WO2024227806A1 - Antigen-specific physiologic dendritic cells for therapy - Google Patents
Antigen-specific physiologic dendritic cells for therapy Download PDFInfo
- Publication number
- WO2024227806A1 WO2024227806A1 PCT/EP2024/061957 EP2024061957W WO2024227806A1 WO 2024227806 A1 WO2024227806 A1 WO 2024227806A1 EP 2024061957 W EP2024061957 W EP 2024061957W WO 2024227806 A1 WO2024227806 A1 WO 2024227806A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- mrna
- therapeutic composition
- antigenic protein
- antigen
- phdcs
- 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
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/19—Dendritic cells
-
- 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
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
-
- 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
- A61K2039/555—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
- A61K2039/57—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
Definitions
- the invention further relates to therapeutic compositions comprising physiologic dendritic cells and at least one mRNA, as well as therapeutic compositions for use in therapeutic treatment against infectious diseases and hyper- proliferative diseases such as cancers or tumors.
- DCs Dendritic cells
- T-cells and B-cells As DCs thereby activate na ⁇ ve, effector, and memory immune cells, they are a promising therapeutic agent against diseases such as cancer or infectious diseases.
- the potent activation of de novo T-cell and B-cell responses also suggests dendritic cells as promising agents for prophylactic purposes.
- dendritic cells in the human body represent only about 0.3% of total circulating leukocytes, and consist of a heterogeneous population consisting of different maturation levels and capabilities of stimulating tolerogenic or stimulatory immune responses.
- Techniques for the isolation and maturation of dendritic cells are described in a number of documents, and there are various methods, including bringing monocytes into contact with hematopoietic growth factors and cytokines such as IFN- ⁇ , GM-CSF, DB:csh TNF- ⁇ , IL-3 or combinations thereof (see e.g. EP 922,758 or EP 663,930, WO 95/28479).
- Extracorporeal Photopheresis has been used successfully to treat cutaneous T- cell lymphoma (CTCL) in subsets of patients.
- CCP Extracorporeal Photopheresis
- PBMC peripheral blood mononuclear cells
- 8-MOP UVA 8-methoxypsoralen and UVA
- the underlying mechanism of action includes dendritic cells taking up antigens that are being released by the exposure of malignant T cells to 8-MOP UVA and presenting these to the patient ⁇ s immune system. According to their mode of action, these DCs have been termed immuno-stimulatory DCs.
- ECP leads to the conversion of passaged blood monocytes to DCs.
- antigens are exactly taken up and presented.
- immuno- stimulatory DCs which are moreover, equipped with defined antigens in order to provoke a desired and targeted immuno-stimulatory response.
- OBJECTIVES AND SUMMARY OF THE INVENTION One objective of the present invention is to provide a therapeutic composition comprising phDCs and at least one mRNA. It is another objective of the present invention to provide a therapeutic composition comprising antigen-specific phDCs and at least one mRNA.
- a further objective of the invention is to provide a therapeutic composition comprising antigen-specific monocytes and at least one mRNA. It is another objective of the present invention to use the therapeutic composition for therapeutic treatment. It is still another objective to provide therapeutic compositions for use in a method of treating a disease in a subject. Finally, it is an objective of the invention to provide a kit comprising phDCs and at least one mRNA encoding at least one antigenic protein.
- the present invention as illustratively described in the following may suitably be practiced in the absence of any element or elements, limitation or limitations, not specifically disclosed herein.
- the present invention is described with respect to particular embodiments below and with reference to certain figures but the invention is not limited thereto but only by the claims.
- the present invention is based to some extent on data and experiments presented hereinafter, which lead to the insight that dendritic cells loaded with selected disease antigens can be used effectively for the therapeutic treatment of diseases such as cancer or infectious diseases.
- dendritic cells need to be (i) obtained and (ii) equipped with disease specific antigens.
- so-called physiologic dendritic cells (phDCs) can be used.
- phDCs can inter alia be produced in vitro by applying a shear force to monocytes (e.g. from a blood sample), see e.g. Ventura et al., 2018 “Extracorporeal Photochemotherapy Drives Monocyte-to-Dendritic Cell Maturation to Induce Anticancer Immunity”, WO 2014/106629, WO 2016/001405 or Hanlon et al., 2020 “Rapid Production of Physiologic Dendritic Cells (phDC) for Immunotherapy”).
- monocytes e.g. from a blood sample
- phDC can be characterized by marker expression including markers such as HLA-DR, CD83, CD86, ICAM or PLAUR and/or no increased expression of GILZ (see, e.g., Ventura et al., 2018 “Extracorporeal Photochemotherapy Drives Monocyte-to-Dendritic Cell Maturation to Induce Anticancer Immunity” or WO 2014/106629).
- dendritic cells which are obtained by subjecting monocytes to a physical force more closely resemble the properties of naturally-occurring dendritic cells which is why dendritic cells which are obtained by subjecting monocytes to a physical force will be designated as physiolgical dendritic cells (phDCs) hereinafter.
- phDCs physiolgical dendritic cells
- a physical force to induce differentiation of monocytes into phDCs can be applied by passing monocytes through a flow chamber which can take the form of, e.g., a plate or, e.g., a bag (e.g. flexible bag or plastic bag).
- a flow chamber which can take the form of, e.g., a plate or, e.g., a bag (e.g. flexible bag or plastic bag).
- a bag is described, e.g., in Buechler et al., 2004 “Generation of Dendritic Cells Using Cell Culture Bags - Description of a Method and Review of Literature”.
- phDC physiological dendritic cells
- phDCs which are preferably produced in the absence of apoptotic signals and subsequently loaded with antigens will be effective in stimulating an immune response.
- transfection with e.g. at least one mRNA encoding for at least one antigenic protein
- monocytes can be combined with disease specific antigens (in the form of e.g.
- phDC are capable of internalizing antigens and presenting them on their surface.
- the antigens can be provided in different forms including in the form of mRNA-containing lipid nanoparticles (LNP).
- PhDCs are capable of expressing the encoded antigens such as disease associated proteins or peptides.
- the loaded antigen-specific phDCs e.g.
- the antigen-specific phDCs of the invention are capable of eliciting a therapeutic immune response by their unique ability of presenting the specific antigen on MHC molecules leading to activation, proliferation and differentiation of T cells into armed effector T cells.
- effector T cells is the killing of infected cells by CD8+ cytotoxic T cells and the activation of macrophages by Th1 cells, which together make up cell-mediated immunity, and the activation of B cells by both Th2 and Th1 cells to produce different classes of antibody, thus driving the humoral immune response.
- T cells (such as e.g.
- antigen-specific phDCs e.g. therapeutic composition comprising phDCs and at least one mRNA
- antigen-specific phDCs can also be considered a safe therapeutic approach as the immune system is activated by a very defined set of cells as compared to other therapeutics, which randomly scatter into systemic circulation providing a larger probability of missing target cells and eliciting unwanted side effects.
- antigen-specific phDCs e.g.
- therapeutic composition comprising phDCs and at least one mRNA
- the present invention also provides for significant dose sparing possibilities.
- Still another advantage of antigen-specific phDCs e.g. therapeutic composition comprising phDCs and at least one mRNA
- Antigen-specific phDCs are able to directly trigger clonal expansion of the subject ⁇ s already preexisting T-cells into effector cytotoxic T-cells.
- the present invention relates to a therapeutic composition
- a therapeutic composition comprising a pharmaceutically effective amount of phDCs and at least one RNA, which comprises a coding sequence encoding for at least one antigenic protein.
- the RNA is a single stranded RNA, an mRNA, a circular RNA, a self-amplifying RNA, and/or a synthetic RNA.
- the RNA is self-amplifying mRNA.
- the RNA is an mRNA.
- the present invention relates to a therapeutic composition comprising a pharmaceutically effective amount of phDCs and at least one mRNA (e.g. one mRNA molecule or more than one mRNA molecule), which comprises a coding sequence encoding for at least one antigenic protein.
- the RNA e.g. mRNA
- the RNA comprises at least one chemical modification. If more than one RNA (e.g. mRNA) is used, each RNA (e..g mRNA) independently comprises at least one chemical modification (i.e. the chemical modification from one RNA to another can be different).
- chemical modification means that one of the four naturally- occuring standard nucleosides which occur in RNA (adenosine (A), guanosine (G), uridine (U), and cytidine (C)) are replaced by modified forms thereof wherein the modification affects the base moiety within the nucleoside.
- Modified nucleosides can be naturally-occurring or non-naturally-occurring modified nucleosides. Naturally- occurring modified nucleosides are preferred.
- RNA e.g. mRNA
- the chemical modification is a substitution of one or more nucleosides of the RNA (e.g. mRNA) by one or more modified nucleosides.
- Naturally occurring modified nucleosides comprise 1-methyladenosine (m 1 A), N 6 - methyladenosine (m 6 A), 2'-O-methyladenosine (Am), 5-methylcytidine (m 5 C), 2'-O- methylcytidine (Cm), 2-thiocytidine (s 2 C), N 4 -acetylcytidine (ac 4 C), 5-formylcytidine (f 5 C), 2'-O-methylguanosine (Gm), inosine (I), pseudouridine ( ⁇ ), 5-methyluridine (m 5 U), 2'-O-methyluridine (Um).1-methylpseudouridine (m1 ⁇ ), 2-thiouridine (s2U), 4-thiouridine (s4U), 5-methoxyuridine (mo5U) and 3-methyluridine (m3U).
- RNA e.g. mRNA
- RNA e.g. mRNA
- the above-mentioned effects on the TLR- mediated immune reaction and/or the expression level of the RNA (e.g. mRNA) encoded antigen depend on the extent of replacement of uridine by naturally-occurring modified uridine nucleosides.
- all uridine nucleosides within an antigen-encoding RNA e.g. mRNA
- the present invention also considers less than 90%, less than 80%, less than 70%, less than 60%, or less than 50% of the uridine nucleosides present in the RNA (e.g. mRNA) encoding the antigenic protein are replaced with naturally-occurring modified uridine nucleosises. Where uridine nucleosides are replaced by naturally-occuring modified forms thereof, it is preferred to use pseudouridine and more preferred to use N1-methylpseudouridine or N1- ethylpseudouridine. N1-methylpseudouridine is most preferred.
- the invention thus considers as a particularly preferred embodiment antigen-encoding RNAs (e.g.
- RNA in which all uridines are replaced N1-methylpseudouridine.
- the invention also considers to replace, instead of and preferably in addition to uridine nucleosides, A, C, and/or G nucleosides by modified forms thereof. It is preferred to use naturally-occurring modified forms of A, C, and/or G for such replacements. If such additional replacements are considered, it is preferred that all of A, C, and/or G nucleosides are replaced by modified forms thereof.
- the RNA e.g. mRNA
- the RNA (e.g. mRNA) contains all of these elements.
- the 5' cap is a Cap1 structure or a m7GpppG cap.
- the 5’cap is a Cap1 structure.
- the sequence of the RNA (e.g. mRNA) is optimized.
- the sequence of the 5’ UTR, 3’UTR, and/or coding sequence for the antigenic protein is optimized.
- the sequence of the RNA (e.g. mRNA) has optimized codon usage or optimized G/C content. In one embodiment, codon usage, G/C content and structural elements are optimized.
- optimization of the sequence of the structural elements 3’ and/or 5’UTR can additionally comprise using one or more of a heterologous UTR, a Kozak sequence, a FI element, removed AURES elements and an enzymatically added tail (e.g. poly(A) tail).
- a poly(A) tail is added enzymatically.
- the at least one RNA e.g., mRNA
- Nanoparticles comprise lipid nanoparticles, poly(amine-co-ester) particles (PACE), poly-beta- amino-ester particles, PACE polyplex particles, lipoplexes and poly(N,N- cystaminebis(acrylamide)-co-4-amino-1-butanol) (pABOL) particles.
- the nanoparticle is a lipid nanoparticle (LNP).
- the RNA e.g. mRNA
- LNP lipid nanoparticle
- a lipid nanoparticle can comprise a cationic lipid, a PEG- modified lipid, a cholesterol, a DSPE-PEG-maleimide, DSPN-PEG-azide and/or a non-cationic lipid.
- a lipid nanoparticle comprises a cationic lipid, a PEG-modified lipid, a cholesterol, and/or a non-cationic lipid.
- the lipid nanoparticle comprises a cationic lipid, a PEG-modified lipid, a cholesterol and a non-cationic lipid.
- Cationic lipids comprise cKK-E12, cKK-E14, LP01, SM102, Lipid 5, etc.
- the cationic lipid is a cKK-E12 lipid. In one embodiment, the cationic lipid is a SM102 lipid. In one embodiment, the cationic lipid is a MC3 lipid (DLin-MC3-DMA). In one embodiment, the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE. In one embodiment, the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 30-40:41-51:1.0-4.0:12-21.
- the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 33-37:44- 48:2.0-3.0:14-18. In one embodiment, the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 34-36:45-47:2.2-2.8:15-17. In one embodiment, the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 35:46.5:2.5:16. In one embodiment, the lipid nanoparticle comprises SM-102, cholesterol, C14-PEG 2000-PE and DOPE.
- the lipid nanoparticle comprises SM-102, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 30-40:41-51:1.0-4.0:12-21. In one embodiment, the lipid nanoparticle comprises SM-102, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 33-37:44-48:2.0-3.0:14-18. In one embodiment, the lipid nanoparticle comprises SM102, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 34-36:45-47:2.2-2.8:15-17. In one embodiment, the lipid nanoparticle comprises SM-102, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 35:46.5:2.5:16.
- the lipid nanoparticle comprises SM102, cholesterol, DMG-PEG-2K, DSPC. In one embodiment, the lipid nanoparticle comprises SM102, cholesterol at a ratio of 30-50:35-45:1.0-4.0:10-20, DMG-PEG- 2K, DSPC at a ratio of 50:38.5:1.5:10. In one embodiment, the lipid nanoparticle comprises MC3, cholesterol, C14-PEG 2000-PE and DOPE. In one embodiment, the lipid nanoparticle comprises MC3, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 30-40:41-51:1.0-4.0:12-21.
- the lipid nanoparticle comprises MC3, cholesterol, C14-PEG 2000- PE and DOPE at a ratio of 33-37:44-48:2.0-3.0:14-18. In one embodiment, the lipid nanoparticle comprises MC3, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 34-36:45-47:2.2-2.8:15-17. In one embodiment, the lipid nanoparticle comprises MC3, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 35:46.5:2.5:16. In one embodiment, the at least one mRNA encoding for at least one antigenic protein is heterologous or exogenous to the phDCs.
- the antigenic protein is exogenous to the phDCs.
- heterologous or exogenous as used herein, in relation to a specific cell type, e.g., phDCs, can refer to a nucleic acid sequence, e.g. mRNA, or a protein, that originates from a source other than the specified cell type, e.g., the phDC.
- a heterologous mRNA or exogenous mRNA to phDCs thus refers to an mRNA that is not endogenous to the phDC, but has been introduced to the phDCs, e.g. by transfection or other means.
- the antigenic protein is an infectious disease associated antigen or a tumor associated antigen. In one embodiment, the antigenic protein is an infectious disease associated antigen.
- the therapeutic composition comprises phDCs and at least one mRNA, which comprises a coding sequence encoding at least one infectious disease associated antigenic protein.
- the infectious disease associated antigen is a viral antigen, a bacterial antigen, a fungal antigen or a parasite antigen. In one embodiment, the infectious disease associated antigen is a bacterial antigen. In one embodiment, the bacterial antigen is derived from Borrelia spp. or Mycobacteria spp.. In another embodiment, the infectious disease associated antigen is a fungal antigen.
- the fungal antigen is derived from Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis or Candida albicans.
- the infectious disease associated antigen is a parasite antigen.
- the parasite antigen is derived from Plasmodium malariae.
- the infectious disease associated antigen is a viral antigen.
- the therapeutic composition comprises phDCs and at least one mRNA, which comprises a coding sequence encoding at least one viral antigenic protein.
- the viral antigen is a coronavirus antigenic protein or HIV antigenic protein.
- the viral antigen is a coronavirus antigenic protein. In one embodiment, the viral antigen is a betacoronavirus antigenic protein. In another embodiment, the viral antigen is a SARS-CoV-2 antigenic protein. In one embodiment, the coronavirus antigenic protein or betacoronavirus antigenic protein is an antigenic protein of the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein or a fragment of each of the aforementioned. In one embodiment, the SARS-CoV-2 antigen is an antigenic protein of the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein or a fragment of each of the aforementioned.
- the SARS-CoV-2 antigen is an antigenic protein of the spike protein or a fragment thereof.
- a fragment comprises at least 10, at least 50, at least 100, at least 200, at least 400 or at least 800 amino acid residues.
- the RNA e.g.
- RNA comprises a sequence which is at least 75%, 85%, 90%, 95%, 98% or 99% or a 100% identical to a sequence or a part of a sequence selected from the group comprising SEQ ID NO: 1 (spike protein transcript), SEQ ID NO: 2 (envelope protein transcript), SEQ ID NO: 3 (nucleocapsid protein transcript), SEQ ID NO: 4 (membrane protein transcript) and/or SEQ ID NO: 5 (Orf1ab polyprotein transcript).
- the RNA e.g. mRNA
- the antigenic protein, for which the RNA (e.g. mRNA) encodes comprises at least 100, at least 200, at least 300, at least 400, at least 500, at least 600, at least 700, at least 800, at least 900, at least 1000, at least 1200, at least 2000 or at least 3000 amino acids. In one embodiment, the antigenic protein, for which the RNA (e.g. mRNA) encodes, comprises at least 50 amino acids. In one embodiment, the antigenic protein, for which the RNA (e.g. mRNA) encodes, comprises at least 500 amino acids. In one embodiment, the antigenic protein, for which the RNA (e.g. mRNA) encodes, comprises at least 1000 amino acids.
- the SARS-CoV-2 antigen is a protein or peptide comprising a sequence which is at least 75%, 85%, 90%, 95%, 98% or 99% or a 100% identical to a sequence or a part of a sequence selected from the group comprising SEQ ID NO: 6 (spike amino acid sequence), SEQ ID NO: 7 (envelope amino acid sequence), SEQ ID NO: 8 (nucleocapsid amino acid sequence), SEQ ID NO: 9 (membrane amino acid sequence) and/or SEQ ID NO: 10 (Orf1ab amino acid sequence).
- the protein or peptide comprises a sequence or part of a sequence which is at least 75%, 85%, 90%, 95%, 98% or 99% or a 100% identical to SEQ ID NO: 6.
- the at least one antigenic protein is a SARS-CoV-2 hexapro spike protein, with a foldon domain in its ectodomain right above the transmembrane domain (RNA sequence of which is depicted as bold and underlined in Table 3) or an RNA (e.g. mRNA) encoding therefore.
- the corresponding RNA may be modified with one or more of N1-methylpseudouridine, a cap1 structure and a poly-A tail.
- the RNA (e.g. mRNA) comprises N1-methylpseudouridine (e.g. each uridine is replaced by N1-methylpseudouridine), a cap1 structure and a poly- A tail.
- the codons may be optimized to GC enrich the RNA (e.g. mRNA) and optimized 5’ and 3’ UTRs may be used.
- the RNA (e.g. mRNA) comprises a sequence or part of a sequence corresponding to SEQ ID NO: 19 or a sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to SEQ ID NO: 19.
- the RNA (e.g.
- the viral antigen is derived from HIV.
- the HIV antigen is derived from the envelope protein (env), group antigens polyprotein (gag), reverse transcriptase (pol) and/or negative factor protein (nef).
- the RNA e.g.
- the HIV antigen is a protein or peptide comprising a sequence which is at least 75%, 85%, 90%, 95%, 98% or 99% or a 100% identical to a sequence or a part of a sequence selected from the group comprising SEQ ID NO: 15 (env amino acid sequence), SEQ ID NO: 16 (gag amino acid sequence), SEQ ID NO: 17 (pol amino acid sequence) and/or SEQ ID NO: 18 (nef amino acid sequence).
- the therapeutic composition of the present invention may comprise a pharmaceutically effective amount of physiological DCs (phDCs) and at least one mRNA, which comprises a coding sequence encoding for at least one antigenic protein, wherein, the mRNA may be at least partially comprised in the phDCs. In some embodiments, substantially all of the mRNA is comprised in the phDCs.
- the mRNA not incorporated into the phDCs may be removed by washing the phDCs or other means known in the art.
- the resulting therapeutic composition may thus comprise phDCs, wherein the phDCs have taken up mRNA encoding for at least one antigenic protein.
- the mRNA may be translated to the antigenic protein.
- the phDCs express the antigenic protein.
- the antigenic protein may be exogenous or heterologous to the phDCs.
- the free, i.e., the non-incorporated mRNA is not removed from the therapeutic composition, and the composition thus comprises unincorporated mRNA.
- the therapeutic composition of the invention may comprise a pharmaceutically effective amount of physiological DCs (phDCs) and at least one mRNA, which comprises a coding sequence encoding for at least one antigenic protein, wherein the mRNA is at least partially not incorporated into the phDCs.
- only a subset of the mRNA comprised in the therapeutic composition is taken up by the phDCs present in the composition.
- the at least one antigenic protein is a tumor associated antigen.
- the therapeutic composition comprises phDCs and at least one mRNA, which comprises a coding sequence encoding at least one tumor associated antigenic protein.
- the tumor associated antigen is a leukemia antigen, melanoma antigen, lymphoma antigen, endometrial cancer antigen, kidney cancer antigen, brain cancer antigen, cervical cancer antigen, liver cancer antigen, head and neck cancer antigen, gastrointestinal cancer antigen, lymph node cancer antigen, pancreas cancer antigen, ear, nose and throat (ENT) cancer antigen, breast cancer antigen, prostate cancer antigen, ovarian cancer antigen or lung cancer antigen.
- the at least one antigenic protein is a blood cancer associated antigen.
- a blood cancer antigenic protein can be a leukemia antigenic protein, lymphoma antigenic protein or myeloma antigenic protein.
- phDCs are obtainable by subjecting monocytes (e.g. obtained from a donor) to a physical force.
- the physical force can be applied to the monocytes by passing said monocytes through a flow chamber.
- the flow chamber can be a plate, a bag or a flow chamber of a device, e.g.
- a large-scale ECP device e.g., a clinical ECP device (e.g., a THERAKOS® CELLEX® device; Combination of Terumo’s Spectra Optia with UVA PIT; Combination of apheresis device from Fresenius and Macogenix from Macopharma; Single-Needle Option for the Amicus® Extracorporeal Photopheresis Protocol), or a miniaturized ECP device, e.g., a Transimmunization plate as described in WO2017/005700 A1, or a combination thereof (e.g. a hybrid of a bag and a chamber as disclosed herein).
- a miniaturized ECP device e.g., a Transimmunization plate as described in WO2017/005700 A1, or a combination thereof (e.g. a hybrid of a bag and a chamber as disclosed herein).
- phDCs are obtainable by plate-passage of monocytes using an extracorporeal photopheresis (ECP) derived process.
- ECP extracorporeal photopheresis
- the flow chamber is a plate.
- Monocytes e.g. obtained from a donor
- platelets are present in the plate which can be either derived from the donor ⁇ s blood sample or a fraction thereof or provided separately.
- plasma components can be present in the plate which can be either derived from the donor ⁇ s blood sample or a fraction thereof or provided separately.
- the flow chamber is a bag.
- the bag is a flexible bag or a plastic bag.
- the flow chamber is a flexible bag.
- the bag is a plastic bag.
- the material of the flow chamber e.g. plate, flexible bag or plastic bag
- the material of the flow-chamber is non-plastic such as glass, ceramic or silicone.
- plastic materials are considered, one may use acrylics, polycarbonate, polyetherimide, polysulfone, polyphenylsulfone, styrenes, polyurethane, polyethylene, teflon or any other appropriate medical grade plastic.
- the flow chamber is made from an acrylic plastic.
- a bag e.g. flexible bag
- the material may be plastic, rubber or silicone.
- the material is plastic.
- Plastic materials comprise polyolefin, polyethylene, fluoropolymer, polyvinyl chloride, ethylene-vinyl acetate-copolymer, ethylene vinyl alcohol, polyvinylidene fluoride, and/or other plastic comprising materials approved for medical use.
- the flow chamber comprises or consists of a plate.
- the plate can be made of various materials, including but not limited to a plastic material.
- the material of the plate is plastic.
- the material of the plate is non-plastic such as glass, ceramic or silicone.
- Non limiting examples of materials for the plate comprise acrylics, polycarbonate, polyetherimide, polysulfone, polyphenylsulfone, styrenes, polyurethane, polyethylene, teflon or any other appropriate medical grade plastic.
- the plate may be rigid or flexible.
- the material of the plate may comprise or consist of plastic, rubber or silicone.
- the plate is elastic, i.e. is made of an elastic material.
- the elastic material may comprise cyclic olefin copolymer (COC), polyolefin, polyethylene, fluoropolymer, polyvinyl chloride, ethylene-vinyl acetate-copolymer, ethylene vinyl alcohol, polyvinylidene fluoride, polydimethylsiloxane (PDMS), dimethicone, and/or other plastic comprising materials approved for medical use.
- the plate is made of PDMS, e.g., PDMS RTV-615 or PDMS Sylgard 184.
- the plate is made from an acrylic plastic.
- the flow chamber is a hybrid flow chamber.
- the hybrid flow chamber may comprise a chamber and a bag, a chamber and a plate, or a bag and a plate.
- the individual components of the hybrid flow chamber i.e. the bag, plate or chamber, are as defined herein.
- human AB serum, autologous (e.g. human) serum, autologous (e.g. human) plasma, allogeneic (e.g. human) serum, allogeneic (e.g. human) plasma, mouse serum, mouse plasma or FBS is added additionally to the composition.
- the flow chamber, bag, plate or hybrid flow chamber can be coated with human AB serum, autologous (e.g. human) serum, autologous (e.g. human) plasma, allogeneic (e.g. human) serum, allogeneic (e.g. human) plasma, mouse serum, mouse plasma or FBS before the monocytes are added.
- the flow chamber, bag, plate or hybrid flow chamber is coated with autologous (e.g. human) serum, autologous (e.g. human) plasma, allogeneic (e.g. human) serum, allogeneic (e.g.).
- the phDCs are incubated with the at least one antigenic protein or the at least one mRNA encoding for the at least one antigenic protein. Incubation can be performed under standard conditions for culturing of human cells, e.g. at 37° C and 5% CO2 in standard mediums such as in RPMI-1640 medium. Alternatively or additionally, an incubation step can also be inserted after generation of phDCs from monocytes.
- Incubation can be performed for 0.5 h, 1 h, 2 h, 3 h, 4 h, 5 h, 6 h, 8 h, 12 h or 24 h. In another embodiment, the incubation step can be performed for at least 0.5 h, at least 1 h, at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 8h, at least 12 h or at least 20h. In one embodiment, incubation is performed for 6 h. In one embodiment, incubation is performed for 12 h. In one embodiment, incubation is performed for 20 h. In one embodiment, incubation is performed for 6 to 20 hours. In one embodiment, incubation is performed for 8 to 20 hours.
- Monocytes may be obtained by any suitable means, e.g., from a blood sample obtained from a donor or a fraction thereof.
- the blood sample or fraction thereof may be, e.g., a buffy coat including white blood cells and platelets.
- the blood sample or fraction thereof may be isolated peripheral blood mononuclear cells (PMBC).
- the monocytes are autologous.
- the therapeutic composition additionally comprises human AB serum, autologous (e.g. human) serum, autologous (e.g. human) plasma, allogeneic (e.g. human) serum, allogeneic (e.g.).
- the therapeutic composition additionally comprises autologous serum, autologous plasma, allogeneic serum, allogeneic plasma, mouse serum, mouse plasma or FBS. In one embodiment, the therapeutic composition additionally comprises mouse serum, mouse plasma or FBS. In one embodiment, the present invention relates to a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one infectious disease associated antigenic protein or at least one tumor associated antigenic protein. In one embodiment, the present invention relates to a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one viral antigenic protein, optionally a coronavirus (e.g.
- the present invention relates to a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one SARS-CoV-2 antigenic protein. In one embodiment, the present invention relates to a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one tumor associated antigenic protein, optionally a blood cancer antigenic protein.
- the present invention relates to a therapeutic composition
- a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one infectious disease associated antigenic protein or at least one tumor associated antigenic protein, wherein the at least one mRNA is comprised in nanoparticles, and wherein the at least one mRNA is optionally modified with one or more naturally occurring modified nucleosides.
- the present invention relates to a therapeutic composition
- a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one infectious disease associated antigenic protein or at least one tumor associated antigenic protein, wherein the at least one mRNA is comprised in lipid nanoparticles, and wherein the at least one mRNA is optionally modified with one or more naturally occurring modified nucleosides.
- the present invention relates to a therapeutic composition
- a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one viral antigenic protein, optionally a SARS-CoV-2 antigenic protein, wherein the at least one mRNA is comprised in lipid nanoparticles, and wherein the at least one mRNA is optionally modified with one or more naturally occurring modified nucleosides.
- the at least one mRNA encodes a SARS-COV-2 spike protein or a fragment thereof.
- the fragment has a length of at least 100, at least 200, at least 400, at least 600 or at least 800 amino acids.
- the fragment has a length of 400 to 1200 amino acids, 600 to 1200 amino acids or 800 to 1200 amino acids.
- the at least one mRNA, encoding a SARS-COV-2 antigenic protein comprises a sequence of SEQ ID NO:19 or a sequence, which is at least 75%, 85%, 90%, 95%, 98% or 99% or 100% identical to SEQ ID NO:19.
- the present invention relates to a therapeutic composition
- a therapeutic composition comprising phDCs and at least one mRNA, which comprises a coding sequence encoding for at least one blood cancer antigenic protein, wherein the at least one mRNA is comprised in lipid nanoparticles, and wherein the at least one mRNA is optionally modified with one or more naturally occurring modified nucleosides.
- the respective embodiments as mentioned before and below apply mutatis mutandis.
- the lipid nanoarticles can comprise a cationic lipid, a polyethylene glycol (PEG) modified lipid, a cholesterol-based lipid and/or a non-cationic lipid.
- the cationic lipid is present at a molar ratio between 30% and 40%
- said PEG-modified lipid is present at a molar ratio between 1.5% and 4.0%
- said cholesterol-based lipid is present at a molar ratio between 40% and 52%
- said non-cationic lipid is present at a molar ratio between 11% and 21%, wherein all the molar ratios are relative to the total lipid content of the LNP.
- the at least one mRNA optionally modified with one or more naturally occurring modified nucleosides can be fully modified with N1-methyl-pseudouridine in place of every uridine.
- the therapeutic composition comprises (e.g. at least a portion of) antigen-specific phDCs.
- Said antigen-specific phDCs are obtainable by combining phDCs with the at least one mRNA encoding for the at least one antigenic protein or by combining monocytes with the at least one mRNA encoding for the at least one antigenic protein and then subjecting the mixture of monocytes and the at least one mRNA encoding for the at least one antigenic protein to a physical force.
- the present invention further relates to a therapeutic composition comprising antigen- specific monocytes and at least one mRNA encoding for at least one antigenic protein.
- the antigen-specific monocytes are obtainable by - combining monocytes (which have been obtained as described above) with at least one mRNA encoding for at least one antigenic protein; - subjecting the monocytes and the at least one mRNA encoding for at least one antigenic protein to a physical force.
- the present invention relates to a therapeutic composition comprising antigen-specific monocytes, phDCs and at least one mRNA encoding for at least one antigenic protein.
- a physical force e.g.
- the at least one mRNA can be comprised in nanoparticles, lipid nanoparticles in particular. All embodiments, e.g. as regards the at least one mRNA encoding for the at least one antigenic protein, or e.g. nanoparticles (in particular lipid nanoparticles), as disclosed herein apply mutatis mutandis.
- the therapeutic composition according to the first aspect can be used for therapeutic treatment.
- the invention relates to a therapeutic composition for use in a method of treating a disease in a subject, said method comprising administering the therapeutic composition to the subject.
- the subject has been previously been diagnosed as having the disease
- the therapeutic compositions of the invention are suitable for use in the treatment against diseases as caused by the pathogens as listed in the first aspect or against the tumors/cancers as listed in the first aspect.
- the therapeutic compositions of the invention are suitable for use in the treatment of diseases as caused by the pathogens listed in the detailed description, section “pathogens” or the tumors/cancers as listed in the detailed description, section “tumors/cancer”.
- the therapeutic compositions of the invention are suitable for use in the treatment of AIDS, hepatitis A, hepatitis B, hepatitis C, polio, gastroenteritis, dengue fever, yellow fever, encephalitis, meningitis, or meningoencephalitis caused by West nile virus, influenza, ebola, rabies, mumps, measles, herpes, pox, Middle East respiratory syndrome, severe acute respiratory syndrome, Covid-19, borreliosis, pseudomonas, burkholderia, tuberculosis and/or malaria depending on the at least one antigen or the at least one mRNA (encoding the at least one antigenic protein), which is present in the composition.
- the therapeutic compositions of the invention are for use in the treatment of AIDS and/or Covid-19. In one embodiment, the therapeutic compositions of the invention are for use in the treatment of Covid-19. In one embodiment, the therapeutic compositions of the invention are for use in treatment of Covid-19 and are administered intranasally.
- the subject and the donor are identical. In this embodiment, the subject is treated with autologous (antigen-specific) phDCs. In one embodiment, the subject and the donor are related. In another embodiment, the subject and donor are different (i.e. not related). In this embodiment, the subject is treated with allogeneic (antigen-specific) phDCs.
- Administration of the therapeutic compositions of the invention can be subcutaneous, intravenous, intramuscular, inhalative, intra-articular, intra-synovial, intrasternal, intrathecal, intra-hepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, intranasal or sublingual.
- administration is inhalative, intravenous, intradermal, intranasal or intramuscular.
- administration is intradermal or intramuscular.
- administration is intranasal.
- administration is intravenous.
- the therapeutic compositions of the invention are administered more than once, e.g.
- the therapeutic compositions of the invention are able to stimulate an immune response against the disease associated antigen or antigenic protein in the subject. In one embodiment, the therapeutic compositions of the invention are able to stimulate an immune response against an infectious disease associated antigen or infectious disease associated antigenic protein. In one embodiment, the therapeutic compositions of the invention are able to stimulate an immune response against a tumor associated antigen or a tumor associated antigenic protein.
- the disease is caused by disease causing particles, optionally comprising viruses, bacteria, fungi, parasites and/or tumor cells. In one embodiment, the disease causing particles comprise pathogens such as viruses, bacteria, fungi and parasites. In one embodiment, the disease causing particles comprise viruses.
- the disease causing particles comprise tumor cells (or cancer cells).
- the therapeutic compositions as provided herein may be characterized in that subjects, who are treated with such compositions (e.g., with at least one dose, at least two doses, etc) may show reduced and/or more transient presence of disease causing particles in relevant site(s) (e.g. nose and/or lungs, and/or any other tissue susceptible to the corresponding disease or disorder) as compared with an appropriate control (e.g. an established expected level for a comparable subject or population not having been treated and having been exposed to the disease causing particles; or a comparable subject or population having been treated with a different therapeutic such as an RNA therapeutic not comprising dendritic cells).
- relevant site(s) e.g. nose and/or lungs, and/or any other tissue susceptible to the corresponding disease or disorder
- an appropriate control e.g. an established expected level for a comparable subject or population not having been treated and having been exposed to the disease causing particles; or a comparable subject or population having been treated with
- the concentration of the disease causing particles is lower as compared to the concentration of the disease causing particles in a second subject, which has been previously been diagnosed with the same disease, but which has not been administered the therapeutic composition of the invention; or as compared the concentration of the disease causing particles in a second subject, which has been previously been diagnosed with the same disease, and which has been treated against the same disease causing particle with a different therapeutic, preferably a therapeutic not comprising dendritic cells; even more preferred a RNA therapeutic not comprising dendritic cells.
- the concentration of the disease causing particles is effectively lower after a tumor outbreak in the subject as compared to the concentration of the disease causing particles in a second subject, which has been previously been diagnosed with the same tumor, and which has not been administered the therapeutic composition of the invention; or as compared to the concentration of the disease causing particles in a second subject, which has been previously been diagnosed with the same tumor, and which has been treated against the same tumor causing particle with a different therapeutic, preferably a therapeutic not comprising dendritic cells; even more preferred an RNA therapeutic not comprising dendritic cells.
- the anti-antigen antibody titer in the subject is lower after treatment with the therapeutic composition of the invention as compared to the anti- antigen antibody titer in a second subject, which has been treated against the same disease causing particle with a different therapeutic, preferably a therapeutic not comprising dendritic cells; even more preferred an RNA therapeutic not comprising dendritic cells.
- the subject who received the therapeutic composition of the invention is equally well or even better treated as compared to the second subject showing a higher anti-antigen antibody titer (e.g. shows less severe symptoms, a more transient presence of disease causing particles and/or shorter course of disease).
- the concentration of the disease causing particles is lower by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98% or at least 99% in the subject.
- the time period in which the subject shows symptoms of the disease is reduced by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98% or at least 99%.
- the concentration of the disease causing particles is lower in the subject systemically. In one embodiment, the concentration of the disease causing particles is lower locally. In one embodiment, the concentration of the disease causing particles is lower in the brain. In one embodiment, the concentration of the disease causing particles is lower in mucosal tissues (i.e., the therapeutic compositions of the invention induce mucosal immunity).
- the reduction may be at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%.
- Comparison can be made to an appropriate control, e.g., as compared to before outbreak of the disease; as compared to the second subject as mentioned above; as compared to subjects not having been treated with the therapeutic composition of the invention or having been treated with a different therapeutic such as a RNA therapeutic not comprising dendritic cells.
- the subject after treatment with the therapeutic composition of the invention, shows an increased proportion of central memory T cells (Tcm) and/or stem-like T cells, which are specific to the at least one disease associated antigen (or antigenic protein).
- Tcm central memory T cells
- stem-like T cells which are specific to the at least one disease associated antigen (or antigenic protein).
- the subject after treatment with the therapeutic composition of the invention, shows a decreased proportion of effector dominant T cells (Teff), which are specific to the at least one disease associated antigen (or antigenic protein).
- Teff effector dominant T cells
- the subject after treatment with the therapeutic composition of the invention, shows an increased proportion of natural killer cells (NK).
- NK natural killer cells
- the subject after treatment with the therapeutic composition of the invention, shows an increased level of IFN- ⁇ .
- the subject after treatment with the therapeutic composition of the invention, shows a decreased proportion of exhausted effector T cells.
- Comparison can be made to an appropriate control, e.g., as compared to before outbreak of the disease; or as compared to the levels or proportions in subjects not having been treated with the therapeutic composition of the invention, or having been treated with a different therapeutic such as a RNA therapeutic not comprising dendritic cells.
- the increase in proportion of central memory T cells and/or stem-like T cells and/or natural killer cells can be at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%.
- the decrease in proportion of effector dominant T cells and/or exhausted effector T cells can be at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%.
- the increase in level of IFN- ⁇ secretion can be at least 2-fold, at least 10-fold, at least 50-fold, at least 100-fold, at least 500-fold or at least 5000-fold.
- the proportion of Tcm can be determined, e.g., by measuring markers which are associated with a central memory T cell phenotype such as CD44/CD62L. Teff show reduced expression of CD62L as compared to Tcm.
- the proportion of stem-like T cells can be determined by, e.g., measuring markers, which are associated with a stem-like T cell phenotype such as IL7Ra/SCA-1. The expression of both markers is increased for stem-like T cells.
- NK cells can e.g. be determined by positive selection using a NK1.1 cell isolation method.
- the proportion of exhausted effector T cells can be determined by, e.g., measuring expression markers, which are associated with an exhausted state such as PD1.
- Treatment can refer to one dose or to multiple doses such as at least 2, at least 3, at least 4 or at least 5 doses.
- the therapeutic compositions of the invention are administered to a subject, which has been previously been diagnosed to have an infectious disease.
- the subject can have been previously been diagnosed to have a viral infectious disease.
- the subject can have been previously been diagnosed to have coronavirus disease 2019 (Covid-19).
- the subject can have been previously been diagnosed to have a cancer or a tumor.
- said cancer (or tumor) is classifiable as stage I, II, III or IV according to the Tumor Node Metastasis (TNM) anatomic/prognostic group system of the cancer staging system of the American Joint Committee on Cancer.
- TNM Tumor Node Metastasis
- said cancer (or tumor) is classifiable as stage I according to the Tumor Node Metastasis (TNM) anatomic/prognostic group system of the cancer staging system of the American Joint Committee on Cancer.
- said cancer (or tumor) is classifiable as stage II according to the Tumor Node Metastasis (TNM) anatomic/prognostic group system of the cancer staging system of the American Joint Committee on Cancer.
- said cancer (or tumor) is classifiable as stage III according to the Tumor Node Metastasis (TNM) anatomic/prognostic group system of the cancer staging system of the American Joint Committee on Cancer.
- said cancer (or tumor) is classifiable as stage IV according to the Tumor Node Metastasis (TNM) anatomic/prognostic group system of the cancer staging system of the American Joint Committee on Cancer.
- the subject is elderly, pregnant, an infant, has a chronic medical condition, underwent cancer treatment recently, has a pulmonary disease or is immunocompromised.
- the subject is over age 60, under age 18, immunocompromised, pregnant, has an inherited disease that affects the immune system, has been previously been diagnosed as having an infectious disease, has an underlying medical condition, is overweight or obese, and/or has prior diagnosis of COVID-19.
- administration of the therapeutic composition of the invention additionally has a vaccination effect in the subject.
- subjects to be treated with the therapeutic composition of the invention are older subjects (used synonymously with “elderly”, e.g., subjects over age 60, 65, 70, 75, 80, 85, etc, for example subjects of age 65-85).
- subjects to be treated with the therapeutic composition of the invention are age 18 or younger. In some embodiments, subjects to be treated with the therapeutic composition of the invention are age 12 or younger. In some embodiments, subjects to be treated with the therapeutic composition of the invention are age 10 or younger. In some embodiments, subjects to be treated with the therapeutic composition of the invention are infants, e.g. less than 1 year old. In some embodiments, subjects to be treated with the therapeutic composition of the invention are pregnant.
- subjects to be treated with the therapeutic composition of the invention are immunocompromised subjects (e.g., those with HIV/AIDS; cancer and transplant patients who are taking certain immunosuppressive drugs; autoimmune diseases or other physiological conditions expected to warrant immunosuppressive therapy (e.g., within 3 months, within 6 months, or more); and those with inherited diseases that affect the immune system (e.g., congenital agammaglobulinemia, congenital IgA deficiency)).
- subjects to be treated with the therapeutic composition of the invention have been previously been diagnosed as having an infectious disease (e.g. those infected with human immunodeficiency virus (HIV) and/or a hepatitis virus (e.g., HBV, HCV)).
- HIV human immunodeficiency virus
- HCV hepatitis virus
- subjects to be treated with the therapeutic composition of the invention are those with underlying medical conditions (e.g. hypertension, cardiovascular disease, diabetes, chronic respiratory disease, e.g. chronic pulmonary disease, asthma, etc., cancer, and other chronic diseases such as, e.g., lupus, rheumatoid arthritis, chonic liver diseases, chronic kidney diseases (e.g., Stage 3 or worse such as in some embodiments as characterized by a glomerular filtration rate (GFR) of less than 60 mL/min/1.73m 2 ).
- subjects to be treated with the therapeutic composition of the invention are overweight or obese subjects, e.g., specifically including those with a body mass index (BMI) above about 30 kg/m 2 .
- BMI body mass index
- subjects to be treated with the therapeutic composition of the invention have prior diagnosis of COVID-19 or evidence of current or prior SARS-CoV-2 infection, e.g., based on serology or nasal swab.
- the therapeutic composition of the invention are administered to one or more of the above risk groups.
- the present invention relates to the therapeutic composition of the first aspect for use in a method of treating a disease in a subject, wherein the subject has been previously been diagnosed as having the disease, said method comprising administering the therapeutic composition to the subject.
- the therapeutic composition of the invention is for use in a method of treating a disease in a subject, wherein the subject has been previously been diagnosed as having the disease, said method comprising administering the therapeutic composition to the subject, wherein the disease is an infectious disease or a tumor.
- the therapeutic composition of the invention is for use in a method of treating a viral disease in a subject, wherein the subject has been previously been diagnosed as having the viral disease, said method comprising administering the therapeutic composition to the subject.
- the therapeutic composition of the invention is for use in a method of treating Covid-19 disease in a subject, wherein the subject has been previously been diagnosed as having Covid-19, said method comprising administering the therapeutic composition to the subject.
- the therapeutic composition of the invention is for use in a method of treating a blood cancer in a subject, wherein the subject has been previously been diagnosed as having the blood cancer, said method comprising administering the therapeutic composition to the subject.
- the therapeutic composition of the invention is for use in a method of treating a solid tumor in a subject, wherein the subject has been previously been diagnosed as having the solid tumor, said method comprising administering the therapeutic composition to the subject.
- the present invention relates to a method of treating a disease in a subject, wherein the subject has been previously been diagnosed as having the disease, said method comprising administering the therapeutic composition of the first aspect (including all embodiments of the first and second aspect as described above) to the subject.
- Fourth aspect Use of a therapeutic composition for therapeutic treatment
- the present invention relates to the use of a therapeutic composition for therapeutic treatment, wherein the therapeutic composition is the composition of the first aspect.
- the therapeutic composition phDCs, nanoparticles, RNA (e.g. mRNA) and at least one antigenic protein
- the respective embodiments of the first, second and third aspect apply mutatis mutandis.
- Kit comprising phDCs and mRNA
- the present invention relates to a kit comprising phDCs and at least one RNA (e.g. mRNA), which encodes for at least one antigenic protein.
- the kit may further comprise nanoparticles (e.g. lipid nanoparticles).
- the nanoparticles comprise the RNA (e.g. mRNA), which encodes for the at least one antigenic protein.
- the respective embodiments of the first aspect apply mutatis mutandis.
- the present invention also relates to a kit comprising monocytes and at least one RNA (e.g. mRNA), which encodes for at least one antigenic protein.
- the kit may further comprise nanoparticles (e.g. lipid nanoparticles).
- the nanoparticles comprise the RNA (e.g. mRNA), which encodes for the at least one antigenic protein.
- the respective embodiments of the first aspect apply mutatis mutandis.
- FIGURE LEGENDS Figure 1 Murine phDC specifically internalize and express Spike protein encoded by mRNA-containing LNPs.
- A, B FACS plot showing spike protein positivity in CD11b positive phDCs without LNP transfection (A) and with LNP transfection (B).
- C, D Confocal microscopy analysis using same sample showing spike protein expression in CD11b+Ly6G- phDCs (projected Z-stack images).
- E Z-plane slice image of the LNP transfected phDC.
- Figure 2 A C57BL/6 mice were administered i.v. treatment with phDCs transduced with OVA mRNA LNP, or treated with OVA mRNA LNP via i.m.
- FIG. 3 A, phDC[ova] or IM[ova] mice were given five total treatments at 10ng [ova] per mouse, starting two days following EG7-OVA subcutaneous tumor inoculation with 3*10 6 cells per mouse (same experiment as Fig. 3).
- B-C tumor growth was monitored for the duration of the experiment, with cumulative tumor growth per experimental group (B) and individual mouse tumor growth curves (C) represented.
- D-E CD8+ splenic T cells were isolated from treated mice on Day 30 and immediately placed into an 18hr IFN-g Elispot assay at 1*10 5 cells per well in the presence or absence of 10ug/mL SIINFEKL peptide.
- Splenocytes from the treated mice were harvested at the end of the 28-day observation period for T cell analysis and characterization by flow cytometry of A, antigen (SIINFEKL)-specific CD8 T cells via dextramer analysis; Tem (effector)/Tcm (central memory) type phenotype analysis via CD44/CD62L expression; detection of stem- like T cell via IL7Ra/SCA-1 expression, and B, T cell exhaustion marker evaluation via PD-1 expression.
- *[ova] LNP-mRNA encoding ovalbumin.
- Figure 5 phDC[ova] or IM[ova] mice were vaccinated (prime/boost) on days -7 and 0, respectively.
- Undifferentiated splenocytes were harvested post vaccination and immediately placed in an 18hr IFN-g Elispot without added antigenic stimulation (A, B and C).
- IFN-g 18hr IFN-g Elispot without added antigenic stimulation
- isolated CD8+ or NK1.1+ splenocytes were collected post vaccination from phDC[ova] mice and assessed for IFN-g spots (D).
- *[ova] LNP-mRNA encoding ovalbumin.
- Figure 6 A C57BL/6 mice were administered i.v. treatment with phDCs transduced with 1ug/mL OVA mRNA LNP, or treated i.v.
- FIG. 8 Mouse phDCs were incubated overnight with OVA in various antigen form; soluble, expressed in tumor cells or OVA encoding mRNA containing LNPs. Cells were then harvested, and stained for surface CD11b, Ly6G and 25.D1 (ab against SIINFEKL bound MHC I in H- 2Kb strain). Ly6G positive cells were selected out for neutrophil exclusion. FACs plot showing 25.D1 positivity on CD11b+ phDCs.
- Figure 9 PhDC PP PBMCs from B6 mice) were transduced with OVA mRNA LNPs (1 ug/ml) in standard overnight culture protocol.
- Figure 12 phDC from mouse were pulsed with OVA mRNA LNPs at differing concentration, then cultured with OT1 T cells. Proliferation of the OT1 CD8 T cells (CFSE labeled) was evaluated as shown by reduction in CFSE signal.
- Figure 13 phDC from mouse (B6) were pulsed with 1 ug/ml OVA mRNA, then cultured with 100K OT1 T cells at differing phDC number (96 well U bottom plate). Proliferation of the OT1 CD8 T cells (CFSE labeled) were evaluated as readout.
- Figure 14 C57BL/6 mice were administered i.v.
- phDCs can be prepared from monocytes (e.g. obtained from a donor). For instance, phDCs can be generated by subjecting monocytes (e.g. obtained from a donor) to a physical force.
- a physical force can e.g. be generated by passing or moving the monocytes through a flow chamber.
- phDCs are obtained by plate-passage of monocytes using an extracorporeal photopheresis (ECP) derived process.
- ECP extracorporeal photopheresis
- the flow chamber can be a plate in one embodiment.
- a plate for preparing phDCs has been previously described in the literature, see e.g.
- ECP describes a process in which monocytes derived from a blood sample or a fraction thereof are exposed to mechanical stress (e.g., shear forces) and plasma components (e.g., platelets) or derivatives or mimics thereof, thereby activating the monocytes to differentiate into healthy, physiologic dendritic cells which are also termed phDC herein.
- mechanical stress e.g., shear forces
- plasma components e.g., platelets
- ECP and ECP derived processes including the differentiation of monocytes into phDCs, may be performed in a flow chamber (e.g.
- a plate a flow chamber of a device such as a large-scale ECP device, e.g., a clinical ECP device (e.g., a THERAKOS® CELLEX® device); or in a miniaturized ECP device, e.g., a Transimmunization plate as described in WO2017/005700 A1; or in a bag (e.g. flexible bag or plastic bag); or in a combination of any of the afore-mentioned).
- the bag can be made of any material that does not leak liquids such as e.g. rubber, silicone or plastic. Preferably, the material is able to bend easily without breaking.
- the bag or plate is made of a plastic material.
- Suitable plastic materials comprise polyolefin, polyethylene, fluoropolymer, polyvinyl chloride, ethylene-vinyl acetate-copolymer, ethylene vinyl alcohol, polyvinylidene fluoride, and/or other plastic comprising materials approved for medical use.
- the preferred plastic material is ethylen-vinyl acetate-copolymer.
- the bag or plate may be made of a material that provides a degree of transparency such that the sample or cell mixture can be irradiated with visible or UV light.
- the flow chamber can be hybrid flow chamber or a combination of a chamber and a bag, wherein each of the components are as described herein.
- phDC obtained by the method described above are advantageous as compared to DC obtained by other methods such as incubation of blood monocytes with cytokines or direct isolation from a donor, as phDC are generated physiologically (without the need for chemicals such as cytokines and/or apoptotic agents) with greater reproducibility and controllability under precise in vitro laboratory conditions.
- Benefits of the above described method for generating phDCs compared to other methods such as incubation with cytokines include one or more of higher yields, a faster process, better intracellular antigen processing and more effective priming of disease-specific cytotoxic T cells.
- phDC of a donor are obtained by subjecting monocytes contained in a blood sample (obtained from the donor) to a shear force by passing the blood sample or fraction thereof through a flow chamber.
- a blood sample obtained from the donor
- platelets are present in the flow chamber which can be either derived from the donor ⁇ s blood sample or a fraction thereof or provided separately.
- plasma components can be present in the flow chamber which can be either derived from the donor ⁇ s blood sample or a fraction thereof or provided separately.
- a monocyte of a donor may be obtained by any suitable means, e.g., from a blood sample or a fraction thereof.
- the fraction of the blood sample may be, e.g., a buffy coat including white blood cells and platelets.
- the fraction of the blood sample may be an isolated peripheral blood mononuclear cell (PMBC).
- PMBCs may be isolated from a blood sample using, e.g., centrifugation over a Ficoll-Hypaque gradient (Isolymph, CTL Scientific).
- the fraction of the blood sample may be a purified or enriched monocyte preparation.
- Monocytes may be enriched from PBMCs using, e.g., one, two, or all three of plastic adherence; CD14 magnetic bead positive selection (e.g., from Miltenyi Biotec); and a Monocyte Isolation Kit II (Miltenyi Biotec). Any suitable volume of blood can be used.
- the blood sample (e.g., the blood sample from which the fraction is derived) may be between about 1 ⁇ L and about 500 mL, e.g., between about 1 ⁇ L and about 10 mL, between about 1 ⁇ L and about 5 mL, between about 1 ⁇ L and about 1 mL, between about 1 ⁇ L and about 750 ⁇ L, between about 1 ⁇ L and about 500 ⁇ L, between about 1 ⁇ L and about 250 ⁇ L, between about 10 mL and about 450 mL, about 20 mL and about 400 mL, about 30 mL and about 350 mL, about 40 mL and about 300 mL, about 50 mL and about 200 mL, or about 50 mL and about 100 mL.
- mL and about 500 mL e.g., between about 1 ⁇ L and about 10 mL, between about 1 ⁇ L and about 5 mL, between about 1 ⁇ L and about 1 mL, between about 1
- the blood sample or the fraction thereof or the additional blood sample or the fraction thereof is less than or equal to about 100 mL (e.g., about 50 mL to about 100 mL).
- the monocytes are derived from a blood sample obtained from the donor.
- monocytes can be derived from other sources in the donor that provide monocytes, e.g. bone marrow, spleen or other peripheral tissues.
- monocytes are derived from peripheral blood mononuclear cells (PBMC) obtained from the donor.
- PBMC peripheral blood mononuclear cells
- CD80, CD83, and CD86 are markers expressed in higher levels by mature dendritic cells as compared to monocytes.
- the donor is mammalian. Mammals include for example, but are not limited to, humans, non-human primates, pigs, dogs, cats and rodents. In a preferred embodiment, the donor is human. II.
- the antigenic protein phDCs are combined with at least one mRNA encoding at least one antigenic protein.
- the antigenic protein may be a disease associated antigen or a fragment thereof. If it is referred to a “disease associated antigen” or “antigenic protein”, “a fragment thereof” is included in the meaning.
- phDCs Upon combination of phDCs with a disease associated antigen, phDCs are able to take up, process and present the antigen on their surface. This process is also referred to as “loading”. Such loaded phDCs can elicit potent anti-disease, e.g. anti-infectious disease or anti-tumor, immune responses.
- the at least one disease associated antigen can be loaded as the antigen itself (e.g., proteins, peptides, epitopes, cells, cell or tissue lysates, viruses, viral particles, etc., including fragments of each of the aforementioned) or can be loaded (loaded in the meaning of the phDCs taking up the antigen or antigenic protein) as a nucleic acid encoding antigen.
- the phDCs comprise a nucleic acid, preferably a RNA, more preferably an mRNA, encoding the antigenic protein or disease associated antigen.
- the at least one disease associated antigen is loaded as a nucleic acid encoding the antigen.
- the at least one disease associated antigen can be provided directly or stored in frozen or a lyophilized form until use.
- Methods for loading dendritic cells with antigens are known to those of skill in the art.
- the phDCs are loaded by combination, e.g. incubation, of the phDCs with the at least one disease associated antigen.
- the phDCs may be loaded with different antigens to give rise to multi-valent antigen- specific phDCs.
- phDCs are loaded by electroporation, polymer or lipid based nanoparticles or the cell squeeze method.
- phDCs are loaded by encapsulating the at least one disease associated antigen (e.g.
- the at least one antigen is encoded by RNA, in particular mRNA
- the at least one disease associated antigen is referred to as an antigenic protein for the purpose of this invention.
- phDCs are flowed in a solution containing the at least one disease associated antigen or fragment thereof. For loading, phDCs are squeezed through a microfluidic constriction that is smaller than the diameter of the cells. This results in temporary disruption of the cell membrane, enabling the at least one disease-associated antigen to enter the cytosol of the phDCs.
- the mixture can for example be incubated under standard conditions. Culturing can be performed under standard conditions, e.g. at 37° C and 5% CO 2 in standard mediums for culturing of human cells such as in RPMI-1640 medium (obtainable e.g. from GIBCO), supplemented with 15% AB serum (obtainable from e.g. Gemini Bio-Products).
- An incubation step can be added after combining the phDCs with at least one disease associated antigen. Alternatively or additionally, an incubation step can also be inserted after generation of phDCs from monocytes.
- the incubation step can be performed for 0.5 h, 1 h, 2 h, 3 h, 4 h, 5 h, 6 h, 12 h, 20 h or 24 h. In another embodiment, the incubation step can be performed for at least 0.5 h, at least 1 h, at least 2 h, at least 3 h, at least 4 h, at least 5 h, at least 6 h, at least 12 h, at least 20 h or at least 24 h. It needs to be understood that all method steps are performed in vitro. II.A Disease causing particles
- the at least one disease associated antigen or the at least one antigenic protein can be derived from disease causing particles, which comprise pathogens and tumor cells.
- Pathogens are viruses, bacteria, fungi, prions and parasites.
- disease causing particles comprise viruses, bacteria, fungi, prions, parasites and tumor cells.
- antigenic protein is used interchangeably with the term “antigenic protein”.
- an infectious disease antigen or tumor antigen it is also referred to an infectious disease associated antigenic protein or a tumor associated antigenic protein.
- a viral antigen is mentioned, it is also referred to a viral antigenic protein.
- HIV or SARS- CoV-2 antigen it is also referred to a HIV or SARS-CoV-2 antigenic protein.
- the disease associated antigen is an infectious disease associated antigen.
- the disease associated antigen is a viral antigen.
- viruses, from which the disease associated antigen can be derived include: Retroviridae (e.g. human immunodeficiency viruses, such as HIV-1 (also referred to as HTLV-III, LAV or HTLV-III/LAV, or HIV-III; and other isolates, such as HIV-LP, HIV-2; Picornaviridae (e.g. polio viruses, hepatitis A virus; enteroviruses, human coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (e.g.
- Togaviridae e.g. equine encephalitis viruses, rubella viruses
- Flaviridae e.g. dengue viruses, encephalitis viruses, yellow fever viruses
- Coronaviridae e.g. coronaviruses
- Rhabdoviridae e.g. vesicular stomatitis viruses, rabies viruses
- Filoviridae e.g. ebola viruses
- Paramyxoviridae e.g. parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus
- Orthomyxoviridae e.g. influenza viruses
- Bungaviridae e.g.
- the disease associated antigen is a viral antigen, more preferably a Retroviridae or Coronaviridae antigen, and most preferably a HIV or SARS-CoV-2 antigen.
- the disease associated antigen is derived from SARS-CoV-2.
- the disease associated antigen is a bacterial antigen.
- bacteria, from which the disease associated antigen can be derived include: Helicobacter pyloris, Borrelia spp. (e.g. Borrelia burgdorferi), Legionella pneumophilia, Mycobacteria spp. (e.g. M. tuberculosis, M. avium, M. intracellulare, M. kansasii, M.
- the bacterial antigen is a Borrelia spp. or Mycobacteria spp. antigen.
- the disease associated antigen is a fungal antigen. Examples of fungi, from which the disease associated antigen can be derived include: Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis and Candida albicans.
- the disease associated antigen is a parasite antigen. Parasite antigens can be derived from protozoa, helminths or ectoparasites.
- the disease associated antigen is a tumor or cancer antigen, e.g. a tumor associated antigen (TAA) or a tumor specific antigen (TSA).
- TAA tumor associated antigen
- TSA tumor specific antigen
- TAAs comprise the group of TSAs, if not mentioned otherwise.
- Tumor or cancer antigens can be found on tumor or cancer cells.
- the tumor or cancer antigen can be derived from a solid tumor or a blood cancer.
- Examples of tumors or cancers, from which the disease associated antigen can be derived comprise leukemia, melanoma, lymphoma, endometrial cancer, kidney cancer, brain cancer, cervical cancer, liver cancer, head and neck cancer, gastrointestinal cancer, lymph node cancer, pancreas cancer, ear, nose and throat (ENT) cancer, breast cancer, prostate cancer, ovarian cancer or lung cancer.
- the disease associated antigen is a tumor-associated antigen.
- Tumor-associated antigens comprise Her2, prostate stem cell antigen (PSCA), PSMA (prostate-specific membrane antigen), B cell maturation antigen (BCMA), ERK5, alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), cancer antigen-125 (CA- 125), CA19-9, calretinin, MUC-1, epithelial membrane protein (EMA), epithelial tumor antigen (ETA), tyrosinase, melanoma-associated antigen (MAGE), CD34, CD45, CD99, CD117, chromogranin, cytokeratin, desmin, glial fibrillary acidic protein (GFAP), gross cystic disease fluid protein (GCDFP-15), HMB-45 antigen, protein melan-A (melanoma antigen recognized by T lymphocytes; MART-1), myo- D1, muscle-specific actin (MSA), neurofilament, neuron-specific enolase (NSE), placental alkaline phosphata
- the tumor-associated antigen is CD19, CD22, CD27, CD30, CD70, GD2 (ganglioside G2), EGFRvIII (epidermal growth factor variant III), sperm protein 17 (Sp17), mesothelin, PAP (prostatic acid phosphatase), prostein, TARP (T cell receptor gamma alternate reading frame protein), Trp-p8, or STEAP1 (six-transmembrane epithelial antigen of the prostate 1).
- the TAA is a cancer/testis (CT) antigen, e.g., BAGE, CAGE, CTAGE, FATE, GAGE, HCA661, HOM-TES-85, MAGEA, MAGEB, MAGEC, NA88, NY-ESO-1, NY-SAR-35, OY-TES-1, SPANXB1, SPA17, SSX, SYCP1, or TPTE.
- CT cancer/testis
- the TAA is a carbohydrate or ganglioside, e.g., fuc-GMI, GM2 (oncofetal antigen-immunogenic-1; OFA-I-1); GD2 (OFA-I-2), GM3, GD3, and the like.
- the TAA is alpha- actinin-4, Bage-1, BCR-ABL, Bcr-Abl fusion protein, beta-catenin, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, Casp-8, cdc27, cdk4, cdkn2a, CEA, coa-1, dek-can fusion protein, EBNA, EF2, Epstein Barr virus antigens, ETV6- AML1 fusion protein, HLA-A2, HLA-A11, hsp70-2, KIAAO205, Mart2, Mum-1, 2, and 3, neo-PAP, myosin class I, OS-9, pml-RARa fusion protein, PTPRK, K-ras, N- ras, triosephosphate isomerase, Gage 3,4,5,6,7, GnTV, Herv-K-mel, Lü-1, NA-88, NY-E
- tumor associated antigens are known to those in the art and can be combined with the phDCs provided herein to yield the respective antigen-specific phDCs or can be present in the compositions as disclosed herein.
- II.B Means of providing the disease associated antigen II.B.1 Antigen derived from disease causing particles
- the disease associated antigen can be derived from the disease causing particles, e.g. from cells, biopsies, tissue lysates or other particles which comprise disease causing particles.
- the disease associated antigen can be derived from the tumor or cancer cells to be treated.
- a sample of tumor cells can be taken from a subject suffering from a cancer, e.g.
- a biopsy or a sample can be taken from a subject suffering from an infectious disease.
- the sample can further be treated with agents, which are suitable to release antigens such as photoactivatable agents in combination with light (in particular, UV light) such as the combination of 8-MOP and UVA.
- agents which are suitable to release antigens such as photoactivatable agents in combination with light (in particular, UV light) such as the combination of 8-MOP and UVA.
- the treated sample can then be combined with phDCs.
- pathogens can be cultured in a medium. Relevant antigens may be shed into the culture medium by the pathogen and can then be collected for combination with the phDCs.
- the skilled person is aware of different and further methods known in the art to provide antigens derived from the disease causing particles.
- the phDCs can be loaded with a nucleic acid (functional fragments thereof are included) encoding for the at least one disease associated antigen.
- phDCs are loaded with RNA (functional fragments thereof are included) encoding for the at least one disease associated antigen.
- the at least one disease associated antigen corresponds to the at least one antigenic protein.
- phDCs are directly combined with RNA (e.g. mRNA).
- the RNA (e.g. mRNA) may be translated into the respective disease associated antigen (i.e. antigenic protein) after entering the phDCs.
- the RNA can be a single stranded RNA, an mRNA, a self-amplifying RNA, a circular RNA and/or a synthetic RNA.
- the RNA is an mRNA.
- the mRNA is self-amplifying mRNA.
- the basic components of an mRNA molecule typically include at least one coding region, a 5’ untranslated region (UTR), a 3’ UTR, a 5’ cap and a poly-A tail.
- the at least one disease associated antigen i.e. antigenic protein
- a therapeutic composition comprising phDCs and at least one mRNA (which comprises a coding sequence) encoding for at least one antigenic protein.
- the phDCs can be in a composition with multiple mRNAs that encode more than one antigenic protein, such as 2, 3, 4, 5, 6, 7, 8, 9, 10 or more antigenic proteins, from the same or different pathogens or tumors.
- a polycistronic mRNA can be designed that can be translated into more than one antigenic protein.
- the mRNA encodes one or more antigenic proteins of the above listed disease causing particles or the mRNA encodes one or more antigenic proteins of the above listed tumor or infectious disease associated antigens. In one embodiment, the mRNA encodes one or more antigenic proteins of the above listed pathogens. In one embodiment, the mRNA encodes one or more antigenic proteins of the above listed viruses, bacteria, fungi, prions or parasites. In one embodiment, the mRNA encodes one or more antigenic proteins of the above listed viruses, optionally coronaviruses. In one embodiment the mRNA encodes one or more antigenic proteins of the above listed disease causing particles. In one embodiment the mRNA encodes one or more antigenic proteins of the above listed tumors or cancers.
- the mRNA encodes one or more antigenic proteins of the above listed TAAs. In one embodiment, the at least one mRNA encodes for one antigenic protein. SARS-CoV-2 sequences In one embodiment, the at least one mRNA encodes one or more antigenic proteins derived from SARS-CoV, optionally SARS-CoV-2. Antigenic proteins derived from SARS-CoV or SARS-CoV-2 comprise the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein. Thus in one embodiment, the antigenic protein is the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein or a fragment of each of the aforementioned.
- a fragment comprises at least 10, at least 50, at least 100, at least 200, at least 400 or at least 800 amino acid residues. Due to their surface expression properties, RNA polynucleotides encoding structural proteins are believed to have preferred immunogenic activiy and, hence, may be most suitable for the compositions of the invention.
- the mRNA encodes one or more antigenic proteins derived from the spike protein or nucleocapsid protein. Even more preferred, the mRNA encodes one or more antigenic proteins derived from the spike protein. The mRNA may encode for an antigenic protein derived from the S1 subunit or the S2 subunit of the spike protein.
- S-2P Parallelsen et al.2017
- a second-generation spike construct termed “HexaPro” contains four additional prolines at positions 817, 892, 899 and 942.
- the mRNA comprises a sequence or part of a sequence selected from the group comprising SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4 and/or SEQ ID NO: 5.
- the mRNA comprises a sequence or part of a sequence selected from the group comprising SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4 and/or SEQ ID NO: 5 or a sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the selected sequence.
- the mRNA comprises a sequence or part of a sequence selected from the group comprising SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 4 and/or SEQ ID NO: 5 or a sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the selected sequence, wherein the selected RNA sequence is modified with one or more (e.g. all) naturally occuring modified nuclosides as described in the section “Modifications of RNA”.
- the naturally occuring modified nucleoside is 1-methylpseudouridine.
- the mRNA comprises a sequence or part of a sequence corresponding to SEQ ID NO: 19 or a sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to SEQ ID NO: 19 (Table 3).
- mRNA constructs comprising a sequence that encodes a full-length SARS-CoV-2 spike protein (e.g., including embodiments in which such encoded SARS-CoV-2 spike protein may comprise at least one or more amino acid substitutions, e.g., proline substitutions as described herein, and/or embodiments in which the mRNA sequence is optimized e.g., for mammalian, e.g., human, subjects, and/or embodiments, in which the mRNA comprises one or more chemical modifications).
- HIV sequences In one embodiment, the mRNA encodes one or more antigenic proteins derived from HIV.
- Antigenic proteins derived from HIV comprise the envelope protein (env), group antigens polyprotein (gag), reverse transcriptase (pol) and/or negative factor protein (nef). Due to their surface expression properties, RNA polynucleotides encoding structural proteins are believed to have preferred immunogenic activiy and, hence, may be most suitable for the compositions of the invention. Expressly included are also variants of the afore-mentioned HIV proteins.
- the mRNA comprises a sequence or part of a sequence selected from the group comprising SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 and/or SEQ ID NO: 14.
- the mRNA comprises a sequence or part of a sequence selected from the group comprising SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13 and/or SEQ ID NO: 14 or a sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the selected sequence.
- the sequences above are based on the reference genome from NCBI (accession number NC_AF033819, HIV-1) and also displayed in Table 1 below. The corresponding amino acid seuqences are given in Table 2 below.
- mRNA constructs comprising a sequence that encodes a full-length HIV structural protein (e.g., including embodiments in which such encoded HIV structural protein may comprise at least one or more amino acid substitutions, and/or embodiments in which the mRNA sequence is optimized e.g., for mammalian, e.g., human, subjects and/or embodiments, in which the mRNA comprises one or more chemical modifications).
- Table 1 ⁇ ⁇ ⁇ ⁇ Table 2 ⁇ Table 3 Tumor-associated antigen sequences and tumor-specific antigen sequences
- the at least one mRNA encodes one or more antigenic proteins derived from a tumor or cancer.
- the mRNA encodes a TAA polypeptide, e.g. an amino acid sequence of a TAA.
- TAAs comprise Her2, prostate stem cell antigen (PSCA), PSMA (prostate-specific membrane antigen), B cell maturation antigen (BCMA), ERK5, alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), cancer antigen-125 (CA-125), CA19-9, calretinin, MUC-1, epithelial membrane protein (EMA), epithelial tumor antigen (ETA), tyrosinase, melanoma- associated antigen (MAGE), CD34, CD45, CD99, CD117, chromogranin, cytokeratin, desmin, glial fibrillary acidic protein (GFAP), gross cystic disease fluid protein (GCDFP-15), HMB-45 antigen, protein melan-A (melanoma antigen recognized by T lymphocytes; MART-1), myo-D1, muscle-specific actin (MSA
- the TAA is CD19, CD22, CD27, CD30, CD70, GD2 (ganglioside G2), EGFRvIII (epidermal growth factor variant III), sperm protein 17 (Sp17), mesothelin, PAP (prostatic acid phosphatase), prostein, TARP (T cell receptor gamma alternate reading frame protein), Trp-p8, or STEAP1 (six-transmembrane epithelial antigen of the prostate 1).
- the TAA is a cancer/testis (CT) antigen, e.g., BAGE, CAGE, CTAGE, FATE, GAGE, HCA661, HOM-TES-85, MAGEA, MAGEB, MAGEC, NA88, NY-ESO-1, NY-SAR-35, OY-TES-1, SPANXB1, SPA17, SSX, SYCP1, or TPTE.
- CT cancer/testis
- the TAA or TSA is alpha-actinin-4, Bage-1, BCR-ABL, Bcr-Abl fusion protein, beta-catenin, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, Casp-8, cdc27, cdk4, cdkn2a, CEA, coa-1, dek-can fusion protein, EBNA, EF2, Epstein Barr virus antigens, ETV6-AML1 fusion protein, HLA-A2, HLA-A11, hsp70-2, KIAAO205, Mart2, Mum-1, 2, and 3, neo-PAP, myosin class I, OS-9, pml-RARa fusion protein, PTPRK, K-ras, N-ras, triosephosphate isomerase, Gage 3,4,5,6,7, GnTV, Herv-K-mel, Lü-1, NA-88, NY
- the mRNA which encodes an amino acid sequence of a TAA can be a sequence having at least 99%, 98%, 97%, 96%, 95%, 90%, 85%, or 80% identity to the RNA sequence of the selected TAA.
- Modifications of RNA e.g. mRNA
- the RNA is a modified RNA, in particular a stabilized mRNA.
- the RNA e.g. mRNA
- the RNA e.g.
- mRNA independently comprises at least one chemical modification.
- the chemical modification can e.g. be a modified nucleoside.
- the chemical modification comprises a naturally occurring modified nucleoside.
- Naturally occurring nucleosides comprise 1-methyladenosine (m 1 A), N 6 -methyladenosine (m 6 A), 2'-O-methyladenosine (Am), 5-methylcytidine (m 5 C), 2'-O-methylcytidine (Cm), 2-thiocytidine (s 2 C), N 4 -acetylcytidine (ac 4 C), 5- formylcytidine (f 5 C), 2'-O-methylguanosine (Gm), inosine (I), pseudouridine ( ⁇ ), 5- methyluridine (m 5 U), 2'-O-methyluridine (Um).
- the RNA comprises a modified nucleoside in place of at least one uridine.
- the RNA comprises a modified nucleoside in place of each uridine.
- the modified nucleoside is independently selected from pseudouridine, N1-methyl-pseudouridine, 5-methyl- uridine and N1-ethylpseudouridine.
- the modified nucleoside is a N1-methylpseudouridine modification or a N1-ethylpseudouridine modification. In one embodiment, the modified nucleoside is a N1-methylpseudouridine. For instance, N1-methyl-pseudouridine was found to be superior to several other nucleoside modifications and their combinations in terms of translation capacity. In some embodiments, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, or 100% of the uridine nucleosides in the mRNA are chemically modified.
- RNA sequence optimization inter alia comprises codon-optimization, optimization of G/C content and optimization of structural elements (e.g.5’ cap, 5’ UTR, 3’ UTR and poly(A)-tail).
- amino acid sequence of the at least one disease associated antigen (or antigenic protein) is encoded by a coding sequence which is codon- optimized and/or the G/C content of which is increased compared to wild type coding sequence. This also includes embodiments, wherein one or more sequence regions of the coding sequence are codon-optimized and/or increased in the G/C content compared to the corresponding sequence regions of the wild type coding sequence.
- the codon-optimization and/or the increase in the G/C content preferably does not change the sequence of the encoded amino acid sequence.
- the G/C content of the coding region of the RNA is increased by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 55%, or even more compared to the G/C content of the coding region of the wild type RNA.
- the RNA e.g. mRNA
- the RNA comprises a 5' untranslated region (UTR), a 3' UTR, a 5' cap and/or a poly(A) tail.
- the RNA e.g. mRNA
- the RNA contains all of these elements.
- the RNA e.g. mRNA
- the RNA comprises a 5’-UTR and/or a 3’-UTR.
- the RNA comprises a cap.
- the RNA (e.g. mRNA) comprises a 3’-poly(A) sequence.
- the cap is a Cap1 structure or a m7GpppG cap.
- the sequence of the 5’ UTR and/or 3’UTR is optimized.
- the mRNA comprises a 5' or 3' UTR that is derived from a gene distinct from the sequence encoding the at least one disease associated antigen (or antigenic protein), i.e., the UTR is a heterologous UTR.
- the 5’ and/or 3’UTR sequences can be derived from mRNA which are stable (e.g., globin, actin, GAPDH, tubulin, histone, or citric acid cycle enzymes) to increase the stability of the mRNA.
- the 5’-UTR sequence of the human alpha- globin mRNA optionally with an optimized ’Kozak sequence’ to increase translational efficiency may be used.
- the 5’UTR sequence of a human cytochrome mRNA may be used, e.g. the human cytochrome b-245 alpha mRNA or cytochrome p4502E1 mRNA.
- the poly-A sequence comprises at least 100 nucleotides. In one embodiment, the poly-A sequence comprises at least 150 nucleotides. In one embodiment, the poly-A sequence comprises at least 250 nucleotides.
- a poly(A)-tail measuring 110 nucleotides in length, consisting of a stretch of 30 adenosine residues, followed by a 10 nucleotide linker sequence (of random nucleotides) and another 70 adenosine residues may be used.
- This poly(A)-tail sequence enhances RNA stability and translational efficiency.
- a poly(A)-tail measures 300 to 800 nucleotides in length.
- a secretory signal peptide (sec) may be fused to the antigen-encoding regions preferably in a way that the sec is translated as N terminal tag.
- RNA e.g. mRNA
- AURES AU-rich sequences removed. These sequences, sometimes referred to as AURES are destabilizing sequences found in the 3’UTR.
- the AURES may be removed from the RNA (e.g. mRNA). Alternatively the AURES may remain in the RNA (e.g. mRNA).
- Lipid nanoparticles The at least one disease associated antigen (e.g.
- the at least one disease associated antigen can be encapsulated in a lipid nanoparticle (LNP).
- LNP lipid nanoparticle
- the at least one disease associated antigen is an antigenic protein which is encoded by an RNA (e.g. mRNA)
- the RNA can be encapsulated in a lipid nanoparticle for delivery to the phDCs of the invention.
- the encapsulated RNA e.g. mRNA
- the LNP can comprise 3, 4 or 5 classes of lipids.
- LNP with three classes of lipids comprise: (1) an ionizable lipid, (2) a PEGylated lipid and (3) a cholesterol-based lipid.
- LNP with four classes of lipids comprise: (1) an ionizable lipid, (2) a PEGylated lipid, (3) a cholesterol-based lipid and (4) a helper lipid.
- LNPs with five classes of lipids comprise: (1) an ionizable lipid, (2) a PEGylated lipid, (3) a cholesterol-based lipid, (4) a helper lipid and (5) DSPE-PEG-maleimide or DSPN-PEG-azide.
- DSPE-PEG- maleimide or DSPN-PEG-azide allow for adding ligands for targeted delivery.
- Ionizable lipids facilitates mRNA encapsulation and may be a cationic lipid.
- a cationic lipid affords a positively charged environment at low pH to facilitate efficient encapsulation of the negatively charged mRNA.
- the cationic lipid is cKK-E12 ((3,6-bis(4-(bis(2-hydroxydodecyl)amino)butyl)piperazine-2,5-dione); see Dong et al., PNAS (2014) 111(11):3955-60).
- Other cationic lipids that can be used include those described in Dong et al., 2014.
- PEG modified lipid component provides control over particle size and stability of the nanoparticle. The addition of such components may prevent complex aggregation and provide a means for increasing life-time and increasing the delivery of the LNPs to the phDCs.
- Contemplated PEGylated lipids include, but are not limited to, a polyethylene glycol (PEG) chain of up to 5 kDa in length covalently attached to a lipid with alkyl chain(s) of C6-C20 (for example, C8, C10, C12, C14, C16 or C18) length, such as a derivatized ceramide (e.g., N-octanoyl-sphingosine-1- [succinyl(methoxypolyethylene glycol)] (C8 PEG ceramide)).
- PEG polyethylene glycol
- C6-C20 for example, C8, C10, C12, C14, C16 or C18
- the PEGylated lipid is 1,2-dimyristoyl- rac-glycero-3-methoxypolyethylene glycol (DMG-PEG); 1,2-distearoyl-sn-glycero-3- phosphoethanolamine-polyethylene glycol (DSPE-PEG); 1,2-dilauroyl-sn-glycero-3- phosphoethanolamine-polyethylene glycol (DLPE-PEG); or 1,2-distearayl-rac- glycero-polyethelene glycol (DSG-PEG).
- DMG-PEG 1,2-dimyristoyl- rac-glycero-3-methoxypolyethylene glycol
- DSPE-PEG 1,2-distearoyl-sn-glycero-3- phosphoethanolamine-polyethylene glycol
- DLPE-PEG 1,2-dilauroyl-sn-glycero-3- phosphoethanolamine-polyethylene glycol
- DSG-PEG
- the PEG is PEG2000, also known as PEG-2K.
- the PEG-modified lipid is DMG-PEG2000, DSPE-PEG2000, DLPE-PEG2000, DSG-PEG2000 or C8 PEG2000.
- Cholesterol based lipid The cholesterol component provides stability to the lipid bilayer structure within the nanoparticle.
- the LNPs comprise one or more cholesterol-based lipids.
- Suitable cholesterol-based lipids include, for example: N,N-dimethyl-N- ethylcarboxamido-cholesterol, 1,4-bis(3-N-oleylamino-propyl)piperazine , imidazole cholesterol ester, ⁇ -sitosterol, fucosterol, stigmasterol, and other modified forms of cholesterol.
- the cholesterol-based lipid used in the LNPs is cholesterol.
- Helper Lipids A helper lipid enhances the structural stability of the LNP and helps the LNP in endosome escape. It improves uptake and release of the RNA (e.g. mRNA) load. The cross-presenting capacity of DCs can be limited by non-specific degradation during endosome maturation.
- LNPs providing better endosome escape may be useful in some embodiments (e.g. if the antigen is in the form of, for example, a protein, a peptide or mRNA).
- the helper lipid is a non-cationic lipid.
- the helper lipid is a zwitterionic lipid, which has fusogenic properties for enhancing uptake and release of the load.
- helper lipids are 1,2- dioleoyl- SN-glycero-3-phosphoethanolamine (DOPE); 1,2-distearoyl-sn-glycero-3- phosphocholine (DSPC); 1,2-dioleoyl-sn-glycero-3-phospho-L-serine (DOPS); 1,2- dielaidoyl-sn-glycero-3-phosphocholine (DPOC), dipalmitoylphosphatidylcholine (DPPC), 1,2-dilauroyl-sn-glycero-3-phosphocholine (DLPC), 1,2- Distearoylphosphatidylethanolamine (DSPE), and 1,2-dilauroyl-sn-glycero-3- phosphoethanolamine (DLPE).
- DOPE 1,2- dioleoyl- SN-glycero-3-phosphoethanolamine
- DSPC 1,2-distearoyl-sn-glycero-3- phosphocholine
- DOPS 1,
- the lipid nanoparticles comprise cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE.
- the phDCs may be loaded with different antigens to give rise to multi-valent antigen-specific phDCs.
- the antigenic protein is encoded by an mRNA, encapsulated in a LNP
- the LNP may carry mRNAs that encode more than one antigenic protein, such as 2, 3, 4, 5, 6, 7, 8, 9, 10 or more antigenic proteins, from the same or different pathogens or from the same or different tumors.
- the LNP may carry multiple mRNA molecules, each encoding a different antigenic protein.
- the LNP may also carry a polycistronic mRNA that can be translated into more than one antigenic protein. If the LNP carries different mRNA molecules, there will typically be multiple copies of each mRNA molecule.
- Molar Ratios of the Lipid Components Specific molar ratios of the above components may be important for the LNPs’ effectiveness.
- the molar ratio of the cationic lipid in the LNPs relative to the total lipids i.e., A) is 35-50%, optionally 35-45%.
- the molar ratio of the PEGylated lipid component relative to the total lipids is 0.25- 2.75%, optionally about 1.5%. In some embodiments, the molar ratio of the cholesterol-based lipid relative to the total lipids (i.e., C) is 20-46.5%. In some embodiments, the molar ratio of the cholesterol-based lipid relative to the total lipids (i.e., C) is about 46.5%. In another embodiment, the molar ratio of the cholesterol- based lipid relative to the total lipids (i.e., C) is about 38.5%.
- the molar ratio of the helper lipid relative to the total lipids is about 10-35%, such as 10-25%, or 16-35% (e.g., 16-32% such as 16%). In some embodiments, the molar ratio of the helper lipid relative to the total lipids (i.e., D) is about 10%. In another embodiment, the molar ratio of the helper lipid relative to the total lipids (i.e., D) is about 16%. In one embodiment, the ratio of the components is 35: 2.5: 46: 16 (A: B: C: D). In another embodiment, the ratio of the components is 50:38.5:1.5:10 (A: B: C: D).
- the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE. In one embodiment, the lipid nanoparticle comprises cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 35:46.5:2.5:16. In some embodiments, the lipid nanoparticle comprises SM102, cholesterol, DMG- PEG-2K, DSPC. In one embodiment, the lipid nanoparticle comprises SM102, cholesterol, DMG-PEG-2K, DSPC at a ratio of 50:38.5:1.5:10. In some embodiments, the (PEGylated lipid+cholesterol) components have the same molar amount as the helper lipid.
- the LNPs contain a molar ratio of the cationic lipid to the helper lipid that is more than 1.
- the molar amount of the cationic lipid is first determined based on a desired N/P ratio, where N is the number of nitrogen atoms in the cationic lipid and P is the number of phosphate groups in the RNA (e.g. mRNA) to be transported by the LNP.
- the molar amount of each of the other lipids is calculated based on the molar amount of the cationic lipid and the molar ratio selected. These molar amounts are then converted to weights using the molecular weight of each lipid.
- the lipid nanoparticles comprising the antigen can be provided frozen.
- this may be useful if the phDCs and lipid nanoparticles comprising the antigen (e.g. in the form of RNA, particularly mRNA encoding at least one antigenic protein) are provided as a kit. Size and amount of lipid nanoparticles Suitable LNPs may be made in various sizes.
- the majority of purified LNPs i.e., greater than about 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% of the LNPs, have a size of about 50 to 200 nm (e.g., about 145 nm, about 140 nm, about 135 nm, about 130 nm, about 125 nm, about 120 nm, about 115 nm, about 110 nm, about 105 nm, about 100 nm, about 95 nm, about 90 nm, about 85 nm, or about 80 nm).
- nm e.g., about 145 nm, about 140 nm, about 135 nm, about 130 nm, about 125 nm, about 120 nm, about 115 nm, about 110 nm, about 105 nm, about 100 nm, about 95 nm, about 90 nm, about 85 nm, or about 80
- substantially all (e.g., greater than 80 or 90%) of the purified lipid nanoparticles have a size of about 70 to 200 nm (e.g., about 145 nm, about 140 nm, about 135 nm, about 130 nm, about 125 nm, about 120 nm, about 115 nm, about 110 nm, about 105 nm, about 100 nm, about 95 nm, about 90 nm, about 85 nm, or about 80 nm).
- greater than about 70%, 80%, 85%, 90%, 95%, 97%, 98%, 99% of the LNPs in the present composition have a size ranging from about 85 to 100 nm (e.g., about 45-85 nm, about 50-80 nm, about 55-75 nm, about 60-70 nm) or about 50-70 nm (e.g., 55-65 nm).
- the smaller sizes are particular suitable for inhalative delivery via nebulization.
- the phDCs are combined with at least about 1 ⁇ g, 2 ⁇ g, 3 ⁇ g, 4 ⁇ g, 5 ⁇ g, 10 ⁇ g or 20 ⁇ g of enapsulated RNA, particularly mRNA. In some embodiments, the phDCs are combined with about 10 ⁇ g of encapsulated RNA, particularly mRNA. In some embodiments, the phDCs are combined with about 20 ⁇ g of encapsulated RNA, particularly mRNA. Lipoplexes
- the at least one disease associated antigen e.g. at least one mRNA encoding the at least one antigenic protein
- the liposomes typically contain at least two components: a cationic lipid and a neutral lipid. Lipoplexes have extensively be described in the art, see, e.g. Nanomedicine: Nanotechnology, Biology and Medicine, 2009.
- the neutral lipid is a helper lipid as defined above.
- the cationic lipid is DOTMA and the neutral lipid is DOPE.
- the molar ratio of the at least one cationic lipid to the at least one neutral lipid is from about 10:0 to about 1:9, about 4:1 to about 1:2, or about 3:1 to about 1:1.
- the molar ratio may be about 3:1, about 2.75:1, about 2.5:1, about 2.25:1, about 2:1, about 1.75:1, about 1.5:1, about 1.25:1, or about 1:1.
- the molar ratio of the at least one cationic lipid to the at least one additional lipid is about 2:1.
- RNA lipoplex particles described herein have an average diameter that in one embodiment ranges from about 200 nm to about 1000 nm, from about 200 nm to about 800 nm, from about 250 to about 700 nm, from about 400 to about 600 nm, from about 300 nm to about 500 nm, or from about 350 nm to about 400 nm.
- the RNA lipoplex particles have an average diameter of about 200 nm, about 225 nm, about 250 nm, about 275 nm, about 300 nm, about 325 nm, about 350 nm, about 375 nm, about 400 nm, about 425 nm, about 450 nm, about 475 nm, about 500 nm, about 525 nm, about 550 nm, about 575 nm, about 600 nm, about 625 nm, about 650 nm or about 700 nm.
- the RNA lipoplex particles have an average diameter that ranges from about 250 nm to about 700 nm.
- the RNA lipoplex particles have an average diameter that ranges from about 300 nm to about 500 nm. In an exemplary embodiment, the RNA lipoplex particles have an average diameter of about 400 nm.
- the (mRNA) lipoplex particles comprise at least one cationic lipid and at least one neutral lipid. In one embodiment, the at least one cationic lipid comprises 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA) and/or 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP).
- DOTMA 1,2-di-O-octadecenyl-3-trimethylammonium propane
- DOTAP 1,2-dioleoyl-3-trimethylammonium-propane
- the at least one neutral lipid comprises 1,2-di-(9Z-octadecenoyl)-sn-glycero-3- phosphoethanolamine (DOPE), cholesterol (Chol) and/or 1,2-dioleoyl-sn-glycero-3- phosphocholine (DOPC).
- the at least one cationic lipid comprises 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA) and the at least one neutral lipid comprises 1,2-di-(9Z-octadecenoyl)-sn-glycero-3- phosphoethanolamine (DOPE).
- DOPE 1,2-di-(9Z-octadecenoyl)-sn-glycero-3- phosphoethanolamine
- the lipoplex particles comprise 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA) and 1,2-di-(9Z- octadecenoyl)-sn-glycero-3-phosphoethanolamine (DOPE).
- DOTMA 1,2-di-O-octadecenyl-3-trimethylammonium propane
- DOPE 1,2-di-(9Z- octadecenoyl)-sn-glycero-3-phosphoethanolamine
- Spleen targeting RNA lipoplex particles are described in WO 2013/143683, herein incorporated by reference. It has been found that lipoplex particles having a net negative charge may be used to preferentially target spleen tissue or spleen cells such as antigen- presenting cells, in particular dendritic cells.
- Oligomers, polymers or lipidoids comprising oligo(alkylene amine) moieties The at least one antigen (
- At least one mRNA encoding the at least one antigen can be comprised in oligomers, polymers or lipidoids comprising oligo(alkylene amine) moieties, for example, the characteristic oligo(alkylene amine) moieties as described in PCT/EP2014/063756.
- the at least one antigen e.g. at least one mRNA encoding the at least one antigen
- oligomers, polymers or lipidoids comprising oligo(alkylene amine) moieties may be selected from: a) an oligomer or polymer comprising a plurality of groups of formula (II) as a side chain and/or as a terminal group: wherein the variables a, b, p, m, n and R 2 to R 6 are defined as follows, independently for each group of formula (II) in a plurality of such groups: a is 1 and b is an integer of 2 to 4; or a is an integer of 2 to 4 and b is 1, p is 1or 2, m is 1 or 2; n is 0 or 1 and m+n is ⁇ 2; and R 2 to R 5 are, independently of each other, selected from hydrogen; a group –CH 2 - CH(OH
- oligomers, polymers or lipidoids comprising oligo(alkylene amine) moieties are selected from a) and b), wherein a) is an oligomer or polymer comprising a plurality of groups of formula (IIa) as a side chain and/or as a terminal group: wherein a, b, m, n, and R 2 to R 6 are defined as described above, and wherein one or more of the nitrogen atoms indicated in formula (IIa) may be protonated to provide a cationic oligomer or polymer structure; and b) is an oligomer or polymer comprising a plurality of groups of formula (IIIa) as repeating units: wherein a, b, m, n, and R 2 to R 5 are defined as described above, and wherein one or more of the nitrogen atoms indicated in formula (IIIa) may be protonated to provide a cationic oligomer or polymer structure.
- lipidoids comprising oligo(alkylene amine) moieties may be selected from a lipidoid having the structure of formula (IVa): wherein a, b, m, n, and R 1 to R 6 are defined as described above, and wherein one or more of the nitrogen atoms indicated in formula (IVa) may be protonated to provide a cationic lipidoid.
- polymers or lipidoids comprising oligo(alkylene amine) moieties, in formula (II), (IIa), (III), (IIIa), (IV) or (IVa) n may be 1; or m may be 1 and n may be 1.
- polymers or lipidoids comprising oligo(alkylene amine) moieties in formula (II), (IIa), (III), (IIIa), (IV) or (IVa) a may be 1 and b may be 2; or a may be 2 and b may be 1.
- oligomer, polymer or lipidoid comprising oligo(alkylene amine) moieties
- a cationic lipid which was prepared by mixing 100mg N,N’-Bis(2-aminoethyl)-1,3-propanediamine (0.623mmol) with 575.07mg 1,2-Epoxydodecane (3.12mmol, (N-1) eq. where N is 2x amount of primary amine plus 1x amount secondary amine per oligo(alkylene amine)) and mixed for 96h at 80°C under constant shaking.
- Such an oligomer, polymer or lipidoid is also referred to as lipidoid "C12-(2-3-2)".
- An oligomer, polymer or lipidoid comprising oligo(alkylene amine) moieties, in particular a polymer can be a copolymer, in particular a statistical copolymer.
- a copolymer may be a copolymer which contains a statistical/random arrangement of alkylene amine repeating units of alternating length (e.g. in contrast to a less preferred polymer which contains analogous arrangements of alkylene amine repeating units of non-alternating length).
- the copolymer may be a cationic (e.g. protonated) copolymer.
- Copolymers to be employed are known in the art and are, for example, described in EP 14199439.2, WO 01/00708, EP-A11198489 and CA-A12,377,207.
- the copolymer may be a statistical copolymer comprising a plurality of repeating units (a) independently selected from repeating units of the following formulae (a1) and (a2): and a plurality of repeating units (b) independently selected from repeating units of the following formulae (b1) to (b4): wherein the molar ratio of the sum of the repeating units (a) to the sum of the repeating units (b) lies within the range of 0.7/1.0 to 1.0/0.7, and wherein one or more of the nitrogen atoms of the repeating units (a) and/or (b) contained in the copolymer may be protonated to provide a cationic copolymer.
- the copolymer may be a statistical copolymer, wherein any repeating units (a) and any repeating units (b) are statistically distributed in the copolymer macromolecule. It is typically obtained from the copolymerization of a mixture of monomers yielding, during the polymerization reaction, the repeating units (a) with monomers yielding, during the polymerization reaction, the repeating units (b).
- the copolymer is a random copolymer wherein any repeating units (a) and any repeating units (b) are randomly distributed in the polymer macromolecule.
- Such a copolymer can be a linear, branched or dendritic copolymer.
- a repeating unit of the formula (a1), (b1) or (b3) with two valencies leads to a propagation of the copolymer structure in a linear manner.
- a linear copolymer may comprise repeating units of formula (a1) and one or more types of the repeating units of formulae (b1) and (b3), but no repeating units of formula (a2), (b2) or (b4).
- the presence of a repeating unit of formula (a2), (b2) or (b4) with three valencies provides a branching point in the copolymer structure.
- a branched copolymer comprises one or more types of the repeating units of formulae (a2), (b2) and (b4), and may further comprise one or more types of the repeating units of formulae (a1), (b1) and (b3).
- Such a copolymer may comprise a plurality of repeating units (a) independently selected from repeating units of formulae (a1) and (a2) defined above, and a plurality of repeating units (b) independently selected from repeating units of formulae (b1) to (b4) defined above.
- copolymers comprising a plurality of repeating units (a) independently selected from repeating units of formulae (a1) and (a2) defined above, and a plurality of repeating units (b) independently selected from repeating units of formulae (b1) and (b2) defined above.
- such a copolymer is a branched copolymer comprising one or more types of repeating units selected from repeating units (a2), (b2) and (b4), and which optionally further comprises one or more types of the repeating units of formulae (a1), (b1) and (b3), and in particular a copolymer which comprises repeating units of the formula (a2) and one or more type of the repeating units of formulae (b2) and (b4), and which optionally further comprises one or more types of the repeating units of formulae (a1), (b1) and (b3).
- a more preferred copolymer is thus a branched copolymer which comprises repeating units of the formula (a2) and repeating units of formula (b2), and which optionally further comprises one or more types of the repeating units of formulae (a1) and (b1).
- the total number of the repeating units (a) and repeating units (b) is typically 20 or more, preferably 50 or more and more preferably 100 or more.
- the total number of the repeating units (a) and repeating units (b) is 10,000 or less, preferably 5,000 or less, more preferably 1,000 or less.
- repeating units (a) and (b) account for 80 mol% or more, more preferably 90 mol% or more of all repeating units in the copolymer. Further preferred are copolymers wherein repeating units (a) selected from (a1) and (a2) and repeating units (b) selected from (b1) and (b2) account for 80 mol% or more, more preferably 90 mol% or more of all repeating units in the copolymer.
- all of the repeating units in the copolymer are repeating units (a) or (b), in particular that all of the repeating units in the copolymer are repeating units (a) selected from (a1) and (a2) or repeating units (b) selected from (b1) and (b2).
- the weight average molecular weight of the copolymer as measured e.g. via size exclusion chromatography relative to linear poly(ethylene oxide) standards, generally ranges from 1,000 to 500,000 Da, preferably from 2,500 to 250,000 Da and more preferably 5,000-50,000 less.
- the terminal groups of such a copolymer typically comprise one or more types of groups (c) independently selected from groups of the formulae (c1) to (c3) below, preferably from groups of the formulae (c1) and (c2) below:
- the terminal groups in the copolymer consist of one or more types of groups (c) independently selected from groups of the formulae (c1) to (c3) below, preferably from groups of the formulae (c1) and (c2).
- the number of terminal groups depends on the structure of the copolymer. While a linear copolymer has only two terminals, larger numbers of terminal groups are contained in a branched, in particular in a dendritic copolymer.
- the nitrogen atoms of the terminal groups (c) contained in the copolymer may be protonated to provide a cationic copolymer.
- the molar ratio of the sum of the repeating units (a) to the sum of the repeating units (b) lies within the range of 0.7/1.0 to 1.0/0.7, and preferably within the range of 0.8/1.0 to 1.0/0.8. This molar ratio can be determined, e.g., via NMR.
- the ratio is usually determined for a plurality of macromolecules of the copolymer, and typically indicates the overall ratio of the sum of repeating units (a) to the sum of repeating units (b) in the plurality of macromolecules.
- one or more of the nitrogen atoms of the copolymer may be protonated to result in a copolymer in a cationic form, typically an oligocationic or polycationic form.
- the primary, secondary, or tertiary amino groups in the repeating units (a) or (b) or in the terminal groups (c) can act as proton acceptors, especially in water and aqueous solutions, including physiological fluids.
- Such copolymers typically have an overall positive charge in an aqueous solution at a pH of below 7.5.
- An aqueous solution is a solution wherein the solvent comprises 50 % (vol./vol.) or more, preferably 80 or 90 % or more, and most preferably 100 % of water.
- the compositions are in contact with a physiological fluid having a pH of below 7.5, including e.g. blood and lung fluid, they typically contain repeating units (a) and (b) wherein the nitrogen atoms are protonated.
- the pK values of the copolymers used in the compositions can be determined by acid-base titration using an automated pK titrator.
- the net charge at a given pH value can then be calculated e.g. from the Henderson-Hasselbach equation. Any charge may be shared across several of the basic centres and cannot necessarily be attributed to a single point.
- the copolymers used in the compositions comprise repeating units with amino groups in protonated state and repeating units with amino groups in unprotonated state.
- the copolymers may also be provided as a dry salt form which contains the copolymer in a cationic form.
- counterions (anions) for the positive charges of protonated amino groups in compositions comprising the copolymer and nucleic acid, in particular mRNA are typically provided by anionic moieties contained in the nucleic acid. If the positively charged groups are present in excess compared to the anionic moieties in the nucleic acid, positive charges may be balanced by other anions, in particular those typically encountered in physiological fluids, such as Cl or HCO 3 -.
- a preferred copolymer is a random copolymer, wherein 80 mol% or more of all repeating units, more preferably all repeating units, are formed by a plurality of repeating units (a) independently selected from repeating units of the following formulae (a1) and (a2) and a plurality of repeating units (b) independently selected from repeating units of the following formulae (b1) and (b2): wherein the molar ratio of the sum of the repeating units (a) to the sum of the repeating units (b) lies within the range of 0.7/1.0 to 1.0/0.7, more preferably within the range of 0.8/1.0 to 1.0/0.8; 35 wherein the terminal groups of the copolymer are formed by groups (c) independently selected from groups of the formulae (c1) and (c2): and wherein one or more of the nitrogen atoms of the repeating units (a) and/or (b) and/or of the terminal groups (c) contained in the copolymer may be protonated to provide a
- the copolymer is a branched copolymer, comprising units (a2) and (b2), optionally together with units (a1) and/or (b1).
- a lipidoid is a preferred nanoparticle to be employed, in particular as compared to an oligomer and, more particular, to a polymer.
- the at least one disease associated antigen may be an antigenic protein which is encoded by an RNA (e.g. mRNA, modified or unmodified), wherein the RNA can be encapsulated in a lipid nanoparticle (LNP) for delivery to the phDCs of the invention.
- the encapsulated RNA (e.g. mRNA) is combined with the phDCs.
- the uptake of the RNA (e.g., mRNA, optionally modified mRNA) by phDCs may be monitored or assessed by means known in the art. These means include, but are not limited to, fluorescent LNPs, luminescent LNPs, radioactive LNPs or cascading reactions or detection of expressed antigenic proteins that are encoded by the RNA encapsulated in the LNPs.
- the transfection efficacy of phDC is assessed by co-incubating phDC generated from isolated PBMCs with mRNA encoding GPI-anchored nanoluciferase (NLuc) (LNP[NLuc]), and observing the luminescent signals.
- NLuc GPI-anchored nanoluciferase
- the spatio-temporal distribution of LNPs is monitored.
- the pharmaceutical composition or therapeutic composition also provides a pharmaceutical composition or therapeutic composition, comprising phDCs and at least one RNA (e.g. mRNA), which comprises a coding sequence encoding for at least one antigenic protein.
- RNA e.g. mRNA
- the at least one disease associated antigen may be in any form described under the first aspect and in the detailed description.
- the pharmaceutical composition of the invention in one embodiment, contains at least 1, at least 10, at least 100, at least 1000, at least 1 x 10 4 , at least 1 x 10 5 or at least 1 x 10 6 phDCs.
- the number of phDCs can e.g. be estimated from the volume of the whole blood, which became apheresed and flow chamber (e.g. plate or bag or hybrid) passed or flow chamber (e.g. plate or bag or hybrid) passed directly.
- the at least one mRNA can be provided as RNA, in particular mRNA, encapsulated in nanoparticles (e.g.
- a therapeutic composition according to the present invention contains at least 1, at least 10, at least 100, at least 1000, at least 1 x 10 4 , at least 1 x 10 5 or at least 1 x 10 6 antigen-specific phDCs.
- the dose of nanoparticles e.g.
- LNPs comprising RNA, in particular mRNA
- RNA in particular mRNA
- a typical dose for an mRNA therapeutic can be 0.1 to 100 ⁇ g, e.g., 1 ⁇ g, 5 ⁇ g, 10 ⁇ g, 30 ⁇ g or 50 ⁇ g.
- the phDCs of the invention are combined with a maximum of 4.5 ⁇ g of nanoparticles (e.g. LNPs) comprising RNA, in particular mRNA.
- the pharmaceutical composition or therapeutic composition of the invention optionally comprises a pharmaceutically acceptable carrier and/or diluent. Additionally, the pharmaceutical composition or therapeutic composition can comprise adjuvants and/or immuno-modulators to boost the activity of the pharmaceutical composition or therapeutic composition and the subject’s response. Such adjuvants and/or immuno-modulators are understood by those skilled in the art, and are readily described in available published literature.
- the pharmaceutical composition of the invention may contain one or more T cell activating agents. As contemplated herein, and depending on the type of composition being generated, the production of the antigen-specific phDCs can, if desired, be scaled up by culturing cells in bioreactors or fermentors or other such vessels or devices suitable for the growing of cells in bulk.
- the pharmaceutical composition or therapeutic composition is used for the treatment of a disease, such as cancer and/or infectious diseases.
- the pharmaceutical composition or therapeutic composition of the invention can induce antigen-specific T-cell and/or high titer antibody responses, thereby eliciting an immune response that is directed to or reactive against the disease (e.g. cancer or infectious disease) associated with the expression of the respective antigen.
- the induced or elicited immune response can be a cellular, humoral, or both cellular and humoral immune response.
- the induced or elicited cellular immune response includes induction or secretion of interferon-gamma (IFN- ⁇ ) and/or tumor necrosis factor alpha (TNF- ⁇ ).
- the pharmaceutical composition or therapeutic composition acts by one or more of the following: (i) inducing humoral immunity via B cell responses to generate antibodies; (ii) increasing cytotoxic T lymphocytes such as CD8+ (CTL) to attack and kill the disease causing particles which express the respective antigen; (iii) increasing T helper cell responses; (iv) increasing inflammatory responses via IFN- ⁇ and/or TNF- ⁇ ; (v) increasing natural killer cell responses; (vi) increasing central memory T cell and stem-like T cell subsets.
- the pharmaceutical composition or therapeutic composition acts by all of the aforementioned. In one embodiment, the pharmaceutical composition or therapeutic composition acts at least by (ii), (v) and (vi). In one embodiment, the pharmaceutical composition or therapeutic composition acts at least by (vi). In other embodiments, the induced or elicited immune response can reduce or inhibit one or more immune suppression factors that promote the onset of the disease.
- the induced or elicited immune response can reduce or inhibit one or more immune suppression factors that promote growth of the tumor or cancer expressing the antigen, for example, but not limited to, factors that downregulate MHC presentation, factors that upregulate antigen-specific regulatory T cells, PD-L1, FasL, cytokines such as IL-10 and TFG- ⁇ , tumor associated macrophages, tumor associated fibroblasts, soluble factors produced by immune suppressor cells, CTLA-4, PD-1, MDSCs, MCP- 1 and an immune checkpoint molecule.
- the pharmaceutical composition can be combined with other pharmaceutical products or compounds.
- the pharmaceutical composition of the present invention can be used to treat a disease such as a cancer or an infectious disease in individuals.
- the pharmaceutical composition of the invention may be particularly useful for subjects with an impaired immune system.
- subjects treated with the pharmaceutical composition or therapeutic composition of the invention experience less adverse effects after administration compared to subjects treated with therapeutics not comprising dendritic cells, in particular RNA therapeutics (control subjects).
- subjects show less fatigue, pain, headache and/or fever than control subjects. Comparison can be made to a control group of subjects treated with a therapeutic(s) not comprising dendritic cells, in particular an RNA therapeutic.
- the pharmaceutical composition can be administered by different routes such as subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intra-hepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, or sublingual.
- Preferred administration routes are intradermal or intramuscular.
- the composition can also be administered by inhalative administration.
- the composition of the invention is administered intradermally, subcutaneously or intramuscularly to the extremities, arms and legs, of the subjects being treated.
- the composition of the invention is administered intravenously.
- the pharmaceutical composition of the invention can be administered once or in multiple doses.
- the pharmaceutical composition of the invention is administered once. In one embodiment, the pharmaceutical composition of the invention is administered twice, three times, four times or five times. In one embodiment, the pharmaceutical composition of the invention is administered twice. In one embodiment, the pharmaceutical composition of the invention is administered five times.
- the present invention provides an article of manufacture, such as a kit, that provides phDCs in one container and the at least one disease associated antigen (e.g. the at least one mRNA encoding the at least one antigenic protein, optionally encapsulated in nanoparticles) in another container.
- the container may be pre-treated glass or plastic vials or ampules.
- the article of manufacture may include instructions for use. IV.
- the term “comprising” is used in the present description and claims, it does not exclude other elements.
- the term “consisting” is considered to be a preferred embodiment of the term “comprising”. If hereinafter a group is defined to comprise at least a certain number of embodiments, this is also to be understood to disclose a group, which preferably consists only of these embodiments.
- the term “obtained” is considered to be a preferred embodiment of the term “obtainable”. If hereinafter e.g. phDCs are defined to be obtainable by a specific method, this is also to be understood to disclose phDCs, which are obtained by this method.
- first”, “second”, “third” or “(a)”, “(b)”, “(c)”, “(d)” or “(i)”, “(ii)”, “(iii)”, “(iv)” etc. relate to steps of a method or use or assay there is no time or time interval coherence between the steps unless indicated otherwise, i.e. the steps may be carried out simultaneously or there may be time intervals of seconds, minutes, hours, days, weeks, months or even years between such steps, unless otherwise indicated in the application as set forth herein above or below.
- the term “about” means a quantity, level, value, number, frequency, percentage, dimension, size, amount, weight, or length that varies by as much as 30, 25, 20, 15, 30 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1% to a reference quantity, level, value, number, frequency, percentage, dimension, size, amount, weight or length.
- the term “about” in the context of the present invention denotes an interval of accuracy that the person skilled in the art will understand to still ensure the technical effect of the feature in question. In general, the term “about” is intended to modify a numerical value above and below the stated value by a variance of ⁇ 10%.
- nucleic acids and proteins are intended to include DNA and RNA such as genomic DNA, cDNA, mRNA, recombinantly produced and chemically synthesized molecules.
- a nucleic acid may be single-stranded or double- stranded.
- RNA includes in vitro transcribed RNA or synthetic RNA.
- the mRNA encoding the antigenic protein is expressed in phDCs, obtained from monocytes of a donor or the subject to be treated.
- RNA relates to a nucleic acid molecule, which includes ribonucleotide residues. In preferred embodiments, the RNA contains all or a majority of ribonucleotide residues.
- ribonucleotide refers to a nucleotide with a hydroxyl group at the 2’-position of a ⁇ -D-ribofuranosyl group.
- RNA encompasses without limitation, double stranded RNA, single stranded RNA, isolated RNA such as partially purified RNA, essentially pure RNA, mRNA, circular RNA, synthetic RNA, recombinantly produced RNA, synthetic RNA, self-amplifying RNA as well as modified RNA (e.g. modified mRNA) that differs from naturally occurring RNA by the addition, deletion, substitution and/or alteration of one or more nucleotides.
- modified RNA e.g. modified mRNA
- a fragment of an RNA sequence comprises e.g. at least 50 %, at least 60 %, at least 70 %, at least 80%, at least 90% of the residues from an RNA sequence.
- a fragment of an RNA sequence preferably comprises at least 18, in particular at least 24, at least 36, at least 45, at least 60, at least 90, at least 150, or at least 300 consecutive residues from an RNA sequence. Alterations may refer to addition of nonnucleotide material to internal RNA nucleotides or to the end(s) of RNA.
- RNA may be non-standard nucleotides, such as chemically synthesized nucleotides, naturally occurring modified nucleotides or deoxynucleotides.
- these altered RNAs are considered analogs of naturally-occurring RNA.
- the RNA is messenger RNA (mRNA) that relates to a RNA transcript which encodes a peptide or protein.
- mRNA messenger RNA
- mRNA generally contains a 5’ untranslated region (5’-UTR), a coding region and a 3’ untranslated region (3’-UTR).
- the RNA is produced by in vitro transcription or chemical synthesis.
- the mRNA is produced by in vitro transcription using a DNA template where DNA refers to a nucleic acid that contains deoxyribonucleotides.
- RNA is in vitro transcribed RNA (IVT-RNA) and may be obtained by in vitro transcription of an appropriate DNA template.
- the promoter for controlling transcription can be any promoter for any RNA polymerase.
- a DNA template for in vitro transcription may be obtained by cloning of a nucleic acid, in particular cDNA, and introducing it into an appropriate vector for in vitro transcription.
- the cDNA may be obtained by reverse transcription of RNA.
- the RNA, in particular mRNA, described herein may have modified nucleosides.
- the RNA, in particular mRNA comprises a modified nucleoside in place of at least one (e.g., every) uridine.
- "Pseudouridine” is one example of a modified nucleoside that is an isomer of uridine, where the uracil is attached to the pentose ring via a carbon-carbon bond instead of a nitrogencarbon glycosidic bond.
- Another exemplary modified nucleoside is N1- methyl-pseudouridine.
- Another exemplary modified nucleoside is 5-methyl-uridine.
- one or more uridine in the RNA e.g.
- RNA described herein is replaced by a modified nucleoside.
- the RNA comprises other modified nucleosides or comprises further modified nucleosides, e.g., modified cytidine.
- modified cytidine is substituted partially or completely, preferably completely, for 5-methyl-cytidine.
- the RNA comprises 5-methylcytidine and one or more selected from pseudouridine, N1- methyl-pseudouridine, and 5-methyl-uridine.
- the RNA comprises 5-methylcytidine and N1-methyl-pseudouridine.
- RNA comprises 5-methylcytidine in place of each cytidine and N1-methyl-pseudouridine in place of each uridine.
- RNA e.g. mRNA
- RNA may comprise more than one type of modified nucleoside.
- the modified nucleoside is a modified uridine.
- nucleobases and nucleosides having a modified uracil include pseudouridine ( ⁇ ), pyridin-4-one ribonucleoside, 5-aza-uridine, 6-aza-uridine, 2-thio-5-aza-uridine, 2- thio-uridine (s2U), 4-thio-uridine (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5-hydroxy-uridine (ho5U), 5-aminoallyl-uridine, 5-halo-uridine (e.g., 5-iodo-uridine or 5-bromo-uridine), 3-methyl-uridine (m3U), 5-methoxy-uridine (mo5U), uridine 5- oxyacetic acid (cmo5U), uridine 5-oxyacetic acid methyl ester (mcmo5U), 5- carboxymethyl-uridine (cm5U), 1-carboxymethyl-pseudouridine, 5- carboxyhydroxymethyl-uridine
- the modified nucleoside is a modified cytidine.
- exemplary nucleobases and nucleosides having a modified cytosine include 5-aza-cytidine, 6-aza- cytidine, pseudoisocytidine, 3-methyl-cytidine (m3C), N4-acetyl-cytidine (ac4C), 5- formyl-cytidine (f5C), N4-methyl-cytidine (m4C), 5-methyl-cytidine (m5C), 5-halo- cytidine (e.g., 5-iodo-cytidine), 5-hydroxymethyl-cytidine (hm5C), 1-methyl- pseudoisocytidine, pyrrolo-cytidine, pyrrolo-pseudoisocytidine, 2-thio-cytidine (s2C), 2-thio-5-methyl-cytidine, 4-thio-pseudoisocytidine
- the modified nucleoside is a modified adenosine.
- exemplary nucleobases and nucleosides having a modified adenine include 2-amino-purine, 2,6- diaminopurine, 2-amino-6-halo-purine (e.g., 2-amino-6-chloro-purine), 6-halo-purine (e.g., 6-chloro-purine), 2-amino-6-methyl-purine, 8-azido-adenosine, 7-deaza- adenine, 7-deaza-8-aza-adenine, 7-deaza-2-amino-purine, 7-deaza-8-aza-2-amino- purine, 7-deaza-2,6-diaminopurine, 7-deaza-8-aza-2,6-diaminopurine, 1-methyl- adenosine (m1A), 2-methyl-adenine (m2A), N6-methyl-adeno
- the modified nucleoside is a modified guanidine.
- exemplary nucleobases and nucleosides having a modified guanine include inosine (I), 1-methyl- inosine (m1l), wyosine (imG), methylmyosine (mimG), 4-demethyl-wyosine (imG- 14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (02yW), hydroxywybutosine (OhyW), under-modified hydroxywybutosine (OhyW*), 7-deaza- guanosine, queuosine (Q), epoxyqueuosine (oQ), galactosyl-queuosine(galQ), mannosyl-queuosine (manQ), 7-cyano-7-deaza-guanosine (preQo), 7-aninomethyl-7- deaza-guanosine
- the mRNA comprises a 5’-cap. In one embodiment, the mRNA does not have uncapped 5’-triphosphates. In one embodiment, the mRNA may be modified by a 5’- cap analog.
- the term "5’-cap” refers to a structure found on the 5’- end of an mRNA molecule and generally consists of a guanosine nucleotide connected to the mRNA via a 5’- to 5’-triphosphate linkage. In one embodiment, this guanosine is methylated at the 7-position.
- Providing an mRNA with a 5’-cap or 5’-cap analog may be achieved by in vitro transcription, in which the 5’-cap is co-transcriptionally expressed into the mRNA strand, or may be attached to mRNA post-transcriptionally using capping enzymes.
- the term "untranslated region" or "UTR” relates to a region in a DNA molecule which is transcribed but is not translated into an amino acid sequence, or to the corresponding region in an RNA molecule, such as an mRNA molecule.
- An untranslated region (UTR) can be present 5’ (upstream) of an open reading frame (5’-UTR) and/or 3’ (downstream) of an open reading frame (3’-UTR).
- a 5’-UTR if present, is located at the 5’ end, upstream of the start codon of a protein-encoding region.
- a 5’-UTR is downstream of the 5’-cap (if present), e.g. directly adjacent to the 5’-cap.
- a 3’-UTR if present, is located at the 3’ end, downstream of the termination codon of a protein- encoding region, but the term "3’-UTR" does preferably not include the poly(A) sequence.
- the 3’-UTR is upstream of the poly(A) sequence (if present), e.g. directly adjacent to the poly(A) sequence.
- poly(A) sequence or "poly-A tail” refers to an uninterrupted or interrupted sequence of adenylate residues which is typically located at the 3’-end of an RNA molecule (e.g. an mRNA molecule).
- Poly(A) sequences are known to those of skill in the art and may follow the 3’-UTR in the RNAs (e.g. mRNAs) described herein.
- An uninterrupted poly(A) sequence is characterized by consecutive adenylate residues. In nature, an uninterrupted poly(A) sequence is typical.
- RNAs (e.g. mRNAs) according to the invention can have a poly(A) sequence attached to the free 3’-end of the RNA (e.g.
- poly(A) sequences of about 120 A nucleotides have a beneficial influence on the RNA-levels in transfected eukaryotic cells, as well as on the protein-levels, wherein the protein is translated from an open reading frame that is present upstream (5’) of the poly(A) sequence (Holtkamp et al., 2006, Blood, vol.108, pp.4009-4017).
- the poly(A) sequence may be of any length.
- a poly(A) sequence comprises, essentially consists of, or consists of at least 20, at least 30, at least 40, at least 80, or at least 100 and up to 800, up to 400, up to 300, up to 200, or up to 150 A nucleotides. In one embodiment, the poly(A) sequence comprises, essentially consists of, or consists of 300 nucleotides.
- nucleotides in the poly(A) sequence typically at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% by number of nucleotides in the poly(A) sequence are A nucleotides, but permits that remaining nucleotides are nucleotides other than A nucleotides, such as U nucleotides (uridylate), G nucleotides (guanylate), or C nucleotides (cytidylate).
- consists of means that all nucleotides in the poly(A) sequence, i.e., 100% by number of nucleotides in the poly(A) sequence, are A nucleotides.
- a nucleotide or “A” refers to adenylate.
- a poly(A) sequence is attached during RNA transcription, e.g., during preparation of in vitro transcribed RNA (e.g. mRNA), based on a DNA template comprising repeated dT nucleotides (deoxythymidylate) in the strand complementary to the coding strand.
- the DNA sequence encoding a poly(A) sequence (coding strand) is referred to as poly(A) cassette.
- coding regions are preferably codon- optimized for optimal expression in a subject to be treated using the RNA (e.g. mRNA) molecules described herein. Codon-optimization is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells. Thus, the sequence of RNA (e.g. mRNA) molecules described herein.
- the guanosine/cytosine (G/C) content of the coding region of the RNA (e.g. mRNA) described herein is increased compared to the G/C content of the corresponding coding sequence of the wild type RNA, wherein the amino acid sequence encoded by the RNA (e.g. mRNA) is preferably not modified compared to the amino acid sequence encoded by the wild type RNA.
- This modification of the RNA (e.g. mRNA) sequence is based on the fact that the sequence of any RNA region to be translated is important for efficient translation of that RNA (e.g.
- RNA RNA sequence having an increased G/C content
- A adenosine
- U uracil
- the most favourable codons for the stability can be determined (so- called alternative codon usage).
- alternative codon usage Depending on the amino acid to be encoded by the RNA, there are various possibilities for modification of the RNA (e.g. mRNA) sequence, compared to its wild type sequence.
- codons which contain A and/or U nucleotides can be modified by substituting these codons by other codons, which code for the same amino acids but contain no A and/or U or contain a lower content of A and/or U nucleotides.
- transcription relates to a process, wherein the genetic code in a DNA sequence is transcribed into RNA (e.g. mRNA). Subsequently, the RNA (e.g. mRNA) may be translated into peptide or protein.
- the term “transcription” comprises “in vitro transcription”, wherein the term “in vitro transcription” relates to a process wherein RNA, in particular mRNA, is in vitro synthesized in a cell-free system, preferably using appropriate cell extracts.
- cloning vectors are applied for the generation of transcripts.
- These cloning vectors are generally designated as transcription vectors and are according to the present invention encompassed by the term “vector”.
- RNA e.g. mRNA
- expression or “translation” relates to the process in the ribosomes of a cell by which a strand of mRNA directs the assembly of a sequence of amino acids to make a peptide or protein.
- RNA e.g. mRNA
- RNA e.g. mRNA
- lipid particles e.g., lipid particles
- RNA e.g. mRNA
- the RNA is translated by the phDCs to produce the peptide or protein it encodes.
- RNA (e.g. mRNA) particles such as RNA (e.g. mRNA) lipid particles described herein may be used for delivering RNA to phDCs.
- Encoding refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, RNA or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
- a RNA (e.g. mRNA) sequence may encode a protein (e.g. antigen) if translation of the RNA (e.g. mRNA) occurs in a cell.
- peptide comprises oligo- and polypeptides and refers to substances which comprise about two or more, about 3 or more, about 4 or more, about 6 or more, about 8 or more, about 10 or more, about 13 or more, about 16 or more, about 20 or more, and up to about 50, about 100 or about 150, consecutive amino acids linked to one another via peptide bonds.
- protein or “polypeptide” refers to large peptides, in particular peptides having at least about 150 amino acids, but the terms "peptide", “protein” and “polypeptide” are used herein as synonyms if not mentioned otherwise.
- “Fragment” can relate to a part of an antigen such as a disease associated antigen, an amino acid sequence, an antigenic protein, an RNA (including the RNA species listed above) and in particular mRNA.
- fragment relates to an amino acid sequence (e.g. antigenic protein)
- the term refers to a part of a amino acid sequence, i.e. a sequence which represents the amino acid sequence (e.g. of the antigenic protein) shortened at the N-terminus and/or C-terminus.
- a fragment shortened at the C-terminus (N-terminal fragment) is obtainable e.g. by translation of a truncated open reading frame that lacks the 3’-end of the open reading frame.
- a fragment shortened at the N-terminus is obtainable e.g. by translation of a truncated open reading frame that lacks the 5’-end of the open reading frame, as long as the truncated open reading frame comprises a start codon that serves to initiate translation.
- a fragment of an amino acid sequence comprises e.g. at least 50 %, at least 60 %, at least 70 %, at least 80%, at least 90% of the amino acid residues from an amino acid sequence.
- a fragment of an amino acid sequence preferably comprises at least 6, in particular at least 8, at least 12, at least 15, at least 20, at least 30, at least 50, or at least 100 consecutive amino acids from an amino acid sequence.
- variant herein is meant an amino acid sequence that differs from a parent amino acid sequence by virtue of at least one amino acid modification.
- the parent amino acid sequence may be a naturally occurring or wild type (WT) amino acid sequence, or may be a modified version of a wild type amino acid sequence.
- the variant amino acid sequence has at least one amino acid modification compared to the parent amino acid sequence, e.g., from 1 to about 20 amino acid modifications, from 1 to about 10 or from 1 to about 5 amino acid modifications compared to the parent.
- wild type or WT or “native” herein is meant an amino acid sequence that is found in nature, including allelic variations.
- a wild type amino acid sequence, peptide or protein has an amino acid sequence that has not been intentionally modified.
- “Variants” of an amino acid sequence comprise amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants.
- variants includes all mutants, splice variants, posttranslationally modified variants, conformations, isoforms, allelic variants, species variants, and species homologs, in particular those which are naturally occurring.
- variant includes, in particular, fragments of an amino acid sequence.
- the degree of similarity (or identity) between a given amino acid sequence and an amino acid sequence which is a variant of said given amino acid sequence will be at least about 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
- the alignment for determining sequence similarity, preferably sequence identity can be done with art known tools, preferably using the best sequence alignment, for example, using Align, using standard settings, preferably EM-BOSS::needle, Matrix: Blosum62, and the like.
- Sequence similarity indicates the percentage of amino acids that either are identical or that represent conservative amino acid substitutions.
- Sequence identity indicates the percentage of amino acids that are identical between the sequences.
- Sequence identity between two nucleic acid sequences indicates the percentage of nucleotides that are identical between the sequences. Percentage identity is obtained by determining the number of identical positions at which the sequences to be compared correspond, dividing this number by the number of positions compared (e.g., the number of positions in the reference sequence) and multiplying this result by 100.
- a fragment or “variant” is preferably a "functional fragment” or "functional variant”.
- the term "functional fragment” or “functional variant” relates to any fragment or variant exhibiting one or more functional properties identical or similar to those of the unfragmented agent (e.g. unfragmented amino acid sequence, unfragmented RNA sequence, unfragmented mRNA sequence, etc.) from which it is derived, i.e., it is functionally equivalent.
- the unfragmented agent e.g. unfragmented amino acid sequence, unfragmented RNA sequence, unfragmented mRNA sequence, etc.
- one particular function is one or more immunogenic (“antigenic” synonymous herein) activities displayed by the amino acid sequence from which the fragment or variant is derived.
- the modifications in the amino acid sequence of the parent molecule or sequence do not significantly affect or alter the characteristics of the molecule or sequence.
- the function of the functional fragment or functional variant may be reduced but still significantly present, e.g., immunogenicity of the functional fragment or variant may be at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the parent agent, molecule or sequence. However, in other embodiments, immunogenicity of the functional fragment or functional variant may be enhanced compared to the parent molecule or sequence.
- the therapeutic compositions of the invention induce an immune response against the at least one disease associated antigen (e.g. antigenic protein) in a cell, tissue or subject (e.g., a human).
- the therapeutic compositions of the invention are administered in order to treat a disease or disorder or to alleviate the severity of symptoms of the disease or disorder.
- “treat”, “treating” or “treatment” of a disease or disorder occurs after the initiation of a pathologic event (e.g. outbreak or onset of a disease or disorder).
- treatment or “therapeutic treatment” includes treatment of a subject (e.g. a mammal, such as a human) or a cell to alter the current course of the subject or cell.
- treatment refers to a subject or cell, which has been previously been diagnosed or determined to have the disease or disorder (as opposed to prevention). Treatment includes, but is not limited to, administration of the therapeutic compositions of the invention, and is performed subsequently to the initiation of the pathologic event or contact with an infectious agent.
- the next administration is typically performed at least 2, at least 3, at least 4, at least 5, at least 7, at least 14, at least 21, at least 28, at least 35, at least 42, at least 49 or at least 56 or more days after the preceding administration.
- prophylactic administration or vaccination is delineated from therapeutic treatment with regard to the time of administration of the agent or composition to the subject. Vaccination occurs prior to the initiation of a pathologic event, whereas therapeutic treatment or treatment occurs afterwards, e.g. after the subject has been previously been diagnosed as having a disease.
- inducing/stimulating an immune response may indicate that no immune response against a particular antigen was present before induction/stimulation or it may indicate that there was a basal level of immune response against a particular antigen before induction, which was enhanced after induction. Therefore, “induces/stimulates (or inducing/stimulating) an immune response” includes “enhances (or enhancing) an immune response”.
- immunoity As used herein, the term “antigen” or “antigenic protein” refers to a compound, composition, or substance that can stimulate the production of antibodies or a T cell response in an animal, including compositions that are injected or absorbed into an animal.
- an antigen or antigenic protein reacts with the products of specific humoral or cellular immunity, including those induced by heterologous immunogens.
- the term antigen is used interchangeably with the term “immunogen”.
- the term "antigen” or “antigenic protein” include all related antigenic epitopes.
- the terms “antigen”, “antigenic molecule/protein” or “immunogen” include fragments thereof that are still capable of acting as an antigen.
- An “antigenic protein” comprises a number of amino acids ranging from small peptides to large proteins, e.g.4 to 2000 amino acids or more, 4 to 1800 amino acids, 4 to 1600 amino acids or 4 to 1400 amino acids.
- the antigenic protein for which the RNA (e.g.
- the antigenic protein, for which the RNA (e.g. mRNA) encodes comprises at least 100, at least 200, at least 300, at least 400, at least 500, at least 600, at least 700, at least 800, at least 900, at least 1000, at least 1200, at least 2000 or at least 3000 amino acids.
- the antigenic protein, for which the RNA (e.g. mRNA) encodes comprises at least 50 amino acids.
- the antigenic protein, for which the RNA (e.g. mRNA) encodes comprises at least 500 amino acids.
- the antigenic protein, for which the RNA (e.g. mRNA) encodes comprises at least 1000 amino acids.
- the antigenic protein comprises 100 to 1500 amino acids.
- the antigenic protein comprises 200 to 1300 amino acids. In one embodiment, the antigenic protein comprises 400 to 1300 amino acids.
- Epipe refers to a site on an antigen to which B and/or T cells respond.
- immunogenicity refers to the ability of a substance, a cell or a part thereof, such as an antigen or antigenic protein, to provoke an immune response in the body of a human or animal.
- Cell-mediated immunity “cellular immunity”, “cellular immune response”, or similar terms are meant to include a cellular response directed to cells characterized by expression of an antigen or antigenic protein, in particular characterized by presentation of an antigen with class I or class II MHC.
- the cellular response relates to immune effector cells, in particular to cells called T cells or T lymphocytes which act as either "helpers” or “killers".
- the helper T cells also termed CD4+ T cells
- the killer cells also termed cytotoxic T cells, cytolytic T cells, CD8+ T cells or CTLs
- kill diseased cells such as virus-infected cells, preventing the production of more diseased cells.
- “Humoral immunity” or “humoral immune response” is the aspect of immunity that is mediated by macromolecules found in extracellular fluids such as secreted antibodies, complement proteins, and certain antimicrobial peptides. It contrasts with cell- mediated immunity. Its aspects involving antibodies are often called antibody- mediated immunity.
- Humoral immunity refers to antibody production and the accessory processes that accompany it, including: Th2 activation and cytokine production, germinal center formation and isotype switching, affinity maturation and memory cell generation.
- DCs are antigen-presenting immune cells that process antigenic material and present it to other cells of the immune system, most notably to T cells. DCs function to capture and process antigens. When DCs endocytose antigens, they process the antigens into smaller fragments, generally peptides, that are displayed on the DC surface, where they are presented to, for example, antigen-specific T cells through MHC molecules. After uptake of antigens, DCs migrate to the lymph nodes.
- DCs can be prompted by various signals, including signaling through Toll-like receptors (TLR), to express co- stimulatory signals that induce cognate effector T cells (Teff) to become activated and to proliferate, thereby initiating a T-cell mediated immune response to the antigen.
- TLR Toll-like receptors
- DCs can present an antigen to antigen-specific T cells without providing co-stimulatory signals (or while providing co-inhibitory signals), such that Teff are not properly activated.
- Such presentation can cause, for example, death or anergy of T cells recognizing the antigen, or can induce the generation and/or expansion of regulatory T cells (Treg).
- dendritic cells includes differentiated dendritic cells, immature, and mature dendritic cells. These cells can be characterized by expression of certain cell surface markers (e.g., CD11c, MHC class II, and at least low levels of CD80 and CD86), CD11b, CD304 (BDCA4)). In some embodiments, DCs express CD8, CD103, CD1d, etc. Other DCs can be identified by the absence of lineage markers such as CD3, CD14, CD19, CD56, etc. In addition, dendritic cells can be characterized functionally by their capacity to stimulate alloresponses and mixed lymphocyte reactions (MLR).
- MLR mixed lymphocyte reactions
- phDCs that are combined with a SARS- CoV-2 derived spike protein antigen (antigenic protein) or an mRNA encoding therefore, will process and present spike protein associated antigens, i.e. the phDCs will be antigen-specific to the spike protein.
- AB serum or “human AB serum” relates to a well known in the art cell culture reagent for some human cell types providing growth factors, vitamins, nutrients as well as trace elements and transport factors. Human AB serum is collected from healthy volunteer male donors of the AB serotype.
- FBS relates to a widely-used growth supplement for cell culture media. It typically has a high content of embryonic growth-promoting factors.
- disease refers to any disease or disorder which implicates an antigen or antigenic protein, e.g. a disease which is characterized by the presence of an antigen or antigenic protein.
- the disease can, e.g., be an infectious disease or a tumor disease (cancer).
- the antigen may be a disease-associated antigen, such as a viral antigen or a tumor antigen.
- a disease involving an antigen is a disease involving cells expressing an antigen, preferably on the cell surface.
- diseases are caused by disease causing particles.
- the concentration of disease causing particles in a subject can be determined by any suitable means known to the skilled person.
- the viral load may be determined by quantitative PCR and expressed as copy number per ng tissue. If suitable, expression markers may also be used for the quantification of disease causing particles.
- hyper-proliferative diseases encompasses tumors and cancer.
- tumor refers to a disease in which some of the body’s cells grow uncontrollably and spread to other parts of the body. Tumors can be cancerous or not cancerous (benign). In the context of the present invention, the terms tumor and cancer are used interchangeably. Cancerous tumors spread into, or invade, nearby tissues and can travel to distant places in the body to form new tumors (metastasis). Cancerous tumors may also be called malignant tumors.
- cancers of the blood such as leukemias, do not. Benign tumors do not spread into, or invade, nearby tissues.
- Blood cancer includes leukemia, lymphoma, myelodysplastic syndromes (MDS), myeloproliferative disorder (MPD), multiple myeloma and all subtypes thereof.
- infectious disease refers to any disease, which can be transmitted from individual to individual or from organism to organism, and is caused by a microbial agent (e.g. common cold).
- infectious diseases include, for example, a viral disease, a bacterial disease, a fungal disease or a parasitic disease, which diseases are caused by a virus, a bacterium, a fungus, and a parasite, respectively.
- infectious disease can be, for example, hepatitis, sexually transmitted diseases (e.g. chlamydia or gonorrhea), tuberculosis, HIV/acquired immune deficiency syndrome (AIDS), diphtheria, hepatitis B, hepatitis C, cholera, severe acute respiratory syndrome (SARS), Covid-19, the bird flu, and influenza.
- sexually transmitted diseases e.g. chlamydia or gonorrhea
- tuberculosis e.g. chlamydia or gonorrhea
- tuberculosis e.g. chlamydia or gonorrhea
- COVID-19 is a contagious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2).
- SARS-CoV-2 severe acute respiratory syndrome coronavirus-2
- MN908947.3 belongs to betacoronavirus lineage B. It has at least 70% sequence similarity to SARS-CoV.
- COVID-19 refers to a disease as defined in the current international classification of diseases (ICD-11, World Health Organization, Version: 09/2020).
- COVID-19 is used to denote the disease, diagnosed clinically, epidemiologically or otherwise, irrespective of whether laboratory testing is conclusive, inconclusive or not available.
- coronaviruses have four structural proteins, namely, envelope (E), membrane (M), nucleocapsid (N), and spike (S).
- E and M proteins have important functions in the viral assembly, and the N protein is necessary for viral RNA synthesis.
- the critical glycoprotein S is responsible for virus binding and entry into target cells.
- the S protein is synthesized as a singlechain inactive precursor that is cleaved by furin-like host proteases in the producing cell into two noncovalently associated subunits, S1 and S2.
- the S1 subunit contains the receptor-binding domain (RBD), which recognizes the host-cell receptor.
- the S2 subunit contains the fusion peptide, two heptad repeats, and a trans- membrane domain, all of which are required to mediate fusion of the viral and host-cell membranes by undergoing a large conformational rearrangement.
- the S1 and S2 subunits trimerize to form a large prefusion spike.
- the S precursor protein of SARS-CoV-2 can be proteolytically cleaved into S1 (685 aa) and S2 (588 aa) subunits.
- the S1 subunit consists of the receptor-binding domain (RBD), which mediates virus entry into sensitive cells through the host angiotensin-converting enzyme 2 (ACE2) receptor.
- Therapeutically treating COVID-19 may include treating, ameliorating or healing at least one of lung fibrosis, interstitial pneumonia, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), aveolar damage, kidney injury, vasculopathy, cardiac injury, acute myocardial injury, chronic damage to the cardiovascular system, thrombosis and venous thromboembolism, in a subject with COVID-19.
- lung fibrosis interstitial pneumonia, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), kidney injury, such as proteinuria and acute kidney injury, and vasculopathy are triggered by COVID-19.
- SARS-CoV-2 is used to denote all variants of a virus, according to ICTV belonging to realm Riboviria, kingdom Orthornavirae, phylum Pisuviricota, class Pisoniviricetes, order Nidovirales, family Coronaviridae, genus Betacoronavirus, subgenus Sarbecovirus, species Severe acute respiratory syndrome-related coronavirus, strain Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
- HIV refers to human immunodeficiency viruses. HIV are a member of the genus Lentivirus, part of the family Retroviridae. Two types of HIV, HIV-1 and HIV- 2, have been characterized. HIV-1 is the virus that was initially discovered and termed both lymphadenopathy associated virus (LAV) and human T-lymphotropic virus 3 (HTLV-III). HIV-1 is more virulent and more infective than HIV-2, and is the cause of the majority of HIV infections globally. HIV infect humans and cause acquired immunodeficiency syndrome (AIDS) over time. AIDS manifests in progressive failure of the immune system, allowing life-threatening opportunistic infections and cancers to thrive.
- LAV lymphadenopathy associated virus
- HTLV-III human T-lymphotropic virus 3
- the term "animal” or “mammal” encompasses all mammals, including humans.
- the mammal of the present invention is a human subject.
- the subject is a human.
- the terms "individual” and “subject” are used herein interchangeably. They refer to a human or another mammal (e.g. mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate) that can be afflicted with or is susceptible to a disease or disorder but may or may not have the disease or disorder.
- the individual is a human being.
- the terms “individual” and “subject” do not denote a particular age, and thus encompass adults, elderlies, children, and newborns.
- the term “subject” includes humans of age of at least 50, at least 55, at least 60, at least 65, at least 70, or older.
- the term “subject” includes humans of age of at least 65, such as 65 to 80, 65 to 75, or 65 to 70.
- the "individual” or “subject” is a "patient”.
- patient means an individual or subject for treatment.
- the invention further relates to the following embodiments: 1.
- a therapeutic composition comprising a pharmaceutically effective amount of physiological DCs (phDCs) and at least one mRNA, which comprises a coding sequence encoding for at least one antigenic protein.
- the therapeutic composition according to 1 or 2 wherein said at least one mRNA is comprised in lipid nanoparticles.
- the therapeutic composition according to any one of 1 to 3, wherein said at least one mRNA comprises at least one modified nucleoside.
- said at least one modified nucleoside is a modified uridine, optionally wherein all uridine residues are replaced with modified uridine. 6.
- said at least one mRNA comprises a sequence which is optimized; optionally, wherein the sequence of said 5’ UTR, 3’UTR and/or coding sequence for the antigenic protein is optimized.
- said lipid nanoparticles comprise a cationic lipid, a polyethylene glycol (PEG) modified lipid, a cholesterol-based lipid and/or a non-cationic lipid. 12.
- the therapeutic composition according to 14, wherein said antigenic protein associated with a disease is an infectious disease associated antigenic protein.
- said infectious disease associated antigenic protein is a viral antigenic protein, a bacterial antigenic protein, a fungal antigenic protein, a prion antigenic protein or a parasite antigenic protein.
- said viral antigenic protein is a coronavirus antigenic protein or human immunodeficiency virus (HIV) antigenic protein.
- said coronavirus antigenic protein is derived from the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein. 19.
- said bacterial antigenic protein is an antigenic protein of Borrelia spp. or Mycobacteria spp.. 26.
- the therapeutic composition according to 16, wherein said fungal antigenic protein is an antigenic protein of Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis or Candida albicans.
- said parasite antigenic protein is an antigenic protein of Plasmodium malariae.
- said antigenic protein associated with a disease is a tumor associated antigenic protein.
- 29. The therapeutic composition according to 28, wherein said tumor associated antigenic protein is a blood cancer antigenic protein.
- said solid tumor antigenic protein is a melanoma antigenic protein, endometrial cancer antigenic protein, kidney cancer antigenic protein, brain cancer antigenic protein, cervical cancer antigenic protein, liver cancer antigenic protein, head and neck cancer antigenic protein, gastrointestinal cancer antigenic protein, lymph node cancer antigenic protein, pancreas cancer antigenic protein, ear, nose and throat (ENT) cancer antigenic protein, breast cancer antigenic protein, prostate cancer antigenic protein, ovarian cancer antigenic protein or lung cancer antigenic protein.
- the phDCs are obtainable by subjecting monocytes to a physical force.
- the therapeutic composition according to 33 wherein said physical force is applied by passing said monocytes through a flow chamber.
- said flow chamber is a plate or flexible bag, optionally a flexible plastic bag.
- said monocytes are autologous.
- 37. The therapeutic composition of any one of 1 to 36 for use in a method of treating a disease in a subject, wherein the subject has been previously been diagnosed as having the disease, said method comprising administering said therapeutic composition to the subject.
- 38. The therapeutic composition of any one of 15 to 27 for use in a method of treating an infectious disease in a subject, wherein the subject has been previously been diagnosed as having the infectious disease, said method comprising administering said therapeutic composition to the subject.
- 44. The therapeutic composition for use according to any one of 37 to 43, wherein the administration of the therapeutic composition is intradermal, intravenous or intramuscular.
- 45. The therapeutic composition for use according to any one of 37 to 43, wherein the administration of the therapeutic composition is inhalative.
- 46. The therapeutic composition for use according to any one of 37 to 45, wherein the therapeutic composition is administered once or multiple times. 47.
- 49. A method for preparing antigen-specific physiologic dendritic cells (phDCs), the method comprising combining phDCs obtained from a donor with at least one mRNA encoding at least one antigenic protein. 50. The method according to embodiment 49, wherein said phDCs are generated by subjecting monocytes obtained from said donor to a physical force. 51.
- said at least one modified nucleoside is a substitution of some or all uridine residues with at least one modified uridine, optionally wherein some or all uridine residues are substituted with N1-methyl-pseudouridine.
- said some or all modified uridine residues comprise one or more uniquely modified uridine residues.
- said one or more uniquely modified uridine residues comprise N1-methyl-pseudouridine.
- 62. The method according to any of embodiments 49 to 61, wherein said at least one mRNA is partially modified with N1-methyl-pseudouridine.
- said at least one mRNA comprises a sequence which is optimized; preferably, wherein the sequence of the said 5’ UTR, 3’UTR, and/or coding sequence for the antigenic protein is optimized.
- said at least one mRNA encoding the at least one antigenic protein is provided in a lipid nanoparticle.
- said lipid nanoparticle comprises a cationic lipid, a PEG-modified lipid, a cholesterol and/or a non-cationic lipid.
- said antigenic protein is associated with a disease, optionally an infectious disease associated antigenic protein or a tumor associated antigenic protein.
- said infectious disease associated antigenic protein is a viral antigenic protein, a bacterial antigenic protein, a fungal antigenic protein, a prion antigenic protein or a parasite antigenic protein.
- said viral antigenic protein is a coronavirus or HIV antigenic protein.
- said coronavirus antigenic protein is a SARS-CoV antigenic protein, optionally a SARS- CoV-2 antigenic protein.
- SARS-CoV or SARS-CoV-2 antigenic protein is derived from a spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein.
- said bacterial antigenic protein is an antigenic protein of Borrelia spp. or Mycobacteria spp..
- said fungal antigenic protein is an antigenic protein of Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis or Candida albicans.
- said parasite antigenic protein is an antigenic protein of Plasmodium malariae.
- said tumor associated antigenic protein is a leukemia antigenic protein, melanoma antigenic protein, lymphoma antigenic protein, endometrial cancer antigenic protein, kidney cancer antigenic protein, brain cancer antigenic protein, cervical cancer antigenic protein, liver cancer antigenic protein, head and neck cancer antigenic protein, gastrointestinal cancer antigenic protein, lymph node cancer antigenic protein, pancreas cancer antigenic protein, ear, nose and throat (ENT) cancer antigenic protein, breast cancer antigenic protein, prostate cancer antigenic protein, ovarian cancer antigenic protein or lung cancer antigenic protein. 79.
- mRNA comprises a nucleotide sequence which is at least 75%, 85%, 90%, 95%, 98% or 99% or a 100% identical to a sequence or a part of a sequence selected from SEQ ID NO: 1 (spike protein transcript), SEQ ID NO: 2 (envelope protein transcript), SEQ ID NO: 3 (nucleocapsid protein transcript), SEQ ID NO: 4 (membrane protein transcript) and/or SEQ ID NO: 5 (Orf1ab polyprotein transcript).
- SEQ ID NO: 1 spike protein transcript
- SEQ ID NO: 2 envelope protein transcript
- SEQ ID NO: 3 nucleocapsid protein transcript
- SEQ ID NO: 4 membrane protein transcript
- SEQ ID NO: 5 Orf1ab polyprotein transcript
- a therapeutic composition comprising a pharmaceutically effective amount of antigen-specific phDCs and at least one mRNA, which encodes for at least one antigenic protein.
- the therapeutic composition according to embodiment 81 further comprising a pharmaceutically acceptable carrier or diluent.
- the therapeutic composition according to embodiment 81 or 82, wherein said mRNA is comprised in lipid nanoparticles.
- 84. The therapeutic composition according to any one of embodiments 81 to 83, wherein said at least one mRNA comprises at least one modified nucleoside.
- said at least one modified nucleoside is a substitution of some or all uridine residues. 86.
- the therapeutic composition according to embodiment 85 wherein said some or all modified uridine residues comprise one or more uniquely modified uridine residues.
- said 5' cap is a Cap1 structure or a m7GpppG cap.
- the mRNA comprises a coding sequence encoding the at least one antigenic protein.
- said lipid nanoparticles comprise a cationic lipid, a PEG-modified lipid, a cholesterol and/or a non-cationic lipid.
- said antigenic protein is associated with a disease, optionally an infectious disease associated antigenic protein or a tumor associated antigenic protein.
- infectious disease associated antigenic protein is a viral antigenic protein, a bacterial antigenic protein, a fungal antigenic protein, a prion antigenic protein or a parasite antigenic protein.
- said viral antigenic protein is a coronavirus or HIV antigenic protein, preferably a SARS-CoV-2 antigenic protein.
- SARS-CoV-2 antigenic protein is derived from the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein.
- the therapeutic composition according to embodiment 96 wherein said bacterial antigenic protein is an antigenic protein of Borrelia spp. or Mycobacteria spp.. 100.
- said fungal antigenic protein is an antigenic protein of Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis or Candida albicans.
- said parasite antigenic protein is an antigenic protein of Plasmodium malariae. 102.
- tumor associated antigenic proteinic protein is a leukemia antigenic protein, melanoma antigenic protein, lymphoma antigenic protein, endometrial cancer antigenic protein, kidney cancer antigenic protein, brain cancer antigenic protein, cervical cancer antigenic protein, liver cancer antigenic protein, head and neck cancer antigenic protein, gastrointestinal cancer antigenic protein, lymph node cancer antigenic protein, pancreas cancer antigenic protein, ear, nose and throat (ENT) cancer antigenic protein, breast cancer antigenic protein, prostate cancer antigenic protein, ovarian cancer antigenic protein or lung cancer antigenic protein.
- leukemia antigenic protein melanoma antigenic protein
- lymphoma antigenic protein endometrial cancer antigenic protein
- kidney cancer antigenic protein brain cancer antigenic protein
- cervical cancer antigenic protein liver cancer antigenic protein
- head and neck cancer antigenic protein gastrointestinal cancer antigenic protein
- lymph node cancer antigenic protein lymph node cancer antigenic protein
- pancreas cancer antigenic protein pancreas cancer antigenic protein
- ENT nose and throat
- composition according to any of embodiments 81 to 102, wherein said at least one mRNA comprises a nucleotide sequence which is at least 75%, 85%, 90%, 95%, 98% or 99% or 100% identical to a sequence or a part of a sequence selected from the group comprising SEQ ID NO: 1 (spike protein transcript), SEQ ID NO: 2 (envelope protein transcript), SEQ ID NO: 3 (nucleocapsid protein transcript), SEQ ID NO: 4 (membrane protein transcript) and/or SEQ ID NO: 5 (Orf1ab polyprotein transcript). 104.
- the therapeutic composition according to any of embodiments 81 to 103, wherein said phDCs, before combining them with said at least one mRNA, are generated by subjecting monocytes obtained from a donor to a physical force.
- 105. The therapeutic composition according to embodiment 104, wherein said physical force is applied by passing said monocytes obtained from said donor through a flow chamber.
- 106. The therapeutic composition according to embodiment 104 or 105, wherein said monocytes obtained from said donor are derived from an extracorporeal blood sample or PBMCs from said donor.
- said flow chamber is a plate.
- the antigen-specific phDCs or the therapeutic composition for use according to embodiment 111 wherein the administration of the therapeutic composition is subcutaneous, intravenous, intramuscular, intra- articular, intra-synovial, intrasternal, intrathecal, intra-hepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, intranasal or sublingual; preferably, intradermal, intravenous or intramuscular. 113.
- the antigen-specific phDCs or the therapeutic composition for use according to embodiment 113 wherein the concentration of said disease causing particles is lower in the subject as compared to a concentration of disease causing particles in a second subject, which had previously been diagnosed as having the same disease and which had been vaccinated against the same disease and which has been against the same disease causing particle with a different.therapeutic treatment.
- 116. The antigen-specific phDCs or the therapeutic composition for use according to embodiment 115, wherein said different therapeutic does not comprise dendritic cells.
- 119. The antigen-specific phDCs or the therapeutic composition for use according to embodiment 113, wherein said concentration of the disease causing particles is lower in the subject systemically upon infection.
- TPM Tumor Node Metastasis
- the antigen-specific phDCs or the therapeutic composition for use according to any of embodiments 111 to 127, wherein administration of the antigen-specific phDCs of embodiment 80 or the therapeutic composition of any of embodiments 81 to 110 additionally has a vaccination effect in the subject.
- a kit comprising physiologic dendritic cells (phDCs) and at least one mRNA, wherein said at least one mRNA encodes for at least one antigenic protein.
- phDCs physiologic dendritic cells
- said at least one mRNA encodes for at least one antigenic protein.
- said viral antigenic protein is a coronavirus antigenic protein, preferably a SARS-CoV-2 antigenic protein.
- kits according to embodiment 131 wherein said SARS-CoV-2 antigenic protein is derived from the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein.
- SARS-CoV-2 antigenic protein is derived from the spike protein, envelope protein, nucleocapsid protein, membrane protein and/or Orf1ab polyprotein.
- 136 The kit according to any of embodiments 127 to 132, wherein said at least one mRNA is formulated in a lipid nanoparticle.
- a method of treating a disease in a subject comprising administering the antigen-specific phDCs of embodiment 80 or therapeutic composition according to any of embodiments 81 to 110 to the subject, wherein the subject has been previously diagnosed as having the disease. 138.
- the administration of the therapeutic composition is subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intra-hepatic, intralesional, intracranial, transdermal, intradermal, intrapulmonal, intraperitoneal, intracardial, intraarterial, or sublingual; preferably, intradermal, intravenous or intramuscular.
- the disease is caused by disease causing particles, optionally wherein the disease causing particles are viruses, bacteria, fungi, parasites, prions and/or tumor cells.
- the method according to embodiment 138, wherein said concentration of the disease causing particles is lower in the subject as compared to the concentration of the disease causing particles before administration of the therapeutic composition.
- said concentration of disease causing particles is lower in the subject as compared to the concentration of disease causing particles in a second subject, which has been previously diagnosed as having the same disease and which has been treated against the same disease causing particle with a different therapeutic treatment.
- the different therapeutic treatment does not comprise dendritic cells.
- the method according to embodiment 141 or 142, wherein the different therapeutic treatment is an RNA therapeutic treatment. 144.
- 150. The method according to any one of embodiments 137 to 148, wherein the subject has been previously diagnosed to have a viral infectious disease; preferably, wherein the subject is diagnosed to have corona disease 2019 (Covid-19).
- 151. The method according to any one of embodiments 137 to 139, wherein the subject has been previously diagnosed to have a tumor or cancer.
- the cancer is classifiable as stage I, II, III or IV according to the Tumor Node Metastasis (TNM) anatomic/prognostic group system of the cancer staging system of the American Joint Committee on Cancer. 153.
- TPM Tumor Node Metastasis
- PBMC peripheral blood mononuclear cells
- Transimmunization chamber The miniaturized ECP device suitable for work in animal models, called the Transimmunization (TI) chamber, was designed and created for Dr. Edelson's laboratory by Transimmune AG in collaboration with Fraunhofer Institute for Biomedical Engineering, Saarland, Germany.
- the sterile polystyrene TI chamber has the external dimensions of 25 * 75 mm, with the flow path of 18 * 66 mm, and the flow passage height of 290 +/- 15um.
- PBMC TI treatment protocol Isolated murine platelet-containing PBMC are resuspended in fetal bovine serum (FBS).
- the cells are then is incubated in the TI chamber for 1 hour at 37C. This step allows for platelet-activating plasma protein deposition in the chamber, and platelet adherence to the coated chamber surfaces, as confirmed by light microscopy.
- the cells are subsequently passed through the TI chamber using a syringe pump, at a rate of 0.09 mL/min. Following plate passage, cells are collected, and the TI chamber washed with 100% FBS at 0.49 mL/min while being physically perturbed by flicking or tapping the plate surface to help detach any adherent cells from the chamber.
- the collected cells are washed and cultured overnight at standard conditions in RPMI without phenol red (Gibco) supplemented with 15% autologous mouse serum and 1% penicillin/streptomycin/L-glutamine (Invitrogen).
- mRNA-containing LNP transfection of phDC The desired amount of LNP (cKK-E12, cholesterol, C14-PEG 2000-PE and DOPE at a ratio of 35:46.5:2.5:16 (Santangelo laboratory, Emory University) containing mRNA for the antigen of interest (such as SARS-CoV-2 Spike protein, or a sample tumor antigen), is added directly to the overnight phDC culture, at the time of setting up the culture.
- the antigen of interest such as SARS-CoV-2 Spike protein, or a sample tumor antigen
- LNP amounts can vary from 20ug to 10ng, depending on the LNP and antigen used.
- phDC re-introduction into experimental animals The following day, LNP-transfected cells are harvested by scraping, washed, resuspended in sterile PBS (Gibco), and administered intravenously at 100uL/animal via the retro-orbital plexus.
- sterile PBS Gibco
- phDC internalization of viral antigen-containing LNP Methods phDC are prepared from healthy C57BL/6 donor mouse blood by standard protocol described in Example 1.
- LNP containing mRNA for the viral antigen of interest SARS-CoV-2 Spike protein mRNA
- LNP cKK-E12 based LNP containing SARS-CoV-2 Spike protein
- Antigen expression is monitored after overnight incubation by using an appropriate fluorescently labeled detection antibody (anti-human SARS-CoV-2 Spike protein antibody, kindly provided by Santangelo laboratory, Emory University), as well as any necessary antibodies to identify cells of interest (eg CD11b + , Ly6G- for murine phDC within the plate-passed PBMC; CD11b Biolegend clone M1/70; Ly6G Biolegend clone 1A8).
- Antibody binding to phDC can be detected by either flow cytometry (Cytoflex cytometer, analysis using FlowJo v10 software) or by confocal microscopy.
- Therapeutic phDC administration in the EG7-OVA lymphoma mouse model using tumor antigen mRNA-containing LNP (ovalbumin (OVA) antigen) The experimental design is schematically described in Figure 2A.
- 3x10 6 EG7-OVA tumor cells are injected subcutaneously in 100 mL into the right flanks of recipient wild-type C57BL/6J mice.
- Therapeutic treatment is initiated on day 2 following tumor implantation.
- phDC are prepared from the blood of respective experimental group (200uL blood collected per animal) by standard protocol described in Example 1. Blood from groups not treated with phDC is discarded.
- cKK- E12 based LNP containing mRNA for the tumor antigen of interest here the model antigen ovalbumin (OVA) that has been introduced into the EL4 lymphoma cells to produce the EG7-OVA tumor model (SEQ ID NO: 20, also used for all following experiments relating to OVA mRNA containing LNPs).
- OVA ovalbumin
- Mice are usually bled for phDC therapeutic manufacture on Mondays and Thursdays, with phDC or i.m. treatments taking place on Tuesdays and Fridays. Typically, 5-6 bi-weekly therapeutic immunization treatments can be carried out.
- Tumor volume is monitored via biweekly measurement of perpendicular tumor diameters and height using a caliper, and tumor volume calculated as (tumor length x width x height)/2.
- Splenocytes are harvested at the end of the experiment (Day 30 post tumor inoculation) from all treatment and control groups for further analysis, to characterize the resulting anti-tumor immune response.
- Elispot analysis CD8+ splenic T cells from treated mice (negative selection using a Miltenyi CD8 T cell isolation kit) are immediately placed into an 18hr IFN-g Elispot assay at 1*10 5 cells per well in the presence or absence of 10ug/mL SIINFEKL peptide.
- Tem (effector)/Tcm (central memory) type phenotype analysis staining for CD44 (Biolegend clone IM7) and CD62L (Biolegend clone MEL-14) expression on antigen-specific T cells followed by flow cytometry (Cytoflex).
- Stem-like T cell analysis staining for IL7Ra (Biolegend clone A7R34) and SCA-1 (Biolegend clone E13-161.7) expression on antigen-specific T cells followed by flow cytometry (Cytoflex).
- T cell exhaustion marker evaluation PD-1 (Biolegend clone 29F.1A12) expression on antigen-specific T cells followed by flow cytometry (Cytoflex).
- cKK-E12 based LNP containing mRNA for the tumor antigen of interest here the model antigen ovalbumin (OVA). All LNPs prepared and provided by the Santangelo laboratory, Emory University. Plate-passed PBMC are incubated with the LNP overnight, to allow for LNP uptake and antigen expression. On the following day, the PBMC are collected and prepared as described for Example 1, and administered intravenously at 100uL/animal via the retro-orbital plexus to the “phDC [ova]”-vaccinated experimental group.
- OVA model antigen ovalbumin
- an equivalent dose of LNP containing OVA mRNA is injected in 10-50ul volume of sterile PBS into the thigh of each animal in the intramuscularly (“IM [ova]”)- vaccinated experimental group.
- the animals in the “Untreated” group do not receive any therapy.
- Mice are bled on experiment Days -8 and -1, and prophylactically vaccinated with either phDC or with intramuscular LNP on Days -7 and 0.
- whole undifferentiated splenocytes are collected for analysis of the spontaneous immune response by IFNg Elispot.
- Elispot analysis whole undifferentiated splenocytes from all experimental groups are immediately placed into an 18hr IFN-g Elispot assay at 1*10 5 cells per well in the absence of any additional stimulation.
- To further identify cells responsible for IFNg production some Day 13 samples were further fractionated into CD8 T cells (negative selection using a Miltenyi CD8 T cell isolation kit), or into NK cells (positive selection using Miltenyi NK1.1 cell isolation kit). Results Interestingly, animals prophylactically vaccinated and boosted with phDC, but not with intramuscular LNP injection, demonstrate spontaneous IFNg production in the absence of any additional stimulation (Figure 5A-C). This spontaneous response is detectable at least as long as 13 days post vaccination (Figure 5C).
- phDC vaccination not only initiates the antigen-specific T and B cell responses ( Figures 4, 5), but also broadly engages the innate immune system, such as NK cells.
- Figure 6A Prophylactic phDC vaccination in the EG7-OVA lymphoma mouse model using tumor antigen mRNA-containing LNP (ovalbumin (OVA) antigen)
- OVA ovalpha-ovalbumin
- soluble OVA protein in 100uL volume of sterile PBS is injected into the retro-orbital plexus of the “Soluble Ova”- treated experimental group animals.
- the initial vaccination treatment is followed 1 week later (Day -7) by a booster vaccination, carried out in the identical manner as that described above.
- 1x10 6 EG7-OVA tumor cells are injected subcutaneously in 100 mL into the right flanks of mice from all treatment groups. Tumor volume is monitored via biweekly measurement of perpendicular tumor diameters and height using a caliper, and tumor volume calculated as (tumor length x width x height)/2.
- LNP containing mRNA for antigen of interest such as 1ug/mL of cKK-E12 based LNP containing OVA or SARS-CoV-2 Spike protein mRNA (LNP prepared and provided by the Santangelo laboratory, Emory University). Plate-passed PBMC are incubated with the LNP overnight, to allow for LNP uptake and antigen expression.
- Antigen expression is monitored after overnight incubation by staining the cells intracellularly with fluorescently labeled anti-OVA antibody (Rockland Immunochemicals), as well as any necessary antibodies to identify cells of interest (eg CD11b+ for murine phDC within the plate-passed PBMC; CD11b Biolegend clone M1/70).
- Antibody binding to phDC can be detected by flow cytometry (Cytoflex cytometer, analysis using FlowJo v10 software).
- Results Murine phDC, among all other immune cell subsets contained in the PBMC, specifically express OVA protein encoded by mRNA contained in cKK-E12 LNP.
- the antigen source such as soluble OVA protein (10ug/mL; 50ug/mL); EG7-OVA tumor cells treated with 8-MOP/UVA (200ng/mL 8-MOP, UVADEX, Therakos; 2 or 4 J/cm2 UVA); or LNP containing OVA protein mRNA (1ug/mL or 5ug/mL of cKK-E12 based LNP) (LNP prepared and provided by the Santangelo laboratory, Emory University). Plate-passed PBMC are incubated with the respective antigen sources overnight, to allow for antigen uptake and/or expression, and processing.
- the antigen source such as soluble OVA protein (10ug/mL; 50ug/mL); EG7-OVA tumor cells treated with 8-MOP/UVA (200ng/mL 8-MOP, UVADEX, Therakos; 2 or 4 J/cm2 UVA); or LNP containing OVA protein mRNA (1ug/m
- Antigenic OVA peptide SIINFEKL presentation on phDC H-2Kb MHCI molecules is detected after overnight incubation by staining the cells with fluorescently labeled 25.D1 TCR-like antibody (Biolegend, clone 25-D1.16), as well as any necessary antibodies to identify cells of interest (eg CD11b+, Ly6G- for murine phDC within the plate-passed PBMC; CD11b Biolegend clone M1/70; Ly6G Biolegend clone 1A8).
- Antibody binding to phDC can be detected by flow cytometry (Cytoflex cytometer, analysis using FlowJo v10 software).
- Murine phDC among all other immune cell subsets contained in the PBMC, specifically process and present antigenic OVA SIINFEKL peptide in their MHCI molecules. Antigenic peptide presentation is significantly higher when OVA protein is expressed via mRNA LNP, as opposed to delivery via soluble OVA protein, or dying tumor cells expressing OVA protein ( Figure 8).
- Figure 8 Time kinetics of surface SIINFEKL-MHC I complex expression (via 25.D1 ab stain) in phDC transduced with OVA mRNA LNP Methods phDC are prepared from healthy C57BL/6 donor mouse blood by standard protocol described in Experiment 1.
- LNP containing mRNA for antigen of interest such as 1ug/mL of cKK-E12 based LNP containing OVA protein mRNA (LNP prepared and provided by the Santangelo laboratory, Emory University). Plate-passed PBMC are incubated with the LNP for 2, 4, 6, 8, 12, or 20hrs, then washed and fixed for staining.
- Antigenic OVA peptide SIINFEKL expression on phDC H-2Kb MHCI molecules is detected after 2-20hr incubation by staining the cells with fluorescently labeled 25.D1 TCR-like antibody (Biolegend, clone 25-D1.16), as well as any necessary antibodies to identify cells of interest (eg CD11b+ for murine phDC within the plate-passed PBMC; CD11b Biolegend clone M1/70).
- Antibody binding to phDC can be detected by flow cytometry, and antibody-positive cell percentages/mean fluorescence intensity (MFI) of positive cells quantitated (Cytoflex cytometer, analysis using FlowJo v10 software).
- BMDC bone marrow cells were cultured from healthy C57BL/6 donor mouse bone marrow according to standard protocol. Briefly, bone marrow cells were plated in cell culture media containing GM-CSF (20 ng/ml); after 5 days, nonadherent cells were washed and replated in fresh GM-CSF–supplemented media and incubated for 48 hours more; subsequently nonadherent BMDC cells were removed, washed, and used immediately for experiments.
- GM-CSF 20 ng/ml
- LNP containing mRNA for antigen of interest such as 0.1ug/mL of cKK-E12 based LNP containing OVA protein mRNA (LNP prepared and provided by the Santangelo laboratory, Emory University). Plate-passed PBMC or cultured BMDC are incubated with the LNP overnight, to allow for LNP uptake and antigen expression.
- Antigenic OVA peptide SIINFEKL expression on phDC H-2Kb MHCI molecules is detected after incubation by staining the cells with fluorescently labeled 25.D1 TCR- like antibody (Biolegend, clone 25-D1.16), as well as any necessary antibodies to identify cells of interest (eg CD11b+ for murine phDC within the plate-passed PBMC; CD11b Biolegend clone M1/70; CD11c for murine BMDC, Biolegend clone N418).
- Antibody binding to phDC or BMDC can be detected by flow cytometry (Cytoflex cytometer, analysis using FlowJo v10 software).
- LNP containing mRNA for antigen of interest such as 1ug/mL of cKK-E12 based LNP containing OVA protein mRNA, or 0.05, 0.1ug, 0.5ug, 1ug, or 5ug of LNP containing immunogenic OVA SIINFEKL peptide mRNA (LNP prepared and provided by the Santangelo laboratory, Emory University).
- LNP immunogenic OVA SIINFEKL peptide mRNA
- Antigenic OVA peptide SIINFEKL expression on phDC H-2Kb MHCI molecules is detected after overnight incubation by staining the cells with fluorescently labeled 25.D1 TCR-like antibody (Biolegend, clone 25-D1.16), as well as any necessary antibodies to identify cells of interest (eg CD11b+ for murine phDC within the plate- passed PBMC; CD11b Biolegend clone M1/70).
- Antibody binding to phDC or BMDC can be detected by flow cytometry (Cytoflex cytometer, analysis using FlowJo v10 software).
- OT1 proliferation assay phDC pulsed with titrating amounts of LNP or soluble OVA protein cultured with OT1 T cells
- Methods phDC are prepared from healthy C57BL/6 donor mouse blood by standard protocol described in Experiment 1.
- the antigen source such as soluble OVA protein (50ug/mL; 200ug/mL); or LNP containing OVA protein mRNA (1, 0.1, 0.01, or 0.001ug/mL of cKK-E12 based LNP) (LNP prepared and provided by the Santangelo laboratory, Emory University).
- Plate-passed PBMC are incubated with the respective antigen sources overnight, to allow for antigen uptake and/or expression, and processing. After overnight incubation, antigen-loaded cells are harvested and cocultured for 3 days at 2*10 5 cells/mL (of note, phDC are not purified; the cell number is for total PBMC, of which 3-10% are CD11+ phDC; phDC cell numbers within PBMC are therefore at most ⁇ 2*10 4 cells/mL) under standard conditions in 96-well plates with 1*10 5 cells/mL CFSE-labeled OVA-specific OT1 CD8 T cells (isolated from spleens of C57Bl/6-Tg (TcraTcrb)1100Mjb/J mice that recognize OVA peptide residues 257 to 264 in the context of H-2Kb, Jackson Laboratory).
- phDC transduction with the lowest LNP concentration was at least as potent at stimulating CD8 T cell response as phDC loading with the highest soluble OVA protein concentration (200ng/mL), suggesting that mRNA LNP are a significantly superior antigen source.
- OT1 proliferation assay OT1 T cells cultured with titrating amount of phDC (at 1 ug/mL LNP) Methods phDC are prepared from healthy C57BL/6 donor mouse blood by standard protocol described in Experiment 1.
- the antigen source such as LNP containing OVA protein mRNA (1ug/mL of cKK-E12 based LNP) (LNP prepared and provided by the Santangelo laboratory, Emory University). Plate-passed PBMC are incubated with the respective antigen sources overnight, to allow for antigen uptake and/or expression, and processing.
- phDC are not purified; the cell number is for total PBMC, of which 3-10% are CD11+ phDC; phDC cell numbers within PBMC are therefore at most ⁇ 2*10 4 , 1*10 4 , 5*10 3 , 2.5*10 3 , or 1.2*10 3 cells/mL) under standard conditions in 96-well plates with 1*10 5 cells/mL CFSE- labeled OVA-specific OT1 CD8 T cells (isolated from spleens of C57Bl/6-Tg (TcraTcrb)1100Mjb/J mice that recognize OVA peptide residues 257 to 264 in the context of H-2Kb, Jackson Laboratory).
- the experimental groups therefore are: - Mock (SARS-CoV-2 Spike protein) mRNA LNP intramuscular vaccination - OVA mRNA LNP intramuscular vaccination - OVA mRNA LNP phDC retro-orbital vaccination With treatments administered on days 4, 8, 11, 15, 18, and 25 post EG7-OVA tumor implantation ( Figure 14 a).
- OVA mRNA phDC therapeutic phDC administration
- 10ng OVA mRNA-containing LNPs 0.5ug/kg of LNP, dose equivalent to COVID-19 Pfizer mRNA LNP vaccine in current human vaccine setting
- OVA mRNA IM no phDC
- SIINFEKL tetramer analysis and adoptive T cell transfer Methods Splenocytes are harvested at the end of Experiment 13 described above (were harvested at the end of the in vivo tumor monitoring period (Day 32 after tumor inoculation) from all treatment and control groups for further analysis, to characterize the resulting anti-tumor immune response.
- Adoptive T cell transfer CD3+ T cells isolated from splenocytes of the three experimental groups of Experiment 13 (Miltenyi T cell isolation kit) are transferred intravenously at 1.5*10 7 T cells/mouse into antigen-naiive C57BL/6 mice, freshly inoculated with EG7-OVA tumors (tumor inoculation as described for Experiment 3), and tumor development was monitored over the course of 19 days.
- OVA protein mRNA LNP-transduced phDC vaccination (“OVA phDC”) and intramuscular OVA protein mRNA LNP vaccination (“OVA IM”) both successfully induced OVA antigen-specific T cells, as measured by percentage of H- 2Kb SIINFEKL tetramer-positive CD8 T cells ( Figure 15 a). Mock-treated animals (“Mock IM”) also showed some tetramer positivity, reflecting the presence of background, natural T immunity in EG7-OVA tumor bearing mice.
- OVA phDC OVA protein mRNA LNP-transduced phDC
- Experiment 15 Spike protein ELISpot detection of SARS-CoV-2 reactive T cells
- Spike protein mRNA-containing LNP transduction of human phDC Plate-passed PBMC containing nascent phDC from healthy human donors, produced per methods described in Experiment 1, are plated at 5*10 5 cells per well, in triplicate per experimental group, in human IFN ⁇ ELISpot plates.
- RNA sequence corresponds to SEQ ID NO: 19
- culture medium consisting of RPMI without phenol red (Gibco) supplemented with 15% autologous human plasma and 1% penicillin/streptomycin/L-glutamine (Invitrogen).
- Positive control wells are treated with an overlapping pool of Spike Class I & II peptides (Miltenyi Biotec, PepTivatorSARS-CoV-2 Prot-S Complete).
- Negative control wells are treated with 62.5-250ng/well of cKK-E12 based LNP containing an irrelevant (eg Nanoluciferase) protein mRNA.
- Elispot assay read-out After overnight incubation, wells are washed and IFN ⁇ spots are detected with biotinylated anti-human IFN ⁇ mAb, streptavidin-ALP and BCIP/NBT-plus substrate per manufacturer’s protocol (MAbTech, 3420-2AST-2).
- CD4 and CD8 T cell depletions To test the contribution of T cell subsets to cytokine production, CD4 or CD8 T cells may be selectively depleted from PBMC after plate passage, but prior to overnight incubation with LNPs in Elispot wells. T cells are depleted using standard depletion kits (Miltenyi; CellSep).
- FIG. 16A An example of a phDC ELISpot using PBMC isolated from a single human donor, 4 weeks post SARS-CoV2 infection. IFN ⁇ release was significantly elevated in the presence of phDC [Spike], when compared to negative controls. This demonstrates detection of human Spike specific T cells via mRNA transduced phDC in a Covid convalescent donor.
- Figure 16B An example of a phDC ELISpot dose response using PBMC from a single human donor, 4 weeks post SARS-CoV2 infection. IFN ⁇ release increased to levels above 150 SFU/million when LNP [Spike] antigen was added at 62.5ng/well.
- Figure 17B 11 previously vaccinated and convalescent human donors were screened in phDC ELISpot against SARS-CoV-2 Spike antigen. Dotted line represents the standard threshold cutoff for positive response (50 SFU/Million cells). This demonstrates that 1) the response strength is generally negatively correlated w/ convalescent period; 2) the phDC induced IFN ⁇ response is durable and detectable out to 1 year post Covid infection.
- Figure 17C Vaccinated and convalescent donors’ plate passed PBMC were depleted of either CD8 or CD4 T cells prior to incorporation into the standard 18hr phDC [Spike] ELISpot IFN ⁇ assay.
- FIG. 17D An example of a phDC ELISpot using PBMC isolated from a single human donor prior to, and six weeks post, SARS-CoV2 infection. IFN ⁇ release increased above the positive threshold of 50 SFU/Million cells following infection. This demonstrates the phDC induced IFN ⁇ T cell response increases following natural Covid infection, thus showing the feasibility and utility of longitudinal immune monitoring.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Oncology (AREA)
- Immunology (AREA)
- Medicinal Chemistry (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU2024265076A AU2024265076A1 (en) | 2023-05-01 | 2024-04-30 | Antigen-specific physiologic dendritic cells for therapy |
Applications Claiming Priority (8)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363499349P | 2023-05-01 | 2023-05-01 | |
| US202363499343P | 2023-05-01 | 2023-05-01 | |
| US63/499,343 | 2023-05-01 | ||
| US63/499,349 | 2023-05-01 | ||
| EP23193757.4 | 2023-08-28 | ||
| EP23193761 | 2023-08-28 | ||
| EP23193757 | 2023-08-28 | ||
| EP23193761.6 | 2023-08-28 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024227806A1 true WO2024227806A1 (en) | 2024-11-07 |
Family
ID=90971499
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/061957 Pending WO2024227806A1 (en) | 2023-05-01 | 2024-04-30 | Antigen-specific physiologic dendritic cells for therapy |
Country Status (2)
| Country | Link |
|---|---|
| AU (1) | AU2024265076A1 (en) |
| WO (1) | WO2024227806A1 (en) |
Citations (12)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0663930A1 (en) | 1992-10-09 | 1995-07-26 | Cray Valley Sa | Charged sheet mouldable resin compositions transformable by compression and application thereof in the production of moulded parts |
| WO1995028479A1 (en) | 1994-04-14 | 1995-10-26 | KLINIKUM DER ALBERT-LUDWIGS-UNIVERSITäT FREIBURG | Process for producing dendritic cells, cells thus produced and container for carrying out this process |
| EP0922758A2 (en) | 1997-10-27 | 1999-06-16 | Rockefeller University | Methods and compositions for obtaining mature dendritic cells |
| CA2377207A1 (en) | 1999-06-25 | 2001-01-04 | Christian Plank | Combinations for introducing nucleic acids into cells |
| WO2013143683A1 (en) | 2012-03-26 | 2013-10-03 | Biontech Ag | Rna formulation for immunotherapy |
| WO2014106631A1 (en) | 2013-01-03 | 2014-07-10 | Transimmune Ag | Method for obtaining immuno-suppressive dendritic cells |
| WO2014106629A1 (en) | 2013-01-03 | 2014-07-10 | Transimmune Ag | Method for obtaining immuno-stimulatory dendritic cells |
| WO2016001405A1 (en) | 2014-07-03 | 2016-01-07 | Transimmune Ag | Method for obtaining globally activated monocytes |
| WO2017005700A1 (en) | 2015-07-03 | 2017-01-12 | Transimmune Ag | Device and method for obtaining immuno-stimulatory antigen-presenting cells |
| WO2021239880A9 (en) * | 2020-05-29 | 2022-01-13 | Curevac Ag | Nucleic acid based combination vaccines |
| CN114478798A (en) * | 2021-01-19 | 2022-05-13 | 康希诺生物股份公司 | Protein or protein composition and preparation method and use thereof |
| EP4223306A2 (en) | 2019-02-14 | 2023-08-09 | Ethris GmbH | Treatment of ciliopathies |
-
2024
- 2024-04-30 AU AU2024265076A patent/AU2024265076A1/en active Pending
- 2024-04-30 WO PCT/EP2024/061957 patent/WO2024227806A1/en active Pending
Patent Citations (14)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0663930A1 (en) | 1992-10-09 | 1995-07-26 | Cray Valley Sa | Charged sheet mouldable resin compositions transformable by compression and application thereof in the production of moulded parts |
| WO1995028479A1 (en) | 1994-04-14 | 1995-10-26 | KLINIKUM DER ALBERT-LUDWIGS-UNIVERSITäT FREIBURG | Process for producing dendritic cells, cells thus produced and container for carrying out this process |
| EP0922758A2 (en) | 1997-10-27 | 1999-06-16 | Rockefeller University | Methods and compositions for obtaining mature dendritic cells |
| CA2377207A1 (en) | 1999-06-25 | 2001-01-04 | Christian Plank | Combinations for introducing nucleic acids into cells |
| WO2001000708A1 (en) | 1999-06-25 | 2001-01-04 | Christian Plank | Combinations for introducing nucleic acids into cells |
| EP1198489A1 (en) | 1999-06-25 | 2002-04-24 | Christian Plank | Combinations for introducing nucleic acids into cells |
| WO2013143683A1 (en) | 2012-03-26 | 2013-10-03 | Biontech Ag | Rna formulation for immunotherapy |
| WO2014106631A1 (en) | 2013-01-03 | 2014-07-10 | Transimmune Ag | Method for obtaining immuno-suppressive dendritic cells |
| WO2014106629A1 (en) | 2013-01-03 | 2014-07-10 | Transimmune Ag | Method for obtaining immuno-stimulatory dendritic cells |
| WO2016001405A1 (en) | 2014-07-03 | 2016-01-07 | Transimmune Ag | Method for obtaining globally activated monocytes |
| WO2017005700A1 (en) | 2015-07-03 | 2017-01-12 | Transimmune Ag | Device and method for obtaining immuno-stimulatory antigen-presenting cells |
| EP4223306A2 (en) | 2019-02-14 | 2023-08-09 | Ethris GmbH | Treatment of ciliopathies |
| WO2021239880A9 (en) * | 2020-05-29 | 2022-01-13 | Curevac Ag | Nucleic acid based combination vaccines |
| CN114478798A (en) * | 2021-01-19 | 2022-05-13 | 康希诺生物股份公司 | Protein or protein composition and preparation method and use thereof |
Non-Patent Citations (11)
| Title |
|---|
| "NCBI", Database accession no. NC_AF033819 |
| BECK JAN D. ET AL: "mRNA therapeutics in cancer immunotherapy", vol. 20, no. 1, 15 April 2021 (2021-04-15), XP093034772, Retrieved from the Internet <URL:https://link.springer.com/article/10.1186/s12943-021-01348-0/fulltext.html> DOI: 10.1186/s12943-021-01348-0 * |
| BUECHLER ET AL., GENERATION OF DENDRITIC CELLS USING CELL CULTURE BAGS, 2004 |
| DONG ET AL., PNAS, vol. 111, no. 11, 2014, pages 3955 - 60 |
| DURAZZO ET AL., INDUCTION OF MONOCYTE-TO-DENDRITIC CELL MATURATION BY EXTRACORPOREAL PHOTOCHEMOTHERAPY: INITIATION VIA DIRECT PLATELET SIGNALING, 2014 |
| HANLON ET AL., RAPID PRODUCTION OF PHYSIOLOGIC DENDRITIC CELLS (PHDC) FOR IMMUNOTHERAPY, 2020 |
| HOLTKAMP ET AL., BLOOD, vol. 108, 2006, pages 4009 - 4017 |
| TATESHITA NAHO ET AL: "Development of a lipoplex-type mRNA carrier composed of an ionizable lipid with a vitamin E scaffold and the KALA peptide for use as an ex vivo dendritic cell-based cancer vaccine", JOURNAL OF CONTROLLED RELEASE, vol. 310, 1 September 2019 (2019-09-01), AMSTERDAM, NL, pages 36 - 46, XP093127564, ISSN: 0168-3659, DOI: 10.1016/j.jconrel.2019.08.002 * |
| VENTURA ET AL., EXTRACORPOREAL PHOTOCHEMOTHERAPY DRIVES MONOCYTE-TO-DENDRITIC CELL MATURATION TO INDUCE ANTICANCER IMMUNITY, 2018 |
| WANG YANG ET AL: "mRNA vaccine: a potential therapeutic strategy", vol. 20, no. 1, 1 December 2021 (2021-12-01), XP055907500, Retrieved from the Internet <URL:https://molecular-cancer.biomedcentral.com/track/pdf/10.1186/s12943-021-01311-z.pdf> DOI: 10.1186/s12943-021-01311-z * |
| Y. DONG ET AL: "Lipopeptide nanoparticles for potent and selective siRNA delivery in rodents and nonhuman primates", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 111, no. 11, 18 March 2014 (2014-03-18), pages 3955 - 3960, XP055167076, ISSN: 0027-8424, DOI: 10.1073/pnas.1322937111 * |
Also Published As
| Publication number | Publication date |
|---|---|
| AU2024265076A1 (en) | 2025-11-06 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| Qin et al. | A guide to nucleic acid vaccines in the prevention and treatment of infectious diseases and cancers: from basic principles to current applications | |
| US20220265722A1 (en) | Engineering and delivery of therapeutic compositions of freshley isolated cells | |
| Ashley et al. | Bone marrow–generated dendritic cells pulsed with tumor extracts or tumor RNA induce antitumor immunity against central nervous system tumors | |
| JP6134763B2 (en) | Dendritic cells that have been produced using GM-CSF and interferon α and that have taken up cancer cells that have been heat-treated and killed | |
| US20180000912A1 (en) | Virus Vectors Expressing Multiple Epitopes of Tumor Associated Antigens For Inducing Antitumor Immunity | |
| DK2700708T3 (en) | Enhancement of T-cell-stimulating ability of human antigen-presenting cells in vitro and in vivo and their use in vaccination | |
| US11684671B2 (en) | Enhancing the T-cell stimulatory capacity of human antigen presenting cells in vitro and in vivo and its use in vaccination | |
| JP2014043459A (en) | Method for enhancing t cell response | |
| EP2892930A1 (en) | Fusion proteins for promoting an immune response, nucleic acids encoding same, and methods of making and use thereof | |
| US12220451B2 (en) | Cell-based vaccine compositions and methods of use | |
| AU2003265574A1 (en) | Expression vectors encoding epitopes of target-associated antigens | |
| EP2029165B1 (en) | B cell-based vaccine loaded with the ligand of natural killer t cell and antigen | |
| JPWO2010061930A1 (en) | Immunotherapy using CD1d and target antigen co-expressing allocells | |
| Kramer et al. | Functionalisation of Virus‐Like Particles Enhances Antitumour Immune Responses | |
| EP3838915A1 (en) | Tumor immunotherapy composition based on antigen-presenting cells activated by attenuated listeria monocytogenes, preparation method therefor and application thereof | |
| AU2024265451A1 (en) | Antigen-specific physiologic dendritic cells for vaccination | |
| JPH11512724A (en) | Immunization with targeted particulate genes enhances immune response in cell therapy | |
| CA3028168C (en) | Compositions and methods for activating antigen presenting cells with chimeric poliovirus | |
| Smed-Sörensen et al. | Targeting dendritic cells for improved HIV-1 vaccines | |
| Walczak et al. | Heterologous prime-boost immunizations with a virosomal and an alphavirus replicon vaccine | |
| AU2024265076A1 (en) | Antigen-specific physiologic dendritic cells for therapy | |
| Kreiter et al. | FLT3 ligand as a molecular adjuvant for naked RNA vaccines | |
| US20100028380A1 (en) | B cell-based vaccine loaded with the ligand of natural killer t cell and antigen | |
| CN118215489A (en) | Antigen presenting cell/target cell hybridoma-derived vaccines | |
| WO2025083147A1 (en) | Methods for analyzing immune responses and antigen profiling |
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: 24723532 Country of ref document: EP Kind code of ref document: A1 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: AU2024265076 Country of ref document: AU |
|
| WWE | Wipo information: entry into national phase |
Ref document number: P2025-03504 Country of ref document: AE |
|
| ENP | Entry into the national phase |
Ref document number: 2024265076 Country of ref document: AU Date of ref document: 20240430 Kind code of ref document: A |
|
| REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112025023432 Country of ref document: BR |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024723532 Country of ref document: EP |
|
| ENP | Entry into the national phase |
Ref document number: 2024723532 Country of ref document: EP Effective date: 20251201 |