WO2025109008A1 - Nouvelles compositions vaccinales et procédés de traitement du vhs - Google Patents
Nouvelles compositions vaccinales et procédés de traitement du vhs Download PDFInfo
- Publication number
- WO2025109008A1 WO2025109008A1 PCT/EP2024/082999 EP2024082999W WO2025109008A1 WO 2025109008 A1 WO2025109008 A1 WO 2025109008A1 EP 2024082999 W EP2024082999 W EP 2024082999W WO 2025109008 A1 WO2025109008 A1 WO 2025109008A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- fold
- hsv
- seq
- nucleic acid
- immunogenic composition
- Prior art date
- Legal status (The legal status 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 status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
- A61P31/22—Antivirals for DNA viruses for herpes viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/53—DNA (RNA) vaccination
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/545—Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55505—Inorganic adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55555—Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55561—CpG containing adjuvants; Oligonucleotide containing adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
- A61K2039/572—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/16011—Herpesviridae
- C12N2710/16611—Simplexvirus, e.g. human herpesvirus 1, 2
- C12N2710/16622—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/16011—Herpesviridae
- C12N2710/16611—Simplexvirus, e.g. human herpesvirus 1, 2
- C12N2710/16634—Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
Definitions
- the present invention relates to an immunogenic composition and a related vaccine composition comprising one or more nucleic acid(s) encoding structural proteins of Herpes Simplex Virus 2 (HSV-2) orimmunogenic fragments thereof.
- the vaccine composition may be used for the treatment and/or prevention of HSV-2 infection and is particulary beneficial for the immunological control of periodic reactivation of the virus in infected patients.
- Herpes simplex virus is a viral genus of the viral family known as Herpesviridae.
- the species that infect humans are commonly known as Herpes simplex virus 1 (HSV-1) and Herpes simplex virus 2 (HSV-2), wherein their formal names are Human herpesvirus 1 (HHV-1) and Human herpesvirus 2 (HHV-2), respectively.
- HSV-2 HSV-2 genital herpes
- Most patients infected with HSV-2 either have no or mild symptoms and thus do not know they are infected. When symptoms do occur, they typically include small blisters that break open to form painful ulcers. Flu-like symptoms, such as fever, aching, or swollen lymph nodes, may also occur. Onset is typically around 4 days after exposure with symptoms lasting up to 4 weeks. The disease is typically spread by direct genital contact with the skin surface or secretions of someone who is infected. Once infected, there is no cure. Antiviral medications may, however, prevent outbreaks or shorten outbreaks if they occur.
- genital herpes is associated with an increased risk of HIV acquisition by two- to threefold, HIV transmission on a per-sexual act basis by up to fivefold, and may account for 40- 60% of new HIV infections in high HSV-2 prevalence populations (Looker et al., 2008, Bulletin of the World Health Organization, vol. 86, pp. 805-812).
- acyclovir a synthetic acyclic purine-nucleoside analogue
- valacyclovir converted to acyclovir
- famciclovir converted to penciclovir
- the available drugs have a good safety because they are converted by viral thymidine kinase to the active drug only inside virally infected cells.
- HSV-2 can develop resistance to acyclovir through mutations in the viral gene that encodes thymidine kinase by generation of thymidine-kinase- deficient mutants or by selection of mutants with a thymidine kinase unable to phosphorylate acyclovir.
- Most clinical HSV-2 isolates resistant to acyclovir are deficient in thymidine kinase, although altered DNA polymerase has been detected in some.
- HSV-2 can lie latent in neurons for months or years before becoming active, such a therapy may be used to treat symptoms caused by HSV-2 but cannot avoid the periodic reactivation of the virus.
- HSV-2 the most effective and economical way to fight HSV would be a vaccine preventing initial infection and/or periodic reactivation of the virus.
- attempts to develop a potent HSV vaccine have focused on a limited number of antigens that have shown poor performance in clinical trials. Accordingly, there is an urgent need for a vaccine against HSV-2.
- HSV-2 vaccine based on nucleoside modified mRNAs of HSV glycoproteins (disclosed in US2020/0276300), however these are still in an early stage of development.
- Other attempts include vaccines based on surface proteins and glycoproteins of HSV-2 (disclosed in WO17157969).
- the present disclosure provides an immunogenic composition capable of eliciting a HSV- 2-specific immune response in a subject when administered to said subject, wherein said immunogenic composition comprises at least one, such as at least two, such as all three nucleic acid(s) selected from the group consisting of
- a nucleic acid encoding a UL21 protein of HSV-2 or an immunogenic fragment thereof wherein said UL11 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:1, said UL16 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:2 and said UL21 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:3.
- immunogenic composition which comprises all three nucleic acids as defined in i), ii) and iii) is particularly advantageous in the present context.
- immunogenic composition capable of eliciting a HSV-2-specific immune response in a subject when administered to said subject, wherein said immunogenic composition comprises (i) a nucleic acid encoding a UL11 protein of HSV-2 or an immunogenic fragment thereof,
- a nucleic acid encoding a UL21 protein of HSV-2 or an immunogenic fragment thereof wherein said UL11 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:1, said UL16 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:2 and said UL21 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:3.
- the immunogenic composition comprises all three nucleic acids selected from the group consisting of
- the immunogenic composition comprises all three nucleic acids selected from the group consisting of
- Herpes Simplex Virus and "HSV” are used interchangeably herein and refer generally to the viruses of the herpesviral Genus Simplexvirus, i.e. Ateline herpesvirus 1, Bovine herpesvirus 2, Cercopithecine herpesvirus 1, Cercopithecine herpesvirus 2, Cercopithecine herpesvirus 16, Human herpesvirus 1, Human herpesvirus 2, Macropodid herpesvirus 1, Macropodid herpesvirus 2, Saim broadlyne herpesvirus 1.
- Viral species of the Genus Simplex virus include viruses infecting humans, such as Herpes simplex virus 1 (HSV-1) and Herpes simplex virus 2 (HSV-2) which are also known as human herpesvirus 1 and 2 (HHV-1 and HHV-2), respectively.
- the present disclosure particularly relates to the viral species, HSV-2.
- immune response refers to a humoral and/or cell mediated immune response.
- An immune response may be induced in vivo and/or in vitro, preferably at least in vivo.
- a humoral immune response comprises a B-cell mediated antibody response.
- a cell mediated immune response comprises a T-cell mediated immune response, including but not limited to CD4+ T-cells and CD8+ T-cells.
- the ability of an antigen to elicit immune responses is called immunogenicity, which immune responses can be humoral and/or cell-mediated immune responses.
- An immune response of the present invention is preferably an immune response against HSV, in particular against HSV-2, and even more preferably an immune response against a HSV infection, in particular against HSV-2, in a subject.
- the ability to induce a humoral and/or cell mediated immune response in vivo can be determined using a guinea pig model of genital HSV-2 infection, which accurately mirrors the disease in humans and represents a system to examine pathogenesis and therapeutic efficacy of candidate antiviral compounds and vaccines. It also serves as an ideal system to address the nature of both genitalresident and neural tissue-resident immune memory.
- Genital infection of guinea pigs results in a self-limiting vulvovaginitis with neurologic manifestations mirroring those found in human disease.
- Primary disease in female guinea pigs involves virus replication in genital epithelial cells which is generally limited to eight days.
- HSV-2 recurrences may manifest as clinically apparent disease with erythematous and/or vesicular lesions on the perineum or as asymptomatic recurrences characterized by shedding of virus from the genital tract.
- Vaccine efficacy may for example be assessed using the guinea pig genital infection model.
- Animals may be infected intravaginally with SxlO 1 PFU, 5xl0 2 PFU, 5xl0 3 PFU, 5xl0 4 PFU, 5xl0 6 PFU, 5xl0 7 PFU, 5xl0 8 PFU, or 5xl0 9 PFU and preferably 5xl0 5 PFU of HSV-2 (e.g. strain MS). Animals may be immunized prior or post infection one, two, three, four, five or more times. Preferably, at day 15 post infection animals were immunized twice with 15 days interval. In general, any suitable route of administration may be used for immunization.
- animals are preferably immunized intramuscularly.
- Possible control groups are either mock-immunized with adjuvant-only (e.g. CpG 100 pg /Alum 150 pg) or with PBS (both negative controls), or with the HSV-2 dl 5-29 mutant virus strain (positive control).
- Groups that are immunized with vaccine candidates combined with the adjuvant may receive a dose of 0.1 pg, 0.5 pg, 1 pg, 2 pg, 3 pg, 4 pg, 5 pg, 10 pg, 15 pg, 25 pg, 30 pg, 35 pg, 40 pg, 50 pg, 60 pg, 70 pg, 80 pg, 90 pg, 100 pg, 150 pg, 200 pg and preferably 20 pg of the respective mRNA in each immunization round.
- vaginal swabs can be collected for evaluation of the frequency and magnitude of recurrent virus shedding, e.g.
- Vaginal swabs can be collected every 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days.
- vaginal swabs are collected every day, from day 15 post infection to day 85.
- the severity (scores 0 to 4) and duration of recurrent genital herpetic lesions are scored daily.
- the antibody responses as well as the CD4+ and CD8+ T-cell responses are determined.
- immunogenic refers to the properties of an immunogen, which is an entity capable of eliciting humoral and/or cell-mediated immune response.
- an immunogenic agent in other words an immunogen, is capable of eliciting an immune response, which may be a humoral and/or cell- mediated immune response.
- fragment of a protein, such as an immunogenic fragment, is meant to refer to a portion of the amino acid sequence of the full-length polypeptide.
- an immunogenic fragment as defined in any one of the herein discussed embodiments, refers to a fragment of an immunogenic protein which retains the same or a similar degree of immunogenicity as the immunogenicity of said protein.
- the "subject” as used herein relates to an animal, such as a mammal, which can be, for instance, a mouse, rat, guinea pig, hamster, rabbit, dog, cat, or primate.
- the subject may also be a human.
- said subject is a human subject.
- UL11 when used herein relates to a tegument protein of HSV-2.
- SEQ ID NO:1 depicts an exemplarily amino acid sequence of HSV-2 UL11, also deposited with NCBI GenBank under accession number AHG54674.1.
- UL11 also encompasses UL11 polypeptides having an amino acid sequence which shares a certain degree of identity with the amino acid sequence shown in SEQ ID NO:1 and also encompasses polypeptides having mutations relative to the reference sequence shown in SEQ ID NO:1 as described herein.
- certain degree of mismatch between two amino acid sequences has no significant bearing on the structure and function of a protein comprising any of the two amino acid sequences.
- the UL11 protein and/or said immunogenic fragment thereof translated from the nucleic acid as defined in i) is able to form a complex with one or more protein(s) selected from a group consisting of UL16, UL21, an HSV glycoprotein, such as an HSV-2 glycoprotein E (gE), and an immunogenic fragment thereof, such as a cytoplasmic tail of gE.
- the UL11 protein and/or said immunogenic fragment thereof is able to bind to the gE protein and/or said immunogenic fragment thereof.
- the binding of the UL11 protein and/or said immunogenic fragment thereof to the gE protein and/or said immunogenic fragment thereof facilitates the binding of the gE protein and/or said immunogenic fragment thereof to UL16.
- the UL11 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16 and UL21 or UL11, UL16 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL11 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16, UL21 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL11 protein of HSV-2 comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:1 and any amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, identity to SEQ ID NO:1.
- said UL11 protein of HSV-2 comprises or consists of an amino acid sequence according to SEQ ID NO:1.
- UL16 when used herein relates to a tegument protein of HSV-2.
- SEQ ID NO:2 depicts an exemplarily amino acid sequence of HSV-2 UL16, also deposited with NCBI GenBank under accession number AHG54679.1.
- UL16 also encompasses UL16 polypeptides having an amino acid sequence which shares a certain degree of identity with the amino acid sequence shown in SEQ ID NO:2 and also encompasses polypeptides having mutations relative to the reference sequence shown in SEQ ID NO:2 as described herein.
- the UL16 protein and/or said immunogenic fragment thereof translated from the nucleic acid as defined in ii) is able to form a complex with one or more protein(s) selected from a group consisting of UL11, UL21, gE and the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL16 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL16 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE; or UL16 and UL21.
- the UL16 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL16 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16 and UL21.
- the UL16 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16, UL21 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL16 protein of HSV-2 comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:2 and any amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, identity to SEQ ID NO:2.
- said UL16 protein of HSV-2 comprises or consists of an amino acid sequence according to SEQ ID NO:2.
- UL21 when used herein relates to a tegument protein of HSV-2.
- SEQ ID NO:3 depicts an exemplarily amino acid sequence of HSV-2 UL21, also deposited with NCBI GenBank under accession number AHG54684.1.
- UL21 also encompasses UL21 polypeptides having an amino acid sequence which shares a certain degree of identity with the amino acid sequence shown in SEQ ID NO:3 and also encompasses polypeptides having mutations relative to the reference sequence shown in SEQ ID NO:3 as described herein.
- the UL21 protein and/or said immunogenic fragment thereof translated from the nucleic acid as defined in iii) is able to form a complex with one or more protein(s) selected from a group consisting of UL11, UL16, a gE and the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL21 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL16 and UL21.
- the UL21 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL16, UL21 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL21 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16 and UL21.
- the UL21 protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of UL11, UL16, UL21 and gE or the immunogenic fragment of gE, such as the cytoplasmic tail of gE.
- the UL21 protein of HSV-2 comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:3 and any amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, identity to SEQ ID NO:3.
- said UL21 protein of HSV-2 comprises or consists of an amino acid sequence according to SEQ ID NO:3.
- polypeptide refers to a molecule comprising a polymer of amino acids linked together by peptide bonds. Said term is not meant herein to refer to a specific length of the molecule and is interchangeably used with the term “polypeptide”.
- polypeptide or protein also includes a "polypeptide of interest” or “protein of interest” which is expressed by the expression cassettes or vectors or can be isolated from the host cells of the invention.
- a polypeptide comprises an amino acid sequence, and, thus, sometimes a polypeptide comprising an amino acid sequence is referred to herein as a "polypeptide comprising a polypeptide sequence”.
- polypeptide sequence is interchangeably used with the term "amino acid sequence”.
- amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogues and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
- Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, y-carboxyglutamate, and O-phosphoserine.
- Amino acid analogues refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogues have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
- Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid.
- sequence identity refers to the percentage of residue matches between at least two polypeptide sequences aligned using a standardized algorithm. Such an algorithm may insert, in a standardized and reproducible way, gaps in the sequences being compared in order to optimize alignment between two sequences, and therefore achieve a more meaningful comparison of the two sequences.
- sequence identity between two amino acid sequences or between two nucleotide sequences is determined using the NCBI BLAST program version 2.3.0 (Jan-13-2016) (Altschul et al., Nucleic Acids Res. (1997) 25:3389-3402).
- % identity may for example be calculated as follows.
- the query sequence is aligned to the target sequence using the CLUSTAL W algorithm (Thompson et al, Nucleic Acids Research, 22: 4673-4680 (1994)).
- a comparison is made over the window corresponding to the shortest of the aligned sequences.
- the shortest of the aligned sequences may in some instances be the target sequence. In other instances, the query sequence may constitute the shortest of the aligned sequences.
- the amino acid residues at each position are compared, and the percentage of positions in the query sequence that have identical correspondences in the target sequence is reported as % identity.
- the properties of a polypeptide may be dependent on the sequence structure of the polypeptide and the presence and accessibility of immunogenic regions within said polypeptide. It is therefore possible to make minor changes to the sequence of amino acids in a polypeptide without affecting the function thereof.
- the disclosure encompasses uses of nucleic acids encoding modified variants of the immunogenic polypeptide as described herein, which are such that the immunogenic characteristics are retained.
- amino acid residues belonging to a certain functional grouping of amino acid residues could be exchanged for another amino acid residue from the same functional group. It is also possible, that one or several amino acid residues are exchanged for one or several amino acid residues that belong to a different functional group, provided that the resulting polypeptide retains its immunogenic properties.
- nucleic acid refers to a polymeric form of nucleotides which are usually linked from one deoxyribose or ribose to another.
- polynucleotide preferably includes single and double stranded forms of DNA or RNA.
- a nucleic acid molecule of this invention may include both sense and antisense strands of RNA (containing ribonucleotides), cDNA, genomic DNA, and synthetic forms and mixed polymers of the above.
- nucleotide bases may be modified chemically or biochemically or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those of skill in the art.
- modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analogue, internucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen, etc.), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids, etc.)
- synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen bond
- each nucleic acid as defined in i), ii) and/or iii) is present in an individual nucleic acid construct or wherein at least two of said nucleic acid as defined in i), ii) and/or iii) are present in the same nucleic acid construct, such as wherein all three of said nucleic acid as defined in i), ii) and/or iii) are present in the same nucleic acid construct. In one embodiment, at least two of said nucleic acid as defined in i), ii) and/or iii) are present in the same nucleic acid construct.
- nucleic acid as defined in i), ii) and/or iii) are present in the same nucleic acid construct. In one embodiment, each nucleic acid as defined in i), ii) and/or iii) is present in an individual nucleic acid construct.
- the nucleic acid as defined in i), ii) and/or iii) is selected from the group consisting of DNA and RNA.
- the nucleic acid as defined in i), ii) and/or iii) is DNA.
- DNA refers to a deoxyribonucleic acid that carries genetic information for one or more proteins in the context of the present disclosure. Generally, such a DNA encodes a polypeptide and is transcribed into a messenger RNA (mRNA) which in turn is translated into the encoded protein in the target cell.
- mRNA messenger RNA
- the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NO:51 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:51.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:52 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:52.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:53 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:53.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NO:51.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:52.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:53.
- the nucleic acid as defined in i), ii) and/or iii) is inserted into a plasmid or a vector. In one embodiment, the nucleic acid as defined in i), ii) and/or iii) is inserted into a plasmid. In one embodiment, the nucleic acid as defined in i), ii) and/or iii) is inserted into a vector.
- Said vector may be a viral vector. Said viral vector may be an adenoviral vector.
- the nucleic acid as defined in i), ii) and/or iii) is RNA.
- the RNA is a messenger RNA (mRNA). Said RNA may be obtained by an RNA manufacturing method, such as by in vitro transcription.
- mRNA refers to a messenger ribonucleic acid. Generally, such an mRNA encodes a polypeptide and is translated into the protein it encodes in the target cell.
- codon optimality is one feature that contributes greatly to mRNA stability. Stable mRNAs are enriched in codons designated optimal, whereas unstable mRNAs contain predominately non-optimal codons. Substitution of optimal codons with synonymous, non-optimal codons results in dramatic mRNA destabilization, while the converse substitution significantly increases stability. Codon optimality impacts ribosome translocation, connecting the processes of translation elongation and decay through codon optimality (Presnyak et al, Cell, 160(6):llll-24 (2015)). As demonstrated in the appended Examples, codon optimized versions of the herein disclosed mRNAs are useful in the immunogenic composition as disclosed herein.
- the nucleic acid as defined in i) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:4 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:4 and any codon-optimized version thereof.
- the nucleic acid as defined in i) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:4 and SEQ ID NQ:10.
- the nucleic acid as defined in i) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:4. In one particular embodiment, the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NO:10.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:5 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:5 and any codon-optimized version thereof.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:5 and SEQ ID NO:11.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:5. In one particular embodiment, the nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:11.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:6 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:6 and any codon-optimized version thereof.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:6 and SEQ ID NO:12.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:6. In one particular embodiment, the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:12.
- codon optimization refers to experimental approaches designed to improve the codon composition of a recombinant nucleic acid based on various criteria without altering the amino acid sequence encoded by the nucleic acid.
- codon-optimized refers to nucleic acid sequences comprising modified codons compared to the native naturally occurring sequences.
- sequences comprising SEQ ID NO: 10, 11 and 12. are known in the art and appreciates the present disclosure presents non-limiting examples of codon-optimized sequences, such as sequences comprising SEQ ID NO: 10, 11 and 12.
- any codon- optimized sequence of SEQ ID NO:4, 5 and 6 may be used in the context of the present invention.
- the immunogenic composition of the disclosure may further comprise one or more nucleic acid(s) encoding an HSV glycoprotein.
- said one or more nucleic acid(s) encoding an HSV glycoprotein is/are selected from a group consisting of a) a nucleic acid encoding an HSV glycoprotein D (gD) or an immunogenic fragment thereof, b) a nucleic acid encoding an HSV glycoprotein B (gB) or an immunogenic fragment thereof, and c) a nucleic acid encoding an HSV glycoprotein E (gE) or an immunogenic fragment thereof.
- said HSV gD, said HSV gB and/or said HSV gE is/are an HSV-2 glycoprotein(s). In one embodiment, said HSV gD, said HSV gB and said HSV gE are HSV-2 glycoproteins.
- said one or more nucleic acid(s) encoding an HSV glycoprotein is/are selected from a group consisting of a) a nucleic acid encoding an HSV glycoprotein D (gD) or an immunogenic fragment thereof comprising or consisting of an amino acid sequence which is at least 70%, such as at least 80%, or more identical to the amino acid sequence of SEQ ID NO:41, b) a nucleic acid encoding an HSV glycoprotein B (gB) or an immunogenic fragment thereof comprising or consisting of an amino acid sequence which is at least 70%, such as at least 80%, or more identical to the amino acid sequence of SEQ ID NO:40, and c) a nucleic acid encoding an HSV glycoprotein E (gE) or an immunogenic fragment thereof comprising or consisting of an amino acid sequence selected from a group consisting of SEQ ID NO:39, SEQ ID NO:64, SEQ ID NO:65 and any amino acid sequence having at least 70%, such as at least 80%, or more identity
- said HSV gE or the immunogenic fragment thereof as defined in c) comprises or consist of an amino acid sequence selected from a group consisting of SEQ ID NO:39, SEQ ID NO:64 and any amino acid sequence having at least 70%, such as at least 80%, or more identity to SEQ ID NO:39 and/or SEQ ID NO:64.
- said HSV gE or the immunogenic fragment thereof as defined in c) comprises or consist of an amino acid sequence selected from a group consisting of SEQ ID NO:39, SEQ ID NO:65 and any amino acid sequence having at least 70%, such as at least 80%, or more identity to SEQ ID NO:39 and/or SEQ ID NO:65.
- said HSV gE or the immunogenic fragment thereof as defined in c) comprises or consist of an amino acid sequence selected from a group consisting of SEQ ID NO:39 and any amino acid sequence having at least 70%, such as at least 80%, or more identity to SEQ ID NO:39.
- said one or more nucleic acid(s) encoding an HSV glycoprotein is/are selected from a group consisting of a) a nucleic acid encoding an HSV glycoprotein D (gD) or an immunogenic fragment thereof comprising or consisting of an amino acid sequence according to SEQ ID NO:41, b) a nucleic acid encoding an HSV glycoprotein B (gB) or an immunogenic fragment thereof comprising or consisting of an amino acid sequence according to SEQ ID NO:40, and c) a nucleic acid encoding an HSV glycoprotein E (gE) or an immunogenic fragment thereof comprising or consisting of an amino acid sequence selected from a group consisting of amino acid sequences according to SEQ ID NO:39, SEQ ID NO:64 and SEQ ID NO:65.
- said HSV gE or the immunogenic fragment thereof as defined in c) comprises or consist of an amino acid sequence selected from a group consisting of amino acid sequences according to SEQ ID NO:39 and SEQ ID NO:64. In one embodiment, said HSV gE or the immunogenic fragment thereof as defined in c) comprises or consist of an amino acid sequence selected from a group consisting of amino acid sequences according to SEQ ID NO:39 and SEQ ID NO:65. In one embodiment, said HSV gE or the immunogenic fragment thereof as defined in c) comprises or consist of an amino acid sequence according to SEQ ID NO:39.
- said immunogenic fragment of gD as defined in a) comprises or consists of an amino acid sequence according to positions 26 to 331 of the amino acid sequence according to SEQ ID NO:41 or an amino acid sequence having at least 70%, such as at least 80%, or more identity to the amino acid sequence according to SEQ ID NO:41.
- said immunogenic fragment of gD as defined in a) comprises or consists of an amino acid sequence according to positions 26 to 331 of the amino acid sequence according to SEQ ID NO:41.
- the nucleic acid as defined in a), b) and/or c) is selected from the group consisting of DNA and RNA.
- the nucleic acid as defined in a), b) and/or c) is DNA.
- the nucleic acid as defined in a) comprises a nucleic acid sequence according to SEQ ID NO:56 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:56.
- the nucleic acid as defined in b) comprises a nucleic acid sequence according to SEQ ID NO:55 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:55.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:54, SEQ ID NO:57, SEQ ID NO:58 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:54, SEQ ID NO:57 and/or SEQ ID NO:58.
- the nucleic acid as defined in a) comprises a nucleic acid sequence according to SEQ ID NO:56 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:56.
- the nucleic acid as defined in b) comprises a nucleic acid sequence according to SEQ ID NO:55 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:55.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:54, SEQ ID NO:57 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:54 and/or SEQ ID NO:57.
- the nucleic acid as defined in a) comprises a nucleic acid sequence according to SEQ ID NO:56 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:56.
- the nucleic acid as defined in b) comprises a nucleic acid sequence according to SEQ ID NO:55 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:55.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:57, SEQ ID NO:58 or any sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:57 and/or SEQ ID NO:58.
- the nucleic acid as defined in a) comprises a nucleic acid sequence according to SEQ ID NO:56. In one embodiment, the nucleic acid as defined in b) comprises a nucleic acid sequence according to SEQ ID NO:55. In one embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:54, SEQ ID NO:57 or SEQ ID NO:58. In one embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:54 or SEQ ID NO:57. In one embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:57 or SEQ ID NO:58. In one embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:57.
- the nucleic acid as defined in a), b) and/or c) is RNA.
- the RNA is a messenger RNA (mRNA). Said RNA may be obtained by an RNA manufacturing method, such as by in vitro transcription.
- the nucleic acid as defined in a) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:44 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:44 and any codon- optimized version thereof.
- the nucleic acid as defined in a) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:44 and SEQ ID NQ:50.
- the nucleic acid as defined in a) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:44. In one particular embodiment, the nucleic acid as defined in a) comprises a nucleic acid sequence according to SEQ ID NQ:50.
- the nucleic acid as defined in b) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:43 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:43 and any codon- optimized version thereof.
- the nucleic acid as defined in b) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:43 and SEQ ID NO:49.
- the nucleic acid as defined in b) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:43. In one particular embodiment, the nucleic acid as defined in b) comprises a nucleic acid sequence according to SEQ ID NO:49.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:42, SEQ ID NO:59, SEQ ID NO:66 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:42, SEQ ID NO:59, SEQ ID NO:66 and any codon-optimized version thereof.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:42, SEQ ID NO:59, SEQ ID NO:66, SEQ ID NO:48, SEQ ID NQ:60 and SEQ ID NO:62.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:42, SEQ ID NO:59 and/or SEQ ID NO:66.
- the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:48, SEQ ID NQ:60 and/or SEQ ID NO:62.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:42, SEQ ID NO:66 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:42, SEQ ID NO:66 and any codon-optimized version thereof.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:42, SEQ ID NO:66, SEQ ID NO:48 and SEQ ID NO:62. In one embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:42 and/or SEQ ID NO:66. In one particular embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:48 and/or SEQ ID NO:62.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:59, SEQ ID NO:66 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:59, SEQ ID NO:66 and any codon-optimized version thereof.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:59, SEQ ID NO:66, SEQ ID NO:48 and SEQ ID NQ:60. In one embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:59 and/or SEQ ID NO:66. In one particular embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:48 and/or SEQ ID NQ:60.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:66 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:66 and any codon- optimized version thereof.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:66 and SEQ ID NO:48.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:66. In one particular embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:48.
- SEQ ID NO:41 depicts an exemplarily an amino acid sequence of HSV-2 gD.
- the term “gD” also encompasses gD polypeptides having an amino acid sequence which shares a certain degree of identity with the amino acid sequence shown in SEQ ID NO:41 and also encompasses polypeptides having mutations relative to the reference sequence shown in SEQ ID NO:41 as described herein.
- the gD protein and/or said immunogenic fragment thereof translated from the nucleic acid sequence as defined in a) is able to form a complex with one or more protein selected from a group consisting of UL11, UL16 and UL21.
- said complex is a protein complex comprising or consisting of the gD protein or said immunogenic fragment thereof, UL11, UL16 and UL21.
- SEQ ID NQ:40 depicts an exemplarily amino acid sequence of HSV- 2 gB.
- the term “gB” also encompasses gB polypeptides having an amino acid sequence which shares a certain degree of identity with the amino acid sequence shown in SEQ ID NQ:40 and also encompasses polypeptides having mutations relative to the reference sequence shown in SEQ ID NQ:40 as described herein.
- the gB protein and/or said immunogenic fragment thereof translated from the nucleic acid sequence as defined in b) is able to form a complex with one or more protein selected from a group consisting of UL11, UL16 and UL21.
- said complex is a protein complex comprising or consisting of the gB protein or said immunogenic fragment thereof, UL11, UL16 and UL21.
- GE when used herein may sometimes be referred to as "glycoprotein E”.
- SEQ ID NO:39 depicts an exemplarily an amino acid sequence of HSV-2 gE, also deposited with NCBI GenBank under accession number AHG54732.1.
- the term “gE” also encompasses gE polypeptides having an amino acid sequence which shares a certain degree of identity with the amino acid sequence shown in SEQ ID NO:39 and also encompasses polypeptides having mutations relative to the reference sequence shown in SEQ ID NO:39 as described herein.
- the gE protein and/or said immunogenic fragment thereof translated from the nucleic acid sequence as defined in c) is able to bind UL11.
- the binding of the gE protein and/or said immunogenic fragment thereof to UL11 facilitates binding of the gE protein and/or said immunogenic fragment to UL16.
- the gE protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of the gE protein and/or said immunogenic fragment thereof, UL11 and UL16.
- the gE protein and/or said immunogenic fragment thereof is able to form a protein complex with UL16 and UL21 comprising or consisting of the gE protein and/or said immunogenic fragment thereof, UL16 and UL21.
- the gE protein and/or said immunogenic fragment thereof is able to form a protein complex comprising or consisting of the gE protein and/or said immunogenic fragment thereof, UL11, UL16 and UL21.
- said immunogenic fragment of gE according to c) comprises or consists of the cytoplasmic domain (i.e. cytoplasmic tail) of a HSV, such as an HSV-2, gE polypeptide.
- said cytoplasmic domain of the gE polypeptide comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:65 and any amino acid sequence having at least 70%, such as at least 80%, or more identity to SEQ ID NO:65.
- the nucleic acid as defined in c) encodes said cytoplasmic domain of the gE polypeptide and comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:59 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:59 and any codon-optimized version thereof.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:59 and SEQ ID NQ:60.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:59.
- the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NQ:60.ln one embodiment, said immunogenic fragment of gE according to c) comprises or consists of the extracellular domain of a HSV, such as an HSV-2, gE polypeptide.
- said cytoplasmic domain of the gE polypeptide comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:64 and any amino acid sequence having at least 70%, such as at least 80%, or more identity to SEQ ID NO:64.
- the nucleic acid as defined in c) encodes said extracellular domain of the gE polypeptide and comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:42 and any codon-optimized version thereof, such as a group consisting of SEQ ID NO:42 and any codon-optimized version thereof.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:42 and SEQ ID NO:62.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from any codon-optimized version of SEQ ID NO:42. In one particular embodiment, the nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:62.
- nucleoside(s) may be replaced by other naturally modified nucleosides or by synthetic nucleoside analogues during RNA, such as mRNA, manufacturing.
- Nucleoside modified mRNAs encode the same protein as the protein encoded by the non-nucleoside modified mRNA variant. Certain modified nucleosides are considered to enhance the stability of the mRNA and/or increase the efficiency of its translation which in turn enhances the production of the desired protein in the cell comprising the mRNA.
- nucleoside modifications are considered to alter the immunogenicity profile of vaccines which comprise such nucleoside modified mRNA, and can be particularly useful to reduce the innate immune response often elicited by non-nucleoside modified mRNA variants. This may be achieved by decreasing the recognition of the mRNA as foreign by pattern recognition receptors which would normally result in the activation of innate immunity.
- nucleoside modifications include for example 5-methoxyuridine, which is a modified nucleoside triphosphate (NTP) for incorporation into mRNA using T7 RNA polymerase. Incorporation of 5-methoxyuridine can reduce the immunogenicity of the resulting mRNA.
- 5-methylcytidine is also one example of such nucleoside modifications that involves the addition of a methyl group to cytidine.
- Additional non-limiting examples are N6-methyladenosine (m6A), which involves the addition of a methyl group to adenosine, 5-methyluridine (m5U), which involves the addition of a methyl group to uridine, 2-thiouridine (s2U), which contains a sulfur group and is known to increase mRNA stability, 2'-O-methylguanosine (m2,2G), which involves the addition of a methyl group to the 2'-oxygen of guanosine, 2'-O-methylcytidine (m2,2C), which involves the addition of a methyl group to the 2'-oxygen of cytidine, 2'-O- methyluridine (m2,2U), which involves the addition of a methyl group to uridine at the 2'-oxygen, and 5-methoxycytidine (m5A),
- Pseudouridine (abbreviated by the Greek letter psi, UJ) is an isomer of the nucleoside uridine in which the uracil is attached via a carbon-carbon instead of a nitrogencarbon glycosidic bond.
- Pseudouridine which is the most abundant RNA modification in cellular RNA, can regulate RNA expression post-transcriptionally and plays a variety of roles in the cell including translation, localization and stabilization of RNA.
- nucleoside modified in relation to a nucleic acid sequence is meant to be understood that said nucleic acid sequence comprises at least one nucleoside modification as discussed above.
- at least one uridine in a RNA sequence is replaced by pseudouridine.
- all uridines in a RNA sequence are replaced by pseudouridines.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) is a nucleoside modified mRNA comprising one or more modified nucleoside(s).
- the nucleoside modified mRNA comprises one or more pseudouridine residue(s).
- said one or more modified nucleoside(s) is/are selected from a group consisting of 5- methoxyuridine, 5-methylcytidine (m5C), N6-methyladenosine (m6A), 5- methyluridine (m5U), 2-thiouridine (s2U), 2'-O-methylguanosine (m2,2G), 2'-O- methylcytidine (m2,2C), 2'-O-methyluridine (m2,2U), 5-methoxycytidine (m5oC), 1- methylpseudouridine (m 1 ⁇ ), l-methyl-3-(3-amino-5-carboxypropyl)pseudouridine (m 1 acp 3 UJ), 2'-0-methylpseudouridine (UJm), 5-methyldihydrouridine (m 5 D) and (3- methylpse
- said one or more modified nucleoside(s) is/are modified uridine residue(s).
- said modified uridine residue(s) is/are selected from a group consisting of 5- methoxyuridine, 5-methyluridine (m5U), 2-thiouridine (s2U) and 2'-O-methyluridine (m2,2U).
- said one or more modified nucleoside(s) is/are pseudouridine residue(s).
- said pseudouridine residue(s) is/are selected from a group consisting of 1-methylpseudouridine (mlUJ), 1-methyl- 3-(3-amino-5-carboxypropyl)pseudouridine (mlacp3UJ), 2'-0-methylpseudouridine (UJm), 5-methyldihydrouridine (m5D) and (3-methylpseudouridine (m3UJ).
- said one or more modified nucleoside(s) is/are modified cytidine residue(s).
- said modified cytidine residue(s) is/are selected from a group consisting of 5-methylcytidine (m5C), 2'-O-methylcytidine (m2,2C) and 5-methoxycytidine (m5oC).
- said one or more modified nucleoside(s) is/are modified adenosine residue(s).
- said modified adenosine residue(s) is/are N6-methyladenosine(s) (m6A).
- said one or more modified nucleoside(s) is/are modified guanosine residue(s).
- said modified guanosine residue(s) is/are 2'-O- methylguanosine(s) (m2,2G).
- nucleoside modified nucleic acid sequences as defined in i) are SEQ ID NO:68 and 71; nucleoside modified nucleic acid sequences as defined ii) are SEQ ID NO:69 and 72; nucleoside modified nucleic acid sequences as defined iii) are SEQ ID NQ:70 and 73. It will be appreciated by those skilled in the art that nucleoside modifications which are not listed above but are expected to result in the above discussed advantageous characteristics of the nucleoside modified mRNA may also be applied according to the present disclosure.
- the nucleic acid as defined in i) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:4 and any codon-optimized or nucleoside modified version of SEQ ID NO:4, such as a group consisting of SEQ ID NO:4 and any codon-optimized or nucleoside modified version thereof.
- the nucleic acid as defined in i) comprises a nucleic acid sequence selected from any codon-optimized or nucleoside modified version of SEQ ID NO:4, such as any codon-optimized and nucleoside modified version of SEQ ID NO:4, such as any nucleoside modified version of SEQ ID NQ:10.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NQ:10 or SEQ ID NO:68.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according SEQ ID NO:68 or any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99% identity to SEQ ID NO:68.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:5 and any codon-optimized or nucleoside modified version of SEQ ID NO:5, such as a group consisting of SEQ ID NO:5 and any codon-optimized or nucleoside modified version thereof.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence selected from any codon-optimized or nucleoside modified version of SEQ ID NO:5, such as any codon-optimized and nucleoside modified version of SEQ ID NO:5, such as any nucleoside modified version of SEQ ID NO:11.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:11 or SEQ ID NO:69.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according SEQ ID NO:69 or any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99% identity to SEQ ID NO:69.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence selected from a group consisting of any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:6 and any codon-optimized or nucleoside modified version of SEQ ID NO:6, such as a group consisting of SEQ ID NO:6 and any codon-optimized or nucleoside modified version thereof.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence selected from any codon-optimized or nucleoside modified version of SEQ ID NO:6, such as any codon-optimized and nucleoside modified version of SEQ ID NO:6, such as any nucleoside modified version of SEQ ID NO:12.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:12 or SEQ ID NQ:70.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according SEQ ID NQ:70 or any nucleic acid sequence having at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99% identity to SEQ ID NQ:70.
- said immunogenic composition comprises a nucleic acid comprising a sequence selected from a group consisting of any codon-optimized and nucleoside modified version of SEQ ID NQ:10; a nucleic acid comprising a sequence selected from a group consisting of any codon-optimized and nucleoside modified version of SEQ ID NO:11; and a nucleic acid comprising a sequence selected from a group consisting of any codon-optimized and nucleoside modified version of SEQ ID NO:12.
- said immunogenic composition comprises a nucleic acid comprising SEQ ID NO:68, a nucleic acid comprising SEQ ID NO:69 and a nucleic acid comprising SEQ ID NQ:70.
- Polyadenylation is the addition of a poly-A tail (i.e. polyadenylated 3'-ends) to an RNA transcript, typically an mRNA.
- a poly-A tail consists of multiple adenosine monophosphates - in other words, it is a stretch of RNA that has only adenine bases.
- the poly-A tail is known to be advantageous for the nuclear export, translation and/or stability of mRNA. The tail is shortened over time in the cells and when it is short enough, the mRNA is enzymatically degraded. Without being bound by theory, such poly-A tail may thus be beneficial in the context of the present disclosure.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) comprises a poly-A tail.
- said poly-A tail comprises a nucleic acid sequence according to SEQ ID NO:36.
- an untranslated region refers to either of two sections, one on each side of a coding sequence on a strand of mRNA. If it is found on the 5' side, it is called the 5' UTR (also known as leader sequence) or if it is found on the 3' side, it is called the 3' UTR (also known as trailer sequence).
- the 5' UTR also known as leader sequence
- the 3' UTR also known as trailer sequence.
- a sequence region is recognized by the ribosome which allows the ribosome to bind and initiate translation.
- the 3' UTR which is thus found immediately following the translation stop codon, plays a critical role in translation termination as well as post- transcriptional modifications. These regions are thus involved in translation regulation and maintaining mRNA stability.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) comprises a 5' UTR, such as wherein said 5' UTR enhances translation.
- the 5' UTR comprises a nucleic acid sequence according to SEQ ID NO:37.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) comprises a 3' UTR, such as wherein said 3' UTR enhances translation.
- the 3' UTR comprises a nucleic acid sequence according to SEQ ID NO:38.
- the nucleic acid as defined in i), ii) and/or iii) comprise both a 5' UTR and a 3' UTR.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) comprises both a 5' UTR comprising a nucleic acid sequence according to SEQ ID NO:37 and a 3' UTR comprising a nucleic acid sequence according to SEQ ID NO:38.
- a 5' UTR and/or a 3' UTR into an mRNA incorporating each of these into one or more mRNA(s) as disclosed herein, may be particularly beneficial.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NO:7.
- the nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:8.
- the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:9.
- the nucleic acid may be nucleoside modified as described herein, for example pseudouridine modified, for example with 1-methyl-pseudouridine modified.
- the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NO:71.
- nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:72. In yet another particular embodiment, the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:73. In one particular embodiment of the present disclosure, the nucleic acid as defined in a) comprises a nucleic acid sequence according to SEQ ID NO:47. In one particular embodiment of the present disclosure, the nucleic acid as defined in b) comprises a nucleic acid sequence according to SEQ ID NO:46.
- the nucleic acid as defined in c) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:45, SEQ ID NO:61 and SEQ ID NO:63, such as wherein said nucleic acid as defined in c) comprises a nucleic acid sequence according to SEQ ID NO:45.
- mRNA capping is highly regulated and vital in the creation of stable and mature mRNA, which is able to undergo translation during protein synthesis.
- An mRNA cap also known as a five-prime cap (5' cap) is a specially altered nucleotide on the 5' end of the mRNA.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) comprises an mRNA cap.
- Said mRNA cap may for example be an m7GpppG cap, a 3'-0-methyl-m7GpppG cap or an anti-reverse cap analog.
- the nucleic acid as defined in i), ii) and/or iii) and/or the nucleic acid sequence as defined in a), b) and/or c) comprises a cap-independent translational enhancer.
- the present immunogenic composition differs from naturally occurring phenomena and therefore is part of patent eligible subject matter according to US patent practice.
- the present immunogenic composition differs from naturally occurring virus as it does not comprise a viral envelope and/or comprises non-naturally occurring substitution modification or non-naturally occurring nucleoside modification in comparison to the naturally occurring nucleic acids which encode for the UL11, UL16 and U21 proteins of HSV-2.
- an immunogenic composition as defined herein wherein the composition does not comprise a viral envelope.
- the immunogenic composition as defined herein does not comprise a viral capsid.
- the immunogenic composition does not comprise any HSV-2 glycoproteins or does not comprise any nucleic acids encoding such any HSV-2 glycoproteins.
- said immunogenic composition does not comprise any gE protein (SEQ ID NO:39) or fragment thereof.
- said immunogenic composition does not comprise any nucleic acid encoding gE protein (SEQ ID NO:66) or fragment thereof.
- said immunogenic composition does not comprise any gB protein (SEQ ID NQ:40) or fragment thereof.
- said immunogenic composition does not comprise any nucleic acid encoding gB protein (SEQ ID NO:43) or fragment thereof.
- said immunogenic composition does not comprise any gD protein (SEQ ID NO:41) or fragment thereof.
- said immunogenic composition does not comprise any nucleic acid encoding gD protein (SEQ ID NO:44) or fragment thereof.
- said nucleic acid as defined in i), ii) and/or iii) is selected from the group consisting of DNA and RNA, such as wherein said DNA or RNA comprises at least one non-naturally occurring substitution modification or non- naturally occurring nucleoside modification in comparison to the naturally occurring nucleic acids which encode for the UL11, UL16 and U21 proteins of HSV-2, respectively.
- said nucleic acid as defined in i) is RNA selected from the group consisting of nucleic acid sequenced that have at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99% identity to SEQ ID NO:4 and contains at least one non-naturally occurring nucleoside modification relative to SEQ ID NO:4; said nucleic acid as defined in ii) is RNA selected from the group consisting of nucleic acid sequences that have at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99 % identity to SEQ ID NO:5 and contains at least one non-naturally occurring nucleoside modification relative to SEQ ID NO:5; and/or said nucleic acid as defined in iii) is RNA selected from the group consisting of nucleic acid sequences that have at least 80%, such as at least 85%, such as at least 90%, such as
- said nucleic acid as defined in i) is RNA selected from the group consisting of nucleic acid sequences that have at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99% identity to SEQ ID NO:4 and contains at least one non-naturally occurring substitution modification relative to SEQ ID NO:4;
- said nucleic acid as defined in ii) is RNA selected from the group consisting of nucleic acid sequences that have at least 80%, such as at least 85%, such as at least 90%, such as at least 95%, such as at least 98%, such as at least 99% identity to SEQ ID NO:5 and contains at least one non-naturally occurring substitution modification relative to SEQ ID NO:5; and/or said nucleic acid as defined in iii) is RNA selected from the group consisting of nucleic acid sequences that have at least 80%, such as at least 85%, such as at least 90%, such as at least 95%,
- the present inventors have surprisingly found that the immunogenic composition as disclosed herein elicits a particularly advantageous HSV-2 specific immune response in a subject when administered to said subject.
- said advantageous HSV-2 specific immune response may be evaluated in an in vivo experimental animal model and in comparison to various types of controls, such as naive controls and controls which have been administered an alternative HSV-2 immunogenic composition known in the prior art.
- controls such as naive controls and controls which have been administered an alternative HSV-2 immunogenic composition known in the prior art.
- the immunogenic composition as disclosed herein elicits an HSV-2-specific antibody response in the subject who has been administered the immunogenic composition.
- said HSV-2-specific antibody response is a HSV-2-specific serum IgG response.
- said serum IgG response is detectable in a serum sample obtained from said subject at a dilution of at least about 10 3 , such as at least about 10 5 , such as at least about 10 6 , optionally as measured by ELISA.
- said serum IgG response is measured in comparison to serum IgG levels detectable in a control serum sample.
- control serum sample refers to a serum sample which may be obtained from a subject who has not been administered any immunogenic composition and/or vaccine composition against HSV, such as HSV-2, nor has been infected with HSV, such as HSV-2. Such subject may thus be a naive control. Said control serum sample may also be obtained from a subject who has been vaccinated against HSV, such as HSV-2, using a vaccine that does not comprise UL11, UL16 and/or UL21 proteins and/or the immunogenic composition according to the present disclosure. It is appreciated by those skilled in the art, that these subjects are considered naive with respect to the immunogenic composition according to the present disclosure.
- control serum sample is obtained from a naive control.
- control serum sample is obtained from a subject prior to the administration of the immunogenic composition as disclosed herein to the subject.
- control serum sample is also known as a "pre-bleed sample", which may be used as an internal control for evaluating the effect of administering an immunogenic composition to a subject, and is well known in the art.
- the immunogenic composition as disclosed herein is particularly superior in eliciting an HSV-2 antigenspecific T cell response upon restimulation of said subject with one or more HSV-2- specific antigens. It is to be understood that such superior HSV-2 antigen-specific T cell response is also expected to occur upon restimulation by a HSV-2 infection.
- the immunogenic composition according to the present disclosure induces an HSV-2 antigen-specific T cell response upon restimulation of said subject with one or more HSV-2-specific antigens.
- said one or more HSV-2-specific antigens comprise one or more overlapping peptides, such as a pool of overlapping peptides, derivable from amino acid sequence(s) of UL11, UL16 and/or UL21 proteins.
- said one or more overlapping peptides comprise overlapping peptides from each of said UL11, UL16 and UL21 proteins.
- said overlapping peptides are 15-mer peptides.
- said 15-mer peptides are overlapping by at least 5 amino acids, such as at least 6 amino acids, such as at least 7 amino acids, such as at least 8 amino acids, such as at least 9 amino acids, such as at least 10 amino acids, such as at least 11 amino acids. In one embodiment, said 15-mer peptides are overlapping by 8 to 11 amino acids. In one embodiment, said 15-mer peptides are overlapping by 8 amino acids. In one embodiment, said 15-mer peptides are overlapping by 11 amino acids.
- said one or more HSV-2 specific antigens may be selected based on the experimental model used for testing said HSV-2 antigenspecific T cell response.
- said one or more HSV-2-specific antigens may be non-overlapping peptides derivable from amino acid sequence(s) of UL11, UL16 and/or UL21 proteins, such as nonoverlapping peptides as defined by amino acid sequences according to SEQ ID NO:13-29.
- said one or more HSV-2-specific antigens are selected from a group consisting of peptides comprising the amino acid sequences according to SEQ ID NO:13-29.
- said one or more HSV-2-specific antigens comprise each of said peptides comprising the amino acid sequences according to SEQ ID NO:13-29.
- said skilled person is aware to design overlapping peptides covering the full amino acid sequences according to SEQ ID NO:l-3. The skilled person appreciates the concept and is able to perform the design of suitable overlapping peptides without undue burden for evaluating a human immune response based on SEQ ID NO:l-3.
- said one or more HSV-2-specific antigens are selected from a group consisting of overlapping peptides designed for ex vivo evaluation of the immune response elicited by the immunogenic composition in the subject administered said immunogenic composition, such as wherein said subject is a human subject.
- the skilled person is familiar with the concept of peptide design for stimulating an immune response in a subject and knows that it refers to the use of bioinformatical and experimental approaches for selecting suitable peptide sequences for such purpose.
- These peptides are known to be designed for efficient binding to MHC class I and/or MHC class II proteins and are known to be preferably designed for covering the entire amino acid sequences of antigenic proteins in question.
- said one or more overlapping peptides are selected from a group consisting of peptides designed for ex vivo evaluation of the immune response elicited by the immunogenic composition in the subject administered said immunogenic composition, wherein said peptides are designed based on one or more amino acid sequences according to SEQ ID NO:l-3.
- said overlapping peptides cover all three amino acid sequences according to SEQ ID NO:l-3.
- the HSV-2 antigen-specific T cell response comprises inducing an increase in the number of I N F- y, IL-2 and/or TNF-a secreting HSV-2 antigen-specific T cells. In one embodiment, the HSV-2 antigen-specific T cell response comprises inducing an increase in the number of INF-y and/or TNF-a secreting HSV-2 antigen-specific T cells. In one embodiment, the HSV-2 antigen-specific T cell response comprises inducing an increase in the number of INF-y secreting HSV-2 antigen-specific T cells.
- said increase in the number of INF-y, IL-2 and/or TNF-a secreting HSV-2 antigen-specific T cells may be measured in comparison to various types of controls.
- the skilled person will appreciate that the degree of said increase may be dependent on the in vitro or in vivo experimental model used for testing said HSV-2 antigenspecific T cell response.
- said increase may be assessed in an ex vivo experimental model, for example measured in a sample obtained by biopsy from the subject, such as wherein said subject is a human subject. For example, while an extreme increase may be observed in a laboratory setting, in a real-life situation, this increase may be substantially lower.
- control sample refers to a sample which may be obtained from a subject who has not been administered any immunogenic composition and/or vaccine composition against HSV, such as HSV-2, nor has been HSV, such as HSV-2, infected. Such subject may thus be a naive control.
- Said control sample may also be obtained from a subject who has been vaccinated against HSV, such as HSV-2, using a vaccine that does not comprise UL11, UL16 and/or UL21 proteins and/or the immunogenic composition according to the present disclosure. It is appreciated by those skilled in the art, that these subjects are considered naive with respect to the immunogenic composition according to the present disclosure.
- said control sample is obtained from a naive control.
- said control sample is obtained from a subject prior to the administration of the immunogenic composition as disclosed herein to the subject.
- Such control sample is also known as a "blank", which may be used as an internal control for evaluating the effect of administering an immunogenic composition to a subject, and is well known in the art.
- said increase in the number of INF-y secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 2.5-fold, such as at least about 5-fold, such as at least about 7.5-fold, such as at least about 10-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 50-fold, such as at least about 75-fold, such as at least about 100-fold, such as at least about 200-fold, such as at least about 250-fold, such as at least about 500-fold, such as at least about 750-fold, such as at least about 1000-fold, such as at least about 1500-fold, such as at least about 2000-fold, such as at least about 2500-fold, such as at least about 3000-fold, such as at least about 3500-fold, such as at least about 4000-fold, such as at least about 5000-fold, such as at least about 6000-fold, such as at least about 7000-fold, as measured in comparison to a control sample.
- 2-fold such as
- said increase in the number of IL-2 secreting HSV-2 antigenspecific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, as measured in comparison to said control sample.
- said increase in the number of TNF-a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, as measured in comparison to said control sample.
- the immunogenic composition as disclosed herein is unexpectedly superior in comparison to a composition comprising UL11, UL16 and UL21 proteins, such a composition comprising a multimeric protein complex comprising UL11, UL16 and UL21 proteins, as known in the prior art.
- said increase in the number of INF-y secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30- fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 50-fold, such as at least about 60-fold, such as at least about 70-fold, as measured in comparison to a control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said increase in the number of IL-2 secreting HSV-2 antigen-specific T cells is at least about 1.5-fold, such as at least about 2-fold, such as at least about 2.5-fold, such as at least about 3-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said increase in the number of TNF-a secreting HSV-2 antigen-specific T cells is at least about 1.5-fold, such as at least about 2-fold, such as at least about 2.5-fold, such as at least about 3-fold, such as at least about 3.5-fold, such as at least about 4-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- control sample obtainable from a subject administered UL11, UL16 and UL21 proteins differs from the control sample discussed above in that the subject from which the control sample is obtained has been administered a composition comprising UL11, UL16 and UL21 proteins but not the immunogenic composition according to the present disclosure.
- the immunogenic composition as disclosed herein is unexpectedly superior in terms of the HSV-2 antigen-specific T cell response.
- this superiority is particularly outstanding with respect to inducing multifunctional HSV-2 antigen-specific T cells, especially wherein said multifunctional HSV-2 antigen-specific T cells are INF-y/IL-2, INF-y/TNF-a or INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells.
- the increase in the number of multifunctional HSV-2 antigen-specific T cells may be measured in comparison to various controls, such as the control sample and the control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, as explained above.
- the present inventors have found that this remarkable superiority of the immunogenic composition as disclosed herein is particularly advantageous with respect to the therapeutic and/or prophylactic treatment of, such as the prevention of, a reactivation of a latent HSV-2 infection.
- said HSV-2 antigen-specific T cell response comprises inducing multifunctional HSV-2 antigen-specific T cells, such as wherein said HSV-2 antigen-specific T cell response comprises inducing an increase in the number of multifunctional HSV-2 antigen-specific T cells.
- said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/IL-2, IL-2/TNF-a, INF-y/TNF-a and INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells.
- said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/IL-2, INF- y/TNF-a and INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells. In one embodiment, said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/IL-2 and INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells. In one embodiment, said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/TNF-a and INF-y/IL-2/TNF-a secreting HSV- 2 antigen-specific T cells.
- said multifunctional HSV-2 antigen-specific T cells are INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells. In one embodiment, said multifunctional HSV-2 antigen-specific T cells are IL-2/TNF- a secreting HSV-2 antigen-specific T cells. In one particular embodiment, said multifunctional HSV-2 antigen-specific T cells are INF-y/IL-2 secreting HSV-2 antigenspecific T cells. In another particular embodiment, said multifunctional HSV-2 antigen-specific T cells are INF-y/TNF-a secreting HSV-2 antigen-specific T cells.
- said increase in the number of INF-y/IL-2 secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 45-fold, such as at least about 50-fold, such as at least about 100-fold, such as at least about 150- fold, such as at least about 200-fold, such as at least about 250-fold, such as at least about 300-fold, such as at least about 350-fold, such as at least about 400-fold, such as at least about 450-fold, such as at least about 500-fold,
- said increase in the number of INF-y/TNF-a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8- fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 45-fold, such as at least about 50-fold, such as at least about 100-fold, such as at least about 150-fold, such as at least about 200-fold, such as at least about 250-fold, such as at least about 300-fold, such as at least about 350-fold, such as at least about 400-fold, such as at least about 450-fold, such as 500-fold, such
- said increase in the number of IL-2/TNF- a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, as measured in comparison to said control sample.
- said increase in the number of INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells is at least about 10-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 50-fold, such as at least about 75- fold, such as at least about 100-fold, such as at least about 125-fold, such as at least about 150-fold, such as at least about 175-fold, such as at least about 200-fold, such as at least about 300-fold, such as at least about 400-fold, such as at least about 450-fold, such as at least about 500-fold, as measured in comparison to said control sample.
- said increase in the number of INF-y/IL-2 secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 11-fold, such as at least about 12-fold, such as at least about 13-fold, such as at least about 14-fold, such as at least about 15-fold, such as at least about 16-fold, such as at least about 17-fold, such as at least about 18-fold, such as at least about 19-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 40-fold, such as at least about 50-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such
- said increase in the number of INF-y/TNF-a secreting HSV-2 antigen-specific T cells is at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 45-fold, such as at least about 50-fold, such as at least about 55-fold, such as at least about 60-fold, such as at least about 65-fold, such as at least about 70-fold, such as at least about 75-fold, such as at least about 80-fold, such as at least about 100-fold, such as at least about 150-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said increase in the number of INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 10-fold, such as at least about 15-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- the present inventors have surprisingly found that the immunogenic composition as disclosed herein is particularly superior in inducing HSV-2 antigen-specific INF-y producing CD4+ and CD8+ T cells upon restimulation of the subject who has been administered the composition with one or more HSV-2-specific antigens - as discussed above. This surprising effect appears to be particularly outstanding in case of CD8+ T cells. Accordingly, in one embodiment of the present disclosure, the immunogenic composition as disclosed herein induces HSV-2 antigen-specific INF-y producing CD4+ and/or CD8+ T cells upon restimulation of the subject with one or more HSV-2-specific antigens.
- the induction of said HSV-2 antigen-specific INF-y producing CD4+ and/or CD8+ T cells may be measured in comparison to various controls.
- the present inventors have surprisingly found and demonstrated in the appended Examples, that the immunogenic composition as disclosed herein is superior in inducing HSV-2 antigen-specific INF-y producing CD4+ and/or CD8+ T cells in comparison to both naive controls as well as controls administered a composition comprising UL11, UL16 and UL21 proteins.
- the immunogenic composition as disclosed herein induces an increase in the number of HSV-2 antigen-specific INF-y producing CD4+ and/ CD8+ T cells upon restimulation of the subject with one or more HSV-2-specific antigens in comparison to the control sample.
- the immunogenic composition as disclosed herein induces an increase in the number of HSV-2 antigen-specific INF-y producing CD4+ and/ CD8+ T cells upon restimulation of the subject with one or more HSV-2-specific antigens in comparison to the control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said increase in the number of HSV-2 antigen-specific INF- y producing CD4+ T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold in comparison to the control sample.
- said increase in the number of HSV-2 antigen-specific INF-y producing CD8+ T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 11-fold, such as at least about 12-fold in comparison to the control sample.
- said increase in the number of HSV-2 antigenspecific INF-y producing CD8+ T cells is at least about 5-fold, such as at least about 6- fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 11-fold, such as at least about 12-fold in comparison to the control sample.
- said increase in the number of HSV-2 antigen-specific INF-y producing CD8+ T cells is at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 11-fold, such as at least about 12-fold in comparison to the control sample.
- said increase in the number of HSV-2 antigen-specific INF- Y producing CD4+ T cells is at least about 1.5-fold, such as at least about 2-fold, such as at least about 2.5-fold in comparison to the control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said increase in the number of HSV-2 antigen-specific INF-y producing CD8+ T cells is at least about 1.5-fold, such as at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold in comparison to the control sample to a control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said increase in the number of HSV-2 antigen-specific INF-y producing CD8+ T cells is at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold in comparison to the control sample to a control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- said HSV-2 antigenspecific INF-y producing CD8+ T cells correspond to about 14% of the total pool of CD8+ T cells in the spleen of the subject administered said composition.
- the skilled person appreciates that the proportion of INF-y producing CD8+ T cells in the spleen may be seen as representative of the entire pool of INF-y producing CD8+ T cells in said subject.
- a peripheral blood sample may be used for such analysis.
- said HSV-2 antigen-specific INF-y producing CD8+ T cells correspond to at least about 10%, such as at least about 12%, such as at least about 13%, such as at least about 14% of the total pool of CD8+ T cells in the subject administered said composition.
- said HSV-2 antigen-specific INF- y producing CD8+ T cells correspond to about 14% of the total pool of CD8+ T cells in the subject administered said composition. In one embodiment, said HSV-2 antigenspecific INF-y producing CD8+ T cells correspond to at least about 10%, such as at least about 12%, such as at least about 13%, such as at least about 14% of the total pool of CD8+ T cells in a peripheral blood sample from the subject administered said composition. In one embodiment, said HSV-2 antigen-specific INF-y producing CD8+ T cells correspond to about 14% of the total pool of CD8+ T cells in a peripheral blood sample from the subject administered said composition.
- HSV-2 antigen-specific T cell response is measured by a Fluorospot analysis.
- the present inventors found that the immunogenic composition as disclosed in the first aspect of the disclosure is surprisingly advantageous for a vaccine composition against an HSV-2 infection.
- a vaccine composition comprising the above described immunogenic composition elicits a superior therapeutic effect, such as a superior protective effect, in comparison to known HSV-2 vaccines in the prior art.
- an agent with adjuvant properties may be provided to said subject together with immunogenic compositions as disclosed herein.
- said immunogenic composition as described herein further comprises an agent with adjuvant effect.
- the agent with an adjuvant effect may be present in an immune-effective amount.
- the immunogenic compositions as disclosed herein may be useful as a medicament, as discussed below in relation to the third aspect of the disclosure.
- a vaccine composition comprising the immunogenic composition as defined in the first aspect of the present disclosure and a pharmaceutically acceptable carrier or excipient.
- Said vaccine composition may be used in the therapeutic and/or prophylactic treatment of a HSV-2 infection as described above for the immunogenic composition and as further explained below in relation to the fourth aspect of the present disclosure.
- vaccine composition as used herein relates to a composition comprising the immunogenic composition of the present disclosure which is able to prevent or treat a pathological condition associated with HSV-2 in a subject.
- a “vaccine composition” is capable of eliciting an effective therapeutic immune response in a subject in need thereof.
- the "vaccine composition” comprises the immunogenic composition as disclosed herein and may or may not include one or more additional components that enhance the immunological activity of the active component or such as buffers, reducing agents, stabilizing agents, chelating agents, bulking agents, osmotic balancing agents (tonicity agents); surfactants, polyols, anti-oxidants; lyoprotectants; anti-foaming agents; preservatives; and colorants, detergents, sodium salts, and/or antimicrobials etc.
- the vaccine composition may additionally comprise further components typical to pharmaceutical compositions.
- the vaccine of the present invention is, preferably, for human and/or veterinary use.
- the vaccine composition may be sterile and/or pyrogen-free.
- the vaccine composition may be isotonic with respect to humans.
- the vaccine composition comprises a therapeutically effective amount of the nucleic acids, such as mRNAs, of the invention as defined in i), ii) and iii).
- carrier and “excipient” are used interchangeably herein.
- Pharmaceutically acceptable carriers include, but are not limited to diluents (fillers, bulking agents, e.g. lactose, microcrystalline cellulose), disintegrants (e.g. sodium starch glycolate, croscarmellose sodium), binders (e.g. PVP, HPMC), lubricants (e.g. magnesium stearate), glidants (e.g. colloidal SiO2), solvents/co- solvents (e.g. aqueous vehicle, Propylene glycol, glycerol), buffering agents (e.g. citrate, gluconates, lactates), preservatives (e.g.
- diluents fillers, bulking agents, e.g. lactose, microcrystalline cellulose
- disintegrants e.g. sodium starch glycolate, croscarmellose sodium
- binders e.g. PVP, HPMC
- lubricants e.g. magnesium stearate
- glidants
- BHT e.g. BHT, BHA, Ascorbic acid
- wetting agents e.g. polysorbates, sorbitan esters
- anti-foaming agents e.g. Simethicone
- thickening agents e.g. methylcellulose or
- Further pharmaceutically acceptable carriers are (biodegradable) liposomes; microspheres made of the biodegradable polymer poly(D,L)-lactic-coglycolic acid (PLGA), albumin microspheres; synthetic polymers (soluble); nanofibers, protein- DNA complexes; protein conjugates; erythrocytes; or virosomes.
- Various carrier based dosage forms comprise solid lipid nanoparticles (SLNs), polymeric nanoparticles, ceramic nanoparticles, hydrogel nanoparticles, copolymerized peptide nanoparticles, nanocrystals and nanosuspensions, nanocrystals, nanotubes and nanowires, functionalized nanocarriers, nanospheres, nanocapsules, liposomes, lipid emulsions, lipid microtubules/microcylinders, lipid microbubbles, lipospheres, lipopolyplexes, inverse lipid micelles, dendrimers, ethosomes, multicomposite ultrathin capsules, aquasomes, pharmacosomes, colloidosomes, niosomes, discomes, proniosomes, microspheres, microemulsions and polymeric micelles.
- said vaccine composition further comprises an agent with adjuvant effect. In one embodiment, said vaccine composition further comprises an immune-effective amount of an agent with adjuvant effect.
- immune-effective refers a sufficient amount of an adjuvant to increase the vaccine's immunogenicity to a level high enough to effectively vaccinate a typical patient.
- an immunogenic composition as disclosed herein and/or a vaccine composition as disclosed herein may comprise an agent with adjuvant effect in an amount that is im mu no-effective.
- said adjuvant stimulates systemic or mucosal immunity. The skilled person is aware of suitable adjuvants.
- Non-limiting examples of suitable adjuvant in the context of the present disclosure include polymers of acrylic or methacrylic acid, maleic anhydride and alkenyl derivative polymers, immunostimulating sequences (ISS), an oil in water emulsion, cation lipids containing a quaternary ammonium salt, cytokines, aluminum hydroxide or aluminum phosphate, saponin or nanoparticles or any combinations or mixtures thereof.
- adjuvant refers to a substance that enhances, augments or potentiates the host's immune response (antibody and/or cell-mediated) to an antigen or fragment thereof.
- exemplary adjuvants for use in accordance with the present disclosure further include inorganic compounds such as alum, aluminum hydroxide, aluminum phosphate, calcium phosphate hydroxide, the TLR9 agonist CpG oligodeoxynucleotide, the TLR4 agonist monophosphoryl lipid (MPL), the TLR4 agonist glucopyranosyl lipid (GLA), the water in oil emulsions Montanide ISA 51 and 720, mineral oils, such as paraffin oil, virosomes, bacterial products, such as killed bacteria Bordetella pertussis, Mycobacterium bovis, toxoids, nonbacterial organics, such as squalene, thimerosal, detergents (Quil A), cytokines, such as IL
- pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the immunogenic composition according to the present disclosure.
- the term "pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, in Alfonso R Gennaro, Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472).
- the skilled person is aware of potential manners suitable for the delivery of a vaccine composition comprising nucleic acids, for example, based on Hou et al, Nature Review Materials, 6: 1078- 1094 (2021), Kowalski et al, Molecular Therapy, 27(4): 710-728 (2019), Kimura et al, Molecular Therapy, 31(8): 2360-2375 (2023) and Pardi et al, Nature Reviews, Drug Discovery, 17(4): 261-279 (2016).
- the immunogenic composition is encapsulated, attached to a carrier surface or is formulated for delivery to said subject by electroporation.
- said immunogenic composition is encapsulated, such as encapsulated in a particle.
- said particle is made of a lipid, a polymer, cholesterol and/or a cell penetrating peptide.
- said particle is a lipid particle and/or a liposome.
- said particle is a nanoparticle, such as a lipid nanoparticle.
- the vaccine composition of the present disclosure is formulated in a form suitable for physiological administration.
- the vaccine composition is formulated for intramuscular administration, subcutaneous administration, intradermal administration, intranasal administration, intravaginal administration, intrarectal administration or topical administration.
- the vaccine composition is formulated for intramuscular administration or subcutaneous administration.
- the vaccine composition is formulated for subcutaneous administration.
- the vaccine composition is formulated for intramuscular administration.
- the vaccine composition is formulated for injection.
- the vaccine composition as disclosed herein comprises the immunogenic composition in an effective amount and, as demonstrated in the appended Examples, is capable of eliciting a HSV-2 specific immune response in the subject when administered to said subject as discussed above.
- the skilled person will appreciate that the embodiments discussed above in relation to the first aspect of the present disclosure, are equally relevant and applicable to the second aspect disclosed herein. This particularly applies to embodiments relating to the HSV-2 specific immune response including the HSV-2-specific antibody response, the HSV-2 antigen-specific T cell response upon restimulation of said subject with one or more HSV-2-specific antigens and the induction of multifunctional HSV-2 antigen-specific T cells. For the sake of brevity these will not be repeated in relation to the second aspect or will only be briefly mentioned.
- the present inventors have found that the immunogenic composition and/or the vaccine composition as disclosed herein is advantageous as a medicament.
- the immunogenic composition and/or the vaccine composition according to the present disclosure including the embodiments discussed above in relation to the first and second aspect of the disclosure for use as a medicament.
- the present inventors have found that the immunogenic composition and/or the vaccine composition as disclosed herein is particularly advantageous in the therapeutic treatment and/or prophylactic treatment of an HSV-2 infection in a subject in need thereof.
- the immunogenic composition and/or the vaccine composition according to the present disclosure including the embodiments discussed above in relation to the first and second aspect of the disclosure for use in the therapeutic treatment and/or prophylactic treatment of an HSV-2 infection.
- Said use comprises administration of the immunogenic composition or the vaccine composition to a subject, such as a subject in need thereof.
- the subject in need thereof may be a subject susceptible to the HSV-2 infection or a subject already infected with HSV-2.
- said therapeutic treatment comprises ameliorating and/or eliminating symptoms associated with said HSV-2 infection.
- said prophylactic treatment comprises preventing symptoms associated with said HSV-2 infection.
- said HSV-2 infection is selected from a group consisting of a primary HSV-2 infection and a reactivation of a latent HSV-2 infection, such as wherein said HSV-2 infection is a reactivation of a latent HSV-2 infection.
- said reactivation of a latent HSV-2 infection is a flare, a recurrence and/or a HSV-2 labialis following a primary HSV-2 infection.
- said HSV-2 infection is a genital HSV-2 infection and/or an oral HSV-2 infection.
- said HSV-2 infection causes HSV-2 encephalitis.
- said therapeutic treatment and/or prophylactic treatment comprises preventing the development of HSV-2 encephalitis in said subject.
- said HSV-2 infection is a neonatal infection.
- the immunogenic composition and/or the vaccine composition of the present disclosure may be used for clearing the virus in a subject, i.e. after treatment no HSV-2 can be detected in a suitable sample obtained from the subject using suitable methods known to those of ordinary skill in the art, e.g. PCR, ELISA etc.
- the immunogenic composition and/or the vaccine composition of the present invention may be used to block primary infection, stop primary disease, block virus reactivation and re-infection, and to block latency.
- a prophylactic vaccine to prevent the first HSV-2 infection of the mother is desirable, whereas an effective therapy is needed in the case a mother is diagnosed with an active HSV-2 infection. This is important in order to prevent neonatal HSV-2 infection.
- the immunogenic composition and/or the vaccine composition according to the present disclosure may be applied as a prophylactic vaccine, e.g. for expectant mothers or children, or as a therapeutic vaccine in seropositive women to prevent subclinical reactivation at the time of delivery.
- the administration of the vaccine composition comprising at least one, such as at least two, such as all three nucleic acid(s) as defined in (i),(ii) and (iii) according to the present invention was well tolerated and induced significant IgG responses after prime and boost vaccination in an animal model system.
- the vaccine candidate showed surprisingly beneficial effects in terms of T cell response after immunization. Based on the extraordinary strong T cell responses, the present inventors consider that the immunogenic composition and/or the vaccine composition will be superior to prior art vaccine compositions. It is known that T cells are crucial for the immunologic control of HSV-2 reactivation, and play several key roles in the immune response against the virus.
- CD4+ T cells are critical for the activation of B cells and antibody class-switching, as well as for "licensing" DCs to activate CD8+ T cells.
- CD8+ T cells secrete IFN-y which performs a number of antiviral roles including as discussed in more detail in the Example section and leads to limitation of HSV viral replication, restoration of HSV-induced MHC class and chemokine production, which results in the recruitment of CD8+ T cells to the site of infection, where the cytotoxic CD8+ T cells kill cells infected by the virus.
- the present inventors found that the administration of the immunogenic composition and/or the vaccine composition as disclosed herein induced very potent induction of IFN-y secretion in subjects, which was significantly higher than responses elicited by corresponding protein vaccination, as well as lead to higher IL- 2 and TNF-a secretion. Importantly, administration of the vaccine composition as disclosed herein elicited significantly more multifunctional T cells that produced more than one cytokine: I FN-y/l L-2; IFN-y/TNF-a and IFN-y /IL-2/TNF-a.
- the immunogenic composition comprising nucleic acid(s) as defined in i), ii) and iii) and related vaccine compositions are particularly useful for superior protection against disease/initial infection as well as provide improved immunological control of HSV-2 infection in already infected patients.
- this effect is associated with the surprisingly strong induction of antigen-specific T cells, including secretion of IFN-y and large numbers of polyfunctional T cells, observed upon administration of the immunogenic composition comprising the nucleic acids as defined in i), ii) and iii), as shown in the appended Examples.
- the immunogenic composition and/or the vaccine composition according to the present disclosure including the embodiments discussed above in relation to the first and second aspect of the disclosure is provided for use in the prophylactic treatment of an HSV-2 infection in said subject.
- said prophylactic treatment comprises preventing a reactivation of a latent HSV-2 infection, such as preventing a flare, a recurrence and/or a HSV-2 labialis following a primary HSV-2 infection.
- said prophylactic treatment comprises preventing the development of HSV-2 encephalitis in said subject.
- the exact dose of the immunogenic composition and/or vaccine composition which is administered to the subject may depend on the purpose of the treatment (e.g. treatment of acute disease vs. prophylactic vaccination), route of administration, age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition, and will be ascertainable with routine experimentation by those skilled in the art.
- said administration is administration of a therapeutically effective amount of said immunogenic composition or vaccine composition.
- said effective amount of said immunogenic composition or said vaccine composition is an amount which elicits an immune response resulting in therapeutic treatment and/or prophylactic treatment of HSV-2, for example resulting in ameliorating and/or eliminating symptoms associated with said HSV-2 infection or preventing symptoms associated with said HSV-2 infection.
- said immunogenic composition or vaccine composition comprises at least one, such as at least two, such as all three nucleic acid(s) as defined in (i), (ii) and (iii) as disclosed herein.
- said immunogenic composition or vaccine composition comprises all three nucleic acids as defined in (i),(ii) and (iii) as disclosed herein.
- the above mentioned therapeutically effective dose is in the range of about 10 to 250 pg total amount of nucleic acids per administration.
- said use comprises administration of the immunogenic composition or the vaccine composition to said such subject, such as wherein said use comprises the administration of the immunogenic composition or the vaccine composition in a single dose to said subject, wherein said single dose corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- the immunogenic composition and/or the vaccine composition of the present invention may be administered to the subject one or more times, such as 2, 3, 4, 5, 6, 7, 8, 9, 10 or more times. Accordingly, in one embodiment, said use comprises multiple administration of the immunogenic composition or the vaccine composition to said subject.
- the vaccine composition of the disclosure may be used in a prime-boost regimen.
- a prime/boost vaccine is used which is composed of two or more types of vaccines including a vaccine used in primary immunization (prime or priming) and a vaccine used in booster immunization (boost or boosting).
- the vaccine used in primary immunization and the vaccine used in booster immunization may differ from each other or may be the same.
- Primary immunization and boosting immunization may be performed sequentially, this is, however, not mandatory.
- the prime/boost regimen includes, without limitation, e.g. mRNA prime/protein boost.
- the boosting composition may also be used as priming composition and said priming composition may be used as boosting composition.
- said multiple administration comprises a prime vaccination and at least one boost vaccination, such as wherein said multiple administration is a two-dose prime-boost regimen comprising said prime vaccination and one of said at least one boost vaccination.
- the time interval between said prime vaccination and said at least one boost vaccination is less than about 10 weeks, such as less than about 9 weeks, such as less than about 8 weeks, such as a time interval between about 2 weeks and about 8 weeks.
- said two-dose prime-boost regimen is administered repeatedly on an annual basis to said subject.
- said two-dose prime-boost regimen is administered within a year from the administration of the prime dose and only the boost dose is administered repeatedly on an annual basis to said subject.
- a boost dose is administered on annual or semi-annual basis to said subject.
- a prime dose and optionally a first boost dose are administered year 1
- a second boost dose is administered year 2
- subsequent doses are administered annually 1, 2 or 3 years after administration of the second boost dose.
- said boost doses can be administered year 2, 3, 4, 5 etc; or year 2, 4, 6, 8 etc; or year 5, 8, 11, 14 etc; or year 3, 6, 9, 12 etc; or year 2, 3, 5, 8; or according to any other regimen.
- the time interval between said prime vaccination and said first boost vaccination is less than about 10 weeks, such as less than about 9 weeks, such as less than about 8 weeks, such as a time interval between about 2 weeks and about 8 weeks. In one embodiment the time interval between said prime vaccination and said first boost vaccination is about 14 to about 42 days, such as is about 28 days.
- the dose of said prime vaccination corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- the dose of said at least one boost vaccination corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- the dose of said prime vaccination and of dose of said at least one boost vaccination are the same.
- the dose of said prime vaccination and of dose of said at least one boost vaccination are each between about 50 and about 150 ng, such as is about 100 ng.
- a method for therapeutic treatment and/or prophylactic treatment of an HSV-2 infection comprising administration of the immunogenic composition as disclosed herein or the vaccine composition as disclosed herein to a subject in need thereof.
- a use of the immunogenic composition as disclosed herein or the vaccine composition as disclosed herein in the manufacture of a medicament for the therapeutic treatment and/or prophylactic treatment of an HSV-2 infection in a subject susceptible thereto is provided. It is to be understood that embodiments discussed above in relation to the fourth aspect of the disclosure relating to the immunogenic composition as disclosed herein or the vaccine composition as disclosed herein for use in the therapeutic treatment and/or prophylactic treatment of an HSV-2 infection in a subject susceptible thereto are equally relevant for seventh aspect of the disclosure. For the sake of brevity, these will not be repeated herein.
- an mRNA RBT26 composition such as an mRNA RBT26 vaccine composition, comprises RBT26.1 mRNA, RBT26.2 mRNA and RBT26.3 mRNA and a protein RBT26 composition, such as a protein RBT26 vaccine composition, comprises RBT26.1 protein, RBT26.2 protein and RBT26.3 protein.
- an expression cassette includes one or more of the expression cassettes disclosed herein and reference to “the method” includes reference to equivalent steps and methods known to those of ordinary skill in the art that could be modified or substituted for the methods described herein.
- Figure 1 shows exemplary amino acid sequences with corresponding SEQ ID NO: 1
- RBT26.1 protein of HSV-2 (SEQ ID NO:1)
- RBT26.2 protein of HSV-2 (SEQ ID NO:2)
- RBT26.3 protein of HSV-2 (SEQ ID NO:3)
- exemplary nucleotide sequences with corresponding SEQ ID NOs of the following nucleotides RBT26.1 of HSV-2 RNA sequence(SEQ ID NO:4), RBT26.2 of HSV- 2 RNA sequence (SEQ ID NO:5), RBT26.3 of HSV-2 RNA sequence(SEQ ID NO:6), Codon-optimized RBT26.1 of HSV-2 RNA sequence including UTRs and polyA and without the UTRs and polyA (SEQ ID NO:7 and 10, respectively), Codon-optimized RBT26.2 of HSV-2 RNA sequence including UTRs and polyA and without the UTRs and polyA (SEQ ID NO:8 and 11, respectively), Codon-optimized RBT26.3 of HSV- 2 RNA sequence including UTRs and polyA
- Figure 2 (a) is an image of RBT26.1 (lane 4 and 5), RBT26.2 (lane 8) and RBT26.3 (lane 9) (corresponding to SEQ ID NOs: 71, 72 and 73) mRNA on nondenaturing agarose gel electrophoresis. Lanes 1 and 7 show the ladder (cat no N0362S, New England Biolabs).
- Figure 2 (b) shows Western Blot analysis of exemplary proteins derived from the mRNAs RBT26.1, RBT26.2 and RBT26.3 (SEQ ID NOs: 71, 72 and 73).
- Figure 2 (c) shows the results from SDS-PAGE gel electrophoretic analysis. Lane 1: MW marker (Cat no 161-0363, BioRad Switzerland), following lanes are different fractions collected from purification. Lanes 13 and 14 are positive controls from two independent batches. Figure 2 (d) shows Western Blot analysis of
- Figure 2 shows an example of the Western Blot performed using a polyclonal anti-RBT26 serum from previous animal studies to detect protein UL11, UL16 and UL21 (also referred to herein as protein RBT26.1, RBT26.2 and RBT26.3, respectively).
- Lane 1 is a MW marker (Cat no 161-0385, BioRad, Switzerland) and lanes 2 and 3 show purified protein at two different concentrations.
- Figure 3 (a) shows the immunization schedule and Figure 3 (b) and (c) show graphs of absolute (b) and relative (c) body weight development during in-life phase of the study of animals administered RBT26 mRNA vaccine, RBT26 protein vaccine and naive animals as indicated in the figure.
- One asterisk (*) indicates p ⁇ 0.05 and **** indicate p ⁇ 0.0001.
- Figure 4 shows graphs representing serum IgG responses after the prime (a) and boost (b) immunization as assesd by ELISA.
- Statistical analysis was performed using the non-parametric Kruskal-Wallis followed by Dunn's test for multiple comparisons.
- One asterisk (*) indicates statistical differences of p ⁇ 0.05, ** indicate p ⁇ 0.01, *** indicate p ⁇ 0.001 and **** indicate p ⁇ 0.0001.
- Figure 5 shows graphs representing secretion of IFN-y (a), IL-2 (b), and TNF-a (c) from splenocytes restimulated with the pool of RBT26-derived peptides as determined by Fluorospot analysis from mice immunized with 2, 5 or 15 pg RBT26 mRNA formulated in LNPs or with 1 or 10 g of RBT26 protein together with CpG 1826 and alum, respectively, as well as naive controls.
- Statistical analysis was performed using the non-parametric Kruskal-Wallis followed by Dunn's test for multiple comparisons.
- One asterisk (*) indicates statistical differences of p ⁇ 0.05, ** indicate p ⁇ 0.01, *** indicate p ⁇ 0.001 and **** indicate p ⁇ 0.0001.
- Figure 6 shows graphs representing secretion of I FN-y/l L-2 (a), IFN-y/TNF-a (b), and IL-2/TNF-a (c) from splenocytes restimulated with the pool of RBT26-derived peptides as determined by Fluorospot analysis from mice immunized with 2, 5 or 15 pg RBT26 mRNA formulated in LNPs or with 1 or 10 g of RBT26 protein together with CpG 1826 and alum, respectively, as well as naive controls.
- Statistical analysis was performed using the non-parametric Kruskal-Wallis followed by Dunn's test for multiple comparisons.
- One asterisk (*) indicates statistical differences of p ⁇ 0.05, ** indicate p ⁇ 0.01, *** indicate p ⁇ 0.001 and **** indicate p ⁇ 0.0001.
- Figure 7 shows graphs representing secretion of IFN-y/IL-2/TNF-a from splenocytes restimulated with the pool of RBT26-derived peptides as determined by Fluorospot analysis from mice immunized with 2, 5 or 15 pg RBT26 mRNA formulated in LNPs or with 1 or 10 g of RBT26 protein together with CpG 1826 and alum, respectively, as well as naive controls.
- Statistical analysis was performed using the non-parametric Kruskal-Wallis followed by Dunn's test for multiple comparisons.
- One asterisk (*) indicates statistical differences of p ⁇ 0.05, ** indicate p ⁇ 0.01, *** indicate p ⁇ 0.001 and **** indicate p ⁇ 0.0001.
- Figure 8 is a bargraph representing the total accumulated antigen-specific T cells determined in groups of mice immunized with 2, 5 or 15 pg RBT26 mRNA formulated in LNPs or with 1 or 10 g of RBT26 protein together with CpG 1826 and alum, respectively, as well as naive controls.
- Statistical analysis was performed using the non-parametric Kruskal-Wallis followed by Dunn's test for multiple comparisons.
- One asterisk (*) indicates statistical differences of p ⁇ 0.05, ** indicate p ⁇ 0.01, *** indicate p ⁇ 0.001 and **** indicate p ⁇ 0.0001.
- Figure 9 shows graphs representing the results obtained from intracellular staining of T cells for IFN-y and shows the fraction of CD4+ (a) and CD8+ (b) T cells that produce IFN-y after restimulation, as determined for groups of mice immunized with 2, 5 or 15 pg RBT26 mRNA formulated in LNPs or with 1 or 10 g of RBT26 protein together with CpG 1826 and alum, respectively, as well as naive controls.
- * sign indicates p ⁇ 0.05, ** indicates a p ⁇ 0.01, *** indicates a p ⁇ 0.001 as determined by Kruskal-Wallis analyses followed by Dunn's post hoc analysis for multiple comparisons.
- Figure 10 shows the shows the challenge and two dose administration schedule used in the challenge study described in Example 10.
- mRNA HSV-2 vaccine candidate RBT26 comprises the mRNAs comprising nucleotide sequences SEQ ID NO:68, 69 and 70 which encode the polypeptides RBT26.1 (SEQ ID NO:1), RBT26.2 (SEQ ID NO:2) and RBT26.3 (SEQ ID NO:3), respectively.
- mRNA RBT26 vaccine or “mRNA RBT26 vaccine candidate” refers to a composition comprising nucleotide sequences encoding the three polypeptides RBT26.1 (SEQ ID NO:1), RBT26.2 (SEQ ID NO:2) and RBT26.3 (SEQ ID NO:3).
- said three polypeptides are also referred to as UL11, UL16 and UL21, respectively.
- protein RBT26 vaccine or “protein RBT26 vaccine candidate” refers to a composition comprising three polypeptides RBT26.1 (SEQ ID NO:1), RBT26.2 (SEQ ID NO:2) and RBT26.3 (SEQ ID NO:3).
- mRNA vaccination induced very potent induction of IFN-y secretion, which was significantly higher than responses elicited by protein vaccination.
- mRNA vaccination stimulated significantly more T cells to secrete IL-2 and TNF-a than the protein vaccine. It was surprisingly found that mRNA vaccination elicited significantly more multifunctional T cells that produce more than one cytokine: I FN-y/l L-2; IFN-y/TNF-a and IFN-y/IL-2/TNF-a.
- Such polyfunctional HSV-specific T cells have been implicated as an important factor for immunologic control of herpes virus infections.
- CD8+ T cell responses were very strong consisting of up to 14 % of the total pool of CD8+ T cells in the spleen in some animals.
- RNA RBT26.1 encodes protein RBT26.1 (SEQ ID NO:1).
- Example RNA sequences encoding said protein are SEQ ID NO:4 and 10.
- SEQ ID NQ:10 is a codon optimized variant of SEQ ID NO:4.
- SEQ ID NO:68 is a nucleoside modified variant of SEQ ID NQ:10.
- the present study utilized RNA sequence according to SEQ ID NO:71, which comprises SEQ ID NO:68, untranslated regions (UTRs) and a polyA- tail.
- mRNA RBT26.2 encodes protein RBT26.2 (SEQ ID NO:2).
- Example RNA sequences encoding said protein are SEQ ID NO:5 and 11.
- SEQ ID NO:11 is a codon optimized variant of SEQ ID NO:5.
- SEQ ID NO:69 is a nucleoside modified variant of SEQ ID NO:11.
- the present study utilized RNA sequence according to SEQ ID NO:72, which comprises SEQ ID NO:69, untranslated regions (UTRs) and a polyA- tail.
- mRNA RBT26.3 encodes protein RBT26.3 (SEQ ID NO:3).
- Example RNA sequences encoding said protein are SEQ ID NO:6 and 12.
- SEQ ID NO:12 is a codon optimized variant of SEQ ID NO:6.
- SEQ ID NQ:70 is a nucleoside modified variant of SEQ ID NO:12.
- the present study utilized RNA sequence according to SEQ ID NO:73, which comprises SEQ ID NQ:70 and untranslated regions (UTRs) and a polyA-tail.
- RBT26.1, RBT26.2 and RBT26.3 mRNAs were prepared from linear DNA templates generated by PCR from plasmids encoding HSV-2 UL11, UL16 and UL21 coding sequences and 5' and 3' UTRs, respectively.
- the transcription reaction was assembled with the replacement of uridine nucleotide triphosphate with the triphosphate derivative 1- methylpseudouridine (m 1 ⁇ ).
- Custom made primers (TWIST bioscience, USA) encoding the T7 promoter region, and parts of the 5' and 3' UTRs, and the polyA sequences were used for the PCR amplification.
- the primers used were manufactured by TWIST bioscience and had the following sequences:
- DNA templates were amplified on a thermocycler using the primers described above for 35 cycles and an annealing temperature of 60°C. PCR products were verified on agarose gel electrophoresis and used as templates for the in vitro transcription (IVT) reaction using the HiScribe T7 mRNA Kit with CleanCap Reagent AG (Cat no NEB-E2080S, New England Biolab, BioNordika, Sweden) according to the manufacturer's instructions. At the end of the IVT reaction, DNase I was added to digest template DNA. Then, mRNAs were purified using LiCI precipitation according to the manufacturer's instructions and stored at -80°C until analysis.
- IVTT in vitro transcription
- the mRNAs RBT26.1, RBT26.2 and RBT26.3 (SEQ ID NOs:71, 72 and 73) were analysed on nondenaturing agarose gel electrophoresis according to standard protocol and the gel images are shown in Figure 2a.
- mRNAs were considered pure based on the A 260/280 ratio of approximately 2. mRNA aliquots were resuspended in Mi 11 i-Q. Water (Milli-Q® Type 1 Ultrapure Water Systems, Merck-Millipore, Sweden). The three mRNAs were mixed according to Table 2 below and aliquots were stored at -80 °C for subsequent use.
- LNPs were obtained from Precision Nanosystems, Vancouver, Canada, and mRNAs were formulated at their site. Briefly, a GenVoy ILM LNP composition using NanoAssemblr Ignite for intramuscular delivery of mRNA RBT26 vaccine candidate as prepared. One LNP formulation was prepared at the scale of 0.9 mg formulated mRNA at target concentration of 0.4 mg/mL. Ultra-centrifugation with Amicon device was used for downstream process. Final formulation was analyzed for size, PDI, total RNA concentration, encapsulation efficiency (%EE), and zeta potential. Stability of LNP formulation was evaluated after one freeze/thaw cycle. The final formulation was then aliquoted for in vivo studies, and stored at -80 °C.
- GenVoy-ILM lipid mix (cat. no. NWW0042, Precision Nanosystems, Canada) was used for preparation of the mRNA LNP formulation. 25 mM lipid mix was heated at 55°C to fully dissolve (5-10 min) and cooled down to RT prior to use. The three mRNAs (RBT26.1, RBT26.2, and RBT26.3) were mixed (referred to as tri-mix RNA) to prepare total amount of 995 pg at final concentration of 1 mg/mL. Next, aqueous phase of the tri-mix RNA was prepared by dilution RNase free water (cat. no. 02- 0201-0500, VWR, Canada) and PNI formulation buffer (pH 4.0) (lot. No.
- RNA stock solution having a concentration of 0.0927 mg/mL. Concentration was measured using the Nano Drop system (model AZY1915880 ThermoFisher, Canada). mRNA LNP preparation was performed using the Ignite system (NanoAssmblr
- the LNPs were processed using centrifugal filtration. Briefly, the LNP formulation volume of 37.3 mL was diluted with of PBS to (5x bulk dilution) to reach the final volume of 186.5 mL and transferred to eight 30kDa Amicon tubes (15mL) (cat. no. UFC903096, Mi llipore Sigma, Canada). This bulk diluted formulation was centrifuged at 2500 x g 4 °C for 30 mins. The centrifugation process was repeated until all the bulk diluted formulation were concentrated to total remaining volume of approximately 8 mL (approximately 1 ml in each Amicon). The formulations were diluted 5x by adding 4 mL CB1 (lot no.
- the LNP formulations were sterile filtered manually through 0.2 pm 13 mm EZflow disc syringe filters 371-2115-OEM (cat. no. 76018-866, VWR, Canada) in a biosafety cabinet into a sterile vial.
- concentration of mRNA in the formulation was confirmed after filtration using Quant-iTTM RiboGreenTM RNA Reagent kit (cat no. R11491, ThermoFisher, Canada) according to manufacturer's recommendations and subsequently adjusted in sterile CB1 buffer to three target concentrations of 0.300 mg/mL, 0.100 mg/mL and 0.040 mg/mL.
- the final products were aliquoted and transferred to manufacturing -80 °C freezer.
- Particle size, polydispersity index (PDI) and zeta potential measurements was carried out using Dynamic Light Scattering (DLS) on Zetasizer Nano-ZS or Ultra (Malvern Instruments, UK) according to manufacturer's instructions.
- DLS Dynamic Light Scattering
- 5-20 pL of a sample was suspended in 280-295 pL of PBS buffer.
- For zeta potential analyzes 5-20 pL of LNP was diluted to 1000 pL with 0.1 x PBS, pH 7.4.
- mRNA encapsulation efficiency (EE%) measurements were performed using the fluorescence-based RiboGreen 96-well plate assay (Quant-iTTM RiboGreenTM RNA Reagent kit (cat no.
- a standard curve was prepared using provided mRNA and was used to estimate the total mRNA concentration. All readings and measurements were performed using a BioTek Synergi-Hl Hybrid multi-mode reader (Agilent, Canada) at an excitation wavelength of 485 nm and an emission wavelength of 528 nm.
- the particle size of final mRNA RBT26-LNP product was 127 nm with polydispersity index (PDI) of 0.11.
- the EE% was 94% and LNPs were stable after one freeze/thaw cycle.
- Test items (LNP-formulated RBT26 mRNA) were provided in vials of 0.04 mg/mL, 0.1 mg/mL and 0.3 mg/mL in dilution buffer, to be used without further diliutions.
- HEK 293T cells (ATCC, USA) were seeded at a concentration of 0.4xl0 6 /mL in 12 well plates (Cat no 83.3921.300, Sarstedt, Switzerland) containing RPMI medium (Cat no 21875034, ThermoFisher, Switzerland) and 10 % FBS (Cat no 10500064, ThermoFisher, Switzerland), and incubated at 37 °C and 5 % CO2. The next day the cells were transfected using Invitrogen Lipofectamine MessengerM AX Transfection kit (Cat no LMRNA003, ThermoFisher, Switzerland). 2-4.5 pl of 1 pg/pl mRNA (SEQ ID NOs:71, 72 and 73) was added per well. The empty transfection wells had only the transfection reagent added, nothing was added to the negative control wells.
- the cells were harvested over the following days. To do this, the media was removed from the wells and 70 pl of chilled Thermo Scientific RIPA Lysis and Extraction Buffer (Cat no 89900, ThermoFisher, Switzerland), along with 10 pl 7x complete, EDTA-free Protease Inhibitor Cocktail (Cat no 4693132001, Sigma-Aldrich, Switzerland) was added to each well. The plate was incubated at 4 °C for 2-3 minutes, and the cells were then detached using a cell scraper. The cell-buffer mix was transferred to a 1.5 ml Eppendorf tube and incubated on ice.
- Thermo Scientific RIPA Lysis and Extraction Buffer Cat no 89900, ThermoFisher, Switzerland
- EDTA-free Protease Inhibitor Cocktail Cat no 4693132001, Sigma-Aldrich, Switzerland
- the membrane was then blocked overnight at 4 °C in 5 % BSA (Cat no 810531, Sigma-Aldrich, Switzerland) TBS containing 0.1 % Tween-20 (Cat no J60497.K2, ThermoFisher, Switzerland).
- RBT26 protein used as comparative control in the present disclosure.
- the production of the RBT26 protein was as described in WO201757969, wherein the RBT26.1 (SEQ ID NO:1), RBT26.2 (SEQ. ID N0:2) and RBT26.3 (SEQ ID N0:3) are referred as UL11, UL16 and UL21, respectively.
- RBT26.1, RBT26.2 and RBT26.3 in the form of a His-tagged trimer was expressed in Hi-5 insect cells and released from cell pellets after proper lysis.
- the trimer was subsequently purified using IMAC and a 0-500 mM imidazole buffer (Cat no J62593.AK, ThermoFisher, Switzerland) system (50 mM Hepes (Cat no 15630056, ThermoFisher, Switzerland), 500 mM NaCI (Cat no S1679, Sigma-Aldrich, Switzerland), pH 7.0, 1 mM TCEP (Cat no 75259, Sigma-Aldrich, Switzerland), 10 % glycerol (Cat no G5516, Sigma-Aldrich, Switzerland)).
- Impurities were washed out by applying 25 mM imidazole to the column.
- the trimer was then eluted with 250 mM imidazole, followed by dialysis in Hepes buffer without imidazole (50 mM Hepes, 150 mM NaCI, pH 7.0, 0.5 mM TCEP, 10 % glycerol).
- Figure 2 (c) showns an example of an SDS- PAGE gel electrophoretic analysis is shown.
- Lane 1 MW marker (Cat no 161-0363, BioRad, Switzerland), following lanes different fractions collected from purification. Lanes 13 and 14 are positive controls from two independent batches.
- Figure 2 (d) shows an example of the Western Blot performed using a polyclonal anti-RBT26 serum from previous animal studies to detect protein UL11, UL16 and UL21 (also referred to herein as protein RBT26.1, RBT26.2 and RBT26.3, respectively).
- the first lane is the MW marker (Cat no 161-0385, BioRad, Switzerland) and the following lanes show purified protein at two different concentrations.
- the protein RBT26 vaccine candidate composition was prepared as follows:
- (10) pg (8 pL) of Test item was further diluted in 142 pL PBS to a concentration of 67 pg/mL on the day of immunisation to a final volume of 150 pL. Then, 100 pL of 4 mg/mL CpG 1826 (cat. no. vac-1826-1, InvivoGen, France) was added to the solution. The capped bottle of Alhydrogel® adjuvant 2 % (cat no. vac- alu-250, InvivoGen, France) was shook well before 250 pL Alhydrogel® adjuvant 2 % was added to the antigen solution. The vaccine was mixed well by pipetting up and down for at least 5 minutes to allow Alhydrogel adjuvant 2 % to effectively adsorb to the adjuvant.
- test item For the high dose group, one hundred (100) pg (80 pL) of test item was further diluted in 70 pL PBS to a concentration of 670 pg/mL on the day of immunisation to a final volume of 150 pL. Then, 100 pL 4 mg/mL CpG 1826 will be added to the solution. The capped bottle of Alhydrogel® adjuvant 2 % was shook well before 250 pL Alhydrogel® adjuvant 2 % was added to the antigen solution. The vaccine was mixed well by pipetting up and down for at least 5 minutes to allow Alhydrogel adjuvant 2 % to effectively adsorb to the adjuvant.
- mice 45 female BALB/cAnNCrl mice were obtained from Charles River, Germany and were randomized into six groups of eight or five animals per group to receive test item according to the Table below. Animals were acclimatized to the new environment before the initiation of the study for at least 5 days. The animals were subjected to an intramuscular (i.m.) injection of vaccine candidate, with adjuvant (as shown in Table 4a and 4b below).
- Table 4a Summary of information about animals used in study.
- Vaccine administration was performed on Days 0 and 28 as outlined in Table 5 and shown in Figure 3a. Animals were injected in the gastrocnemius muscle using an 29G insulin syringe (BD micro fine + insulin syringes 0.3 ml, cat. no. 324826, VWR).
- 29G insulin syringe BD micro fine + insulin syringes 0.3 ml, cat. no. 324826, VWR.
- mice were weighed, and the health status was noted prior to each administration and at termination. As shown in Figure 3a, blood for the preparation of serum, was collected at day 21 and at termination. Animals were euthanized on day 42, whereupon terminal blood samples were collected for the preparation of serum, and spleens were excised. Thereafter animals were disposed of.
- the blood collection sites were Vena saphena or retro-orbital plexus (at termination). Prior to collection from the orbital plexus, the animals were anaesthesized with isoflurane (Cat no 170579, Apoteket, Sweden). A volume of 0.1 ml of blood was collected at day 21 or as much as possible at day 42.
- the effect of vaccination with the mRNA vaccine candidate RBT26 according to the present disclosure or the comparative protein RBT26 vaccine on health status and body weight is compared. Different doses of the vaccine compositions are investigated and compared to naive controls.
- RBT26 mRNA vaccine and RBT26 protein vaccine had no negative effect of the health status of the animals and thus was well tolerated. Importantly, all doses of the RBT26 mRNA vaccine were equally well tolerated.
- mice were immunized with 2, 5 or 15 mg of RBT26 mRNA formulated in LNPs or with 1 or 10 mg of RBT26 protein together with CpG 1826 (cat. no. vac-1826-1, InvivoGen, France) and alum (Alhydrogel adjuvant 2%, cat no. vac-alu-250, InvivoGen, France) as described above and serum IgG responses after the prime and boost immunizations were analyzed. Total antigen-specific IgG titers were determined in serum samples collected three weeks post the priming immunization and two weeks post boost by ELISA.
- ELISA was performed as follows: ELISA plates (Nunc Polysorp, cat. no. 475094 ThermoFisher, Sweden) were coated with RBT26 protein antigen diluted in PBS (cat no. 14190250, ThermoFisher, Sweden), 100 pl/well. Plates were covered and stored overnight (o.n.) at 4 °C, washed three times with PBS-Tween (0.05%) (Tween20: cat no P1379, Sigma-Aldrich, Sweden) and blocked with 5 % milk (cat no. 70166-500 G, Sigma-Aldrich, Sweden) overnight (o.n.) at 4°C or 2 hours at 37°C.
- Anti-RBT26 IgG were elicited in all animals in all vaccinated groups after one single immunization, with the highest titres observed in serum obtained from animals treated with 10 pg protein antigen, which had significantly higher titres than all mRNA vaccinated groups ( Figure 4a) after the priming immunization. After the second immunization, titers were increased in all animals approaching or exceeding end-point titers of 10 6 in all groups. Again, the highest titers were elicited in the animals treated with 10 pg protein antigen formulated in alhydrogel and CpG1826, although the differences were small and only significant compared to the naive control group and the lowest mRNA dose used (Figure 4b).
- RBT26 vaccine composition elicited anti-RBT26 antibodies at all doses tested after a single immunization. After the second immunization, animals administered the mRNA RBT26 vaccine composition exhibited end-point titers approaching or exceeding 10 6 , in line with the animals administered the protein RBT26 vaccine composition. Thus, the RBT26 mRNA vaccine and the RBT26 protein vaccines are able to elicit antibody responses to the same extent.
- mice were injected with the vaccine intramuscularly according to a predefined schedule according to Table 4b and 5.
- Analysis of T cell responses was performed on lymphocytes isolated from spleens (referred to herein as splenocytes) from immunized mice. Cells were prepared and cultured with and without stimulants (a pool of predicted MHC class I and II binding peptides derived from RBT26 as defined in SEQ ID NO:13-29). Detection of cytokines IFN-y, IL-2, and TNF-a producing lymphocytes from the spleen (splenocytes) was performed using Flourospot analysis.
- splenocytes Animals were euthanized and spleens were dissected and placed individually in PBS on ice. Each spleen was put in a 70 pm Falcon cell strainer (cat no. 352350/734-0003, VWR , Sweden) in a Petri dish, mashed, 5 ml complete RPMI-1640 (cat no. 21875-034, ThermoFisher, Sweden) was added and the cells were washed through the strainer two times. Cell suspension was collected, centrifuged at 350 x g for 5 minutes and subsequently, the supernatant was discarded and the cells were resuspended. The cells were lysed in 1 ml red blood cell lysis buffer (cat no.
- 96-well plates (supplied in Fluorospot kit Mouse FluoroSpot Plus Mouse IFN-y/IL-2/TNF-a; Cat no. FSP-414245-10, Mabtech, Sweden) were washed 3 times with sterile PBS 200 pl/well and PBS was removed; blocking was performed with 200 pl complete RPMI-1640/well for >30 min at RT.
- Antigens peptides (SEQ ID NO:13-29) and positive control (Concanavalin A (ConA), cat no C0412, Sigma-Aldrich, Sweden) were diluted in complete RPMI-1640 to a working concentration of 4 pg/ml and 100 pl of peptide antigen or ConA was added per well.
- the peptide antigens were prepared as follows:
- lyophilized antigen peptide (SEQ ID NO:13-19, obtained from GenScript, the Netherlands) was dissolved in 200 pl DMSO. When the peptides were completely dissolved, 800 pl of PBS was added. After thorough mixing, aliquots of 50 pl were made and stored at -20 °C until use. At use, peptides were diluted from stock in complete RPMI-1640 to a working concentration of 4 pg/ml.
- Detection antibodies (as specified below in Table 7) were diluted in PBS containing 0.1 % BSA. Table 7. The antibodies were diluted according to the table in PBS + 0.1% BSA.
- Mouse FluoroSpot Plus Mouse IFN-y/IL-2/TNF-a kit from Mabtech (cat no. FSP-414245-10) was used.
- Fluorescence enhancer provided in kit (cat no. FSP- 414245-10, Mabtech, Sweden) was added and left for 10 ⁇ 5 minutes at RT. Fluorescence enhancer as removed and the bottom of the plate was removed to allow the plate to dry completely and was protected from light to avoid fluorophore fading. Plates were stored in the dark and subsequently read in the IRIS plug and play automated Fluorospot reader (FluoroSpot reader (IRIS), Mabtech Plate evaluation Service) using the RAWspot technology analysis software package (Mabtech, Sweden).
- kit catalog no. FSP- 414245-10, Mabtech, Sweden
- IFN-y, IL-2, and TNF-a was determined by Fluorospot analysis after restimulation of splenocytes with a pool of 17 predicted RBT26-derived MHC class I and Il-binding peptides (SEQ ID NO:13-29) (GenScript, the Netherlands). These peptides bind efficiently to MHC class I and MHC class II without need for processing and they do not contain potentially immune activating impurities of recombinant proteins, and were not used for the immunization.
- the peptide pool containing predicted BALB/c MHC class I and II epitopes was very efficient at restimulating secretion of cytokines, especially from the mRNA vaccinated groups.
- the number of IFN-y spot forming cells was the highest in T cells isolated from animals administered the 2 and 5 g doses of the mRNA RBT26, which animals showed significantly higher numbers of IFN-y, IL-2 and TNF-a spot forming cells compared to T cells obtained from animals adminstered with 1 pg of protein RBT26 vaccine plus adjuvant, and significantly more IFN-y and TNF-a compared to the administered 10 pg of protein RBT26 vaccine plus adjuvant ( Figure 5).
- the 15 pg dose of RBT26 mRNA induced more IFN-y and TNF-a SFC compared the animals adminstered the protein RBT26 vaccine.
- the present inventors found that immunization with mRNA RBT26 vaccine candidate elicited significantly more multifunctional T cells that produced more than one cytokine: I FN-y/IL-2; IFN-y/TNF-a ( Figure 6) and IFN-y /IL- 2/TNF-a ( Figure 7) in contrast to immunization with the protein RBT26 vaccine candidate.
- polyfunctional HSV 1 or 2-specific T cells have been implicated as an important factor for immunologic control of herpes virus infections (review in Troung et al, 2019; Egan et al., 2013, Khan et al., 2017 and Kuo et al., 2014).
- the combined T cell responses ( Figure 8) after peptide restimulation indicate that protein vaccination elicited significantly fewer vaccine induced T cells.
- the data shows that adminstration of the mRNA RBT26 vaccine candidate drove very strong induction of antigen-specific T cells, including secretion of IFN-y and large numbers of polyfunctional T cells that secreted two or three of the cytokines measured.
- administration of the protein RBT26 vaccine candidate plus adjuvant elicited very few IFN-y spot forming cells and most T cells produced only single cytokines or a combination of IL-2 and TNF-a.
- the two vaccine types elicited IL-2 and TNF-a producing T cells in comparable numbers.
- the mRNA RBT26 vaccine candidate can elicit very strong induction of antigen-specific T cells, including secretion of IFN-y and large numbers of polyfunctional T cells, in contrast to the protein RBT26 vaccine candidate plus adjuvant. It was concluded that a mRNA RBT26 vaccine is expected to provide superior protection against disease/initial infection, as well as provide improved immunological control of HSV-2 infection in already infected patients.
- Splenocytes were prepared as described in Example 8. Obtained splenoctes were resuspended in cell culture media (supplier) at 2.5xl0 6 cells/ml. The splenocytes were seeded 1 ml per well into wells of a 24 well plate (Cat no. 83.3922.300, Sarstedt, Sweden) and either left unstimulated or stimulated with peptides (SEQ ID NO:13-29) as described above.
- protein transport inhibitor (cocktail of Monensin and Brefeldin A, cat. no. 00-4980-03, eBioscience, ThermoFisher, Sweden) was added and the cells were incubated an additional 6 hours. Cells were harvested and stained with anti-CD8a-PerCP-Cy5.5 (cat. no. 45-0081-82, ThermoFisher, Sweden) and anti-CD4-APC (cat no. 51-0042-82, ThermoFisher, Sweden) for 30 minutes at 4 °C, then were washed after which they were fixed and permeabilized with Intracellular Fixation & Permeabilization buffer (cat.
- Intracellular cytokine staining (ICS) for IFN-y of peptide restimulated T cells obtained from all animals showed a strong induction of IFN-y in T cells from animals administered 2 and 5 pg RBT26 mRNA vaccine candidate ( Figure 8). IFN-y production was only detected in T cells from animals administered RBT26 mRNA vaccine candidate, thus confirming the findings in the Fluorospot analysis. Further analysis revealed that both CD4+ and CD8+ T cells were responsible for the IFN-y secretion.
- the mice immunized with the lowest mRNA RBT26 dose (2 pg) had significantly higher frequencies of IFN-y producing CD4+ T cells as compared to T cells from animals immunized with the protein RBT26 vaccine candidate.
- the frequencies of vaccine-specific CD8+ T cells in the animals administered 2 and 5 pg RBT26 mRNA vaccine candidate were very high and significantly higher as compared to the protein vaccinated animals.
- the present data shows that immunization with mRNA RBT26 vaccine candidate according to the present invention and with the comparative protein RBT26 vaccine candidate are well tolerated by the treated animals, as indicated by growth curves and visual inspections during the in-life phase of the study. This conclusion was further supported by the relative body weight curves, indicating highly similar body weight development between groups as defined in Table 4b expressed as a percentage relative to the original weight of each animal. Thus, the immunization with the mRNA RBT26 vaccine candidate according to the present invention and protein RBT26 vaccine candidate did not have any negative impact on the health status or body weight of the animals.
- mRNA RBT26 vaccine candidate according to the present invention was well tolerated and induced significant IgG responses after prime and boost vaccination.
- mRNA RBT26 vaccine candidate surprisingly showed benificial effects in terms of T cell response after immunization.
- T cells are crucial for the immunologic control of HSV reactivation, and play several key roles in the immune response. Thus, the provision of a vaccine against HSV-2 which elicits strong T cell response is is particularly desirable.
- CD4+ T cells are critical for the activation of B cells and antibody classswitching, as well as for "licensing" DCs to activate CD8+ T cells.
- IFN-y which performs a number of antiviral roles including: (i) limiting HSV viral replication the induction of antiviral genes such as PKR, which inhibits translation within infected cells; (ii) restores HSV-induced MHC class I downregulation; and (iii) stimulates epithelial cell CXCL9 and CXCL10 production, which recruits CD8+ T cells to the site of infection.
- CD8+ T cells have the important role of killing virally infected cells via their cytotoxic components (e.g. perforin and granzyme), mediated through the engagement of MHC class I molecules presenting viral peptides on target cells.
- CD4+ and CD8+ T cells surround the neurons and adherent satellite cells of trigeminal ganglia and can control latency and reactivation.
- Activated (CD69+) HSV-specific effector memory CD4+ and CD8+ T cells expressing IFN-y, TNF and CCL5 are found in HSV infected ganglia and around neurons, and that CD8 T cells initially clear active lesions, then become TRM cells, that ensure reactivation is a rare occurrence through high killing efficiency and IFN-y production (reviewed Troung et al, supra).
- the present inventors have found that the mRNA RBT26 vaccine candidate according to the present invention as well as the protein RBT26 vaccine candidate elicited antigen-specific T cells, however surprisingly the adminstration of the mRNA RBT26 vaccine candidate induced very potent induction of IFN-y secretion, which was significantly higher than responses elicited by protein vaccination. In addition, adminstration of the mRNA RBT26 vaccine candidate stimulated more IL-2 and TNF-a secretion than the protein RBT26 vaccine candidate. Importantly, adminstration of the mRNA RBT26 vaccine candidate elicited significantly more multifunctional T cells that produced more than one cytokine: IFN-y/l L-2;
- IFN-y/TNF-a and IFN-y /IL- 2/TNF-a are examples of multifunctional T cells, including bifunctional and trifunctional T cells.
- Multifunctional HSV-specific T cells have been implicated as an important factor for immunologic control of herpes virus infections (Srivastava et al., 2018).
- the ability of a vaccine candidate to elicit multifuntional T cell response is particularly beneficial.
- CD8+ T cell responses were very strong consisting of up to 14 % of the total pool of CD8+ T cells in the spleen. It is further surprising that low and intermediate mRNA doses were more efficient at eliciting T cell responses than the highest dose tested in the present study.
- the present inventors surprisingly found that the mRNA RBT26 vaccine candidate according to the present disclosure is superior to its protein counterpart as evidenced by the data presented herein. Moreover, it is expected that the mRNA RBT vaccine will efficiently improve immunologic control of herpes virus infections and thus improve control of reoccurring reactivation of the latent virus.
- This example describes an in vivo study in guinea pigs comprising an HSV-2 challenge followed by therapeutic vaccination with mRNA RBT26 vaccine candidate as described herein and with the comparative protein RBT26 vaccine.
- mice 40 female 5-6 weeks of age Hartley strain guinea pigs from a commercial breeder are randomized and divided into four groups of ten individuals. After one week of acclimatization, animals are infected by intravaginal inoculation with 200 pL of a suspension containing 5 x 10 5 PFU of HSV-2 strain MS (Cat no VR-540, ATCC, USA). Once acute infection is resolved, latently infected animals are vaccinated intramuscularly twice, in the right hind calf muscle on day 15 and on day 25 postinfection.
- RNA consisting of equimolar amounts of RBT26.1 (SEQ ID NO:71), RBT 26.2 (SEQ ID NO:72) and RBT26.3 (SEQ ID NO:73) mRNA formulated in LNPs essentially as described in Example 2 (Genvoy ILM, Precision Nanosystems, Canada) by intramuscular injection of 50 pL vaccine.
- a second group is vaccinated with 20 pg RNA consisting of equimolar amounts of RBT26.1 (SEQ ID NO:71), RBT 26.2 (SEQ ID NO:72) and RBT26.3 (SEQ ID NO:73) mRNA formulated in LNPs essentially as described in Example 2 (Genvoy ILM, Precision Nanosystems, Canada) by intramuscular injection of 50 pL vaccine.
- the comparator group is immunized with 10 pg of RBT26 proteins mixed with 100 pg CpG and 150pg alum (CpG oligonucleotide (5'-TCGTCGTTGTCGTTTTGTCGTT-3' (SEQ ID NO:30)), cat no tlrl-2007, Invivogen, France; and Alhydrogel, cat no. vac-alu-250, InvivoGen, France) by intramuscular injection of 50 pL vaccine.
- the comparative protein RBT26 vaccine is prepared essentially as described in Example 4.
- a negative control group is vaccinated with mRNA/LNP diluent (Precision Nanosystems, Canada) by intramuscular injection of 50 pL.
- Guinea pigs are examined for vaginal lesions and were recorded for each individual animal on a daily basis on a scale of 0 to 4, where 0 reflects no disease, 1 reflects redness, 2 reflects a single lesion, 3 reflects coalesced lesions, and 4 reflects ulcerated lesions. Observations are carried out starting right after second immunization and throughout the study until study end at day 90 post infection. Vaginal swabs are collected weekly using a Dacron swab (cat no. dacroswab type 1; Spectrum Laboratories, USA) starting from day 0, day 2, day 7 and thereafter on a weekly basis until the end of the study.
- Dacron swab catalog no. dacroswab type 1; Spectrum Laboratories, USA
- HSV-2 DNA copy numbers in the individual samples are then determined by quantitative PCR.
- DNA is isolated from 300 pl of guinea pig vaginal swab material using DNeasy blood and tissue kits (Cat no 69504, Qiagen, Germany) .
- HSV-2 DNA copy number was determined using purified HSV-2 DNA (cat no 17-922-500, Advanced Biotechnologies, USA) and based on a standard curve that is generated with 50,000, 5,000, 500, 50, and 5 copies of DNA and run in triplicates. Each guinea pig sample is analyzed in duplicate.
- Primer and probe sequences for HSV-2 Us9 are: primer forward, 5'-GGCAGAAGCCTACTACTCGGAAA-3' (SEQ ID NO:31), and reverse 5'-CCATGCGCACGAGGAAGT-3'(SEQ ID NO:32), and probe with reporter dye 5'-FAM-CGAGGCCGCCAAC-MGBNFQ-3' (FAM, 6- carboxyfluorescein, custom made, ThermoFisher, Sweden) - wherein the reporter dye is defined by a DNA sequence region according to SEQ ID NO:33 and an amino acid sequence region according to SEQ ID NO:67.
- An immunogenic composition capable of eliciting a Herpes Simplex Virus 2 (HSV- 2) -specific immune response in a subject when administered to said subject, wherein said immunogenic composition comprises at least one, such as at least two, such as all three nucleic acid(s) selected from the group consisting of
- a nucleic acid encoding a UL21 protein of HSV-2 or an immunogenic fragment thereof wherein said UL11 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:1, said UL16 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:2 and said UL21 protein comprises an amino acid sequence having at least 80 % identity to SEQ ID NO:3.
- HSV-2 comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:1 and any amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, identity to SEQ ID NO:1.
- said UL16 protein of HSV-2 comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:2 and any amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, identity to SEQ ID NO:2.
- said UL21 protein of HSV-2 comprises or consists of an amino acid sequence selected from a group consisting of SEQ ID NO:3 and any amino acid sequence having at least 80%, such as at least 81%, such as at least 82%, such as at least 83%, such as at least 84%, such as at least 85%, such as at least 86%, such as at least 87%, such as at least 88%, such as at least 89%, such as at least 90%, such as at least 91%, such as at least 92%, such as at least 93%, such as at least 94%, such as at least 95%, such as at least 96%, such as at least 97%, such as at least 98%, such as at least 99%, identity to SEQ ID NO:3.
- each nucleic acid as defined in i), ii) and/or iii) is present in an individual nucleic acid construct or wherein at least two of said nucleic acid as defined in i), ii) and/or iii) are present in the same nucleic acid construct, such as wherein all three of said nucleic acid as defined in i), ii) and/or iii) are present in the same nucleic acid construct.
- each nucleic acid as defined in i), ii) and/or iii) is present in an individual nucleic acid construct.
- nucleic acid as defined in i), ii) and/or iii) is selected from the group consisting of DNA and RNA.
- nucleic acid as defined in i), ii) and/or iii) is DNA, such as wherein the nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NO:51, the nucleic acid as defined in ii) comprises a nucleic acid sequence according to SEQ ID NO:52 and/or the nucleic acid as defined in iii) comprises a nucleic acid sequence according to SEQ ID NO:53.
- RNA such as mRNA
- RNA obtained by an RNA manufacturing method, such as by in vitro transcription.
- nucleic acid as defined in i) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:4 and any codon-optimized and/or nucleoside modified version thereof, such as wherein the nucleic acid as defined in i) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:4 and SEQ ID NQ:10.
- nucleic acid as defined in i) comprises a nucleic acid sequence selected from any codon-optimized or nucleoside modified version of SEQ ID NO:4, such as any codon-optimized and nucleoside modified version of SEQ ID NO:4, such as any nucleoside modified version of SEQ ID NQ:10.
- nucleic acid as defined in i) comprises a nucleic acid sequence according to SEQ ID NQ:10 or SEQ ID NO:68.
- nucleic acid as defined in ii) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:5 and any codon-optimized and/or nucleoside modified version thereof, such as wherein the nucleic acid as defined in ii) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:5 and SEQ ID NO:11.
- nucleic acid as defined in iii) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:6 and any codon-optimized and/or nucleoside modified version thereof, such as wherein the nucleic acid as defined in iii) comprises a nucleic acid sequence selected from a group consisting of SEQ ID NO:6 and SEQ ID NO:12.
- nucleic acid as defined in iii) comprises a nucleic acid sequence selected from any codon-optimized or nucleoside modified version of SEQ ID NO:6, such as any codon-optimized and nucleoside modified version of SEQ ID NO:6, such as any nucleoside modified version of SEQ ID NO:12.
- nucleic acid as defined in i), ii) and/or iii) is a nucleoside modified mRNA comprising one or more modified nucleoside(s), such as a nucleoside modified mRNA comprising one or more pseudouridine residue(s).
- the immunogenic composition according to item 28 wherein said poly-A tail comprises a nucleic acid sequence according to SEQ ID NO:36.
- 30 The immunogenic composition according to any one of items 15 to 29, wherein the nucleic acid as defined in i), ii) and/or iii) comprises a 5' untranslated region, such as wherein said 5' untranslated region enhances translation.
- a vaccine composition comprising the immunogenic composition as defined in any one of items 1 to 39 and a pharmaceutically acceptable carrier or excipient.
- an HSV-2-specific antibody response such as a serum IgG response
- said serum IgG response is detectable in a serum sample obtained from said subject at a dilution of at least about 10 3 , such as at least about 10 5 , such as at least about 10 6 , optionally as measured by ELISA.
- immunogenic composition according to any one of items 1 to 39 and 50 to 52 or the vaccine composition according to any one of items 40 to 52, wherein the immunogenic composition or the vaccine composition induces an HSV-2 antigenspecific T cell response upon restimulation of said subject with one or more HSV-2-specific antigens.
- HSV-2 antigen-specific T cell response comprises inducing an increase in the number of INF-y, IL-2 and/or TNF-a secreting HSV-2 antigen-specific T cells.
- the immunogenic composition or the vaccine composition according to any one of items 53 to 56, wherein said increase in the number of INF-y secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 2.5-fold, such as at least about 5-fold, such as at least about 7.5-fold, such as at least about 10-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 50-fold, such as at least about 75-fold, such as at least about 100-fold, such as at least about 200-fold, such as at least about 250-fold, such as at least about 500-fold, such as at least about 750-fold, such as at least about 1000-fold, such as at least about 1500-fold, such as at least about 2000-fold, such as at least about 2500-fold, such as at least about 3000-fold, such as at least about 3500-fold, such as at least about 4000-fold, such as at least about 5000-fold, such as at least about 6000-fold, such as at least about
- the immunogenic composition or the vaccine composition according to any one of items 53 to 54, wherein said increase in the number of IL-2 secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, as measured in comparison to said control sample.
- the immunogenic composition or the vaccine composition according to any one of items 53 to 55, wherein said increase in the number of TNF-a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, as measured in comparison to said control sample.
- the immunogenic composition or the vaccine composition according to any one of items 53 to 56, wherein said increase in the number of INF-y secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 50-fold, such as at least about 60-fold, such as at least about 70-fold, as measured in comparison to a control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- immunogenic composition or the vaccine composition according to any one of items 53 to 54, wherein said increase in the number of IL-2 secreting HSV-2 antigen-specific T cells is at least about 1.5-fold, such as at least about 2-fold, such as at least about 2.5-fold, such as at least about 3-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- HSV-2 antigen-specific T cell response comprises inducing multifunctional HSV-2 antigen-specific T cells, such as wherein said HSV-2 antigen-specific T cell response comprises inducing an increase in the number of multifunctional HSV-2 antigen-specific T cells.
- the immunogenic composition or the vaccine composition according to item 63 wherein said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/IL-2, IL-2/TNF-a, INF-y/TNF-a and INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells.
- said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/IL-2, INF-y/TNF-a and INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells.
- immunogenic composition or the vaccine composition according to any one of items 63 to 65, wherein said multifunctional HSV-2 antigen-specific T cells are selected from a group consisting of INF-y/IL-2 and INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells.
- the immunogenic composition or the vaccine composition according to any one of items 64 to 66 and 70, wherein said increase in the number of INF-y/IL-2 secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6- fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 45-fold, such as at least about 50-fold, such as at least about 100-fold, such as at least about 150-fold, such as at least about 200-fold, such as at least about 250-fold, such as at least about 300-fold, such as at least about 350-fold, such as at least about 400-
- the immunogenic composition or the vaccine composition according to any one of items 64, 65, 67 and 71, wherein said increase in the number of INF-y/TNF-a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30- fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 45-fold, such as at least about 50-fold, such as at least about 100-fold, such as at least about 150-fold, such as at least about 200-fold, such as at least about 250- fold, such as at least about 300-fold, such as at least about 350-fold, such
- the immunogenic composition or the vaccine composition according to item 64 or 69, wherein said increase in the number of IL-2/TNF-a secreting HSV-2 antigenspecific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6-fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, as measured in comparison to said control sample.
- the immunogenic composition or the vaccine composition according to any one of items 64 to 68, wherein said increase in the number of INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells is at least about 10-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 50-fold, such as at least about 75-fold, such as at least about 100-fold, such as at least about 125-fold, such as at least about 150-fold, such as at least about 175-fold, such as at least about 200-fold, such as at least about 300-fold, such as at least about 400-fold, such as at least about 450-fold, such as at least about 500-fold, as measured in comparison to said control sample.
- the immunogenic composition or the vaccine composition according to any one of items 64 to 66 and 70, wherein said increase in the number of INF-y/IL-2 secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6- fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 9-fold, such as at least about 10-fold, such as at least about 11-fold, such as at least about 12-fold, such as at least about 13-fold, such as at least about 14-fold, such as at least about 15-fold, such as at least about 16-fold, such as at least about 17-fold, such as at least about 18-fold, such as at least about 19-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 40-fold, such as at least about 50-fold, as measured in comparison to said control
- the immunogenic composition or the vaccine composition according to any one of items 64, 65, 67 and 71, wherein said increase in the number of INF-y/TNF-a secreting HSV-2 antigen-specific T cells is at least about 10-fold, such as at least about 15-fold, such as at least about 20-fold, such as at least about 25-fold, such as at least about 30-fold, such as at least about 35-fold, such as at least about 40-fold, such as at least about 45-fold, such as at least about 50-fold, such as at least about 55-fold, such as at least about 60-fold, such as at least about 65-fold, such as at least about 70-fold, such as at least about 75-fold, such as at least about 80-fold, such as at least about 100-fold, such as at least about 150-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21
- immunogenic composition or the vaccine composition according to any one of items 64 to 68, wherein said increase in the number of INF-y/IL-2/TNF-a secreting HSV-2 antigen-specific T cells is at least about 2-fold, such as at least about 3-fold, such as at least about 4-fold, such as at least about 5-fold, such as at least about 6- fold, such as at least about 7-fold, such as at least about 8-fold, such as at least about 10-fold, such as at least about 15-fold, as measured in comparison to said control sample obtainable from a subject administered UL11, UL16 and UL21 proteins, such as administered a multimeric protein complex comprising UL11, UL16 and UL21 proteins.
- a latent HSV-2 infection such as preventing a flare, a recurrence and/or a HSV-2 labialis following a primary HSV-2 infection.
- the immunogenic composition or the vaccine composition for use according to any one of items 81 to 89, wherein said use comprises administration of the immunogenic composition or the vaccine composition to said such subject, such as wherein said uses comprises the administration of the immunogenic composition or the vaccine composition in a single dose to said subject, wherein said single dose corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- a method for therapeutic treatment and/or prophylactic treatment of an HSV-2 infection comprising administration of the immunogenic composition according to any one of items 1 to 39 and 50 to 79 or the vaccine composition according to any one of items 40 to 79 to a subject in need thereof.
- HSV-2 infection is selected from a group consisting of a primary HSV-2 infection and a reactivation of a latent HSV-2 infection, such as wherein said HSV-2 infection is a reactivation of a latent HSV-2 infection.
- prophylactic treatment comprises preventing a reactivation of a latent HSV-2 infection, such as preventing a flare, a recurrence and/or a HSV-2 labialis following a primary HSV-2 infection.
- any one of items 97 to 105 wherein said method comprises administration of the immunogenic composition or the vaccine composition in a single dose to said subject, wherein said single dose corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- the dose of said at least one boost vaccination corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- time interval between said prime vaccination and said at least one boost vaccination is less than about 10 weeks, such as less than about 9 weeks, such as less than about 8 weeks, such as a time interval between about 2 weeks and about 8 weeks.
- prophylactic treatment comprises preventing symptoms associated with said HSV-2 infection.
- HSV-2 infection is selected from a group consisting of a primary HSV-2 infection and a reactivation of a latent HSV-2 infection, such as wherein said HSV-2 infection is a reactivation of a latent HSV-2 infection.
- HSV-2 infection is a genital HSV-2 infection and/or an oral HSV-2 infection.
- HSV-2 infection is a neonatal infection.
- prophylactic treatment comprises preventing a reactivation of a latent HSV-2 infection, such as preventing a flare, a recurrence and/or a HSV-2 labialis following a primary HSV-2 infection.
- said therapeutic treatment and/or prophylactic treatment comprises administration of the immunogenic composition or the vaccine composition in a single dose to said subject, wherein said single dose corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg. 124.
- said therapeutic treatment and/or prophylactic treatment comprises multiple administration of the immunogenic composition or the vaccine composition to said subject.
- said multiple administration comprises a prime vaccination and at least one boost vaccination, such as wherein said multiple administration is a two-dose prime-boost regimen comprising said prime vaccination and one of said at least one boost vaccination.
- the dose of said prime vaccination corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- the dose of said at least one boost vaccination corresponds to the total amount of nucleic acids as defined in i), ii) and iii) and is less than about 200 pg, such as less than about 150 pg, such as less than about 100 pg, such as is between about 10 pg and about 100 pg.
- time interval between said prime vaccination and said at least one boost vaccination is less than about 10 weeks, such as less than about 9 weeks, such as less than about 8 weeks, such as a time interval between about 2 weeks and about 8 weeks.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Virology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Molecular Biology (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Oncology (AREA)
- Biophysics (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Communicable Diseases (AREA)
- Biotechnology (AREA)
- Engineering & Computer Science (AREA)
- Gastroenterology & Hepatology (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Genetics & Genomics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Immunology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
La présente invention concerne une composition immunogène et une composition vaccinale associée comprenant un ou plusieurs acides nucléiques codant pour des protéines structurales du virus de l'herpès simplex 2 (HSV-2) ou des fragments immunogènes de ceux-ci. La composition vaccinale peut être utilisée pour le traitement et/ou la prévention d'une infection par le HSV-2 et est particulièrement bénéfique pour le contrôle immunologique de la réactivation périodique du virus chez des patients infectés.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23210940.5 | 2023-11-20 | ||
| EP23210940 | 2023-11-20 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025109008A1 true WO2025109008A1 (fr) | 2025-05-30 |
Family
ID=88874715
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/082999 Pending WO2025109008A1 (fr) | 2023-11-20 | 2024-11-20 | Nouvelles compositions vaccinales et procédés de traitement du vhs |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025109008A1 (fr) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2017057969A1 (fr) | 2015-10-02 | 2017-04-06 | 씨제이제일제당(주) | Récipient de stockage d'aliments fermentés |
| WO2017157969A1 (fr) | 2016-03-14 | 2017-09-21 | Redbiotec Ag | Moyens et méthodes pour le traitement du vhs |
| US20200276300A1 (en) | 2017-08-17 | 2020-09-03 | The Trustees Of The University Of Pennsylvania | Modified mrna vaccines encoding herpes simplex virus glycoproteins and uses thereof |
| WO2022189634A1 (fr) * | 2021-03-11 | 2022-09-15 | Redbiotec Ag | Compositions vaccinales et procédés de traitement du vhs |
-
2024
- 2024-11-20 WO PCT/EP2024/082999 patent/WO2025109008A1/fr active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2017057969A1 (fr) | 2015-10-02 | 2017-04-06 | 씨제이제일제당(주) | Récipient de stockage d'aliments fermentés |
| WO2017157969A1 (fr) | 2016-03-14 | 2017-09-21 | Redbiotec Ag | Moyens et méthodes pour le traitement du vhs |
| US20200276300A1 (en) | 2017-08-17 | 2020-09-03 | The Trustees Of The University Of Pennsylvania | Modified mrna vaccines encoding herpes simplex virus glycoproteins and uses thereof |
| WO2022189634A1 (fr) * | 2021-03-11 | 2022-09-15 | Redbiotec Ag | Compositions vaccinales et procédés de traitement du vhs |
Non-Patent Citations (25)
| Title |
|---|
| "GenBank", Database accession no. AHG54732.1 |
| "Handbook of Biochemistry: Section A Proteins", 1976, CRC PRESS |
| "Remington's Pharmaceutical Sciences", 1991, MACK PUBLISHING CO. |
| ALFONSO R GENNARO, REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY |
| ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402 |
| AUSUBEL ET AL.: "Current Protocols in Molecular Biology, J", 1992, GREENE PUBLISHING ASSOCIATES |
| BAUER ET AL.: "Pharmazeutische Technologie", 1997, GOVI-VERLAG |
| EGAN KPWU 5WIGDAHL BJENNINGS SR: "Immunological control of herpes simplex virus infections", J NEUROVIROL, vol. 19, no. 4, August 2013 (2013-08-01), pages 328 - 45 |
| HOU ET AL., NATURE REVIEW MATERIALS, vol. 6, 2021, pages 1078 - 1094 |
| HOU XZAKS TLANGER RDONG Y: "Lipid nanoparticles for mRNA delivery", NAT REV MATER, vol. 6, no. 12, 2021, pages 1078 - 1094, XP037634156, DOI: 10.1038/s41578-021-00358-0 |
| KHAN AASRIVASTAVA RCHENTOUFI AAKRITZER ECHILUKURI 5GARG 5YU DCVAHED HHUANG LSYED SA: "Bolstering the Number and Function of HSV-1-Specific CD8+ Effector Memory T Cells and Tissue-Resident Memory T Cells in Latently Infected Trigeminal Ganglia Reduces Recurrent Ocular Herpes Infection and Disease", J IMMUNOL., vol. 199, no. 1, 1 July 2017 (2017-07-01), pages 186 - 203 |
| KIMURA ET AL., MOLECULAR THERAPY, vol. 31, no. 8, 2023, pages 2360 - 2375 |
| KIMURA TLEAL JMSIMPSON AWARNER NLBERUBE BJARCHER JFPARK 5KURTZ RHINKLEY TNICHOLES K: "A localizing nanocarrier formulation enables multi-target immune responses to multivalent replicating RNA with limited systemic inflammation", MOL THER, vol. 31, no. 8, 2 August 2023 (2023-08-02), pages 2360 - 2375 |
| KOWALSKI ET AL., MOLECULAR THERAPY, vol. 27, no. 4, 2019, pages 710 - 728 |
| KOWALSKI PSRUDRA AMIAO LANDERSON DG: "Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery", MOL THER, vol. 27, no. 4, 10 April 2019 (2019-04-10), pages 710 - 728, XP055634628, DOI: 10.1016/j.ymthe.2019.02.012 |
| KUO TWANG CBADAKHSHAN TCHILUKURI 5BENMOHAMED L: "The challenges and opportunities for the development of a T-cell epitope-based herpes simplex vaccine", VACCINE, vol. 32, no. 50, 28 November 2014 (2014-11-28), pages 6733 - 45 |
| LOOKER ET AL., BULLETIN OF THE WORLD HEALTH ORGANIZATION, vol. 86, 2008, pages 805 - 812 |
| P PARDI NHOGAN MJPORTER FWWEISSMAN D: "mRNA vaccines - a new era in vaccinology", NAT REV DRUG DISCOV, vol. 17, no. 4, April 2018 (2018-04-01), pages 261 - 279, XP055524319, DOI: 10.1038/nrd.2017.243 |
| PARDI ET AL.: "Nature Reviews", DRUG DISCOVERY, vol. 17, no. 4, 2018, pages 261 - 279 |
| PRESNYAK ET AL., CELL, vol. 160, no. 6, 2015, pages 1111 - 24 |
| PRESNYAK VALHUSAINI NCHEN YHMARTIN 5MORRIS NKLINE NOLSON 5WEINBERG DBAKER KEGRAVELEY BR: "Codon optimality is a major determinant of mRNA stability", CELL, vol. 160, no. 6, 12 March 2015 (2015-03-12), pages 1111 - 24, XP029203792, DOI: 10.1016/j.cell.2015.02.029 |
| RUCHI SRIVASTAVAPIERRE-GRÉGOIRE COULONSOUMYABRATA ROYSRAVYA CHILUKURISUMIT GARGLBACHIR BENMOHAMED: "Phenotypic and Functional Signatures of Herpes Simplex Virus-Specific Effector Memory CD73+CD45RAhighCCR7lowCD8+ TEMRA and CD73+CD45RAlowCCR7lowCD8+ TEM Cells Are Associated with Asymptomatic Ocular Herpes", J IMMUNOL, vol. 201, no. 8, 15 October 2018 (2018-10-15), pages 2315 - 2330 |
| SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS |
| THOMPSON ET AL., NUCLEIC ACIDS RESEARCH, vol. 22, 1994, pages 4673 - 4680 |
| TRUONG NRSMITH JBSANDGREN KJCUNNINGHAM AL: "Mechanisms of Immune Control of Mucosal HSV Infection: A Guide to Rational Vaccine Design", FRONT IMMUNOL, vol. 10, 6 March 2019 (2019-03-06), pages 373 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20230346907A1 (en) | Sexually transmitted disease vaccines | |
| US20200054737A1 (en) | Herpes simplex virus vaccine | |
| US8501194B2 (en) | Vaccine for viruses that cause persistent or latent infections | |
| JP6109165B2 (ja) | ヘルペスウイルスワクチンおよび使用方法 | |
| WO2023107999A2 (fr) | Vaccins à arnm contre le virus de l'herpès simplex | |
| BE1025121B1 (fr) | Constructions antigeniques du virus zika | |
| US9408905B2 (en) | Herpes simplex virus vaccines | |
| US20240156951A1 (en) | Vaccine compositions and methods for treating hsv | |
| US20240350619A1 (en) | Vaccines and compositions based on sars-cov-2 s protein | |
| US8282939B2 (en) | Attenuated live triple G protein recombinant rabies virus vaccine for pre- and post-exposure prophylaxis of rabies | |
| JP2019512501A (ja) | Hsvを処置するための手段及び方法 | |
| US20230372473A1 (en) | Vaccine compositions | |
| US20240156954A1 (en) | Vaccine compositions and methods for treating hsv | |
| WO2025109008A1 (fr) | Nouvelles compositions vaccinales et procédés de traitement du vhs | |
| JPWO2008065752A1 (ja) | diRNAを有効成分とする免疫治療用薬剤 | |
| WO2010005474A1 (fr) | Virus de la vaccine ankara (mva) recombinant modifié exprimant des antigènes polypeptidiques de chlamydia | |
| US20170224808A1 (en) | Therapeutic compositiojns and methods for inducing an immune response to herpes simplex virus type 2 (hsv-2) | |
| JP2003528816A (ja) | ヒトおよび動物の病原体のゲノム由来の構築物に基づいたdnaワクチン | |
| ES2577704T3 (es) | Vacunas de virus herpes simple | |
| HK1175807B (en) | Herpes simplex virus vaccines |
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: 24809366 Country of ref document: EP Kind code of ref document: A1 |