . diphtheria or tetanus toxoid, KLH etc.)
  • T-helper epitopes shorter peptides
  • Details about such T-helper epitopes can e.g . be found in WO
  • the carrier is a virus like particle, i .e. a particle sharing properties with virions without being infectious.
  • virus-like particles may be provided chemically (e.g . Jennings and Bachmann Ann. Rev. Pharmacol . Toxicol. 2009. 49 : 303-26 Immunodrugs: Therapeutic VLP-based vaccines for chronic diseases) or using cloning techniques to generate fusion proteins (e.g. Peabody et al. J. Mol. Biol . 2008; 380 : 252-63. Immunogenic display of diverse peptides on virus-like particles of RNA phage MS2) .
  • an immunogenic composition (such as a vaccine composition) comprising a composition of at least one HIV-specific peptides, in combination with a pharmaceutically acceptable diluent, excipient or vehicle and optionally one or more immunological adjuvant.
  • the at least one HIV-specific peptide is selected from the group of amino acid sequences of SEQ ID NOs: 1, 4, 9 and 15, as defined above; wherein the terminal ends of each HIV specific peptide may be free carboxyi- or amino- groups, amides, acyls or acetyls; and in the form of an acetate salt.
  • amino acid sequence of SEQ ID NO : 1 is selected from the group of SEQ ID NO: 2 and SEQ ID NO: 3.
  • amino acid sequence of SEQ ID NO : 4 is selected from the group of SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
  • amino acid sequence of SEQ ID NO : 9 is selected from the group of SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 and SEQ ID NO: 14.
  • amino acid sequence of SEQ ID NO : 15 is selected from the group of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and SEQ ID NO: 20.
  • the at least one HIV-specific peptide comprises at least, two, three, or four peptides selected from each of the groups of SEQ ID NO : 1, SEQ ID NO: 4, SEQ ID NO: 9 and SEQ ID NO: 15.
  • the at least one HIV-specific peptide consists of or comprises the peptides of SEQ ID NO: 3, SEQ ID NO: 6, SEQ ID NO: 11 and SEQ ID NO: 18.
  • immunogenic compositions include the use of state-of-the-art constituents such as immunological adjuvants. Apart from these adjuvants, which are detailed, by way of example, below, immunogenic compositions are prepared as generally taught in the art:
  • vaccines which contain peptide sequences as active ingredients is generally well understood in the art, as exemplified by U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, all incorporated herein by reference.
  • such vaccines are prepared as injectables either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared.
  • the preparation may also be emulsified.
  • the active immunogenic ingredient(s), such as each one peptide or composition comprising these peptides described herein is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient.
  • Suitable excipients are, for example, water, buffers, antioxidants, chelating agents, sugars, polyols, organic co-solvents, preservatives, isotonicity agents, stabilizers surfactants, saline, dextrose, glycerol, ethanol, ethylene glycol, propylene glycol, or the like, and combinations thereof.
  • the vaccine may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants which enhance the effectiveness of the vaccines; cf. the detailed discussion of adjuvants below.
  • excipients such as buffers, antioxidants, preservatives, isotonicity agents, stabilizers, chelating agents and surfactants in pharmaceutical compositions are well-known to the skilled person.
  • excipients such as buffers, antioxidants, preservatives, isotonicity agents, stabilizers, chelating agents and surfactants
  • Remington The Science and
  • Buffers suitable for use in pharmaceutical compositions include, but are not limited to, orhto-phosphate, TRIS, glycine, N-glycylglycine, citrate, sodium acetate, sodium carbonate, glycylglycine, histidine, lysine, arginine, sodium phosphate, and sodium citrate or mixtures thereof.
  • Antioxidants for use as excipients include thioredoxin, glutathione, L-Methionine, ascorbic acid. Ascorbic acid Ectoine, Glutathione, Monothioglycerol, Morin, Poly(ethylenimine), Propyl gallate and Vitamin E.
  • Preservatives for use in pharmaceutical compositions include, but are not limited to, phenol, m-cresol, methyl p-hydroxybenzoate, propyl p- hydroxybenzoate, 2-phenoxyethanol, butyl p-hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, or mixtures thereof.
  • Buffers suitable for use in pharmaceutical compositions include, but are not limited to, orhto-phosphate, TRIS, glycine, N-glycylglycine, citrate sodium acetate, sodium carbonate, glycylglycine, histidine, lysine, arginine, sodium phosphate, and sodium citrate or mixtures thereof.
  • the pharmaceutical composition of the invention may comprise an isotonicity agent, including but not limited to, sodium chloride, xylitol, mannitol, sorbitol, glycerol, glucose, maltose, sucrose, trehalose, raffinose, L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine, dimethyl sulphone, polyethyleneglycol, propylene glycol or mixtures thereof.
  • the compositions according to the invention may contain a stabilizer, for example selected from polyethylene glycol (e.g.
  • PEG 3350 polyvinylalcohol (PVA), polyvinylpyrrolidone, carboxymethylcellulose, different salts (e.g. sodium chloride), L-glycine, L-histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof.
  • PVA polyvinylalcohol
  • PVP polyvinylpyrrolidone
  • carboxymethylcellulose different salts (e.g. sodium chloride)
  • L-glycine L-histidine
  • imidazole imidazole
  • arginine arginine
  • lysine isoleucine
  • aspartic acid tryptophan
  • threonine mixtures thereof.
  • Compositions according to the invention may further comprise a chelating agent, such as ethlenediaminetetraacetic acid (EDTA), desferal, diethylenetriaminepentaacetic acid (DTPA), inositol hexaphosphate, ethylenediamine bis(o-hydroxyphenylacetic acid),
  • EDTA ethlenediaminetetraacetic acid
  • DTPA diethylenetriaminepentaacetic acid
  • inositol hexaphosphate ethylenediamine bis(o-hydroxyphenylacetic acid
  • Tris(hydroxymethyl)aminomethane TMS
  • tartaric acid citric acid and aspartic acid, and mixtures thereof.
  • compositions according to the invention may further comprise a surfactant, which may be selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, poloxamers, such as 188 and 407,
  • a surfactant which may be selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, poloxamers, such as 188 and 407,
  • polyoxyethylene sorbitan fatty acid esters polyoxyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g. Tween-20, or Tween-80), monoglycerides or
  • ethoxylated derivatives thereof diglycerides or polyoxyethylene derivatives thereof, glycerol, cholic acid or derivatives thereof, lecithins, alcohols and phospholipids, glycerophospholipids (lecithins, kephalins, phosphatidyl serine), glyceroglycolipids (galactopyransoide),
  • sphingophospholipids sphingomyelin
  • sphingoglycolipids ceramides, gangliosides
  • DSS docusate sodium, CAS registry no [577-11-7]
  • docusate calcium CAS registry no [128-49- 4]
  • docusate potassium CAS registry no [7491-09-0]
  • SDS sodium dodecyl sulfate or sodium lauryl sulfate
  • dipalmitoyl phosphatidic acid sodium caprylate, bile acids and salts thereof and glycine or taurine conjugates
  • ursodeoxycholic acid sodium cholate, sodium deoxycholate, sodium taurocholate
  • sodium glycocholate N-Hexadecyl-N,N-dimethyl-3- ammonio-l-propanesulfonate
  • anionic (alkyl-arylsulphonates) monovalent surfactants palmitoyl lysophosphatidyl
  • zwitterionic surfactants e.g. N-alkyl- N,N-dimethylammonio-l-propanesulfonates, 3-cholamido-l-propyldimethylammonio-l- propanesulfonate, dodecylphosphocholine, myristoyl lysophosphatidylcholine, hen egg lysolecithin
  • cationic surfactants quarternary ammonium bases
  • non-ionic surfactants e.g. N-alkyl- N,N-dimethylammonio-l-propanesulfonates, 3-cholamido-l-propyldimethylammonio-l- propanesulfonate, dodecylphosphocholine, myristoyl lysophosphatidylcholine, hen egg lysolecithin
  • polyethyleneoxide/polypropyleneoxide block copolymers Pluronics/Tetronics, Triton X-100, Dodecyl ⁇ -D-glucopyranoside
  • polymeric surfactants Tween-40, Tween-80, Brij-35
  • fusidic acid derivatives- e.g. sodium tauro-dihydrofusidate etc.
  • long-chain fatty acids and salts thereof C6-C12 eg.
  • acylcarnitines and derivatives Na- acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, Na-acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, Na-acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof.
  • the vaccines are conventionally administered parenterally, by injection, for example, either subcutaneously, intracutaneously, intradermal ⁇ , subdermally or intramuscularly.
  • Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral, nasal, buccal, sublinqual, intraperitoneal, intravaginal, anal, epidural, spinal, and intracranial formulations.
  • traditional binders and carriers may include, for example, polyalkalene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10% (w/w), preferably 1-2% (w/w).
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and may contain 10-95% (w/w) of active ingredient, preferably 25-70% (w/w).
  • the peptides may be formulated into a vaccine as neutral or salt forms.
  • Pharmaceutically acceptable salts include acid addition salts (formed with the free amino groups of the peptide) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and organic bases such as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • the vaccines are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective and immunogenic.
  • the quantity to be administered depends on the subject to be treated, including, e.g., the capacity of the individual's immune system to mount an immune response, and the degree of immunity desired.
  • Suitable dosage ranges are of the order of several hundred micrograms of active ingredient per vaccination with a preferred range from about 0.1 ⁇ g to 2,000 ⁇ g (even though higher amounts in the 1-10 mg range are contemplated), such as in the range from about 0.5 ⁇ g to 1,000 ⁇ g, preferably in the range from 1 ⁇ g to 500 ⁇ g and especially in the range from about 10 ⁇ g to 100 ⁇ g.
  • Suitable regimens for initial administration and booster shots are also variable but are typified by an initial administration followed by subsequent inoculations or other administrations.
  • immunogenic molecules described herein can therefore be formulated with adjuvants:
  • the adjuvants to be combined are known to induce humoral responses and include: i) Salt suspensions (e.g. varieties of salts containing aluminum ions or calcium ions), ii) Oil- in-water emulsions (e.g. varieties of squalane-based or squalene-based emulsions), iii) Water-in-oil emulsions (e.g. Montanide ISA51 or ISA720), iv) Neutral liposomes, v) Cationic liposomes, vi) Microspheres, vii) Immunostimulating complexes (e.g. ISCOMs or
  • Pattern-recognition receptor agonists e.g. agonists for C-type lectin receptors (CLRs), NOD-like receptors (NLRs), RIG-like helicases (RLHs), Triggering receptor expressed on myeloid cells (TREMs) and Toll-like receptors (TLRs)
  • Saponins i.e. Any saponin derived from Quillaja saponaria or Platycodon grandifiorum
  • Virosomes/Virus-like particles i.e. Cholera toxin, CTA1-DD or Esherichia coli heat-labile enterotoxin
  • vaccine antigenic properties they could be combined with a well known adjuvant with an oral immune modulant or adjuvant such as a Cox-2 inhibitor or a immunomodulating compound.
  • an oral immune modulant or adjuvant such as a Cox-2 inhibitor or a immunomodulating compound.
  • a further apect of the invention is the use of the vaccine combined with adjuvant, with one or more further therapeutic agents, such as an (oral) immunemodulating agent and/or a reservoir purging agent.
  • further therapeutic agents such as an (oral) immunemodulating agent and/or a reservoir purging agent.
  • cytokines such as interferons
  • monoclonal antibodies such as anti-PDl antibodies
  • cyclophosphamide Thalidomide
  • Levamisole Lenalidomide
  • a virus reservoir purging agent includes but is not limited to auranofin, IL-7, prostratin, bryostatin, HDAC inhibitors, such as vorinostat, Disulfiram and any suitable agent disclosed in any one of WO2013050422, WO2012051492 A3 and in Barton et al., Clinical Pharmacology & Therapeutics (2013); 93 1, 46-561, including but not limited to a NF-kappa- B-inducer selected from the group comprising : PMA, prostratin, bryostatin and TNF-alpha, and/or b) a histone deacetylase inhibitor selected from the different families (hydroxamates, cyclic peptides, aliphatic acids, and benzamides) including : TSA, SAHA, MS-275,
  • suitable immunomodulatory compounds or purging agents may be DNA methylation inhibitors selected from the two classes (non-nucleoside and nucleoside demethylating agents) including : 5-azacytidine (azacitidine), Sinefungin, 5-aza-2'- deoxycytidine (5-aza-CdR, decitabine), l-3-Darabinofuranosyl-5-azacytosine (fazarabine) and dihydro-5-azacytidine (DHAC), 5-fluorodeoxycytidine (FdC), oligodeoxynucleotide duplexes containing 2-H pyrimidinone, zebularine, antisense oligodeoxynucleotides (ODNs), MG98, (-)-epigallocatechin-3-gallate, hydralazine, procaine and procainamide.
  • 5-azacytidine azacitidine
  • Sinefungin 5-aza-2'- deoxycytidine
  • Suitable immunomodulatory compounds or purging agents to be used according to the present invention includes histone deacetylase inhibitor selected from the different families of HDACI (hydroxamates, cyclic peptides, aliphatic acids, and benzamides) including TSA, SAHA, MS-275, aminosuberoyl hydroxamic acids, M-Carboxycinnamic acid bishydroxamate, LAQ-824, LBH-589, belinostat (PXD-101 ), Panobinostat (LBH-589), a cinnamic hydroxamic acid analogue of M-carboxycinnamic acid bishydroxamate, IF2357, aryloxyalkanoic acid hydroxamides, depsipeptide, apicidin, cyclic hydroxamic acid-containing peptide group of molecules, FK-228, red FK, cyclic peptide mimic linked by an aliphatic chain to a hydroxamic acid, buty
  • Suitable immunomodulatory compounds or purging agents to be used according to the present invention includes histone methyltransferase inhibitors (chaetocin and BIX-01294); Inhibitors of Enhances of Zeste 2 (EZH2) - such as 3-deazaneplanocin A (DZNep) used alone or in combination with other classes of immunomodulatory compounds or purging agents.
  • histone methyltransferase inhibitors chaetocin and BIX-01294
  • Inhibitors of Enhances of Zeste 2 (EZH2) - such as 3-deazaneplanocin A (DZNep) used alone or in combination with other classes of immunomodulatory compounds or purging agents include histone methyltransferase inhibitors (chaetocin and BIX-01294); Inhibitors of Enhances of Zeste 2 (EZH2) - such as 3-deazaneplanocin A (DZNep) used alone or in combination with other classes of immunomodulatory compounds or purging agents
  • Suitable adjuvants includes response-selective C5a agonists, such as EP54 and EP67 described in Hung CY et al.
  • An agonist of human complement fragment C5a enhances vaccine immunity against Coccidioides infection.
  • Vaccine (2012) and Kollessery G et al. Tumor-specific peptide based vaccines containing the conformationally biased, response- selective C5a agonists EP54 and EP67 protect against aggressive large B cell lymphoma in a syngeneic murine model.
  • Vaccine (2011) 29: 5904-10 are examples of response-selective C5a agonists, such as EP54 and EP67 described in Hung CY et al.
  • An agonist of human complement fragment C5a enhances vaccine immunity against Coccidioides infection.
  • Vaccine (2012) and Kollessery G et al. Tumor-specific peptide based vaccines containing the conformationally biased, response- selective C5a agonists EP54 and EP67 protect against aggressive large B
  • the at least one HIV-specific peptide may be administered in combination with one or more further thereapeutically active agents, such as agents for the treatment and or prevention of HIV and/or AIDS.
  • One aspect of the invention is the use of the vaccine combined with adjuvant, with one or more further therapeutic agents, such as an (oral) immunemodulating agent and/or a reservoir purging agent.
  • further therapeutic agents such as an (oral) immunemodulating agent and/or a reservoir purging agent.
  • therapeutic agent such as “immunomodulating agent” or virus reservoir purging agent as used herein, includes but is not limited to cytokines, such as interferons, monoclonal antibodies, such as ant-PDl antibodies, cyclophosphamide, Thalidomide,
  • a virus reservoir purging agent includes but is not limited to auranofin, IL-7, prostratin, bryostatin, HDAC inhibitors, such as vorinostat, and Disulfiram, and the further agents described herein.
  • ART antiretroviral therapy
  • Latently infected resting CD4+ cells carry transcriptionally silent HIV-1 and represent the predominant reservoir of HIV-1 infection.
  • Other cells may also act as reservoirs (Reviewed in Alexaki et al., 2008, Curr. HIV Res. 6:388-400) such as macrophages, dendritic cells and astrocytes (where HIV-1 infection occurs via a CD4-independent mechanism). It is these latent reservoirs that represent the major challenge to eradication of HIV-1 infection.
  • Approaches towards eradication include attempts to purge reservoirs by selective activation of latently infected cells (such as memory cells) in the presence of ART such that released virus may not infect and replicate in neighbouring cells (Richman et al., 2009, Science 323 : 1304-1307).
  • Agents include histone deacetylase inhibitors, cytokines, such as IL-2 and IL-7, as well as bryostatin, the protein kinase C activator (Kovochich et al ., 2011, PLoS ONE 6 (4) :el8270).
  • Therapeutic vaccines have the advantage of being able to penetrate sanctuary sites less well accessed by ART such as lymphoid tissue (Pantaleo et al., 1991, Proc. Natl. Acad. Sci. USA 88 :9838-42; Fox et al ., 1991, J. Infect. Dis. 164 : 1051-57) and the central nervous system (Alexaki et al., 2008, Curr. HIV Res. 6 :388-400), that represent regions for viral persistence. This relates to therapeutic interventions targeting both the virus itself as well as HIV-associated immune activation.
  • Particular examples include, but are not limited to, the substituted 2-(2,6-dioxopiperidin-3-yl)phthalimides and substituted 2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindoles as described in US 6281230 and US 6316471.
  • Monocyte/macrophage function is part of the Innate Immune System that serves as a first line of defense against an infection. By modulating the host's monocytes and macrophages, immunomodulatory compounds can change the dynamics of the response to a viral infection, such as influenza.
  • Histone deacetylases are a class of enzymes that remove acetyl groups from N-acetylated lysines amino acid on histone proteins.
  • HDACs are a class of enzymes that remove acetyl groups from N-acetylated lysines amino acid on histone proteins.
  • Class I includes HDACs 1, 2, 3, and 8 which are found primarily in the nucleus.
  • Class II HDACs HDACs 4, 5, 6, 7 9, and 10 are found primarily in the cytoplasm but may be able to shuttle between the nucleus and the cytoplasm; class Ila comprises four HDACs (HDACs 4, 5, 7 and 9) while class lib comprises two HDACs (HDACs 6 and 10) which are expressed only in the cytoplasm.
  • HDAC11 which is ubiquitously expressed, shares sequence similarities with both class I and class II HDACs and represents Class IV.
  • Class III also called "sirtuin family” groups NAD+-dependent proteins which do not act primarily on histone
  • the at least one HIV-specific peptide may be administered in combination, such as in a composition comprising an immunomodulatory compound and/or a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor.
  • a histone deacetylase (HDAC) inhibitor such as a histone deacetylase (HDAC) inhibitor.
  • the immunomodulatory compounds may be selected from anti-PDl antibodies, such as MDX-1106 (Merck), THALOMID ® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene), and immunomodulatory compound described in any one of WO2007028047, WO2002059106, and WO2002094180.
  • anti-PDl antibodies such as MDX-1106 (Merck), THALOMID ® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene)
  • the immunomodulatory compound may be selected from 4-(amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-l,3-dione and 3-(4-amino-l-oxo-l,3- dihydro-isoindol-2-yl)-piperidine-2,6-dione.
  • the immunomodulatory compound is lenalidomide.
  • the immunomodulatory compound may be enantiomerically pure.
  • the reservoir purging agent such as a histone deacetylase (HDAC) inhibitor
  • HDAC histone deacetylase
  • M344 (4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide), chidamide (CS055/HBI-800), 4SC-202, (4SC), Resminostat (4SC), hydroxamic acids such as vorinostat (SAHA), belinostat (PXD101), LAQ824, trichostatin A and panobinostat (LBH589);
  • benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), cyclic tetrapeptides (such as trapoxin, such as trapoxin B), and the depsipeptides, such as romidepsin (ISTODAX), electrophilic ketones, and the aliphatic acid compounds such as phenylbutyrate, valproic acid, Oxamflatin, ITF2357 (generic givinostat), Apicidin, MC1293, CG05, and CG06; compounds that activate transcription factors including NF-KappaB, Prostratin, auranofin, bryostatin, a nontumorigenic phorbol ester, DPP (12-deoxyphorbol-13- phenylacetate), PMA, and Phorbol 12-myristate 13-acetate (PMA); Compounds that activate HIV mRNA elongation including P-TEF-b kinase and hex
  • the components of the at least one HIV-specific peptide and/or the one or more further thereapeutically active agents may be administered simultaneously, sequentially or separately in any order.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising one, two or more components of the at least one HIV-specific peptide and/or the one or more further thereapeutically active agents optionally in combination with one or more pharmaceutically acceptable adjuvants, diluents or carriers.
  • the invention also provides a combination product comprising of components of the at least one HIV-specific peptide and/or the one or more further thereapeutically active agents, wherein each of component is formulated in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier.
  • the combination product may be either a single (combination) pharmaceutical formulation or a kit-of-parts. In a kit-of-parts some or all of the components may be formulated separately and may each be provided in a form that is suitable for administration in conjunction with the other(s).
  • the component(s) may also be provided for use, e.g. with instructions for use, in combination with one or more further component(s) as defined above.
  • the peptides for use in the invention may be produced synthetically using art recognised methods. Further details for the synthetic production of such peptides are found in the Examples. Alternatively the peptides may be produced recombinantly. When recombinantly producing the peptides for use in the invention by means of transformed cells, it is convenient, although far from essential, that the expression product is either exported out into the culture medium or carried on the surface of the transformed cell.
  • this stable cell line which carries the vector of the invention and which expresses the nucleic acid fragment of the invention.
  • this stable cell line secretes or carries the peptide expression product, thereby facilitating purification thereof.
  • plasmid vectors containing replicon and control sequences which are derived from species compatible with the host cell are used in connection with the hosts.
  • the vector ordinarily carries a replication site, as well as marking sequences which are capable of providing phenotypic selection in transformed cells.
  • E. coli is typically transformed using pBR322, a plasmid derived from an E. coli species (see, e.g., Bolivar et a/. , 1977).
  • the pBR322 plasmid contains genes for ampicillin and tetracycline resistance and thus provides easy means for identifying transformed cells.
  • the pBR plasmid, or other microbial plasmid or phage must also contain, or be modified to contain, promoters which can be used by the prokaryotic microorganism for expression.
  • promoters most commonly used in recombinant DNA construction include the ⁇ -lactamase (penicillinase) and lactose promoter systems (Chang et a/. , 1978; Itakura et a/. , 1977; Goeddel et a/., 1979) and a tryptophan (trp) promoter system (Goeddel et a/. , 1979; EP-A-0 036 776). While these are the most commonly used, other microbial promoters have been discovered and utilized, and details concerning their nucleotide sequences have been published.
  • eukaryotic microbes such as yeast cultures may also be used, and also here the promoter should be capable of driving expression.
  • Saccharomyces cerevisiase, or common baker's yeast is the most commonly used among eukaryotic microorganisms, although a number of other strains are commonly available.
  • the plasmid YRp7 for example, is commonly used (Stinchcomb et a/., 1979; Kingsman et a/. , 1979; Tschemper et a/. , 1980).
  • Suitable promoting sequences in yeast vectors include the promoters for 3- phosphoglycerate kinase (Hitzman et a/. , 1980) or other glycolytic enzymes (Hess et a/., 1968; Holland et a/., 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
  • the termination sequences associated with these genes are also incorporated into the expression vector 3' of the sequence desired to be expressed to provide polyadenylation of the mRNA and termination.
  • promoters which have the additional advantage of transcription controlled by growth conditions are the promoter region for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, and the aforementioned glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization.
  • Any plasmid vector containing a yeast-compatible promoter, origin of replication and termination sequences is suitable.
  • cultures of cells derived from multicellular organisms may also be used as hosts.
  • any such cell culture is workable, whether from vertebrate or invertebrate culture.
  • useful host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cell lines, and W138, BHK, COS-7 293, Spodoptera frugiperda (SF) cells, Drosophila melanogaster cell lines (such as Schneider 2 (S 2 )), and MDCK cell lines.
  • Expression vectors for such cells ordinarily include (if necessary) an origin of replication, a promoter located in front of the gene to be expressed, along with any necessary ribosome binding sites, RNA splice sites, polyadenylation site, and transcriptional terminator sequences.
  • the control functions on the expression vectors are often provided by viral material.
  • commonly used promoters are derived from polyoma, Adenovirus 2, and most frequently Simian Virus 40 (SV40).
  • SV40 Simian Virus 40
  • the early and late promoters of SV40 virus are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 viral origin of replication (Fiers et al., 1978).
  • Smaller or larger SV40 fragments may also be used, provided there is included the approximately 250 bp sequence extending from the Hindlll site toward the Bgll site located in the viral origin of replication. Further, it is also possible, and often desirable, to utilize promoter or control sequences normally associated with the desired gene sequence, provided such control sequences are compatible with the host cell systems.
  • An origin of replication may be provided either by construction of the vector to include an exogenous origin, such as may be derived from SV40 or other viral (e.g., other Polyoma viruses, Adeno, VSV, BPV) or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • an exogenous origin such as may be derived from SV40 or other viral (e.g., other Polyoma viruses, Adeno, VSV, BPV) or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • nucleic acid vaccines can also be administered intraveneously and intraarterially.
  • nucleic acid vaccines can be administered by use of a so-called gene gun and/or by use of electroporation, and hence also these and equivalent modes of administration are regarded as part of the present invention.
  • nucleic acid fragment is introduced in the form of a vector wherein expression is under control of a viral promoter.
  • vectors according to the invention cf. the discussion above.
  • detailed disclosures relating to the formulation and use of nucleic acid vaccines are available, cf. Donnelly JJ et a/, 1997, Annu. Rev. Immunol. 15: 617-648 and Donnelly JJ et al., 1997, Life Sciences 60: 163- 172. Both of these references are incorporated by reference herein.
  • peptide immunogens or nucleic acid immunogens is the use of live immunogen technology. This entails administering a non-pathogenic microorganism which has been transformed with a nucleic acid fragment or a vector of the present invention.
  • the non-pathogenic microorganism can be any suitable attenuated bacterial strain (attenuated by means of passaging or by means of removal of pathogenic expression products by recombinant DNA technology), e.g. Mycobacterium bovis BCG., non-pathogenic
  • the nucleic acid fragment of the invention can be incorporated in a non-virulent viral vaccine vector such as a vaccinia strain or any other suitable poxvirus.
  • the non-pathogenic microorganism or virus is administered only once to a subject, but in certain cases it may be necessary to administer the microorganism/virus more than once in a lifetime in order to maintain protective immunity. It is even contemplated that immunization schemes as those detailed above for polypeptide vaccination will be useful when using live or virus vaccines.
  • live or virus immunization is combined with previous or subsequent polypeptide and/or nucleic acid immunization.
  • a HIV-specific peptide selected from the group of amino acid sequences:
  • 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
  • Xaa in position 9 is Leu or He
  • 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, and
  • Xaa in position 20 is Gly or Arg; Xaai Xaa 2 Xaa 3 Xaa 4 Xaa 5 Gly Leu Asn Pro Leu Val [Gly] n Xaa i2 Xaa i3 Tyr Xaa i5 Pro Xaa i7 Xaa i8 He Leu Xaa 2 i Xaa 22 (SEQ ID NO : 4)
  • 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 He, Leu, Val or Met
  • Xaa in position 4 is He, Val or Leu
  • Xaa in position 12 is Arg or Lys
  • Xaa in postion 13 is Met or Leu
  • Xaa in position 15 is Ser, Cys or Gin
  • Xaa in position 17 is Thr, Val, He, 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
  • n 0, 1, 2 or 3;
  • 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, lie 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 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 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 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
  • n 1,,2 or 3
  • 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
  • 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, He, Val or Asn
  • Xaa in position 23 is Asn, Tyr, Cys or Gly,
  • Xaa in position 24 is Thr, Met, He, Ala, Val or none,
  • Xaa in postion 25 is Gly or none
  • each HIV specific peptide may be free carboxyl- or amino groups, amides, acyls, acetyls; and wherein each peptide is in the form of an acetate salt.
  • 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.
  • amino acid sequence of SEQ ID NO: 15 is selected from the groups of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and SEQ ID NO: 20.
  • at least one HIV-specific peptide comprises at least , two, three, or four peptides selected from each of the groups of SEQ ID NO: 1, SEQ ID NO: 4, SEQ ID NO: 9 and SEQ ID NO: 15.
  • a peptide consisting of the amino acid sequence shown in SEQ ID NO : 18 (Vacc-10) in the form of an acetate salt.
  • acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 6% and 16%, such as between 7% and 15%, such as between 8% and 14%, such as between 9% and 14%, such as between 9% and 13%, such as between 10% and 14%, such as between 11% and 14%.
  • a peptide consisting of the amino acid sequence shown in SEQ ID NO : 11 (Vacc-11) in the form of an acetate salt.
  • the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 5% and 16%, such as between 5% and 15%, such as between 5% and 14%, such as between 6% and 14%, such as between 6% and 13%, such as between 7% and 12%, such as between 7% and 11%, such as between 8% and 11%, such as between 9% and 11%.
  • the acetate content of the salt is between 3% and 18%, such as between 3% and 17%, such as between 3% and 16%, such as between 3% and 15%, such as between 3% and 14%, such as between 3% and 13%, such as between 3% and 11%, such as between 3% and 10%, such as between 4% and 10%, such as between 4% and 9%, such as between 4% and 8%, such as between 4% and 7%, such as between 4% and 6%, such as between 4% and 5%.
  • composition comprising one, two, three or four peptide acetate salts according to any one of embodiments 1 to 16.
  • composition according to embodiment 17, which comprises said four peptide acetate salts defined in embodiments 9, 11, 13 and 15.
  • composition according to embodiment 21, wherein said liquid is water.
  • the pH of said composition is between between 3 and 8, such as between 3.5 and 7.5, such as between 4 and 7, such as between 4 and 6, such as between 5 and 6, and the osmolality is between 10 and 100, or between 15 and 25, or between 16 and 24 mOsm/kg.
  • immunogenic composition optionally comprising a pharmaceutically acceptable diluent, one or more excipients, such as buffers, antioxidants, preservatives, isotonicity agents, stabilizers, chelating agents and surfactants, a vehicle and/or one or more immunological adjuvants.
  • excipients such as buffers, antioxidants, preservatives, isotonicity agents, stabilizers, chelating agents and surfactants
  • composition according to embodiment 27 for use in the treatment or prophylaxis of HIV or AIDS.
  • composition according to any one of embodiments 17-28 comprising one or more further therapeutically active agents selected from an immunomodulatory compound and a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor.
  • a reservoir purging agent such as a histone deacetylase (HDAC) inhibitor.
  • composition according to embodiment 29, wherein the immunomodulatory compound is selected from anti-PDl antibodies, such as MDX-1106 (Merck), THALOMID® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene), and immunomodulatory compounds described in any one of WO2007028047, WO2002059106, and WO2002094180.
  • anti-PDl antibodies such as MDX-1106 (Merck), THALOMID® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene)
  • composition according to embodiment 30, wherein the immunomodulatory compound is lenalidomide.
  • composition according to embodiment 29, wherein the reservoir purging agent is selected from M344 (4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide), chidamide (CS055/HBI-800), 4SC-202, (4SC), Resminostat (4SC), hydroxamic acids such as vorinostat (SAHA), belinostat (PXDIOI), LAQ824, trichostatin A and panobinostat (LBH589); benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), cyclic tetrapeptides (such as trapoxin, such as trapoxin B), and the depsipeptides, such as romidepsin (ISTODAX), electrophilic ketones, and the aliphatic acid compounds such as phenylbutyrate, valproic acid, Oxamflatin, ITF2357 (gene
  • composition according to embodiment 32, wherein the reservoir purging agent is romidepsin.
  • kits for the detection of antibodies induced by HIV or an HIV specific peptide or protein in a sample of body fluid comprising a peptide according to any of embodiments 1 to 16, or a composition according to any one of embodiments 17-33.
  • HIV immunodeficiency virus I
  • AIDS immunodeficiency syndrome
  • a human infected with HIV the method comprising administering an effective amount of a peptide according to any of embodiments 1 to 16, or a composition according to any one of embodiments 17-33.
  • HDAC histone deacetylase
  • HIV-SPECIFIC PEPTIDES FOR USE ACCORDING TO THE INVENTION
  • the present invention involves the use of HIV-specific peptides based on conserved regions of HIV gag p24, antigens in free or carrier-bound form comprising at least one of the said peptides.
  • the HIV-specific peptides for use according to the invention originate from the four different conserved areas of the HIV-1 core protein p24, having the properties of maintaining the uniqueness (sensitivity and specificity) of the HIV-l-epitope. Further these peptides possess no recognized cytotoxic T lymphocyte (CTL) antagonistic effect and have at least one potential CTL epitope.
  • CTL cytotoxic T lymphocyte
  • HIV-specific peptides for use according to the invention, which have met the above criteria are selected from the group of amino acid sequences of SEQ ID NOs : 1, 4, 9 and 15, as defined above; wherein the terminal ends of each HIV specific peptide may be free carboxyl- or amino- groups, amides, acyls or acetyls; or acetate salts of any of the HIV specific peptides.
  • the HIV-specific peptide sequences have the potential to serve as a particularly good antigen wherein the antigen comprises at least one peptide selected from the group of sequences of SEQ 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 may comprise two or more polypeptide sequences which are either linked by a bridge for instance a disulphide bridge between the Cys residues of the chains or bridges like Ci-C 8 alkylene possibly intervened by one or more heteroatoms like 0, 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 HIV-specific 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 HIV-specific peptide sequences could deviate from the natural sequences by modification of the terminal NH 2 -group and/or COOH-group, they may for instance be acylated, acetylated, amidated or salts thereof; or modified to provide a binding site for a carrier or another molecule.
  • suitable amides included those having the formula -C(0)-NR x R y , wherein R x and R y are independently selected from hydrogen and Ci- 6 al kyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH 3 , -CH 2 CH 3 and -CF 3 , a particular amide group which may be mentioned is -C(0)NH 2 .
  • suitable acetylated N-terminal ends include those of formula -NH-C(0)R z , wherein R z is hydrogen, Ci- 6 al kyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH 3 , -CH 2 CH 3 and -CF 3 , or phenyl .
  • HIV-specific peptides for use according to the invention consist of 6 to 50 amino acids, preferably between 10 and 30 amino acids. They cover all natural variation of amino acids in the identified positions.
  • the polypeptide antigen for use 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 carriers. 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.
  • 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
  • the HIV specific peptides for use according to the present invention comprises antigens containing the peptides of the SEQ ID NOs: 1, 4, 9 and 15, more preferreably the peptides occur in the ratio 1 : 1 : 1 : 1 w/w.
  • HIV specific peptides for use according to the invention comprise the following : RALGPAATLQTPWTASLGVG (SEQ ID NO: 3)
  • RFIIPNIFTALSGGRRALLYGATPYAIG (SEQ ID NO: 18) (Nl in position 6 is Norleucine) or salts thereof, particularly acetate salts.
  • 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.
  • 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.
  • 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.
  • the amino acid derivatives were supplied by Bachem AG, Switzerland.
  • the peptides described herein preferably have a free amino group at the N-terminus and an amidated C-terminus.
  • the counter ion of all peptides described herein is acetate which is bound in ionic form to charged functional groups (i.e. guanidino side chains arginine and the ⁇ -amino groups of lysine [Vacc-11] and the side chains of arginine [Vacc-10, Vacc-12 and Vacc-13]). All amino acid residues except the achiral glycine are in the L-configuration.
  • the peptides described herein were assembled on tricyclic amide linker resins utilising an 9- fluorenylmethyloxycarbonyl (Fmoc) strategy.
  • Steps 3-9 Washing. Each step consists of addition of solvents/reagents, stirring at room temperature and filtration.
  • the peptide resin is treated with cold TFA in the presence of deionised water and 1, 2- Ethanedithiol (EDT), (Vacc-10 and Vacc-13) or triisopropylsilane (TIS) (Vacc-11 and Vacc-12) for approximately two to three hours at room temperature.
  • EDT 2- Ethanedithiol
  • TIS triisopropylsilane
  • the peptide is precipitated in diisopropyl ether (IPE). It is then filtered off, washed with IPE and dried in a desiccator under reduced pressure.
  • ACN acetonitrile
  • UV ultraviolet
  • the last stage of manufacture of Vacc-4x acetate is the exchange from the TFA salt, obtained in stage three, into the acetate salt by ion exchange.
  • the lyophilised material from one or several combined preparative HPLC runs is dissolved in varying concentrations of acetic acid or in purified water according to the properties of the individual peptides.
  • the dissolved peptide is loaded onto the ion exchange resin (acetate form) and equilibrated with 5% acetic acid (or 20% purified water for Vacc-13).
  • the elution is performed with 5% acetic acid (or purified water for Vacc-13), checked by thin-layer chromatography (TLC), filtered through a 0.2 ⁇ membrane filter and lyophilised to yield the final product as a white to off-white powder.
  • 5% acetic acid or purified water for Vacc-13
  • TLC thin-layer chromatography
  • Vacc-4x formulation only optionally contain an ionic excipients
  • the peptides and their counter ions (acetate) account for a certain osmolality.
  • the range of 10 - 100 mOsm/kg was defined based on the result obtained for the technical sample. Potential variability due to the four peptides is taken into account.
  • For the drug product approximately 1 mg of each of the four Vacc-4x peptides was used.
  • the lyophilisate is reconstituted with 0.30 mL of WFI. Taking the acetic acid contents of the peptides listed in table 1 into account, the acetic acid content of Vacc-4x is approximately 0.40 mg in 0.30 mL of solution.
  • the theoretical osmolality is approximately 23 mOsmol/L by calculation, which correlates well with the values determined in the Vacc-4x batches (20-23 mOsmol/kg).
  • 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). Preparation of RALG PAATLQTPWTAS LGVG (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).
  • 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
  • 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
  • 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
  • the peptide was synthesized in amide form, from the corresponding starting materials according to the general description of synthesis. The purity was determined by
  • 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
  • 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).
  • 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).
  • 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).
  • a vaccine comprising the peptides of the SEQ ID NOs: 3, 6, 11 and 18 was prepared (also refered to herein as Vacc-4x).
  • 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 ⁇ .
  • An antigen solution or suspension is mixed with equal parts of Freund ' s adjuvant of
  • the emulsion should remain stable for at least 30 minutes.
  • the antigen-adjuvant emulsions is best injected subcutaneously as a depot.
  • mice and rats on the peptide composition of the vaccine in Example 2 were selected for the study to provide comparative data from a second commonly used rodent species.
  • the test substance was a mixture 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 w/w 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.
  • test animals were divided into four groups of ten 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.
  • mice and rats elicited no adverse reactions and that the no effect level was in excess of 3 mg/kg .
  • 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 ⁇ to 15 ⁇ .
  • 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.
  • 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 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 polypeptides of the invention can be used in a combination of at least one peptide selected from each group of sequences, SEQ ID NOs: 1, 4, 9 and 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.
  • the vaccine composition further contain an adjuvant or vehicle, more preferably the adjuvant or vehicle is Monophosphoryl Lipid A (MPL ®) possibly with alum, Freund ' s adjuvant (complete or incomplete) or aluminum hydroxide.
  • MPL ® Monophosphoryl Lipid A
  • the optimal amount of adjuvant/vehicle will depend on 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 ⁇ g-lmg per kg bodyweight, preferably within 2 ⁇ g-0.15 mg per kg body weight.
  • 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.
  • peptides are dissolved in sterile sodium chloride solution at a final concentration of 1 mg/ml per peptide and 0,9% sodium chloride.
  • an injection volume is 100 ⁇ to 200 ⁇ (2 x 100 ⁇ ) .
  • 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.
  • Vacc-4x immune response resulting from Vacc-4x immunization could in combination with ART potentially improve immune reconstitution in patients who have not fully regained a healthy CD4 level (>600xl0 5 /L) -
  • Potential benefits of Vacc-4x in subjects with incomplete immune reconstitution include a possible sustained improvement in the immune response to p24 and HIV.
  • Potential risks include the discomfort and inconvenience associated with the immunizations and the risk of known or unknown side effects of exposure to Vacc-4x and Leukine (rhu-GM-CSF) including, most commonly, local reactions at the site of injections and fatigue (likelihood not yet determined) .
  • mice and rats The results of non-clinical single-dose studies in mice and rats indicate that the dose levels of intravenous Vacc-4x elicited no adverse reactions and that the no effect level was in excess of 3 mg/kg, which constitutes a 500 fold safety margin over the planned human dose level .
  • Vacc-4x has no limiting toxicology in a model that is relevant to the proposed clinical study.
  • the therapeutic vaccine candidate Vacc-4x has been studies in a Phase I and three Phase II clinical studies.
  • the Phase I study enrolled 11 HIV-positive subjects, including nine subjects on ART. Subjects were maintained on ART (if entered on ART) ; all subjects were treated with 12 immunizations of Vacc-4x at a dose of 0.4 mg/injection over a period of 26 weeks. Immunizations were performed following injection of rhu-GM-CSF (Leucomax®
  • DTH delayed- type hypersensitivity
  • ELISPOT enzyme-linked immunosorbent spot assay
  • the Phase II dose-finding study (CTN B-HIV 2/2001) enrolled 40 HIV positive subjects, of which 38 completed the trial. Subjects were maintained on ART and treated with 10 immunizations at a dose of 0.4 mg (20 subjects) or 1.2 mg (20 subjects) per Vacc 4x injection, over a period of 26 weeks. Immunizations with Vacc 4x were performed following injection of rhu-GM-CSF (Leucomax [molgramostim]) as a local adjuvant. ART was interrupted from Week 26 to Week 30 to allow exposure to the subject's own virus
  • ART autologous immunization
  • ART was resumed from Week 30 to Week 38 to allow maturation of immune responses to the Vacc 4x peptides and to the subject's own virus.
  • ART was discontinued from Week 38 to Week 52 when the study was formally concluded.
  • Treatment- related AEs were observed in 20 subjects (8 subjects in the 0.4mg group and 12 subjects in the 1.2mg group).
  • No SAEs were reported during the period of immunization.
  • One subject experienced a transient vasovagal reaction in conjunction with immunization and the DTH test at Week 26 and Week 38.
  • a second subject experienced a vasovagal reaction in conjunction with the DTH test at Week 52.
  • vital signs, and performance status no changes attributable to immunization were observed. Changes in HIV RNA, CD4 cell counts, and CD8 cell counts showed no safety concerns related to
  • CTN BI Vacc-4x/2009/l was an open-label follow-up of study CTN B-HIV- 2/2001 to detemine whether a re-boost with Vacc-4x could reactivate or increase the immune response obtained during the immunization performed in the CTN B-HIV-2/2001 study.
  • the secondary objectives were to evaluate: the in vivo immunogenicity of Vacc-4x by evaluation of DTH and to compare the DTH response to DTH in the initial study; the effect of Vacc-4x on CD4 counts, CD8 counts and HIV viral RNA; and the safety and tolerability of Vacc-4x. All 26 subjects included in the study received two booster administrations of Vacc-4x.
  • the Phase II Study CT-BI Vacc-4x 2007/1 (EudraCT Number 2007-006302-13) was performed in US and Europe (UK, Germany, Spain and Italy).
  • the study was a randomized, double-blind, multicenter, immunogenicity study of Vacc-4x versus placebo in patients infected with HIV-1 who have maintained an adequate response to ART.
  • the primary objective was to evaluate the effect of Vacc-4x immunizations versus placebo on CD4 counts, T-cell function (ELISPOT, T-cell proliferative responses and intracellular cytokine staining) and the response to interruption of ART.
  • ELISPOT T-cell proliferative responses and intracellular cytokine staining
  • the study was actually planned to include approximately 345 subjects. Due to recruitment difficulties the study was amended to reduce the planned number of subjects to a minimum of 120 subjects. Based on this FDA responded to approval of the amendment "Due to the size (N 130) and nature (no formal hypothesis testing) of this study, please understand that CBER will regard this study as exploratory and for hypothesis-generating purpose only" Based on this it was important to look more in detail into the data by including post-hoc analyses especially related to pre-ART viral load since the two groups showed very different pre-ART viral load values.
  • Vacc-4x did not reduce the proportion of subjects requiring resumption of ART after ART cessation at Week 28 in comparison with placebo. There was also no effect compared with placebo on the percentage change in CD4 count between Week 28 and the last CD4 assessment before resumption of ART. The time to restarting ART was similar in Vacc-4x and placebo-treated subjects.
  • LOCF Long Observation Carried Forward
  • DSMB Data Safety Monitoring Board
  • Expansion of polyfunctional HIV-specific T-cells upon stimulation with Dendritic Cells, pre-incubated with peptides to be used according to the invention, may be studied by methods described by Keersmaecker et al. (J. Virol., 2012 86:9351-9360) and referenced therein, HIV proteins Gag or Nef, they are incubated with peptides to be used according to the invention , before they are used to stimulate T-cells in a co-culture. Keersmaecker et al.
  • IMiDs Lidomide (IMiD3; CC- 5013) and pomalidomide (IMiDl ; CC-4047) during in vitro T-cell stimulation with dendritic cells presenting Gag- or Nef-specific peptides, resulted in a number of improvements in the function of the T-cells.
  • Immunizations (four primary immunizations and two booster immunizations) at Weeks 1, 2, 3 and 4, and booster immunizations at Weeks 12 and 13 with either:
  • Peptide composition 0.6, 0.9, 1.2 and 1.5 mg (Equimolar amount of each peptide)
  • Lenalidomide 5,10, and 25 mg.
  • CC-5013 Subjects randomized to the Lenalidomide (CC-5013) arm will take a single oral dose of Lenalidomide (CC-5013) daily the two preceding days before immunization with the Peptide composition and on the day of each immunization.
  • the Peptide composition used according to this clinical trial setup consists of SEQ ID NO:3, SEQ ID NO:6, SEQ ID NO: ll, and SEQ ID NO: 18.
  • panobinostat LH589
  • days 1, 3, and 5 i.e. 3 times a week
  • ART will be interrupted to evaluate the effect of study treatment on virological control.
  • Enrolment into this part of the study will be optional and determined by the effect of study treatments on the latent HIV-1 reservoir. (Maximum duration of treatment interruption : 16 weeks).
  • Depletion of the viral reservoir as a result of the combination treatments according to the present invention may be quantified by for instance following the procedures set forth in Lehrman et al. (The Lancet (366), 2005, pp. 549-555) and references there in. In brief, this includes measuring in samples of patient blood obtained before, during and after treatment; p24 expression from stimulated latently infected cells, plasma HIV RNA concentration (viral load), and integrated HIV DNA by realtime PCR analysis.
  • Dendritic cells were generated from monocytes isolated from buffy coat preparations from healthy blood donors. Briefly, peripheral blood mononuclear cells were separated by a density gradient centrifugation and the monocytes were then negatively isolated using the Dynabeads Untouched Human Monocytes (Invitrogen, Carlsbad, CA) following the manufacturer's instructions. The monocytes were cultured with IL-4 (20 ng/ml; Immunotools, Friesoythe; Germany) and GM-CSF (100 ng/ml; Immunotools) in X-VIV015 medium (Lonza, Basel, Switerland) for 5-6 days to generate immature DC. Cytokines were replenished every 2-3 days.
  • the maturation of the cells was performed for 24 hours with IFN-Y (1000 IU/ml), TNF-a (50 ng/ml), IL- ⁇ (25 ng/ml) IFN-a (3000 IU/ml).
  • the DC were pulsed for 2 hours at 37°C with peptides at 10 ⁇ g/ml, before extensive washing and co-culture with Peripheral blood mononuclear cells (PBMC) labelled with a fluorescent dye (VPD450, BD biosciences, Sam Jose, CA).
  • PBMC Peripheral blood mononuclear cells
  • IL-2 50U/ml
  • IL-7 50ng/mL
  • wells with or without IMiDs were added at the start of co-culture.
  • the level of T cell proliferation was analysed by flow cytometry.
  • the supernatants from the co-culture wells were investigated with Luminex technology to establish any suppressor activity.
  • peptides according to the invention used in the following examples were synthesized by Schafer-N as c-terminal amides using the Fmoc-strategy of Sheppard, (1978) J.Chem.Soc, Chem. Commun., 539.
  • 96-well U-bottom polystyrene plates (NUNC, cat no: 163320) were used for staining of human PBMCs. Briefly, 8ul of N- or C-terminally biotinylated peptides according to the invention (i .e. 5mM, 2.5mM & 1.25mM tested for each peptide) were incubated at 37°C for 2h with 40ul of PBMC (12.5 x 106 cells/ml) from blood donors.
  • Dendritic cells were then washed 3x with 150ul PermWash, followed by resuspension in staining buffer (BD, cat no : 554656) before flow cytometry.
  • Dendritic cells were gated as CDllc+ events outside lymphocyte region (i.e. higher FSC & SSC signals than lymphocytes). 200 000 total cells were acquired on a FACSCanto II flow cytometer with HTS loader, and histograms for both total cells & dendritic cells with respect to peptide-fluorescence (i.e. GeoMean) were prepared.
  • 96-well U-bottom polystyrene plates (NUNC, cat no: 163320) were used for staining of human PBMCs. Briefly, 8ul of N- or C-terminally biotinylated peptides according to table 1 or table 2 (i .e. 5mM, 2.5mM & 1.25mM tested for each peptide; all peptides manufactured by solid phase synthesis by commercial suppliers) were incubated at 37°C for 2h with 40ul of PBMC (12.5 x 106 cells/ml) from blood donors.
  • NUNC 96-well U-bottom polystyrene plates
  • Cells were then washed 3x with 150ul of Cellwash (BD, cat no: 349524), then stained with Streptavidin-APC (BD, cat no: 554067) & Anti-hCDl lc (eBioscience, cat no: 12-0116) according to manufacturer at 4°C for 30 min aiming to visualize biotinylated peptides & dendritic cells, respectively.
  • Cells were then washed 3x with 150ul of Cellwash (BD, cat no: 349524), followed by resuspension in staining buffer (BD, cat no: 554656) before flow cytometry.
  • Dendritic cells were gated as CDl lc+ events outside lymphocyte region (i.e.
  • the data are geomean-value of each testet peptide, as calculated by the FACS Duva software.
  • Positive CTL response may alternatively be assayed by ELISPOT assay.
  • Human IFN-gamma cytotoxic T-cell (CTL) response by ELISPOT assay Briefly, at day 1, PBMC samples from HCV patients were incubated in flasks
  • PBMCs/cm2 for 2h at 37°C, 5% C02 in covering amount of culture media (RPMI 1640 Fisher Scientific; Cat No. PAAE15-039 supplemented with L- Glutamine, (MedProbe Cat. No. 13E17-605E, 10% Foetal Bovine serum (FBS), Fisher Scientific Cat. No. A15-101) and Penicillin/Streptomycin, (Fisher Acientific Cat. No. Pll-010) in order to allow adherence of monocytes.
  • Non-adherent cells were isolated, washed, and frozen in 10% V/V DMSO in FBS until further usage.
  • Adherent cells were carefully washed with culture media, followed by incubation at 37°C until day 3 in culture media containing 2 ⁇ g/ml final concentration of hrGM-CSF (Xiamen amoytop biotech co, cat no: 3004.9090.90) & ⁇ / ⁇ hrIL-4 (Invitrogen, Cat no: PHC0043) and optionally an immunomodulationg agent (IMiD), and this procedure was then repeated at day 6.
  • hrGM-CSF Xiamen amoytop biotech co, cat no: 3004.9090.90
  • IMD immunomodulationg agent
  • PBMC samples from blood donors were thawed, washed with warm medium and incubated in flasks (250000PBMCs/cm2) for 24 hours at 37°C, 5% C02 in covering amount of culture media (RPMI 1640 with ultra-glutamine, Lonza, BE12-702F701; 10% Foetal Bovine serum (FBS), Fisher Scientific Cat. No. A15-101 ; Penicillin/Streptomycin, Fisher Scientific Cat. No. Pll-010) to allow the cells to recover after thawing.
  • FBS Foetal Bovine serum
  • Fisher Scientific Cat. No. A15-101 Penicillin/Streptomycin, Fisher Scientific Cat. No. Pll-010
  • the cells were added to a Falcon Microtest Tissue Culture plate, 96well flat bottom, at 500 000 cells per well in a volume of 200 ⁇ total medium.
  • Parallel wells were added the indicated stimuli in duplicate and optionally an immunomodulationg agent (IMiD), or left with medium as a control for 6 days at 37°C, 5% C0 2 .
  • IMD immunomodulationg agent
  • ⁇ of the cell suspension were transferred to an ELISPOT (Millipore multiscreen HTS) plate coated with ⁇ g/ml native influenza M2e protein. After a 24 hour incubation, the plate was washed four times with PBS + 0,05% Tween20, and a fifth time with PBS, 200 ⁇ / ⁇ .
  • a mouse Anti- human IgG or IgM biotin (Southern Biotech 9040-08 and 9020-08) was diluted in PBS with 0.5% FBS and incubated for 90 minutes at 37°C. The washing was repeated as described, before 80 ⁇ Streptavidin-Alkaline-Phosphatase (Sigma Aldrich, S289) was added each well and incubated at 60 minutes in the dark, at room temperature. The wells were then washed 2 times with PBS + 0.05% Tween20 and 4 times with PBS, ⁇ / ⁇ , before the substrate, Vector Blue Alkaline Phosphatase Substrate kit III (Vector Blue, SK-5300 ) was added and let to develop for 7 minutes at room temperature. The reaction was stopped with running water, the plates let dry and the sport enumerated by an ELISPOT reader (CTL-ImmunoSpot® S5 UV Analyzer) .
  • Plates are then washed 3x with WB, followed by lh incubation at 37°C with 50-70 ⁇ /well of added human (or rabbit or sheep) sera (serial dilutions ranging from 1 : 5 - 1 : 250 in dilution buffer (PBS + 1% v/v Triton-X100 + 1% w/v BSA; denoted DB)) . Plates are then washed 6x with WB, followed by lh incubation at RT with 70 ⁇ /well of Alkaline Phosphatase-conjugated Protein G in DB; Calbiochem 539305) or goat anti-mouse IgG biotin ( ⁇ / ⁇ , Southern Biotech, 1030-08.
  • the plates were washed one extra step as described, before addition of ⁇ Streptavidin-Alkaline-Phosphatase ( ⁇ / ⁇ , Sigma Aldrich, S289) and incubated 1 hour at RT. Plates are then washed 6x with WB, followed by 10-60 min incubation at room temperature with 100 ⁇ /well of 0.3% w/v of
  • Phenophtalein monophosphate (Sigma P-5758) . Plates are finally quenched by adding 100 ⁇ /well of Quench solution (0.1 M TRIS + 0.1M EDTA + 0.5M NaOH + 0.01% w/v NaN 3 ;

Abstract

The present invention relates to novel compositions of active agents and methods for the treatment of HIV infection and AIDS. In particular, the present invention relates to novel compositions of HIV-specific vaccine peptides.

Description

HIV VACCINE
FIELD OF THE INVENTION
The present invention relates to novel peptides, compositions and methods for the treatment of HIV infections and AIDS. In particular, the present invention relates to novel formulations of HIV-specific vaccine peptides. The novel formulations may further be administered with other therapeutic agents, such as in combination with immunomodulatory compounds and/or reservoir purging agents, such as histone deacetylase (HDAC) inhibitors. BACKGROUND OF THE INVENTION
In recent years a large body of research evidence has accumulated supporting the concept that AIDS is an immunological disease induced by HIV-1 rather than simply being caused by loss of CD4+ T-lymphocytes as a result of chronic cytopathic viral infection. It is therefore important to develop interventions that target the chronic immune stimulation induced by HIV-1 as well as the virus itself, (Sommerfelt 2011, Recent Translational Research in HIV/AIDS Ed. Dr. Yi-Wei Tang. ISBN 979-953-307-189-2. pp 493-510).
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 cells. 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 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 titers (Zwart G., et al. Virology, 201, p. 285-93, June 1994), 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 HIV-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-l-specific proliferative responses to p24 were inversely related to viral load. They conclude that the HIV-l-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 al., 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 (gpl20 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 gpl60. 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.
International Patent Application WO00/52040 discloses methods for treating HIV infections by administering e.g. HIV specific peptides based on conserved regions of HIV gag p24.
There is a need to provide improved treatments for the HIV infections and AIDS.
OBJECT OF THE INVENTION
It is an object of embodiments of the invention to provide effective peptides, combination of such peptides, formulations, and methods which can be used in the prevention and/or treatment of HIV infection and AIDS.
The present invention is based on the finding that a particular formulation of HIV- specific vaccine peptides, provides an effective method for treating HIV infections. Such combinations may also provide other advantageous effects particularly in relation to the properties of pharmaceutical compositions when formulated as a combination therapy. It is a further object of embodiments of the invention to enhance the effect of a therapeutic HIV vaccine by the use of immunomodulatory compounds and/or reservoir purging agents, such as histone deacetylase (HDAC) inhibitors in the prevention and/or treatment and/or eradication of HIV infection and AIDS.
SUMMARY OF THE INVENTION
It has been found that HIV-specific vaccine peptides as a specific salt will provide better and improved vaccines.
So, in a first aspect of the present invention there is provided a HIV-specific peptide selected from the group of amino acid sequences:
Xaai Xaa2 Xaa3 Xaa4 Xaa5 Xaa6 Ala Xaa8 Xaa9 Gin Thr Pro Trp Xaai4 Xaai5 Xaai6 Xaai7 Xaais Val Xaa20 (SEQ ID NO: 1);
wherein Xaa in position 1 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,
Xaa in position 9 is Leu or He,
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, and
Xaa in position 20 is Gly or Arg;
Xaai Xaa2 Xaa3 Xaa4 Xaa5 Gly Leu Asn Pro Leu Val [Gly]n Xaai2 Xaai3 Tyr Xaai5 Pro Xaai7 Xaai8 He Leu Xaa2i Xaa22 (SEQ ID NO: 4);
wherein 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 He, 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 or 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, He, 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, and
n = 0, 1, 2 or 3; Xaai Xaa2 Xaa3 Pro He Pro Xaa7 Xaa8 Xaa9 Xaai0 Xaau Xaai2 [Gly]n Xaai3 Xaai4 Xaai5 Xaai6 Xaai7 Xaai8 Xaai9 Xaa2o Xaa2i 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, Lys, 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 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 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,
n = 1, 2 or 3; and Xaai Xaa2 He He Xaa5 Xaa6 Xaa7 Xaa8 Xaa9 Leu Xaau [Gly]n [Arg]m Xaai2 Xaai3 Xaa^ Xaa^ Xaai6 Xaai7 Xaai8 Xaai9 Xaa20 Xaa2i Xaa22 Xaa23 Xaa24 Xaa25 (SEQ ID NO : 15) ;
wherein 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,
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, He, Val or Asn,
Xaa in position 23 is Asn, Tyr, Cys or Gly,
Xaa in position 24 is Thr, Met, He, Ala, Val or none,
Xaa in postion 25 is Gly or none,
n 1, 2 or 3 and m = 0, 1, 2 or 3 independent of each other; wherein the terminal ends of each HIV specific peptide may be free carboxyl- or amino- groups, amides, acyls or acetyls; and wherein each peptide is in the form of an acetate salt. In a second aspect is provided a composition comprising at least one HIV-specific peptide selected from the group of amino acid sequences of SEQ ID NOs: 1, 4, 9 and 15, as defined in the first embodiment;wherein the terminal ends of each HIV specific peptide may be free carboxyl- or amino- groups, amides, acyls or acetyls; and wherein each peptide is in the form of an acetate salt.
In a third aspect is provided a peptide consisting of the amino acid sequence shown in SEQ ID NO : 18 (Vacc-10) in the form of an acetate salt.
In a further aspect is provided a peptide consisting of the amino acid sequence shown in SEQ ID NO : 11 (Vacc-11) in the form of an acetate salt.
In a further aspect is provided a peptide consisting of the amino acid sequence shown in SEQ ID NO : 6 (Vacc-12) in the form of an acetate salt.
In a further aspect is provided a peptide consisting of the amino acid sequence shown in SEQ ID NO : 3 (Vacc-13) in the form of an acetate salt.
In a further aspect is provided a composition comprising one, two, three or four peptide acetate salts according to the invention.
In a further aspect is provided an immunogenic composition according to the invention optionally comprising a pharmaceutically acceptable diluent, excipient, vehicle and/or one or more immunological adjuvants. In a further aspect is provided an immunoassay kit for the detection of antibodies induced by HIV or an HIV specific peptide or protein in a sample of body fluid, wherein the kit comprises a peptide, or a composition according to the invention.
In a further aspect is provided a method for reducing and/or delaying pathological effects of human immunodeficiency virus I (HIV) or for reducing the risk of developing acquired immunodeficiency syndrome (AIDS) in a human infected with HIV, the method comprising administering an effective amount of a peptide, or a composition according to the invention. DETAILED DISCLOSURE OF THE INVENTION
Definitions
When terms such as "one", "a" or "an" are used in this disclosure they mean "at least one", or "one or more" unless otherwise indicated. Further, the term "comprising" is intended to mean "including" and thus allows for the presence of other constituents, features, conditions, or steps than those explicitly recited.
"HIV" unless otherwise indicated generally denotes human immunodeficiency virus
I.
"HIV disease" is composed of several stages including the acute HIV infection which often manifests itself as a flu-like infection and the early and medium stage symptomatic disease, which has several non-characteristic symptoms such as skin rashes, fatigue, night sweats, slight weight loss, mouth ulcers, and fungal skin and nail infections. Most HIV infected will experience mild symptoms such as these before developing more serious illnesses. It is generally believed that it takes five to seven years for the first mild symptoms to appear. As HIV disease progresses, some individuals may become quite ill even if they have not yet been diagnosed with AIDS (see below), the late stage of HIV disease. Typical problems include chronic oral or vaginal thrush (a fungal rash or spots), recurrent herpes blisters on the mouth (cold sores) or genitals, ongoing fevers, persistent diarrhea, and significant weight loss. "AIDS" is the late stage HIV disease and is a condition which progressively reduces the effectiveness of the immune system and leaves individuals susceptible to opportunistic infections and tumors.
"Reducing and/or delaying pathological effect of HIV" is in the present context meant to denote that use of the methods of the invention provides for a statistically significant reduction and/or delay in morbidity seen in individual infected with HIV which are treated according to the present invention. That is, the time of onset of manifest disease symptoms characterizing AIDS is later compared to non-treated controls and/or the number of pathological manifestations is reduced to controls not receiving the treatment of the present invention.
The term "peptide" is in the present context intended to mean both short peptides of from 2 to 10 amino acid residues, oligopeptides of from 11 to 100 amino acid residues, and polypeptides of more than 100 amino acid residues. When referring to amino acids in peptides, it is intended that the amino acids are L-amino acids, unless other information is provided.
A "variant" or "analogue" of a peptide refers to a peptide having an amino acid sequence that is substantially identical to a reference peptide, typically a native or "parent" polypeptide. The peptide variant may possess one or more amino acid substitutions, deletions, and/or insertions at certain positions within the native amino acid sequence.
"Conservative" amino acid substitutions are those in which an amino acid residue is replaced with an amino acid residue having a side chain with similar physicochemical properties. Families of amino acid residues having similar side chains are known in the art, and include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta- branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). A particular form of conservative amino acid substitutions include those with amino acids, which are not among the normal 20 amino acids encoded by the genetic code. Since preferred embodiments of the present invention entail use of synthetic peptides, it is unproblematic to provide such "non-naturally occurring" amino acid residues in the peptides disclosed herein, and thereby it is possible to exchange the natural saturated carbon chains in the side chains of amino acid residues with shorter or longer saturated carbon chains - for instance, lysine may be substituted with an amino acid having an the side chain -(CH2)nNH3, where n is different from 4, and arginine may be substituted with an amino acid having the side chain -(CH2)nNHC( = NH2) N H2, where n is different from 3, etc. Similarly, the acidic amino acids aspartic acid and glutamic acid may be substituted with amino acid residues having the side chains -(CH2)nCOOH, where n>2.
A "retro form" of a peptide is a form of a peptide where the order of the amino acids in N- to C-terminal direction has been inverted. For instance, the retro form of ALDFR is the peptide RFDLA.
The term "substantially identical" in the context of two amino acid sequences means that the sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, at least about 95, at least about 98, or at least about 99 percent sequence identity. In one embodiment, residue positions that are not identical differ by conservative amino acid substitutions. Sequence identity is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions and other modifications, including conservative amino acid substitutions. For instance, the publicly available GCG software contains programs such as "Gap" and "BestFit" which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild-type protein and a mutein thereof. See, e.g. , GCG Version 6.1. Polypeptide sequences can also be compared using FASTA or ClustalW, applying default or recommended parameters. A program in GCG Version 6.1., FASTA (e.g., FASTA2 and FASTA3) provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 1990; 183 :63-98; Pearson, Methods Mol. Biol. 2000; 132: 185-219). Another preferred algorithm when comparing a sequence to a database containing a large number of sequences from various organisms, or when deducing the is the computer program BLAST, especially blastp, using default parameters. See, e.g. , Altschul et al., J. Mol. Biol. 1990;215:403-410; Altschul et al., Nucleic Acids Res.
1997;25:3389-402 (1997); each herein incorporated by reference. "Corresponding" amino acid positions in two substantially identical amino acid sequences are those aligned by any of the protein analysis software mentioned herein, typically using default parameters.
A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome-binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Generally,
"operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
An "isolated" molecule is a molecule that is the predominant species in the composition wherein it is found with respect to the class of molecules to which it belongs (i.e., it makes up at least about 50% of the type of molecule in the composition and typically will make up at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or more of the species of molecule, e.g., peptide, in the
composition). Commonly, a composition of an antibody molecule will exhibit 98% - 99% homogeneity for antibody molecules in the context of all present peptide species in the composition or at least with respect to substantially active peptide species in the context of proposed use.
In the context of the present invention, "treatment" or "treating" refers to preventing, alleviating, managing, curing or reducing one or more symptoms or clinically relevant manifestations of a disease or disorder, unless contradicted by context. For example, "treatment" of a patient in whom no symptoms or clinically relevant manifestations of a disease or disorder have been identified is preventive or prophylactic therapy, whereas "treatment" of a patient in whom symptoms or clinically relevant manifestations of a disease or disorder have been identified generally does not constitute preventive or prophylactic therapy.
The term antigen denotes a substance of matter which is recognized by the immune system's specifically recognizing components (antibodies, T-cells) .
The term "immunogen" is in the present context intended to denote a substance of matter, which is capable of inducing an adaptive immune response in an individual, where said adaptive immune response targets the immunogen. In other words, an immunogen is an antigen, which is capable of inducing immunity.
The terms "epitope", "antigenic determinant" and "antigenic site" are used interchangeably herein and denotes the region in an antigen or immunogen which is recognized by antibodies (in the case of antibody binding epitopes, also known as "B-cell epitopes") or by T-cell receptors when the epitope is complexed to an MHC molecule (in the case of T-cell receptor binding epitopes, i .e. "T-cell epitopes") .
The term "immunogenically effective amount" has its usual meaning in the art, I.e. an amount of an immunogen, which is capable of inducing an immune response, which significantly engages pathogenic agents, which share immunological features with the immunogen.
The term "vaccine" is used for a composition comprising an immunogen and which is capable of inducing an immune response which is either capable of reducing the risk of developing a pathological condition or capable of inducing a therapeutically effective immune response which may aid in the cure of (or at least alleviate the symptoms of) a pathological condition.
The term "pharmaceutically acceptable" has its usual meaning in the art, I.e. it is used for a substance that can be accepted as part of a medicament for human use when treating the disease in question and thus the term effectively excludes the use of highly toxic substances that would worsen rather than improve the treated subject's condition.
A "T helper lymphocyte epitope" (a TH epitope) is peptide, which binds an MHC Class II molecule and can be presented on the surface of an antigen presenting cell (APC) bound to the MHC Class II molecule. An "immunological carrier" is generally a substance of matter which includes one or many TH epitopes, and which increase the immune response against an antigen to which it is coupled by ensuring that T-helper lymphocytes are activated and proliferate. Examples of known immunological carriers are the tetanus and diphtheria toxoids and keyhole limpet hemocyanin (KLH) .
The term "adjuvant" has its usual meaning in the art of vaccine technology, i.e. a substance or a composition of matter which is 1) not in itself capable of mounting a specific immune response against the immunogen of the vaccine, but which is 2) nevertheless capable of enhancing the immune response against the immunogen . Or, in other words, vaccination with the adjuvant alone does not provide an immune response against the immunogen, vaccination with the immunogen may or may not give rise to an immune response against the immunogen, but the combined vaccination with immunogen and adjuvant induces an immune response against the immunogen which is stronger than that induced by the immunogen alone.
Specific aspects and embodiments of the invention
One aspects of the present invention relates to a composition of at least one HIV- specific peptide as defined above.
In certain embodiments, peptides comprise an N- or C-terminal modification, such as an amidation, acylation, or acetylation. When the C-terminal end of a peptide is an amide, suitable amides included those having the formula -C(0)-NRxRy, wherein Rx and Ry are independently selected from hydrogen and Ci-6 alkyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH3, -CH2CH3 and -CF3, a particular amide group which may be mentioned is -C(0)NH2. When the N-terminal end of the peptide is acetylated, suitable acetylated N-terminal ends include those of formula -NH-C(0)Rz, wherein Rz is hydrogen, Ci-6 alkyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH3, -CH2CH3 and -CF3, or phenyl .
Since the peptides are contemplated as vaccine agents or diagnostic agents, they are in certain embodiments coupled to a carrier molecule, such as an immunogenic carrier. The peptides may thus be linked to other molecules either as recombinant fusions (e.g . via CLIP technology) or through chemical linkages in an oriented (e.g . using heterobifunctional cross-linkers) or nonoriented fashion. Linking to carrier molecules such as for example diphtheria toxin, latex beads (convenient in diagnostic and prognostic embodiments), and magnetic beads (also convenient in diagnostic and prognostic embodiments), polylysine constructs etc, are all possible according to the invention.
The immunogenic carrier is conveniently selected from carrier proteins such as those conventionally used in the art (e.g . diphtheria or tetanus toxoid, KLH etc.), but it is also possible to use shorter peptides (T-helper epitopes) which can induce T-cell immunity in larger proportions of a population. Details about such T-helper epitopes can e.g . be found in WO 00/20027, which is hereby incorporated by reference herein - all immunolgic carriers and "promiscuous" (i .e. universal) T-helper epitopes discussed therein are useful as immunogenic carriers in the present invention.
In certain embodiments, the carrier is a virus like particle, i .e. a particle sharing properties with virions without being infectious. Such virus-like particles may be provided chemically (e.g . Jennings and Bachmann Ann. Rev. Pharmacol . Toxicol. 2009. 49 : 303-26 Immunodrugs: Therapeutic VLP-based vaccines for chronic diseases) or using cloning techniques to generate fusion proteins (e.g. Peabody et al. J. Mol. Biol . 2008; 380 : 252-63. Immunogenic display of diverse peptides on virus-like particles of RNA phage MS2) . Another example is "Remune", an HIV vaccine originally made by Immune Response Corporation, which consists of formalin inactivated HIV that has been irradiated to destroy the viral genome. The company was started by Jonas Salk who used the same technique to generate the killed polio vaccine in widespread use today.
One aspect of the present invention relates to an immunogenic composition (such as a vaccine composition) comprising a composition of at least one HIV-specific peptides, in combination with a pharmaceutically acceptable diluent, excipient or vehicle and optionally one or more immunological adjuvant.
In common for aspects of the invention is that they all include embodiments where the at least one HIV-specific peptide is selected from the group of amino acid sequences of SEQ ID NOs: 1, 4, 9 and 15, as defined above; wherein the terminal ends of each HIV specific peptide may be free carboxyi- or amino- groups, amides, acyls or acetyls; and in the form of an acetate salt.
In some embodiments two or more of the Cys residues of said HIV-specific peptide 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 0, N and S and/or the said peptide sequences are immobilized to a solid support.
In some embodiments the amino acid sequence of SEQ ID NO : 1 is selected from the group of SEQ ID NO: 2 and SEQ ID NO: 3.
In some embodiments the amino acid sequence of SEQ ID NO : 4 is selected from the group of SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
In some embodiments the amino acid sequence of SEQ ID NO : 9 is selected from the group of SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 and SEQ ID NO: 14.
In some embodiments the amino acid sequence of SEQ ID NO : 15 is selected from the group of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and SEQ ID NO: 20.
In some embodiments the at least one HIV-specific peptide comprises at least, two, three, or four peptides selected from each of the groups of SEQ ID NO : 1, SEQ ID NO: 4, SEQ ID NO: 9 and SEQ ID NO: 15.
In some embodiments the at least one HIV-specific peptide consists of or comprises the peptides of SEQ ID NO: 3, SEQ ID NO: 6, SEQ ID NO: 11 and SEQ ID NO: 18.
Preparation of immunogenic compositions includes the use of state-of-the-art constituents such as immunological adjuvants. Apart from these adjuvants, which are detailed, by way of example, below, immunogenic compositions are prepared as generally taught in the art:
The preparation of vaccines which contain peptide sequences as active ingredients is generally well understood in the art, as exemplified by U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, all incorporated herein by reference. Typically, such vaccines are prepared as injectables either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection may also be prepared. The preparation may also be emulsified. The active immunogenic ingredient(s), such as each one peptide or composition comprising these peptides described herein is often mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, buffers, antioxidants, chelating agents, sugars, polyols, organic co-solvents, preservatives, isotonicity agents, stabilizers surfactants, saline, dextrose, glycerol, ethanol, ethylene glycol, propylene glycol, or the like, and combinations thereof. In addition, if desired, the vaccine may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, or adjuvants which enhance the effectiveness of the vaccines; cf. the detailed discussion of adjuvants below.
The use of excipients such as buffers, antioxidants, preservatives, isotonicity agents, stabilizers, chelating agents and surfactants in pharmaceutical compositions is well-known to the skilled person. For convenience reference is made to Remington : The Science and
Practice of Pharmacy, 19th edition, 1995, Jorgensen et al. Expert Opin. Drug deliv. 6 (2009) and references there in; To Rowe et al., eds. Handbook of Pharmaceutical Excipients, 7th edn. London : Pharmaceutical Press, 2012; and to L. Jorgensen, S. Hostrup, E.H. Moeller, H. Grohganz, Recent trends in stabilising peptides and proteins in pharmaceutical formulation - considerations in the choice of excipients, Expert Opin. Drug Deliv. 6 (2009) 1219-30.
Buffers suitable for use in pharmaceutical compositions are known to those skilled in the art and include, but are not limited to, orhto-phosphate, TRIS, glycine, N-glycylglycine, citrate, sodium acetate, sodium carbonate, glycylglycine, histidine, lysine, arginine, sodium phosphate, and sodium citrate or mixtures thereof.
Antioxidants for use as excipients include thioredoxin, glutathione, L-Methionine, ascorbic acid. Ascorbic acid Ectoine, Glutathione, Monothioglycerol, Morin, Poly(ethylenimine), Propyl gallate and Vitamin E.
Preservatives for use in pharmaceutical compositions are known to those skilled in the art and include, but are not limited to, phenol, m-cresol, methyl p-hydroxybenzoate, propyl p- hydroxybenzoate, 2-phenoxyethanol, butyl p-hydroxybenzoate, 2-phenylethanol, benzyl alcohol, chlorobutanol, and thiomerosal, or mixtures thereof.
Buffers suitable for use in pharmaceutical compositions are known to those skilled in the art and include, but are not limited to, orhto-phosphate, TRIS, glycine, N-glycylglycine, citrate sodium acetate, sodium carbonate, glycylglycine, histidine, lysine, arginine, sodium phosphate, and sodium citrate or mixtures thereof.
The pharmaceutical composition of the invention may comprise an isotonicity agent, including but not limited to, sodium chloride, xylitol, mannitol, sorbitol, glycerol, glucose, maltose, sucrose, trehalose, raffinose, L-glycine, L-histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine, dimethyl sulphone, polyethyleneglycol, propylene glycol or mixtures thereof. The compositions according to the invention may contain a stabilizer, for example selected from polyethylene glycol (e.g. PEG 3350), polyvinylalcohol (PVA), polyvinylpyrrolidone, carboxymethylcellulose, different salts (e.g. sodium chloride), L-glycine, L-histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine and mixtures thereof.
Compositions according to the invention may further comprise a chelating agent, such as ethlenediaminetetraacetic acid (EDTA), desferal, diethylenetriaminepentaacetic acid (DTPA), inositol hexaphosphate, ethylenediamine bis(o-hydroxyphenylacetic acid),
tris(hydroxymethyl)aminomethane (TRIS), tartaric acid, citric acid and aspartic acid, and mixtures thereof.
Compositions according to the invention may further comprise a surfactant, which may be selected from a detergent, ethoxylated castor oil, polyglycolyzed glycerides, acetylated monoglycerides, sorbitan fatty acid esters, poloxamers, such as 188 and 407,
polyoxyethylene sorbitan fatty acid esters, polyoxyethylene derivatives such as alkylated and alkoxylated derivatives (tweens, e.g. Tween-20, or Tween-80), monoglycerides or
ethoxylated derivatives thereof, diglycerides or polyoxyethylene derivatives thereof, glycerol, cholic acid or derivatives thereof, lecithins, alcohols and phospholipids, glycerophospholipids (lecithins, kephalins, phosphatidyl serine), glyceroglycolipids (galactopyransoide),
sphingophospholipids (sphingomyelin), and sphingoglycolipids (ceramides, gangliosides), DSS (docusate sodium, CAS registry no [577-11-7]), docusate calcium, CAS registry no [128-49- 4]), docusate potassium, CAS registry no [7491-09-0]), SDS (sodium dodecyl sulfate or sodium lauryl sulfate), dipalmitoyl phosphatidic acid, sodium caprylate, bile acids and salts thereof and glycine or taurine conjugates, ursodeoxycholic acid, sodium cholate, sodium deoxycholate, sodium taurocholate, sodium glycocholate, N-Hexadecyl-N,N-dimethyl-3- ammonio-l-propanesulfonate, anionic (alkyl-arylsulphonates) monovalent surfactants, palmitoyl lysophosphatidyl-L-serine, lysophospholipids (e.g. l-acyl-sn-glycero-3-phosphate esters of ethanolamine, choline, serine or threonine), alkyl, alkoxyl (alkyl ester), alkoxy (alkyl ether)- derivatives of lysophosphatidyl and phosphatidylcholines, e.g. lauroyl and myristoyl derivatives of lysophosphatidylcholine, dipalmitoylphosphatidylcholine, and modifications of the polar head group, that is cholines, ethanolamines, phosphatidic acid, serines, threonines, glycerol, inositol, and the postively charged DODAC, DOTMA, DCP, BISHOP,
lysophosphatidylserine and lysophosphatidylthreonine, zwitterionic surfactants (e.g. N-alkyl- N,N-dimethylammonio-l-propanesulfonates, 3-cholamido-l-propyldimethylammonio-l- propanesulfonate, dodecylphosphocholine, myristoyl lysophosphatidylcholine, hen egg lysolecithin), cationic surfactants (quarternary ammonium bases) (e.g. cetyl- trimethylammonium bromide, cetylpyridinium chloride), non-ionic surfactants,
polyethyleneoxide/polypropyleneoxide block copolymers (Pluronics/Tetronics, Triton X-100, Dodecyl β-D-glucopyranoside) or polymeric surfactants (Tween-40, Tween-80, Brij-35), fusidic acid derivatives- (e.g. sodium tauro-dihydrofusidate etc.), long-chain fatty acids and salts thereof C6-C12 (eg. oleic acid and caprylic acid), acylcarnitines and derivatives, Na- acylated derivatives of lysine, arginine or histidine, or side-chain acylated derivatives of lysine or arginine, Na-acylated derivatives of dipeptides comprising any combination of lysine, arginine or histidine and a neutral or acidic amino acid, Na-acylated derivative of a tripeptide comprising any combination of a neutral amino acid and two charged amino acids, or the surfactant may be selected from the group of imidazoline derivatives, or mixtures thereof. The vaccines are conventionally administered parenterally, by injection, for example, either subcutaneously, intracutaneously, intradermal^, subdermally or intramuscularly.
Additional formulations which are suitable for other modes of administration include suppositories and, in some cases, oral, nasal, buccal, sublinqual, intraperitoneal, intravaginal, anal, epidural, spinal, and intracranial formulations. For suppositories, traditional binders and carriers may include, for example, polyalkalene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10% (w/w), preferably 1-2% (w/w). Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders and may contain 10-95% (w/w) of active ingredient, preferably 25-70% (w/w).
The peptides may be formulated into a vaccine as neutral or salt forms.
Pharmaceutically acceptable salts include acid addition salts (formed with the free amino groups of the peptide) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and organic bases such as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like. The vaccines are administered in a manner compatible with the dosage formulation, and in such amount as will be therapeutically effective and immunogenic. The quantity to be administered depends on the subject to be treated, including, e.g., the capacity of the individual's immune system to mount an immune response, and the degree of immunity desired. Suitable dosage ranges are of the order of several hundred micrograms of active ingredient per vaccination with a preferred range from about 0.1 μg to 2,000 μg (even though higher amounts in the 1-10 mg range are contemplated), such as in the range from about 0.5 μg to 1,000 μg, preferably in the range from 1 μg to 500 μg and especially in the range from about 10 μg to 100 μg. Suitable regimens for initial administration and booster shots are also variable but are typified by an initial administration followed by subsequent inoculations or other administrations.
Some of the peptides are sufficiently immunogenic in a vaccine, but for some of the others the immune response will be enhanced if the vaccine further comprises an adjuvant substance. The immunogenic molecules described herein can therefore be formulated with adjuvants:
The adjuvants to be combined are known to induce humoral responses and include: i) Salt suspensions (e.g. varieties of salts containing aluminum ions or calcium ions), ii) Oil- in-water emulsions (e.g. varieties of squalane-based or squalene-based emulsions), iii) Water-in-oil emulsions (e.g. Montanide ISA51 or ISA720), iv) Neutral liposomes, v) Cationic liposomes, vi) Microspheres, vii) Immunostimulating complexes (e.g. ISCOMs or
ISCOMATRIX), viii) Pattern-recognition receptor agonists (e.g. agonists for C-type lectin receptors (CLRs), NOD-like receptors (NLRs), RIG-like helicases (RLHs), Triggering receptor expressed on myeloid cells (TREMs) and Toll-like receptors (TLRs)), ix) Saponins (i.e. Any saponin derived from Quillaja saponaria or Platycodon grandifiorum), x) Virosomes/Virus-like particles, xi) Enterotoxins (i.e. Cholera toxin, CTA1-DD or Esherichia coli heat-labile enterotoxin), and combinations thereof.
For a further enhancement of the vaccine antigenic properties, they could be combined with a well known adjuvant with an oral immune modulant or adjuvant such as a Cox-2 inhibitor or a immunomodulating compound.
A further apect of the invention is the use of the vaccine combined with adjuvant, with one or more further therapeutic agents, such as an (oral) immunemodulating agent and/or a reservoir purging agent.
The terms "therapeutic agent", such as "immunomodulating agent" or virus reservoir purging agent as used herein, includes but is not limited to cytokines, such as interferons, monoclonal antibodies, such as anti-PDl antibodies, cyclophosphamide, Thalidomide, Levamisole, and Lenalidomide.
"A virus reservoir purging agent", includes but is not limited to auranofin, IL-7, prostratin, bryostatin, HDAC inhibitors, such as vorinostat, Disulfiram and any suitable agent disclosed in any one of WO2013050422, WO2012051492 A3 and in Barton et al., Clinical Pharmacology & Therapeutics (2013); 93 1, 46-561, including but not limited to a NF-kappa- B-inducer selected from the group comprising : PMA, prostratin, bryostatin and TNF-alpha, and/or b) a histone deacetylase inhibitor selected from the different families (hydroxamates, cyclic peptides, aliphatic acids, and benzamides) including : TSA, SAHA, MS-275,
aminosuberoyl hydroxamic acids, M-Carboxycinnamic acid bishydroxamate, LAQ-824, LBH- 589, belinostat (PXD-101 ), Panobinostat (LBH-589), a cinnamic hydroxamic acid analogue of M-carboxycinnamic acid bishydroxamate, IF2357, aryloxyalkanoic acid hydroxamides, depsipeptide, apicidin, cyclic hydroxamic acid- containing peptide group of molecules, FK- 228, red FK, cyclic peptide mimic linked by an aliphatic chain to a hydroxamic acid, butyrate, phenylbutyrate, sodium butyrate, valproic acid, pivaloyloxymethyl butyrate, 5 NOX-275, and MGCD0103. Any of the above virus reservoir purging agents may be used alone or in combination with any one other suitable virus reservoir purging agent, such as with another class of HIV inducers.
DNA methylation, probably together with repressive histone modifications, may also contribute to a "lock" in a silent state of the provirus and makes its return to an active state difficult. These observations suggest that HDAC or HMT or DNA methylation inhibitors together with efficient cART constitute good anti-latency drug candidates aimed at reducing/eliminating the pool of latent reservoirs to a level bearable by the host immune system.
Accordingly suitable immunomodulatory compounds or purging agents may be DNA methylation inhibitors selected from the two classes (non-nucleoside and nucleoside demethylating agents) including : 5-azacytidine (azacitidine), Sinefungin, 5-aza-2'- deoxycytidine (5-aza-CdR, decitabine), l-3-Darabinofuranosyl-5-azacytosine (fazarabine) and dihydro-5-azacytidine (DHAC), 5-fluorodeoxycytidine (FdC), oligodeoxynucleotide duplexes containing 2-H pyrimidinone, zebularine, antisense oligodeoxynucleotides (ODNs), MG98, (-)-epigallocatechin-3-gallate, hydralazine, procaine and procainamide.
Other suitable immunomodulatory compounds or purging agents to be used according to the present invention includes histone deacetylase inhibitor selected from the different families of HDACI (hydroxamates, cyclic peptides, aliphatic acids, and benzamides) including TSA, SAHA, MS-275, aminosuberoyl hydroxamic acids, M-Carboxycinnamic acid bishydroxamate, LAQ-824, LBH-589, belinostat (PXD-101 ), Panobinostat (LBH-589), a cinnamic hydroxamic acid analogue of M-carboxycinnamic acid bishydroxamate, IF2357, aryloxyalkanoic acid hydroxamides, depsipeptide, apicidin, cyclic hydroxamic acid-containing peptide group of molecules, FK-228, red FK, cyclic peptide mimic linked by an aliphatic chain to a hydroxamic acid, butyrate, phenylbutyrate, sodium butyrate, valproic acid,
pivaloyloxymethyl butyrate, 5 NOX-275, and MGCD0103.
Other suitable immunomodulatory compounds or purging agents to be used according to the present invention includes histone methyltransferase inhibitors (chaetocin and BIX-01294); Inhibitors of Enhances of Zeste 2 (EZH2) - such as 3-deazaneplanocin A (DZNep) used alone or in combination with other classes of immunomodulatory compounds or purging agents.
Other suitable adjuvants includes response-selective C5a agonists, such as EP54 and EP67 described in Hung CY et al. An agonist of human complement fragment C5a enhances vaccine immunity against Coccidioides infection. Vaccine (2012) and Kollessery G et al. Tumor-specific peptide based vaccines containing the conformationally biased, response- selective C5a agonists EP54 and EP67 protect against aggressive large B cell lymphoma in a syngeneic murine model. Vaccine (2011) 29: 5904-10.
Various methods of achieving adjuvant effect for the vaccine are thus known.
General principles and methods are detailed in "The Theory and Practical Application of Adjuvants", 1995, Duncan E.S. Stewart-Tull (ed.), John Wiley & Sons Ltd, ISBN 0-471- 95170-6, and also in "Vaccines: New Generationn Immunological Adjuvants", 1995,
Gregoriadis G et al. (eds.), Plenum Press, New York, ISBN 0-306-45283-9, both of which are hereby incorporated by reference herein, but a number of later publications also deal with the technology of incorporating adjuvants: Roestenberg M et a/., PLoS One. 2008;3(12) :e3960. Epub 2008 Dec 18; Relyveld E and Chermann JC, Biomed Pharmacother. 1994;48(2) :79-83; Hsu FJ et a/., Blood. 1997 May l;89(9) :3129-35; Galli G et a/., Proc Natl Acad Sci U S A. 2009 May 12; 106(19) :7962-7. Epub 2009 Apr 27; Bojang KA et a/., Lancet. 2001 Dec 8;358(9297) : 1927-34; Odunsi K et a/., Proc Natl Acad Sci U S A. 2007 Jul
31; 104(31) : 12837-42. Epub 2007 Jul 25; Patel GB and Sprott GD; Crit Rev Biotechnol.
1999; 19(4) :317-57. Review; Agger EM et a/. , PLoS One. 2008 Sep 8;3(9) :e3116; Kirby DJ et al. J Drug Target. 2008 May; 16(4) :282-93; Florindo HF et a/. , Vaccine. 2008 Aug
5;26(33) :4168-77. Epub 2008 Jun 17; Sun HX et a/., Vaccine. 2009 May 28; Guy B, Nat Rev Microbiol. 2007 Jul;5(7) : 505-17. Review.; Vandepapeliere P et a/., Vaccine. 2008 Mar 4;26(10) : 1375-86. Epub 2008 Jan 14; Ghochikyan A et al. Vaccine. 2006 Mar
20;24(13) :2275-82. Epub 2005 Dec 5; Xie Y et a/., Vaccine. 2008 Jun 25;26(27-28) :3452- 60. Epub 2008 May 1; Chung YC et a/. , Vaccine. 2008 Mar 28;26(15) : 1855-62. Epub 2008 Feb 25; Maier M et a/., Vaccine. 2005 Oct 25;23(44) : 5149-59; Sundling C et a/., J Gen Virol. 2008 Dec;89(Pt 12) :2954-64.
In the methods and compositions of the invention the at least one HIV-specific peptide, may be administered in combination with one or more further thereapeutically active agents, such as agents for the treatment and or prevention of HIV and/or AIDS.
One aspect of the invention is the use of the vaccine combined with adjuvant, with one or more further therapeutic agents, such as an (oral) immunemodulating agent and/or a reservoir purging agent.
The terms "therapeutic agent", such as "immunomodulating agent" or virus reservoir purging agent as used herein, includes but is not limited to cytokines, such as interferons, monoclonal antibodies, such as ant-PDl antibodies, cyclophosphamide, Thalidomide,
Levamisole, and Lenalidomide. "A virus reservoir purging agent", includes but is not limited to auranofin, IL-7, prostratin, bryostatin, HDAC inhibitors, such as vorinostat, and Disulfiram, and the further agents described herein.
The failure of antiretroviral therapy (ART) to eradicate HIV-1 infection lies in the observation that HIV-1 remains quiescent in latent reservoirs. Latently infected resting CD4+ cells (either naive or long lived memory cells) carry transcriptionally silent HIV-1 and represent the predominant reservoir of HIV-1 infection. Other cells may also act as reservoirs (Reviewed in Alexaki et al., 2008, Curr. HIV Res. 6:388-400) such as macrophages, dendritic cells and astrocytes (where HIV-1 infection occurs via a CD4-independent mechanism). It is these latent reservoirs that represent the major challenge to eradication of HIV-1 infection. Approaches towards eradication include attempts to purge reservoirs by selective activation of latently infected cells (such as memory cells) in the presence of ART such that released virus may not infect and replicate in neighbouring cells (Richman et al., 2009, Science 323 : 1304-1307). Agents include histone deacetylase inhibitors, cytokines, such as IL-2 and IL-7, as well as bryostatin, the protein kinase C activator (Kovochich et al ., 2011, PLoS ONE 6 (4) :el8270). Therapeutic vaccines have the advantage of being able to penetrate sanctuary sites less well accessed by ART such as lymphoid tissue (Pantaleo et al., 1991, Proc. Natl. Acad. Sci. USA 88 :9838-42; Fox et al ., 1991, J. Infect. Dis. 164 : 1051-57) and the central nervous system (Alexaki et al., 2008, Curr. HIV Res. 6 :388-400), that represent regions for viral persistence. This relates to therapeutic interventions targeting both the virus itself as well as HIV-associated immune activation.
A number of studies have been conducted with the aim of providing compounds that can safely and effectively be used to treat diseases associated with abnormal production of TNF-a. See, e.g., Marriott, J.B., et al, Expert Opin. Biol. Ther. (4) : 1-8 (2001); G.W. Muller, et al, Journal of Medicinal Chemistry, 39(17) : 3238-3240 (1996); and G.W. Muller, et al,
Bioorganic & Medicinal Chemistry Letters, 8 : 2669-2674 (1998). Some studies have focused on a group of compounds selected for their capacity to potently inhibit TNF-a production by LPS stimulated PBMC. L.G. Corral, et al, Ann. Rheum. Dis., 58 (suppl I) : 1107-1113 (1999). These compounds, often referred to as immunomodulatory compounds, show not only potent inhibition of TNF-a, but also marked inhibition of LPS induced monocyte IL1B and IL12 production. LPS induced IL6 is also inhibited by immunomodulatory compounds, albeit partially. These compounds are potent stimulators of LPS induced IL10. Particular examples include, but are not limited to, the substituted 2-(2,6-dioxopiperidin-3-yl)phthalimides and substituted 2-(2,6-dioxopiperidin-3-yl)-l-oxoisoindoles as described in US 6281230 and US 6316471. Monocyte/macrophage function is part of the Innate Immune System that serves as a first line of defense against an infection. By modulating the host's monocytes and macrophages, immunomodulatory compounds can change the dynamics of the response to a viral infection, such as influenza. Histone deacetylases (HDAC) are a class of enzymes that remove acetyl groups from N-acetylated lysines amino acid on histone proteins. Currently 18 HDACs have been identified in mammals. They have been divided into four classes based on cellular localization, function, and sequence similarity. Class I includes HDACs 1, 2, 3, and 8 which are found primarily in the nucleus. Class II HDACs (HDACs 4, 5, 6, 7 9, and 10) are found primarily in the cytoplasm but may be able to shuttle between the nucleus and the cytoplasm; class Ila comprises four HDACs (HDACs 4, 5, 7 and 9) while class lib comprises two HDACs (HDACs 6 and 10) which are expressed only in the cytoplasm. HDAC11, which is ubiquitously expressed, shares sequence similarities with both class I and class II HDACs and represents Class IV. Class III (also called "sirtuin family") groups NAD+-dependent proteins which do not act primarily on histones.
In the methods and compositions of the invention the at least one HIV-specific peptide, may be administered in combination, such as in a composition comprising an immunomodulatory compound and/or a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor.
The immunomodulatory compounds may be selected from anti-PDl antibodies, such as MDX-1106 (Merck), THALOMID® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene), and immunomodulatory compound described in any one of WO2007028047, WO2002059106, and WO2002094180. The immunomodulatory compound may be selected from 4-(amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-l,3-dione and 3-(4-amino-l-oxo-l,3- dihydro-isoindol-2-yl)-piperidine-2,6-dione. In particular the immunomodulatory compound is lenalidomide. The immunomodulatory compound may be enantiomerically pure.
The reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor, may be selected from M344 (4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide), chidamide (CS055/HBI-800), 4SC-202, (4SC), Resminostat (4SC), hydroxamic acids such as vorinostat (SAHA), belinostat (PXD101), LAQ824, trichostatin A and panobinostat (LBH589);
benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), cyclic tetrapeptides (such as trapoxin, such as trapoxin B), and the depsipeptides, such as romidepsin (ISTODAX), electrophilic ketones, and the aliphatic acid compounds such as phenylbutyrate, valproic acid, Oxamflatin, ITF2357 (generic givinostat), Apicidin, MC1293, CG05, and CG06; compounds that activate transcription factors including NF-KappaB, Prostratin, auranofin, bryostatin, a nontumorigenic phorbol ester, DPP (12-deoxyphorbol-13- phenylacetate), PMA, and Phorbol 12-myristate 13-acetate (PMA); Compounds that activate HIV mRNA elongation including P-TEF-b kinase and hexamethylbisacetamide (HMBA); IL-7; T-cell stimulating factors including anti-CD3/CD28 - T-cell stimulating Ab's; Kinase inhibitors including Tyrphostin A, Tyrphostin B, and Tyrphostin C;PTEN (phosphatase and tensin homologue) gene inhibitors including SF1670 (Echelon Bioscience), Disulfiram (DSF), an inhibitor of acetaldehyde dehydrogenase, Protein Tyrosine Phosphatase Inhibitors including bpV(HOpic), bpV(phen), and bpV(pic) (Calbiochem; EMD Millipore), Toll-like receptors agonists including Toll-like receptor-9 (TLR9) and Toll-like receptor-7 (TLR9) agonists, quercetin, lipoic acid, sodium butyrate, TNF-alpha, PHA, Tat. In particular the reservoir purging agent is romidepsin.
In the methods of the invention the components of the at least one HIV-specific peptide and/or the one or more further thereapeutically active agents, may be administered simultaneously, sequentially or separately in any order.
Thus the invention provides a pharmaceutical composition comprising one, two or more components of the at least one HIV-specific peptide and/or the one or more further thereapeutically active agents optionally in combination with one or more pharmaceutically acceptable adjuvants, diluents or carriers.
Similarly, the invention also provides a combination product comprising of components of the at least one HIV-specific peptide and/or the one or more further thereapeutically active agents, wherein each of component is formulated in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier. In this aspect of the invention, the combination product may be either a single (combination) pharmaceutical formulation or a kit-of-parts. In a kit-of-parts some or all of the components may be formulated separately and may each be provided in a form that is suitable for administration in conjunction with the other(s).
The component(s) may also be provided for use, e.g. with instructions for use, in combination with one or more further component(s) as defined above.
The peptides for use in the invention may be produced synthetically using art recognised methods. Further details for the synthetic production of such peptides are found in the Examples. Alternatively the peptides may be produced recombinantly. When recombinantly producing the peptides for use in the invention by means of transformed cells, it is convenient, although far from essential, that the expression product is either exported out into the culture medium or carried on the surface of the transformed cell.
When an effective producer cell has been identified it is preferred, on the basis thereof, to establish a stable cell line which carries the vector of the invention and which expresses the nucleic acid fragment of the invention. Preferably, this stable cell line secretes or carries the peptide expression product, thereby facilitating purification thereof.
In general, plasmid vectors containing replicon and control sequences which are derived from species compatible with the host cell are used in connection with the hosts. The vector ordinarily carries a replication site, as well as marking sequences which are capable of providing phenotypic selection in transformed cells. For example, E. coli is typically transformed using pBR322, a plasmid derived from an E. coli species (see, e.g., Bolivar et a/. , 1977). The pBR322 plasmid contains genes for ampicillin and tetracycline resistance and thus provides easy means for identifying transformed cells. The pBR plasmid, or other microbial plasmid or phage must also contain, or be modified to contain, promoters which can be used by the prokaryotic microorganism for expression.
Those promoters most commonly used in recombinant DNA construction include the β-lactamase (penicillinase) and lactose promoter systems (Chang et a/. , 1978; Itakura et a/. , 1977; Goeddel et a/., 1979) and a tryptophan (trp) promoter system (Goeddel et a/. , 1979; EP-A-0 036 776). While these are the most commonly used, other microbial promoters have been discovered and utilized, and details concerning their nucleotide sequences have been published.
In addition to prokaryotes, eukaryotic microbes, such as yeast cultures may also be used, and also here the promoter should be capable of driving expression. Saccharomyces cerevisiase, or common baker's yeast is the most commonly used among eukaryotic microorganisms, although a number of other strains are commonly available. For expression in Saccharomyces, the plasmid YRp7, for example, is commonly used (Stinchcomb et a/., 1979; Kingsman et a/. , 1979; Tschemper et a/. , 1980).
Suitable promoting sequences in yeast vectors include the promoters for 3- phosphoglycerate kinase (Hitzman et a/. , 1980) or other glycolytic enzymes (Hess et a/., 1968; Holland et a/., 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. In constructing suitable expression plasmids, the termination sequences associated with these genes are also incorporated into the expression vector 3' of the sequence desired to be expressed to provide polyadenylation of the mRNA and termination.
Other promoters, which have the additional advantage of transcription controlled by growth conditions are the promoter region for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, and the aforementioned glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Any plasmid vector containing a yeast-compatible promoter, origin of replication and termination sequences is suitable.
In addition to microorganisms, cultures of cells derived from multicellular organisms may also be used as hosts. In principle, any such cell culture is workable, whether from vertebrate or invertebrate culture. Examples of such useful host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cell lines, and W138, BHK, COS-7 293, Spodoptera frugiperda (SF) cells, Drosophila melanogaster cell lines (such as Schneider 2 (S2)), and MDCK cell lines.
Expression vectors for such cells ordinarily include (if necessary) an origin of replication, a promoter located in front of the gene to be expressed, along with any necessary ribosome binding sites, RNA splice sites, polyadenylation site, and transcriptional terminator sequences. For use in mammalian cells, the control functions on the expression vectors are often provided by viral material. For example, commonly used promoters are derived from polyoma, Adenovirus 2, and most frequently Simian Virus 40 (SV40). The early and late promoters of SV40 virus are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 viral origin of replication (Fiers et al., 1978). Smaller or larger SV40 fragments may also be used, provided there is included the approximately 250 bp sequence extending from the Hindlll site toward the Bgll site located in the viral origin of replication. Further, it is also possible, and often desirable, to utilize promoter or control sequences normally associated with the desired gene sequence, provided such control sequences are compatible with the host cell systems.
An origin of replication may be provided either by construction of the vector to include an exogenous origin, such as may be derived from SV40 or other viral (e.g., other Polyoma viruses, Adeno, VSV, BPV) or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
As for routes of administration and administration schemes of polypeptide based vaccines which have been detailed above, these are also applicable for the nucleic acid vaccines of the invention and all discussions above pertaining to routes of administration and administration schemes for polypeptides apply mutatis mutandis to nucleic acids. To this should be added that nucleic acid vaccines can also be administered intraveneously and intraarterially. Furthermore, it is well-known in the art that nucleic acid vaccines can be administered by use of a so-called gene gun and/or by use of electroporation, and hence also these and equivalent modes of administration are regarded as part of the present invention.
Under normal circumstances, the nucleic acid fragment is introduced in the form of a vector wherein expression is under control of a viral promoter. For more detailed discussions of vectors according to the invention, cf. the discussion above. Also, detailed disclosures relating to the formulation and use of nucleic acid vaccines are available, cf. Donnelly JJ et a/, 1997, Annu. Rev. Immunol. 15: 617-648 and Donnelly JJ et al., 1997, Life Sciences 60: 163- 172. Both of these references are incorporated by reference herein.
An alternative of using peptide immunogens or nucleic acid immunogens is the use of live immunogen technology. This entails administering a non-pathogenic microorganism which has been transformed with a nucleic acid fragment or a vector of the present invention. The non-pathogenic microorganism can be any suitable attenuated bacterial strain (attenuated by means of passaging or by means of removal of pathogenic expression products by recombinant DNA technology), e.g. Mycobacterium bovis BCG., non-pathogenic
Streptococcus spp., E. coli, Salmonella spp., Vibrio cholerae, Shigella, etc. Reviews dealing with preparation of state-of-the-art live vaccines can e.g. be found in Saliou P, 1995, Rev. Prat. 45: 1492-1496 and Walker PD, 1992, Vaccine 10: 977-990, both incorporated by reference herein. For details about the nucleic acid fragments and vectors used in such live vaccines, cf. the discussion below.
As an alternative to bacterial live immunogens, the nucleic acid fragment of the invention can be incorporated in a non-virulent viral vaccine vector such as a vaccinia strain or any other suitable poxvirus.
Normally, the non-pathogenic microorganism or virus is administered only once to a subject, but in certain cases it may be necessary to administer the microorganism/virus more than once in a lifetime in order to maintain protective immunity. It is even contemplated that immunization schemes as those detailed above for polypeptide vaccination will be useful when using live or virus vaccines.
Alternatively, live or virus immunization is combined with previous or subsequent polypeptide and/or nucleic acid immunization. For instance, it is possible to effect primary immunization with a live or virus vaccine followed by subsequent booster immunizations using the polypeptide or nucleic acid approach.
A set of numbered embodiments according to the invention are as follows:
1. A HIV-specific peptide selected from the group of amino acid sequences:
Xaai Xaa2 Xaa3 Xaa4 Xaa5 Xaa6 Ala Xaa8 Xaa9 Gin Thr Pro Trp Xaa^ Xaa^ Xaai6 Xaai7 Xaai8 Val Xaa20 (SEQ ID NO: 1); wherein 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,
Xaa in position 9 is Leu or He,
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, and
Xaa in position 20 is Gly or Arg; Xaai Xaa2 Xaa3 Xaa4 Xaa5 Gly Leu Asn Pro Leu Val [Gly]n Xaai2 Xaai3 Tyr Xaai5 Pro Xaai7 Xaai8 He Leu Xaa2i Xaa22 (SEQ ID NO : 4)
wherein 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 He, 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 or Lys,
Xaa in postion 13 is Met or Leu,
Xaa in position 15 is Ser, Cys or Gin,
Xaa in position 17 is Thr, Val, He, 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, and
n = 0, 1, 2 or 3;
Xaai Xaa2 Xaa3 Pro lie Pro Xaa7 Xaa8 Xaa9 Xaai0 Xaau Xaai2 [Gly]n Xaai3 Xaai4 Xaai5 Xaai6 Xaai7 Xaais Xaai9 Xaa20 Xaa2i 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, lie 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 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 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 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,
n = 1,,2 or 3, and
Xaai Xaa2 He He Xaa5 Xaa6 Xaa7 Xaa8 Xaa9 Leu Xaau [Gly]n [Arg]m Xaai2 Xaai3 Xaa^ Xaa^ Xaai6 Xaai7 Xaai8 Xaai9 Xaa2o Xaa2i Xaa22 Xaa23 Xaa24 Xaa25 (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,
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, He, Val or Asn,
Xaa in position 23 is Asn, Tyr, Cys or Gly,
Xaa in position 24 is Thr, Met, He, Ala, Val or none,
Xaa in postion 25 is Gly or none,
n = 1, 2 or 3 and m= 0, 1, 2 or 3 independent of each other,
the terminal ends of each HIV specific peptide may be free carboxyl- or amino groups, amides, acyls, acetyls; and wherein each peptide is in the form of an acetate salt.
2. The peptide according to embodiment 1, wherein two or more of the Cys residues of said HIV-specific peptide 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.
3. The peptide according to any one of embodiments 1 or 2, wherein the amino acid sequence of SEQ ID NO : 1 is selected from the groups of SEQ ID NO : 2 and SEQ ID NO : 3. 4. The peptide according to any one of the preceding embodiments, wherein the amino acid sequence of SEQ ID NO: 4 is selected from the groups of SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 7 and SEQ ID NO: 8.
5. The peptide according to any one of the preceding embodiments, 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.
6. The peptide according to any one of the preceding embodiments, wherein the amino acid sequence of SEQ ID NO: 15 is selected from the groups of SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO: 18, SEQ ID NO: 19 and SEQ ID NO: 20. 7. The peptide according to any one of the preceding embodiments, wherein at least one HIV-specific peptide comprises at least , two, three, or four peptides selected from each of the groups of SEQ ID NO: 1, SEQ ID NO: 4, SEQ ID NO: 9 and SEQ ID NO: 15.
8. The peptide according to any one of the preceding embodiments, wherein at least one HIV-specific peptide consist of or comprises the peptides of the SEQ ID NO: 3, SEQ ID NO: 6, SEQ ID NO: 11 and SEQ ID NO: 18.
9. A peptide consisting of the amino acid sequence shown in SEQ ID NO : 18 (Vacc-10) in the form of an acetate salt.
10. The peptide of embodiment 9, wherein the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 6% and 16%, such as between 7% and 15%, such as between 8% and 14%, such as between 9% and 14%, such as between 9% and 13%, such as between 10% and 14%, such as between 11% and 14%.
11. A peptide consisting of the amino acid sequence shown in SEQ ID NO : 11 (Vacc-11) in the form of an acetate salt.
12. The peptide of embodiment 11, wherein the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 6% and 16%, such as between
7% and 15%, such as between 8% and 14%, such as between 9% and 14%, such as between 9% and 13%, such as between 10% and 14%, such as between 11% and 13%.
13. A peptide consisting of the amino acid sequence shown in SEQ ID NO: 6 (Vacc-12) in the form of an acetate salt. 14. The peptide of embodiment 13, wherein the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 5% and 16%, such as between 5% and 15%, such as between 5% and 14%, such as between 6% and 14%, such as between 6% and 13%, such as between 7% and 12%, such as between 7% and 11%, such as between 8% and 11%, such as between 9% and 11%.
15. A peptide consisting of the amino acid sequence shown in SEQ ID NO: 3 (Vacc-13) in the form of an acetate salt.
16. The peptide of embodiment 15, wherein the acetate content of the salt is between 3% and 18%, such as between 3% and 17%, such as between 3% and 16%, such as between 3% and 15%, such as between 3% and 14%, such as between 3% and 13%, such as between 3% and 11%, such as between 3% and 10%, such as between 4% and 10%, such as between 4% and 9%, such as between 4% and 8%, such as between 4% and 7%, such as between 4% and 6%, such as between 4% and 5%.
17. A composition comprising one, two, three or four peptide acetate salts according to any one of embodiments 1 to 16.
18. A composition according to embodiment 17, which comprises said four peptide acetate salts defined in embodiments 9, 11, 13 and 15.
19. A composition according to embodiment 17 or 18, wherein the peptides have amide C-terminal ends of formula -C(0)NH2, or acetate salts thereof. 20. A composition according to embodiment 18-19, characterized in that the ratio of peptides is 1 : 1 : 1 : 1 w/w.
21. A composition according to any one of embodiments 17 to 20, wherein said one, two, three or four peptide acetate salts are in a disolved liquid state.
22. A composition according to embodiment 21, wherein said liquid is water. 23. A composition according to embodiment 22, wherein the pH of said composition is between between 3 and 8, such as between 3.5 and 7.5, such as between 4 and 7, such as between 4 and 6, such as between 5 and 6, and the osmolality is between 10 and 100, or between 15 and 25, or between 16 and 24 mOsm/kg. 24. The composition according to any one of embodiments 17-23, which is an
immunogenic composition optionally comprising a pharmaceutically acceptable diluent, one or more excipients, such as buffers, antioxidants, preservatives, isotonicity agents, stabilizers, chelating agents and surfactants, a vehicle and/or one or more immunological adjuvants. 25. The composition according to any one of embodiments 17-24 in the form of a vaccine composition.
26. The composition according to any one of embodiments 17-25 in the form of a vaccine composition where the one or more adjuvants are provided either separately or in
combination with the composition. 27. The composition according to any one of embodiments 17-26 for use as a
pharmaceutical.
28. The composition according to embodiment 27 for use in the treatment or prophylaxis of HIV or AIDS.
29. The composition according to any one of embodiments 17-28 comprising one or more further therapeutically active agents selected from an immunomodulatory compound and a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor.
30. The composition according to embodiment 29, wherein the immunomodulatory compound is selected from anti-PDl antibodies, such as MDX-1106 (Merck), THALOMID® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene), and immunomodulatory compounds described in any one of WO2007028047, WO2002059106, and WO2002094180.
31. The composition according to embodiment 30, wherein the immunomodulatory compound is lenalidomide.
32. The composition according to embodiment 29, wherein the reservoir purging agent is selected from M344 (4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide), chidamide (CS055/HBI-800), 4SC-202, (4SC), Resminostat (4SC), hydroxamic acids such as vorinostat (SAHA), belinostat (PXDIOI), LAQ824, trichostatin A and panobinostat (LBH589); benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), cyclic tetrapeptides (such as trapoxin, such as trapoxin B), and the depsipeptides, such as romidepsin (ISTODAX), electrophilic ketones, and the aliphatic acid compounds such as phenylbutyrate, valproic acid, Oxamflatin, ITF2357 (generic givinostat), Apicidin, MC1293, CG05, and CG06; compounds that activate transcription factors including NF-KappaB, Prostratin, auranofin, bryostatin, a nontumorigenic phorbol ester, DPP (12-deoxyphorbol-13- phenylacetate), PMA, and Phorbol 12-myristate 13-acetate (PMA); Compounds that activate HIV mRNA elongation including P-TEF-b kinase and hexamethylbisacetamide (HMBA); IL-7; T-cell stimulating factors including anti-CD3/CD28 - T-cell stimulating Ab's; Kinase inhibitors including Tyrphostin A, Tyrphostin B, and Tyrphostin C;PTEN (phosphatase and tensin homologue) gene inhibitors including SF1670 (Echelon Bioscience), Disulfiram (DSF), an inhibitor of acetaldehyde dehydrogenase, Protein Tyrosine Phosphatase Inhibitors including bpV(HOpic), bpV(phen), and bpV(pic) (Calbiochem; EMD Millipore), Toll-like receptors agonists including Toll-like receptor-9 (TLR9) and Toll-like receptor-7 (TLR9) agonists, quercetin, lipoic acid, sodium butyrate, TNF-alpha, PHA and Tat.
33. The composition according to embodiment 32, wherein the reservoir purging agent is romidepsin.
34. An immunoassay kit for the detection of antibodies induced by HIV or an HIV specific peptide or protein in a sample of body fluid, wherein the kit comprises a peptide according to any of embodiments 1 to 16, or a composition according to any one of embodiments 17-33.
35. A method for reducing and/or delaying pathological effects of human
immunodeficiency virus I (HIV) or for reducing the risk of developing acquired
immunodeficiency syndrome (AIDS) in a human infected with HIV, the method comprising administering an effective amount of a peptide according to any of embodiments 1 to 16, or a composition according to any one of embodiments 17-33.
36. The method according to embodiment 35, which method comprises the
simultaneously, sequentially or separately in any order administering of one or more immunological adjuvants and/or one or more further therapeutically active agents selected from an immunomodulatory compound and a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor, such as a therapeutically active agent defined in any one of embodiments 30-33.
PREAMBLE TO EXAMPLES
HIV-SPECIFIC PEPTIDES FOR USE ACCORDING TO THE INVENTION
The present invention involves the use of HIV-specific peptides based on conserved regions of HIV gag p24, antigens in free or carrier-bound form comprising at least one of the said peptides.
The HIV-specific peptides for use according to the invention originate from the four different conserved areas of the HIV-1 core protein p24, having the properties of maintaining the uniqueness (sensitivity and specificity) of the HIV-l-epitope. Further these peptides possess no recognized cytotoxic T lymphocyte (CTL) antagonistic effect and have at least one potential CTL epitope.
The HIV-specific peptides, for use according to the invention, which have met the above criteria are selected from the group of amino acid sequences of SEQ ID NOs : 1, 4, 9 and 15, as defined above; wherein the terminal ends of each HIV specific peptide may be free carboxyl- or amino- groups, amides, acyls or acetyls; or acetate salts of any of the HIV specific peptides.
The HIV-specific peptide sequences have the potential to serve as a particularly good antigen wherein the antigen comprises at least one peptide selected from the group of sequences of SEQ 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 may comprise two or more polypeptide sequences which are either linked by a bridge for instance a disulphide bridge between the Cys residues of the chains or bridges like Ci-C8 alkylene possibly intervened by one or more heteroatoms like 0, 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 HIV-specific 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 HIV-specific 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 salts thereof; or modified to provide a binding site for a carrier or another molecule. When the C-terminal end of a peptide is an amide, suitable amides included those having the formula -C(0)-NRxRy, wherein Rx and Ry are independently selected from hydrogen and Ci-6 al kyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH3, -CH2CH3 and -CF3, a particular amide group which may be mentioned is -C(0)NH2. When the N-terminal end of the peptide is acetylated, suitable acetylated N-terminal ends include those of formula -NH-C(0)Rz, wherein Rz is hydrogen, Ci-6 al kyl, which alkyl group may be substituted with one of more fluoro atoms, for example -CH3, -CH2CH3 and -CF3, or phenyl .
The HIV-specific peptides for use according to the invention consist of 6 to 50 amino acids, preferably between 10 and 30 amino acids. They cover all natural variation of amino acids in the identified positions.
The polypeptide antigen for use 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 carriers. 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.
In a preferred embodiment the HIV specific peptides for use according to the present invention comprises antigens containing the peptides of the SEQ ID NOs: 1, 4, 9 and 15, more preferreably the peptides occur in the ratio 1 : 1 : 1 : 1 w/w.
In a further preferred embodiment the HIV specific peptides for use according to the invention comprise the following : RALGPAATLQTPWTASLGVG (SEQ ID NO: 3)
RWLLLGLNPLVGGGRLYSPTSILG (SEQ ID NO: 6)
RAIPIPAGTLLSGGGRAIYKRTAILG (SEQ ID NO: 11)
and
RFIIPNIFTALSGGRRALLYGATPYAIG (SEQ ID NO: 18) (Nl in position 6 is Norleucine) or salts thereof, particularly acetate salts.
In some embodiments the HIV specific peptides for use according to the invention are modified at the C-terminus as follows:
RALGPAATLQTPWTASLGVG- NH2 (SEQ ID NO: 3)
RWLLLGLNPLVGGGRLYSPTSILG-NH2 (SEQ ID NO: 6)
RAIPIPAGTLLSGGGRAIYKRTAILG-N H2 (SEQ ID NO: 11)
and
RFIIPNIFTALSGGRRALLYGATPYAIG-N H2 (SEQ ID NO: 18)
or salts thereof, particularly acetate salts. (In this application also referred to in the examples as Vacc-4x).
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.
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 Int.J. 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.
GENERAL DESCRIPTION OF SYNTHESIS
The amino acid derivatives were supplied by Bachem AG, Switzerland.
The peptides described herein preferably have a free amino group at the N-terminus and an amidated C-terminus. The counter ion of all peptides described herein is acetate which is bound in ionic form to charged functional groups (i.e. guanidino side chains arginine and the ε-amino groups of lysine [Vacc-11] and the side chains of arginine [Vacc-10, Vacc-12 and Vacc-13]). All amino acid residues except the achiral glycine are in the L-configuration.
The peptides described herein were assembled on tricyclic amide linker resins utilising an 9- fluorenylmethyloxycarbonyl (Fmoc) strategy.
In brief the tricyclic amide linker resin is transferred into a solid phase peptide synthesis
(SPPS)-reactor with a stirrer. Synthesis is then started with a 9-fluorenylmethyloxycarbonyl (Fmoc)-deprotection of the resin according to the general description given below, followed by a coupling procedure with Fmoc-Gly-OH. This step is again followed by an Fmoc- deprotection and subsequent coupling of the amino acid derivates, peptides or dipeptides according to the sequence. The last coupling step is performed with side-chain protected
Fmoc-Arg-OH. After final Fmoc-deprotection, the peptide resin is dried in a desiccator under reduced pressure.
Fmoc-deprotecting procedure: Step 1 : Washing;
Step 2: Fmoc-deprotection;
Steps 3-9: Washing. Each step consists of addition of solvents/reagents, stirring at room temperature and filtration.
The peptide resin is treated with cold TFA in the presence of deionised water and 1, 2- Ethanedithiol (EDT), (Vacc-10 and Vacc-13) or triisopropylsilane (TIS) (Vacc-11 and Vacc-12) for approximately two to three hours at room temperature. After filtering off and washing the resin with TFA, the peptide is precipitated in diisopropyl ether (IPE). It is then filtered off, washed with IPE and dried in a desiccator under reduced pressure.
The material obtained in the previous stage is purified by preparative HPLC on reversed phase columns with acetonitrile (ACN) gradient elution and ultraviolet (UV) detection at λ = 220 nano-metres (nm) using a TEAP and/or TFA system. Vacc-10 is only purified using the TFA system.
For Vacc-13, a perchlorate system for preparative HPLC purification prior to using TEAP and TFA sytem has been introduced. Sodium perchlorate is listed as a raw material.
The last stage of manufacture of Vacc-4x acetate is the exchange from the TFA salt, obtained in stage three, into the acetate salt by ion exchange. The lyophilised material from one or several combined preparative HPLC runs is dissolved in varying concentrations of acetic acid or in purified water according to the properties of the individual peptides. The dissolved peptide is loaded onto the ion exchange resin (acetate form) and equilibrated with 5% acetic acid (or 20% purified water for Vacc-13). The elution is performed with 5% acetic acid (or purified water for Vacc-13), checked by thin-layer chromatography (TLC), filtered through a 0.2 μηη membrane filter and lyophilised to yield the final product as a white to off-white powder.
Although the Vacc-4x formulation only optionally contain an ionic excipients, the peptides and their counter ions (acetate) account for a certain osmolality. The range of 10 - 100 mOsm/kg was defined based on the result obtained for the technical sample. Potential variability due to the four peptides is taken into account. For the drug product, approximately 1 mg of each of the four Vacc-4x peptides was used. The lyophilisate is reconstituted with 0.30 mL of WFI. Taking the acetic acid contents of the peptides listed in table 1 into account, the acetic acid content of Vacc-4x is approximately 0.40 mg in 0.30 mL of solution. The theoretical osmolality is approximately 23 mOsmol/L by calculation, which correlates well with the values determined in the Vacc-4x batches (20-23 mOsmol/kg).
Table 1 Acetic acid contents of the four peptides (GMP grade material, two batches each)
Active Peptide batch Acetic acid Peptide batch Acetic acid used for Vacc-4x content used for Vacc-4x content substance
batches 1011584 [%] batch 1018724 [%] and 1012951
Vacc-10 1008290 11.3 1015501 12.2 Acetate
Vacc-11 1009945 17.2 1015502 14.8 Acetate
Vacc-12 1008294 9.9 1015503 10.0 Acetate
Vacc-13 1008296 4.6 1015504 5.1 Acetate
EXAMPLE 1
Preparation of KALG PGATLQTPWTACQGVG - 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). Preparation of RALG PAATLQTPWTAS LGVG (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).
Molecular formula : C88H144O25N26
Preparation ofWI I PG LN PLVGGG KLYS PTS I LCG - 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).
Mass spectral analysis : Theoretical molecular weight : 2454.9 Experimental molecular weight : 2454.8 ES+
Preparation of RW L L L G L N P LV G G G R LY S PTS I L G (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).
Molecular weight (free base) : 2552
Molecular formula : C119H195O29N33
Preparation of K I L L G L N P LV G G G R LY S PTS I LG (SEQ ID NO: 7) , R L L L G L N P LV G G G R LY S PTT I L G (SEQ ID NO: 8) and N I P I P V G D I Y G G G D I Y K RW Q A L C L (SEQ ID NO: 21).
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).
Preparation of R N I P I P V G D I Y G G G D I Y K RW Q A LC 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).
Mass spectral analysis : Theoretical molecular weight : 2817.3
Experimental molecular weight : 2813.7 ES+
Preparation of RA I P I P A G T L L S G G G RA I Y K R W A I L G (SEQ ID NO: 11).
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).
Molecular weight (free base) : 2707
Molecular formula : Ci25H208O29N38 Preparation of A L P I PA G F I Y G G G R I Y K RW Q A LG (SEQ ID NO: 12), K I P I P V G F I G G G W I Y K R W A I L G (SEQ ID NO : 13) and K I P I P V G T L L S G G G R I Y K RW A I L G ( 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).
Preparation of KFI I PNI FSALGGAISYD LNTNI LNCI (SEQ ID NO : 16).
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).
Mass spectral analysis : Theoretical molecular weight : 2783.3
Experimental molecular weight : 2783.3 ES+
Preparation of KFI I PNI FSALSGGGAISYD LNTFLN CIG (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 and mass spectrometry (LDI-MS).
Mass spectral analysis : Theoretical molecular weight : 2932.4
Experimental molecular weight : 2931.8 ES+ Preparation of RFII PNI FTALSGG RRALLYGATPYAIG (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).
Molecular weight (free base) : 2894
Molecular form ula ! C137H217O32N37
Preparation of KI I PNI FSALGGG RLLYGATPYAIG (SEQ ID NO: 19), R I I P Nl F TALSGGG RLLYGATPYAIG (SEQ ID NO: 20) and W I I P Nl F S A L G G A I S Y D L N T Nl L N C I (SEQ ID NO : 22). 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 2
A vaccine comprising the peptides of the SEQ ID NOs: 3, 6, 11 and 18 was prepared (also refered to herein as Vacc-4x). 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 μΙ .
EXAMPLE 3
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 4
Toxicity studies were performed in mice and rats on the peptide composition of the vaccine in Example 2. The mouse was selected for the study to provide comparative data from a second commonly used rodent species. The test substance was a mixture 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 w/w 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 of ten 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 5
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 μΐη to 15 μΐη. 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.
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 μΙ 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 NOs: 1, 4, 9 and 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 composition further contain an adjuvant or vehicle, more preferably the adjuvant or vehicle is Monophosphoryl Lipid A (MPL ®) possibly with alum, Freund ' s adjuvant (complete or incomplete) or aluminum hydroxide. The optimal amount of adjuvant/vehicle will depend on 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 μg-lmg per kg bodyweight, preferably within 2 μg-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 100 μΙ to 200 μΙ (2 x 100 μΙ) . 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.
EXAMPLE 6
The anti-HIV p24 immune response resulting from Vacc-4x immunization could in combination with ART potentially improve immune reconstitution in patients who have not fully regained a healthy CD4 level (>600xl05/L) - Potential benefits of Vacc-4x in subjects with incomplete immune reconstitution include a possible sustained improvement in the immune response to p24 and HIV.
Potential risks include the discomfort and inconvenience associated with the immunizations and the risk of known or unknown side effects of exposure to Vacc-4x and Leukine (rhu-GM-CSF) including, most commonly, local reactions at the site of injections and fatigue (likelihood not yet determined) .
The results of non-clinical single-dose studies in mice and rats indicate that the dose levels of intravenous Vacc-4x elicited no adverse reactions and that the no effect level was in excess of 3 mg/kg, which constitutes a 500 fold safety margin over the planned human dose level .
In a rabbit study the effect of Vacc-4x was evaluated in the presence of concomitant GM-CSF, the adjuvant used in the clinical program. Local intradermal reactions such as erythema and edema were noted, however, similar effects were noted in control animals both macroscopically and histological . These local reactions were slightly more pronounced in the Vacc-4x treated animals. There were no systemic reactions in this study. These data indicate that Vacc-4x has no limiting toxicology in a model that is relevant to the proposed clinical study.
The therapeutic vaccine candidate Vacc-4x, has been studies in a Phase I and three Phase II clinical studies. The Phase I study enrolled 11 HIV-positive subjects, including nine subjects on ART. Subjects were maintained on ART (if entered on ART) ; all subjects were treated with 12 immunizations of Vacc-4x at a dose of 0.4 mg/injection over a period of 26 weeks. Immunizations were performed following injection of rhu-GM-CSF (Leucomax®
[molgramostim]) as adjuvant. All subjects experienced one or more adverse events (AEs) ; nine subjects experienced events judged related to treatment. The adverse reactions reported were mild or moderate in severity except for severe local reactions in one subject. No subjects were withdrawn due to treatment-related AEs or toxicological reactions; no serious adverse events (SAEs) occurred. Treatment related events observed in more than one subject included painful injection (seven subjects), fatigue-vertigo (four subjects), influenza-like symptoms (two subjects), and irritated skin at injection site (two subjects).
All subjects experienced a cell-mediated immune response, measured by delayed- type hypersensitivity (DTH) skin reaction. Some cell-mediated immune response, measured by γ IFN release using enzyme-linked immunosorbent spot assay (ELISPOT), was reported for
45% of the subjects; no antibody response to Vacc-4x peptides was observed.
The Phase II dose-finding study (CTN B-HIV 2/2001) enrolled 40 HIV positive subjects, of which 38 completed the trial. Subjects were maintained on ART and treated with 10 immunizations at a dose of 0.4 mg (20 subjects) or 1.2 mg (20 subjects) per Vacc 4x injection, over a period of 26 weeks. Immunizations with Vacc 4x were performed following injection of rhu-GM-CSF (Leucomax [molgramostim]) as a local adjuvant. ART was interrupted from Week 26 to Week 30 to allow exposure to the subject's own virus
(autologous immunization). ART was resumed from Week 30 to Week 38 to allow maturation of immune responses to the Vacc 4x peptides and to the subject's own virus. ART was discontinued from Week 38 to Week 52 when the study was formally concluded. Treatment- related AEs were observed in 20 subjects (8 subjects in the 0.4mg group and 12 subjects in the 1.2mg group). No SAEs were reported during the period of immunization. One subject experienced a transient vasovagal reaction in conjunction with immunization and the DTH test at Week 26 and Week 38. A second subject experienced a vasovagal reaction in conjunction with the DTH test at Week 52. For the laboratory parameters, vital signs, and performance status, no changes attributable to immunization were observed. Changes in HIV RNA, CD4 cell counts, and CD8 cell counts showed no safety concerns related to
immunization.
Immunological responses reported as DTH positive reactions were observed for all subjects. Overall, positive responses both for induration and erythema were statistically significantly higher in the high dose (HD, 1.2mg Vacc-4x) group compared to the low dose (LD, 0.4 mg Vacc 4x) group. The dose-dependent differences in DTH reactions were maintained throughout the study. T-cell proliferation appeared stable after Week 12 and demonstrated an HD advantage, consistent with the DTH results. ART was interrupted at Week 38 with planned restart when CD4 counts fell to less than 200/μΙ_ or when AIDS- or HIV related events were observed (i.e. clinical practice). DTH responses to Vacc-4x (high versus low response determined at Week 38) were associated with reduced viral loads and correspondingly improved CD4 counts at the end of the study (Week 52).
During the immunization period, CD4 counts were stable or increased. Interruption of ART resulted in reduction of CD4 counts. However, 14 weeks after the last interruption of ART (Week 52), the mean CD4 counts were still above 200 x 106 cells/L. No difference between the LD and the HD groups was observed. The majority of subjects remained off ART following completion of the study (Week 52); permission was given to follow the subjects until they resumed ART. The duration of treatment interruption was linked to immune responsiveness to the peptides. When subjects were compared to similar subjects in the Netherlands that had stopped treatment without Vacc-4x administration, a significantly slower decline in CD4 cells was noted for the Vacc-4x subjects. The median treatment interruption achieved for all the subjects that participated in the Vacc-4x Phase II clinical study was 31 months.
CTN BI Vacc-4x/2009/l was an open-label follow-up of study CTN B-HIV- 2/2001 to detemine whether a re-boost with Vacc-4x could reactivate or increase the immune response obtained during the immunization performed in the CTN B-HIV-2/2001 study. The secondary objectives were to evaluate: the in vivo immunogenicity of Vacc-4x by evaluation of DTH and to compare the DTH response to DTH in the initial study; the effect of Vacc-4x on CD4 counts, CD8 counts and HIV viral RNA; and the safety and tolerability of Vacc-4x. All 26 subjects included in the study received two booster administrations of Vacc-4x.
A total of 74 AEs were reported by 23 subjects. Most adverse events (n = 60) were scored as possibly/probably related to the study treatment. The majority (98%) of the related adverse events were mild. Two adverse events related to study treatment, one headache and one injection site indurations, were scored as moderate intensity. Itching (injection site pruritus) was the most frequent reported adverse event related to the study treatment. Nineteen patients (73%) reported this adverse event at least once. Ten of these patients reported itching related to both immunizations, while for the other nine patients it was only reported once. Five patients reported swelling related to the immunization. For three of these patients swelling was reported after both immunizations. No patient died during the study. No patient reported serious adverse events and no clinically relevant changes were recorded.
The study demonstrated that Vacc-4x peptides induced T cell responses lasting up to seven years. By re-boosting it was possible to increase killing markers, this again indicates that T cells had increased their potential to kill HIV-infected cells. Before re-boosting, all the patients had returned to CD4, CD8 and viral load levels that were similar to those before ART was stopped in the main study. Re-boosting had no negative effect on the CD4, CD8 and viral load of the patients. No safety concern was reported as a result of the re-boost of these patients.
The Phase II Study CT-BI Vacc-4x 2007/1 (EudraCT Number 2007-006302-13) was performed in US and Europe (UK, Germany, Spain and Italy). The study was a randomized, double-blind, multicenter, immunogenicity study of Vacc-4x versus placebo in patients infected with HIV-1 who have maintained an adequate response to ART. The primary objective was to evaluate the effect of Vacc-4x immunizations versus placebo on CD4 counts, T-cell function (ELISPOT, T-cell proliferative responses and intracellular cytokine staining) and the response to interruption of ART. The necessity to resume ART between the interruption of ART at Week 28 and the end of the study at Week 52, due to decreased CD4 count or increased viral loads, was monitored as one of the primary efficacy endpoints.
The study was actually planned to include approximately 345 subjects. Due to recruitment difficulties the study was amended to reduce the planned number of subjects to a minimum of 120 subjects. Based on this FDA responded to approval of the amendment "Due to the size (N = 130) and nature (no formal hypothesis testing) of this study, please understand that CBER will regard this study as exploratory and for hypothesis-generating purpose only" Based on this it was important to look more in detail into the data by including post-hoc analyses especially related to pre-ART viral load since the two groups showed very different pre-ART viral load values.
In the ITT analysis population, it was concluded that Vacc-4x did not reduce the proportion of subjects requiring resumption of ART after ART cessation at Week 28 in comparison with placebo. There was also no effect compared with placebo on the percentage change in CD4 count between Week 28 and the last CD4 assessment before resumption of ART. The time to restarting ART was similar in Vacc-4x and placebo-treated subjects.
The viral load results after ART cessation varied between subjects with evidence of favourable effects of Vacc-4x immunization over placebo. There were no significant differences in the repeated measures ANOVA for viral load over Weeks 4 to 52 when data included all evaluable subjects, irrespective of whether they were or were not taking ART. In the subgroup of subjects who remained off ART until Week 52, the average viral load was lower in the Vacc-4x-treated subjects than the placebo group. A post-hoc analysis showed the Week 52 (Last Observation Carried Forward [LOCF]) viral load to be statistically significantly lower in the Vacc-4x group than the placebo group.
The analysis of change in HIV-1 RNA from Week 28 through to Week 52 revealed a statistically significant difference between groups in favour of Vacc-4x. The AUC in those who remained off ART at Week 52 was lower in the Vacc-4x group than in the placebo group. A post-hoc analysis showed this difference in AUC to be statistically significant.
No safety concern was raised during this study. The study was supervised by a Data Safety Monitoring Board (DSMB).
EXAMPLE 7
Test of peptides together with IMiDs for increased proliferation, polyfunctionality, IL- 2 secretion and IFN-γ production.
Expansion of polyfunctional HIV-specific T-cells upon stimulation with Dendritic Cells, pre-incubated with peptides to be used according to the invention, may be studied by methods described by Keersmaecker et al. (J. Virol., 2012 86:9351-9360) and referenced therein, HIV proteins Gag or Nef, they are incubated with peptides to be used according to the invention , before they are used to stimulate T-cells in a co-culture. Keersmaecker et al. found that the presence of IMiDs (Lenalidomide (IMiD3; CC- 5013) and pomalidomide (IMiDl ; CC-4047) during in vitro T-cell stimulation with dendritic cells presenting Gag- or Nef-specific peptides, resulted in a number of improvements in the function of the T-cells. Among these were; polyfunctional HIV specific CD8+ T cells with enhanced lytic capacity, more Gag antigen epitopes recognized and at lower antigen peptide concentrations, reduced proliferation of CD4+ T cells with increased number of polyfunctional CD4+ T-cells , increased IL-2 production by CD8 T-cells, detectable IFN-γ production by CD8+ T-cells and CD4 T-cells after antigen stimulation.
"Expansion of Polyfunctional HlV-Specific T Cells upon Stimulation with mRNA Electroporated Dendritic Cells in the Presence of Immunomodulatory Drugs"
Brenda De Keersmaecker, Sabine D. Allard, Patrick Lacor, Rik Schots, Kris Thielemans, and Joeri L. Aerts
J. Virol. September 2012 86:9351-9360; published ahead of print 20 June 2012,
doi : 10.1128/JVI.00472-12
EXAMPLE 8
Suggested clinical study protocol for the test of Peptide composition comprising 4 peptides in combination with Lenalidomide and HDAC inhibitor
Immunizations (four primary immunizations and two booster immunizations) at Weeks 1, 2, 3 and 4, and booster immunizations at Weeks 12 and 13 with either:
1) Peptide composition with GM-CSF as adjuvant and Lenalidomide (CC-5013), or
2) Peptide composition with GM-CSF as adjuvant and Placebo for Lenalidomide (CC-5013).
3) Placebo
Suggested doses:
Peptide composition : 0.6, 0.9, 1.2 and 1.5 mg (Equimolar amount of each peptide)
Lenalidomide: 5,10, and 25 mg.
Subjects randomized to the Lenalidomide (CC-5013) arm will take a single oral dose of Lenalidomide (CC-5013) daily the two preceding days before immunization with the Peptide composition and on the day of each immunization.
The Peptide composition used according to this clinical trial setup consists of SEQ ID NO:3, SEQ ID NO:6, SEQ ID NO: ll, and SEQ ID NO: 18.
At week 20 subjects in all study arms will receive 20 mg panobinostat (LBH589) orally on days 1, 3, and 5 (i.e. 3 times a week) every other week for a period of 8 weeks (up to week 28) while maintaining background ART. This will be followed by a 24 week follow up period (up to week 52). Upon completion of the study, subjects may be invited to participate in an additional observational study in which ART will be interrupted to evaluate the effect of study treatment on virological control. Enrolment into this part of the study will be optional and determined by the effect of study treatments on the latent HIV-1 reservoir. (Maximum duration of treatment interruption : 16 weeks).
In summary:
Study arm 1 : Peptide composition + IMiD + HDAC (panobinostat)
Study arm 2: Peptide composition + HDAC (panobinostat)
Study arm 3 : HDAC (panobinostat)
Depletion of the viral reservoir as a result of the combination treatments according to the present invention may be quantified by for instance following the procedures set forth in Lehrman et al. (The Lancet (366), 2005, pp. 549-555) and references there in. In brief, this includes measuring in samples of patient blood obtained before, during and after treatment; p24 expression from stimulated latently infected cells, plasma HIV RNA concentration (viral load), and integrated HIV DNA by realtime PCR analysis.
EXAMPLE 9
DC/ T-CELL PROLIFERATION ASSAY
Dendritic cells (DC) were generated from monocytes isolated from buffy coat preparations from healthy blood donors. Briefly, peripheral blood mononuclear cells were separated by a density gradient centrifugation and the monocytes were then negatively isolated using the Dynabeads Untouched Human Monocytes (Invitrogen, Carlsbad, CA) following the manufacturer's instructions. The monocytes were cultured with IL-4 (20 ng/ml; Immunotools, Friesoythe; Germany) and GM-CSF (100 ng/ml; Immunotools) in X-VIV015 medium (Lonza, Basel, Switerland) for 5-6 days to generate immature DC. Cytokines were replenished every 2-3 days. The maturation of the cells was performed for 24 hours with IFN-Y (1000 IU/ml), TNF-a (50 ng/ml), IL-Ιβ (25 ng/ml) IFN-a (3000 IU/ml). After maturation, the DC were pulsed for 2 hours at 37°C with peptides at 10μg/ml, before extensive washing and co-culture with Peripheral blood mononuclear cells (PBMC) labelled with a fluorescent dye (VPD450, BD biosciences, Sam Jose, CA). Various ratios with DC:T cell were tested alongside with appropriate controls. IL-2 (50U/ml) and IL-7 (50ng/mL) (Both, Immunotools) and wells with or without IMiDs were added at the start of co-culture. At day 6-10, the level of T cell proliferation was analysed by flow cytometry. The supernatants from the co-culture wells were investigated with Luminex technology to establish any suppressor activity.
EXAMPLE 10
The peptides according to the invention used in the following examples were synthesized by Schafer-N as c-terminal amides using the Fmoc-strategy of Sheppard, (1978) J.Chem.Soc, Chem. Commun., 539.
Cell penetration assay Intracellular staining for biotinylated peptides
96-well U-bottom polystyrene plates (NUNC, cat no: 163320) were used for staining of human PBMCs. Briefly, 8ul of N- or C-terminally biotinylated peptides according to the invention (i .e. 5mM, 2.5mM & 1.25mM tested for each peptide) were incubated at 37°C for 2h with 40ul of PBMC (12.5 x 106 cells/ml) from blood donors. Cells were then washed 3x with 150ul of Cellwash (BD, cat no: 349524), followed by resuspension of each cell pellet with lOOul of Trypsin-EDTA (Sigma, cat no: T4424), then incubated at 37°C for 5 min.
Trypsinated cells were then washed 3x with 150ul of Cellwash (BD, cat no: 349524), followed by resuspension with BD Cytofix/Cytoperm™ plus (BD, cat no: 554715), then incubated at 4°C for 20 min according to manufacturer. Cells were then washed 2x with 150ul PermWash (BD, cat no: 554715). Cells were then stained with Streptavidin-APC (BD, cat no: 554067) & Anti-hCDl lc (eBioscience, cat no: 12-0116) according to manufacturer at 4°C for 30 min aiming to visualize biotinylated peptides & dendritic cells, respectively. Cells were then washed 3x with 150ul PermWash, followed by resuspension in staining buffer (BD, cat no : 554656) before flow cytometry. Dendritic cells were gated as CDllc+ events outside lymphocyte region (i.e. higher FSC & SSC signals than lymphocytes). 200 000 total cells were acquired on a FACSCanto II flow cytometer with HTS loader, and histograms for both total cells & dendritic cells with respect to peptide-fluorescence (i.e. GeoMean) were prepared.
Extracellular staining for biotinylated peptides
96-well U-bottom polystyrene plates (NUNC, cat no: 163320) were used for staining of human PBMCs. Briefly, 8ul of N- or C-terminally biotinylated peptides according to table 1 or table 2 (i .e. 5mM, 2.5mM & 1.25mM tested for each peptide; all peptides manufactured by solid phase synthesis by commercial suppliers) were incubated at 37°C for 2h with 40ul of PBMC (12.5 x 106 cells/ml) from blood donors. Cells were then washed 3x with 150ul of Cellwash (BD, cat no: 349524), then stained with Streptavidin-APC (BD, cat no: 554067) & Anti-hCDl lc (eBioscience, cat no: 12-0116) according to manufacturer at 4°C for 30 min aiming to visualize biotinylated peptides & dendritic cells, respectively. Cells were then washed 3x with 150ul of Cellwash (BD, cat no: 349524), followed by resuspension in staining buffer (BD, cat no: 554656) before flow cytometry. Dendritic cells were gated as CDl lc+ events outside lymphocyte region (i.e. higher FSC & SSC signals than lymphocytes). 200 000 total cells were acquired on a FACSCanto II flow cytometer with HTS loader, and histograms for both total cells & dendritic cells with respect to peptide-fluorescence (i.e. GeoMean) were prepared.
It was clearly seen that the CMI peptides according to the invention had improved ability to enter the cell compared to its native counterparts
The data are geomean-value of each testet peptide, as calculated by the FACS Duva software. The Geomean values by trypsinating/Cytofix/Cytoperm. :
EXAMPLE 11
Positive CTL response may alternatively be assayed by ELISPOT assay. Human IFN-gamma cytotoxic T-cell (CTL) response by ELISPOT assay Briefly, at day 1, PBMC samples from HCV patients were incubated in flasks
(430 000 PBMCs/cm2) for 2h at 37°C, 5% C02 in covering amount of culture media (RPMI 1640 Fisher Scientific; Cat No. PAAE15-039 supplemented with L- Glutamine, (MedProbe Cat. No. 13E17-605E, 10% Foetal Bovine serum (FBS), Fisher Scientific Cat. No. A15-101) and Penicillin/Streptomycin, (Fisher Acientific Cat. No. Pll-010) in order to allow adherence of monocytes. Non-adherent cells were isolated, washed, and frozen in 10% V/V DMSO in FBS until further usage. Adherent cells were carefully washed with culture media, followed by incubation at 37°C until day 3 in culture media containing 2μg/ml final concentration of hrGM-CSF (Xiamen amoytop biotech co, cat no: 3004.9090.90) & Ιμς/ηηΙ hrIL-4 (Invitrogen, Cat no: PHC0043) and optionally an immunomodulationg agent (IMiD), and this procedure was then repeated at day 6. At day 7, cultured dendritic cells (5 000-10 000 per well) were added to ELISPOT (Millipore multiscreen HTS) plates coated with 0^g/well anti-human γ Interferon together with thawed autologous non-adherent cells (200 000 per well), antigen samples (l-8ug/ml final concentration for peptide antigens; 5ug/ml final concentration for Concanavalin A (Sigma, Cat no: C7275) or PHA (Sigma, Cat no: L2769)) & anti-Anergy antibodies (0.03-0.05ug/ml final concentration for both anti-PD-1 (eBioscience, cat no: 16- 9989-82) & anti-PD-Ll (eBioscience, cat no: 16-5983-82)). Plates were incubated overnight and spots were developed according to manufacturer. Spots were read on ELISPOT reader (CTL-ImmunoSpot® S5 UV Analyzer).
EXAMPLE 12
ELISPOT assay
At day one, PBMC samples from blood donors were thawed, washed with warm medium and incubated in flasks (250000PBMCs/cm2) for 24 hours at 37°C, 5% C02 in covering amount of culture media (RPMI 1640 with ultra-glutamine, Lonza, BE12-702F701; 10% Foetal Bovine serum (FBS), Fisher Scientific Cat. No. A15-101 ; Penicillin/Streptomycin, Fisher Scientific Cat. No. Pll-010) to allow the cells to recover after thawing. At day two, the cells were added to a Falcon Microtest Tissue Culture plate, 96well flat bottom, at 500 000 cells per well in a volume of 200μΙ total medium. Parallel wells were added the indicated stimuli in duplicate and optionally an immunomodulationg agent (IMiD), or left with medium as a control for 6 days at 37°C, 5% C02. After the six days of incubation, ΙΟΟμΙ of the cell suspension were transferred to an ELISPOT (Millipore multiscreen HTS) plate coated with ^g/ml native influenza M2e protein. After a 24 hour incubation, the plate was washed four times with PBS + 0,05% Tween20, and a fifth time with PBS, 200μΙ/ννεΙΙ. A mouse Anti- human IgG or IgM biotin (Southern Biotech 9040-08 and 9020-08) was diluted in PBS with 0.5% FBS and incubated for 90 minutes at 37°C. The washing was repeated as described, before 80μΙ Streptavidin-Alkaline-Phosphatase (Sigma Aldrich, S289) was added each well and incubated at 60 minutes in the dark, at room temperature. The wells were then washed 2 times with PBS + 0.05% Tween20 and 4 times with PBS, ΖΟΟμΙ/ννεΙΙ, before the substrate, Vector Blue Alkaline Phosphatase Substrate kit III (Vector Blue, SK-5300 ) was added and let to develop for 7 minutes at room temperature. The reaction was stopped with running water, the plates let dry and the sport enumerated by an ELISPOT reader (CTL-ImmunoSpot® S5 UV Analyzer) .
ELISA
100 μΙ of antigen as indicated (pre-incubated in Coating buffer - 0.05M Na2C03 pH9.6; denoted CB - in cold at 8μg/ml 1-3 days) or just CB (background control) was used for coating wells in microtiter plates at 4°C. The microtiter plates are then washed 3x with washing buffer (PBS + 1% v/v Triton-X100; denoted WB), followed by 2h blocking at room temperature (RT) with 200 μΙ/well of blocking buffer (PBS + 1% w/v BSA) . Plates are then washed 3x with WB, followed by lh incubation at 37°C with 50-70 μΙ/well of added human (or rabbit or sheep) sera (serial dilutions ranging from 1 : 5 - 1 : 250 in dilution buffer (PBS + 1% v/v Triton-X100 + 1% w/v BSA; denoted DB)) . Plates are then washed 6x with WB, followed by lh incubation at RT with 70 μΙ/well of Alkaline Phosphatase-conjugated Protein G in DB; Calbiochem 539305) or goat anti-mouse IgG biotin (Ιμς/ιτιΙ, Southern Biotech, 1030-08. In case of the goat anti-mouse IgG biotin, the plates were washed one extra step as described, before addition of ΙΟΟμΙ Streptavidin-Alkaline-Phosphatase (Ιμς/ητιΙ, Sigma Aldrich, S289) and incubated 1 hour at RT. Plates are then washed 6x with WB, followed by 10-60 min incubation at room temperature with 100 μΙ/well of 0.3% w/v of
Phenophtalein monophosphate (Sigma P-5758) . Plates are finally quenched by adding 100 μΙ/well of Quench solution (0.1 M TRIS + 0.1M EDTA + 0.5M NaOH + 0.01% w/v NaN3;
pH 14), followed by a measurement with a ELISA reader (ASYS UVM 340) at 550 nm. The strength of the sera, i .e. the magnitude of the humoral immune response, is then reported as the dilution of sera that result in the described Optical Density (OD) value, or the OD value at the indicated dilution of sera.
Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps.
All patents and patent applications referred to herein are incorporated by reference in their entirety.
The application of which this description and claims forms part may be used as a basis for priority in respect of any subsequent application. The claims of such subsequent application may be directed to any feature or combination of features described herein. They may take the form of product, composition, process, or use claims and may include, by way of example and without limitation, the claims.

Claims

1. A peptide consisting of the amino acid sequence shown in SEQ ID NO: 18 (Vacc-10) in the form of an acetate salt.
2. The peptide of claim 1, wherein the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 6% and 16%, such as between 7% and 15%, such as between 8% and 14%, such as between 9% and 14%, such as between 9% and 13%, such as between 10% and 14%, such as between 11% and 14%.
3. A peptide consisting of the amino acid sequence shown in SEQ ID NO : 11 (Vacc-11) in the form of an acetate salt.
4. The peptide of claim 3, wherein the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 6% and 16%, such as between 7% and 15%, such as between 8% and 14%, such as between 9% and 14%, such as between 9% and 13%, such as between 10% and 14%, such as between 11% and 13%.
5. A peptide consisting of the amino acid sequence shown in SEQ ID NO: 6 (Vacc-12) in the form of an acetate salt.
6. The peptide of claim 5, wherein the acetate content of the salt is between 4% and 18%, such as between 5% and 17%, such as between 5% and 16%, such as between 5% and 15%, such as between 5% and 14%, such as between 6% and 14%, such as between 6% and 13%, such as between 7% and 12%, such as between 7% and 11%, such as between 8% and 11%, such as between 9% and 11%.
7. A peptide consisting of the amino acid sequence shown in SEQ ID NO : 3 (Vacc-13) in the form of an acetate salt.
8. The peptide of claim 7, wherein the acetate content of the salt is between 3% and 18%, such as between 3% and 17%, such as between 3% and 16%, such as between 3% and 15%, such as between 3% and 14%, such as between 3% and 13%, such as between 3% and 11%, such as between 3% and 10%, such as between 4% and 10%, such as between 4% and 9%, such as between 4% and 8%, such as between 4% and 7%, such as between 4% and 6%, such as between 4% and 5%.
9. A composition comprising one, two, three or four peptide acetate salts according to any one of claims 1 to 8.
10. A composition according to claim 9, which comprises said four peptide acetate salts defined in claims 1, 3, 5 and 7.
11. A composition according to claim 9 or 10, wherein the peptides have amide C-terminal ends of formula -C(0)NH2, or acetate salts thereof.
12. A composition according to claim 10 or 11, characterized in that the ratio of peptides is 1 : 1 : 1 : 1 w/w.
13. A composition according to any one of claims 9 to 11, wherein said one, two, three or four peptide acetate salts are in a disolved liquid state.
14. A composition according to claim 13, wherein said liquid is water.
15. A composition according to claim 14, wherein the pH of said composition is between between 3 and 8, such as between 3.5 and 7.5, such as between 4 and 7, such as between 4 and 6, such as between 5 and 6, and the osmolality is between 10 and 100, or between 15 and 25, or between 16 and 24 mOsm/kg.
16. The composition according to any one of claims 9-15, which is an immunogenic composition optionally comprising a pharmaceutically acceptable diluent, excipient, vehicle and/or one or more immunological adjuvants.
17. The composition according to any one of claims 9-16 in the form of a vaccine composition.
18. The composition according to any one of claims 9-17 in the form of a vaccine composition where the one or more adjuvants are provided either separately or in
combination with the composition.
19. The composition according to any one of claims 9-18 for use as a pharmaceutical.
20. The composition according to claim 19 for use in the treatment or prophylaxis of HIV or AIDS.
21. The composition according to any one of claims 9-18 comprising one or more further therapeutically active agents selected from an immunomodulatory compound and a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor.
22. The composition according to claim 21, wherein the immunomodulatory compound is selected from anti-PDl antibodies, such as MDX-1106 (Merck), THALOMID® (thalidomide), anti-PDl antibodies, cyclophosphamide, Levamisole, lenalidomide, CC-4047 (pomalidomide), CC-11006 (Celgene), and CC-10015 (Celgene), and immunomodulatory compounds described in any one of WO2007028047, WO2002059106, and WO2002094180.
23. The composition according to claim 22, wherein the immunomodulatory compound is lenalidomide.
24. The composition according to claim 21, wherein the reservoir purging agent is selected from M344 (4-(dimethylamino)-N-[7-(hydroxyamino)-7-oxoheptyl]benzamide), chidamide (CS055/HBI-800), 4SC-202, (4SC), Resminostat (4SC), hydroxamic acids such as vorinostat (SAHA), belinostat (PXD101), LAQ824, trichostatin A and panobinostat (LBH589); benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), cyclic tetrapeptides (such as trapoxin, such as trapoxin B), and the depsipeptides, such as romidepsin (ISTODAX), electrophilic ketones, and the aliphatic acid compounds such as phenylbutyrate, valproic acid, Oxamflatin, ITF2357 (generic givinostat), Apicidin, MC1293, CG05, and CG06; compounds that activate transcription factors including NF-KappaB, Prostratin, auranofin, bryostatin, a nontumorigenic phorbol ester, DPP (12-deoxyphorbol-13- phenylacetate), PMA, and Phorbol 12-myristate 13-acetate (PMA); Compounds that activate HIV mRNA elongation including P-TEF-b kinase and hexamethylbisacetamide (HMBA); IL-7; T-cell stimulating factors including anti-CD3/CD28 - T-cell stimulating Ab's; Kinase inhibitors including Tyrphostin A, Tyrphostin B, and Tyrphostin C;PTEN (phosphatase and tensin homologue) gene inhibitors including SF1670 (Echelon Bioscience), Disulfiram (DSF), an inhibitor of acetaldehyde dehydrogenase, Protein Tyrosine Phosphatase Inhibitors including bpV(HOpic), bpV(phen), and bpV(pic) (Calbiochem; EMD Millipore), Toll-like receptors agonists including Toll-like receptor-9 (TLR9) and Toll-like receptor-7 (TLR9) agonists, quercetin, lipoic acid, sodium butyrate, TNF-alpha, PHA and Tat.
25. The composition according to claim 24, wherein the reservoir purging agent is romidepsin.
26. An immunoassay kit for the detection of antibodies induced by HIV or an HIV specific peptide or protein in a sample of body fluid, wherein the kit comprises a peptide according to any of claims 1 to 9, or a composition according to any one of claims 9-25.
27. A method for reducing and/or delaying pathological effects of human immunodeficiency virus I (HIV) or for reducing the risk of developing acquired
immunodeficiency syndrome (AIDS) in a human infected with HIV, the method comprising administering an effective amount of a peptide according to any of claims 1 to 9, or a composition according to any one of claims 9-25.
28. The method according to claim 27, which method comprises the simultaneously, sequentially or separately in any order administering of one or more immunological adjuvants and/or one or more further therapeutically active agents selected from an
immunomodulatory compound and a reservoir purging agent, such as a histone deacetylase (HDAC) inhibitor, such as a therapeutically active agent defined in any one of claims 22-25.
PCT/EP2014/077358 2013-12-11 2014-12-11 Hiv vaccine WO2015086738A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP13196620 2013-12-11
EP13196620.2 2013-12-11

Publications (2)

Publication Number Publication Date
WO2015086738A2 true WO2015086738A2 (en) 2015-06-18
WO2015086738A3 WO2015086738A3 (en) 2015-08-06

Family

ID=49725070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/077358 WO2015086738A2 (en) 2013-12-11 2014-12-11 Hiv vaccine

Country Status (1)

Country Link
WO (1) WO2015086738A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106563124A (en) * 2016-10-19 2017-04-19 广西大学 Application of lipoic acid as vaccine adjuvant
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10688047B2 (en) 2015-11-03 2020-06-23 Italfarmaco Spa Physically and chemically stable oral suspensions of givinostat

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NO311807B1 (en) * 1999-03-04 2002-01-28 Bionor Immuno As HIV peptides, antigens, vaccine preparations, immunoassay test kits and a method for detecting antibodies induced by HIV
CA2874936A1 (en) * 2012-06-06 2013-12-12 Bionor Immuno As Vaccine
WO2015007337A1 (en) * 2013-07-19 2015-01-22 Bionor Immuno As Method for the vaccination against hiv

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10688047B2 (en) 2015-11-03 2020-06-23 Italfarmaco Spa Physically and chemically stable oral suspensions of givinostat
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
CN106563124A (en) * 2016-10-19 2017-04-19 广西大学 Application of lipoic acid as vaccine adjuvant

Also Published As

Publication number Publication date
WO2015086738A3 (en) 2015-08-06

Similar Documents

Publication Publication Date Title
AU2013273481B2 (en) HIV vaccine
US20170165321A1 (en) Method for reducing and/or delaying pathological effects of human immunodeficiency virus i (hiv) or for reducing the risk of developing acquired immunodeficiency syndrome (aids)
US20180140694A1 (en) Dosage regimen for hiv vaccine
WO2015110659A1 (en) Methods of immunization with a vaccine inducing a humoral immune response and with a vaccine inducing a cellular immune response
JP2007528368A (en) Immunogenic composition based on cyclophilin A binding site and method for developing vaccine
WO2015086738A2 (en) Hiv vaccine
JP2016208979A (en) POLYPEPTIDE(S)-CARRYING CyaA THAT ELICITS BOTH THERAPEUTIC AND PROPHYLACTIC IMMUNE RESPONSES AND USES THEREOF
WO2015007337A1 (en) Method for the vaccination against hiv
US10449245B2 (en) Method for reducing HIV-1 reservoir size using multivalent immunogen and reservoir purging agent
WO2009110950A1 (en) Attenuated listeria strain and adenovirus strain vaccines and methods of use
US10143737B2 (en) Method for the vaccination against HIV
US10682407B2 (en) Constrained immunogenic compositions and uses therefor
KR20110126636A (en) Splitting gp41
Fiorentini et al. HIV-1 matrix protein p17 modulates in vivo preactivated murine T-cell response and enhances the induction of systemic and mucosal immunity against intranasally co-administered antigens
WO2022271916A1 (en) Sars-cov-2 antigen nanoparticles and uses there of

Legal Events

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

Ref document number: 14812211

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 16.09.2016)

122 Ep: pct application non-entry in european phase

Ref document number: 14812211

Country of ref document: EP

Kind code of ref document: A2