WO2024200823A1 - Nanoparticule à base de lipide ciblant des cellules immunitaires activées pour l'expression d'une molécule d'amélioration de cellule immunitaire et son utilisation - Google Patents

Nanoparticule à base de lipide ciblant des cellules immunitaires activées pour l'expression d'une molécule d'amélioration de cellule immunitaire et son utilisation Download PDF

Info

Publication number
WO2024200823A1
WO2024200823A1 PCT/EP2024/058778 EP2024058778W WO2024200823A1 WO 2024200823 A1 WO2024200823 A1 WO 2024200823A1 EP 2024058778 W EP2024058778 W EP 2024058778W WO 2024200823 A1 WO2024200823 A1 WO 2024200823A1
Authority
WO
WIPO (PCT)
Prior art keywords
lipid
peg
mol
based nanoparticle
virus
Prior art date
Application number
PCT/EP2024/058778
Other languages
English (en)
Inventor
Nicolas Poirier
Damien HABRANT
Aurore MORELLO
Thierry Gautheret
Original Assignee
Ose Immunotherapeutics
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ose Immunotherapeutics filed Critical Ose Immunotherapeutics
Publication of WO2024200823A1 publication Critical patent/WO2024200823A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/77Internalization into the cell

Definitions

  • the invention pertains to the field of immunotherapy.
  • the invention relates to a lipid-based nanoparticle comprising mRNA molecule(s), and its use to treat conditions such as cancers and infections.
  • a solution explored in the prior art to enhance an immune response such as a cytotoxic T- lymphocyte (CTL) response focuses on targeting specific immune cells subsets for the delivery of activation enhancing compounds.
  • CTL cytotoxic T- lymphocyte
  • TEE tumor microenvironment
  • Vaccines based on mRNA-containing lipid nanoparticles are a promising new delivery platform.
  • LNPs are used to deliver mRNA to cells and have led to the expression of the encoded proteins, thus providing immune-protection to the body.
  • Expressing a protein by delivering the encoding mRNA has many benefits over methods that use proteins, plasmid DNA or viral vectors.
  • the coding sequence of the desired protein is the only substance delivered to cells, thus avoiding all the side effects associated with plasmid backbones, viral genes, and viral proteins.
  • the mRNA does not carry the risk of being incorporated into the genome and protein production starts immediately after mRNA delivery.
  • the inventors have developed a targeted lipid-based nanoparticle aimed at activated immune cells which comprises a mRNA molecule encoding proteins enhancing the activity of the immune cells.
  • the invention ensures a specific and localized delivery of mRNA encoding activityenhancing protein to particular immune cells populations, for example located in a tumor microenvironment, for a maximal efficiency with limited or no systemic side effects.
  • the immune cell enhancing protein can therefore be selected even among proteins exhibiting severe side effects, as their action is circumscribed to a precise location, and the protein will not be expressed systemically and causes off-target side effects.
  • the invention also allows the person skilled in the art to easily adjust the mRNA cargo to replace or combine the effect of different immune cells activating proteins, to tailor the invention according to the need of the patient, without the necessity to assess and prove the safety of the delivery lipid-based nanoparticle.
  • the inventors provide herein a method to specifically and selectively target and activate a selected subset of immune cells, instead of using LNP to target tumoral cells to deliver inhibitory compounds or of targeting every cells of a given type and haphazardly activate them, or making them express a specific receptor to activate them upon encountering a given antigen.
  • the target can be selected based on the location of the immune cell harboring it.
  • LAG-3 and PD-1 are over-expressed in the TME of several cancers.
  • Targeting LAG-3 or PD-1 positive immune cells allows to deliver mRNAs to the immune cells located in the most needed location, ensuring a very effective and specific impact.
  • targeting an antigen expressed on numerous cell types with naive or inactivated cells, like CD3 or CD5 does not confer the same level of precision.
  • the protein encoded by the mRNA envisioned herein may be selected among the most potent effectors, it is therefore crucial for the patient’s safety that the risks of off-targets are kept as low as possible, or even purely suppressed.
  • the applicant has developed targeted LNP that are able to target immune cells, but not the whole generic population of immune cells including immune cells that are not activated (notably T cells population, such as patrolling/resting/naive T cells expressing illustrative CD3, CD4, CD5, CD8 markers and present in the general circulation and/or in specific organs not of major interest for the specific targeting of tumors).
  • T cells population such as patrolling/resting/naive T cells expressing illustrative CD3, CD4, CD5, CD8 markers and present in the general circulation and/or in specific organs not of major interest for the specific targeting of tumors.
  • Such specific targeting of activated immune cells will typically allow to avoid a global over-activation with potential side effects of excessive activation and risk of tolerance and a better biodistribution of the injected product.
  • a further aspect of the invention is to provide LNPs with very low uptake by capturing organs, in particular the liver, but also optionally other organs such as the spleen and/or the lungs.
  • organs in particular the liver, but also optionally other organs such as the spleen and/or the lungs.
  • an increased free distribution of the LNPs throughout the body, especially in the blood or other biological fluid, could be achieved, allowing the delivery of the LNPs to the area of interest (e.g., tumor cells and tumor environment).
  • the invention relates to a lipid-based nanoparticle comprising an antigen-binding domain capable of specifically binding to a target expressed on activated immune cells surface and one or several mRNA molecule(s) encoding an activity-enhancing protein of said activated immune cells, wherein the mRNA molecule encodes i) an intracellular protein having an intracellular effect on the activated immune cell and/or ii) a transmembrane protein that is not a chimeric associated receptor (CAR).
  • CAR chimeric associated receptor
  • the invention concerns a lipid-based nanoparticle comprising an antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface and one or several mRNA molecule(s) encoding an activity-enhancing protein of said activated immune cells, wherein the activity-enhancing protein is i) an intracellular protein having an intracellular effect on the activated immune cell or ii) a transmembrane protein that is not a chimeric associated receptor, wherein the antigen binding domain is an antibody or an antigen binding fragment thereof.
  • the lipid-based nanoparticle comprises at least two mRNA molecules, wherein one of said at least two mRNA molecules encodes a transmembrane protein that is a receptor and another of said at least two mRNA molecules encodes a secreted protein that is a ligand of said receptor.
  • the lipid-based nanoparticle comprises at least two mRNA molecules, wherein the first mRNA molecule encodes for IL-7 and the second a mRNA molecule encodes for IL-7R.
  • the activated immune cells are selected from the group consisting of activated T cells, activated B cells, activated myeloid cells activated macrophages and activated dendritic cells, preferably exhausted T cells, tumor infiltrating lymphocytes (TIL) and effector memory stem like T cells.
  • activated T cells activated B cells
  • activated myeloid cells activated macrophages
  • activated dendritic cells preferably exhausted T cells, tumor infiltrating lymphocytes (TIL) and effector memory stem like T cells.
  • the activated immune cells is an activated T cell.
  • the target expressed on activated immune cells surface is selected from the group consisting of BCMA/TNFRSF17, BTLA, CD101/IGSF2, CD103, CD119, CD137/4- 1BB/TNFRSF9, CD150, CD153, CD154, CD223, CD226, CD25, CD254, CD26, CD27, CD275, CD39/ENTPD1, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, GPR35, FPR2, CD80, CD83, CD86, CD95, CMKLR1, CRTAM, CST7, CTLA4, CXCR3, CXCR4, CXCR5, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, TIM3/HAVCR2, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LAG3, TRAILR, OX40L, LY108 /SlamF6, NKG2D, OX40/TNFR
  • TNFRSF 12A/FN14/TWEAKR B AFFR/TNFRSF 13 C/CD268,
  • TNFRSF 19/TROY TNFRSF21/DR6, TNFRSF25/DR3 /TNFRSF 12, CD301, IL4R, CLEC- 1A, CD21, CLEC-9A, CD 180, CD59, CD54, CD71, CD35, CD218a, CD74, CD 165, 4- 1BBL/CD137L, ICOSL and CD160.
  • the target expressed on activated immune cells surface is selected from the group consisting of BCMA/TNFRSF17, BTLA, CD101/IGSF2, CD103, CD119, CD137/4- 1BB/TNFRSF9, CD150, CD153, CD154, CD223, CD226, CD25, CD254, CD26, CD27, CD275, CD30, CD39/ENTPD1, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, GPR35, FPR2, CD80, CD83, CD86, CD95, CMKLR1, CRTAM, CST7, CTLA4, CXCR3, CXCR4, CXCR5, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, TIM3/HAVCR2, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LAG3, TRAILR, OX40L, LY108 /SlamF6, NKG2D, OX40/
  • TNFRSF 12A/FN14/TWEAKR B AFFR/TNFRSF 13 C/CD268,
  • TNFRSF 19/TROY TNFRSF21/DR6, TNFRSF25/DR3 /TNFRSF 12, CD301, IL4R, CLEC- 1A, CD21, CLEC-9A, CD 180, CD59, CD54, CD71, CD35, CD218a, CD74, CD 165, 4- 1BBL/CD137L, ICOSL, CD127, SIRPa and CD160.
  • the activated immune cells are tumor infiltrating lymphocytes (TIL) and the target expressed on activated immune cells surface is selected from the group comprising CD101, CD137 (Tnfrsf9/4-lBBL), CRTAM, CST7, CTLA4, CXCR3, FAS, IL18R1/CXCR1/CD218A, LAG-3 PTPN22, RGS1, TNFSF14 and PD1.
  • TIL tumor infiltrating lymphocytes
  • the antigen binding domain binds to a target selected from the group consisting of PD-1, CD 127, SIRPa and CLEC-1A.
  • the target expressed on activated immune cells surface is PD-1.
  • the antigen binding domain is an anti-PD-1 binding domain comprising: a) (i) a VH comprising HCDR1, HCDR2 and HCDR3, and (ii) a VL comprising LCDR1, LCDR2 and LCDR3, wherein:
  • HCDR1 the heavy chain CDR1 (HCDR1) comprises or consists of an amino acid sequence of SEQ ID NO: 1;
  • HCDR2 the heavy chain CDR2 (HCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 2;
  • HCDR3 the heavy chain CDR3 (HCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 3;
  • LCDR1 the light chain CDR1 (LCDR1) comprises or consists of an amino acid sequence of SEQ ID NO: 4;
  • LCDR2 the light chain CDR2 (LCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 5, and
  • LCDR3 the light chain CDR3 (LCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 6; or b) i) a VH comprising or consisting of an amino acid sequence of SEQ ID NO: 15; and ii) a VL comprising or consisting of an amino acid sequence of SEQ ID NO: 16.
  • the antigen binding domain is an anti-PD-1 binding domain comprising: a) (i) a VH comprising HCDR1, HCDR2 and HCDR3, and (ii) a VL comprising LCDR1, LCDR2 and LCDR3, wherein:
  • HCDR1 the heavy chain CDR1 (HCDR1) comprises or consists of an amino acid sequence of SEQ ID NO: 31;
  • HCDR2 comprises or consists of an amino acid sequence of SEQ ID NO: 32
  • HCDR3 comprises or consists of an amino acid sequence of SEQ ID NO: 33;
  • LCDR1 the light chain CDR1 (LCDR1) comprises or consists of an amino acid sequence of SEQ ID NO: 34;
  • LCDR2 the light chain CDR2 (LCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 35, and
  • LCDR3 the light chain CDR3 (LCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 36; or b) i) a VH comprising or consisting of an amino acid sequence of SEQ ID NO: 37; and ii) a VL comprising or consisting of an amino acid sequence of SEQ ID NO: 38.
  • the antigen binding domain is an anti-PD-1 binding domain comprising: a) (i) a VH comprising HCDR1, HCDR2 and HCDR3, and (ii) a VL comprising LCDR1, LCDR2 and LCDR3, wherein:
  • HCDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 23;
  • HCDR2 the heavy chain CDR2 (HCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 24;
  • HCDR3 the heavy chain CDR3 (HCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 25;
  • LCDR1 the light chain CDR1 (LCDR1) comprises or consists of an amino acid sequence of SEQ ID NO: 26;
  • LCDR2 the light chain CDR2 (LCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 27, and
  • LCDR3 the light chain CDR3 (LCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 28; or b) i) a VH comprising or consisting of an amino acid sequence of SEQ ID NO: 29; and ii) a VL comprising or consisting of an amino acid sequence of SEQ ID NO: 30.
  • the antigen binding domain comprises a Fc domain, preferably an IgG Fc domain.
  • the antigen binding domain is an antibody that comprises an IgG Fc domain and/or that the antigen binding domain comprises an antigen binding fragment of an antibody (such as a Fab or scFv) that is covalently linked to an IgG Fc domain.
  • the antigen binding domain is not covalently bound to any of the lipids of the lipid-based nanoparticle or does not comprise any modification for coupling or grafting the antigen binding domain to a lipid.
  • the lipid-based nanoparticle does not comprise an anchoring moiety comprising a lipidation peptide or motif.
  • the lipid-based nanoparticle comprises an additional antigen binding domain capable of specifically binding to another target expressed on activated immune cells surface.
  • the additional antigen binding domain i) is not covalently bound to any of the lipids of the lipid-based nanoparticle, ii) does not comprise any modification for coupling or grafting the antigen binding domain to a lipid and/or iii) is not covalently bound to a lipidation peptide or motif.
  • the mRNA molecule encodes an intracellular protein having an intracellular effect on the activated immune cells selected from the group consisting of a cytoplasmic protein, an intracellular signaling protein, an enzyme, a transcription factor, an intrabody, a dominant negative receptor or an engineered protein.
  • the activity-enhancing protein is selected from the group consisting of: an enzyme, a cytokine receptor, a chemokine receptor, a lectin receptor, an anchored membrane cytokine, a co-stimulation receptor or ligand, a transcription factor, an intrabody or a dominant negative receptor.
  • the activity-enhancing protein is selected from the group consisting of: TCF1, LEF1, WNT, FRIZZLED, Beta catenin, LRP6, CYCLIN, TOP2A, MUCL1, MDM2, BCL2, BCLXL, BIRC3, MCL1, PGCla, TCF7, NF AT, NFKB, RORgt, TBET, EOMES, RUNX3, GATA3, JUNB, POU2AF1, OCTI, BLIMP-1, XBP-1, FOXO1, PTGS2, CSE, Glutl, Glut3, HK2, FOXO1, arginine resynthesis enzymes, argininosuccinate synthase (ASS), Ornithine transcarbamylase (OTC), GYS, AKT, PLC, SMAD, Blys, BTK, BLK, CD 107a, Lymphotoxin (LT) aip2, granzyme B, perforin, POU2F1, BBS
  • the mRNA molecule encodes an intracellular protein having an intracellular effect on the activated immune cell selected from the group consisting of TCF1, BCL2, IL7R, CXCL9 or CXCL10.
  • the mRNA molecule comprises a modified nucleotide, preferably selected from the group comprising alternative uracils, alternative cytosine, alternative guanine and alternative adenine, preferably selected from the group comprising pseudouridine, 1- methylpseudouridine, 1-ethyl pseudouridine, 2-thiouridine, 4’-thiouridine, 5- methyl cytosine, 2-thio-l -methyl- 1-deaza-pseudouri dine, 2-thio-l-methyl-pseudouridine, 2- thio-5-aza-uridine, 2-thio-dihydropseudouridine, 2-thio-dihydrouridine, 2-thio- pseudouridine, 4-methoxy-2-thio-pseudouridine, 4-methoxy -pseudouridine, 4-thio-l- methyl- pseudouridine, 4-thio-pseudouridine, 5-aza-uridine, dihydropseudourour,
  • the mRNA molecule comprises a 5’UTR, a 5' cap structure, a Kozak sequence, an IRES sequence, a chain terminating nucleotide, a stem loop, a 3’UTR, a poly A sequence and/or a polyadenylation signal.
  • the lipid-based composition of the lipid based nanoparticle comprises or consists of a cationic or ionizable lipid, a helper lipid, a sterol and a PEG-lipid.
  • the ionizable lipid of the lipid based nanoparticle is preferably selected from the group consisting of [(4-hydroxybutyl)azanediyl]di(hexane-6,l-diyl) bis(2-hexyldecanoate) (ALC- 0315), l,2-dioleoyl-3-trimethylammonium propane (DOTAP); N,N-dimethyl-2,3- di oleyloxypropylamine (DODMA), l,2-di-O-octadecenyl-3 -trimethylammoniumpropane (DOTMA), 3-(N-(N',N'-dimethylaminoethane)-carbamoyl)cholesterol (DC-Chol), dimethyldioctadecylammonium (DDAB); l,2-dioleoyl-3 -dimethylammonium -propane (DODAP); l,2-di
  • the sterol of the lipid based nanoparticle is preferably selected from the group consisting of cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, ursolic acid, alpha- tocopherol, and any mixtures thereof, preferably is cholesterol.
  • the helper lipid of the lipid based nanoparticle is preferably selected from DOPE, DOPS, DODMA, DOTAP, DODAP, DDAB, POPE, DSPC, DEPC, DOPC and DSPE, preferably is DOPE or DSPC.
  • the PEG-lipid of the lipid based nanoparticle is preferably selected from PEG-DMG, PEG- DSPE, PEG-c- DOMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, PEG-DPPE, PEG-DAG and PEG-c-DMA, ALC-0159, and any mixture thereof preferably is PEG-DMG, PEG-DSPE or a mixture thereof.
  • the PEG is of between 2000 Daltons and 5000 Daltons, preferably is DSPE- PEG-2000, DMG-PEG-2000, DSPE-PEG-5000, DMG-PEG-5000 or a mixture thereof
  • the lipid-based composition of the lipid-based nanoparticle is preferably selected from the group consisting of: a) ALC-0315, DOPE, cholesterol and DMG-PEG; b) ALC-0315, DDAB, cholesterol and DMG-PEG; c) ALC-0315, POPE, cholesterol and DMG-PEG; d) ALC-0315, DOPE, cholesterol and DSPE-PEG; e) ALC-0315, DSPC, cholesterol and DMG-PEG; f) ALC-0315, DSPC, cholesterol and ALC-0159; g) SM- 102, DSPC, cholesterol and DMG-PEG; h) Dlin-MC3-DMA, DSPC, cholesterol and DMG-PEG;
  • the lipid-based nanoparticle typically comprises from about 35 mol % to about 55 mol % of a cationic or ionizable lipid, from about 5 mol% to about 20 mol % of a helper lipid, from about 30 mol% to about 60 mol% of a sterol, and from about 0.5 mol% to about 4 mol% of a PEG-lipid.
  • the lipid-based nanoparticle comprises from about 45 mol % to about 55 mol % of a cationic or ionizable lipid, from about 5 mol% to about 15 mol % of a helper lipid from about 35 mol% to about 45 mol% of a sterol, and from about 0.5 mol% to about 2.5 mol% of a PEG-lipid.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one lipid- based nanoparticle according to the present invention and optionally a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutical composition further comprises an additional lipid-based nanoparticle comprising an antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface and comprising one or several mRNA molecule(s) encoding an immune cell activity enhancing protein.
  • the invention also relates to the lipid-based nanoparticle according to the present invention, or the pharmaceutical composition according to the present invention, for use as a medicament.
  • the lipid-based nanoparticle or the pharmaceutical composition are for use in the treatment of a disease in a subject in need thereof, wherein the disease is selected from the group consisting of a cancer, an infectious disease and a chronic viral infection; preferably selected from the group comprising metastatic or not metastatic, Melanoma, malignant mesothelioma, Non-Small Cell Lung Cancer, Renal Cell Carcinoma, Hodgkin's Lymphoma, Head and Neck Cancer, Urothelial Carcinoma, Colorectal Cancer, Hepatocellular Carcinoma, Small Cell Lung Cancer Metastatic Merkel Cell Carcinoma, Gastric or Gastroesophageal cancers and Cervical Cancer, Kaposi sarcoma (e.g., associated with Kaposi sarcoma herpes virus); cervical, anal, penile and vulvar squamous cell cancer and oropharyngeal cancers (e.g., associated with human papilloma virus); B cell non
  • the present invention also relates to the use of the lipid-based nanoparticle or the pharmaceutical composition according to the present invention for the manufacture of a medicament for the treatment of a disease as listed herein in a subject in need thereof. It further relates to a method for treating a disease as listed herein in a subject in need thereof comprising administering a therapeutic effective amount of the lipid-based nanoparticle or the pharmaceutical composition according to the present invention to said patient.
  • the cancer to be treated is selected from the group consisting of metastatic or not metastatic, Melanoma, malignant mesothelioma, Non-Small Cell Lung Cancer, Renal Cell Carcinoma, Hodgkin's Lymphoma, Head and Neck Cancer, Urothelial Carcinoma, Colorectal Cancer, Hepatocellular Carcinoma, Small Cell Lung Cancer, Metastatic Merkel Cell Carcinoma, Gastric or Gastroesophageal cancers and Cervical Cancer.
  • metastatic or not metastatic Melanoma
  • Melanoma malignant mesothelioma
  • Non-Small Cell Lung Cancer Renal Cell Carcinoma
  • Hodgkin's Lymphoma Head and Neck Cancer
  • Urothelial Carcinoma Colorectal Cancer
  • Hepatocellular Carcinoma Small Cell Lung Cancer
  • Metastatic Merkel Cell Carcinoma Gastric or Gastroesophageal cancers and Cervical Cancer.
  • the subject suffers from cancer and has a primary or secondary resistance to an immune checkpoint inhibitor, preferably a primary or secondary resistance to an antiprogrammed cell death 1 (PD-1) inhibitor, an anti -programmed cell death 1 ligand 1 (PD- Ll) inhibitor, or a combination of an anti-PDl inhibitor and an anti CTLA-4 inhibitor.
  • an immune checkpoint inhibitor preferably a primary or secondary resistance to an antiprogrammed cell death 1 (PD-1) inhibitor, an anti -programmed cell death 1 ligand 1 (PD- Ll) inhibitor, or a combination of an anti-PDl inhibitor and an anti CTLA-4 inhibitor.
  • the invention also relates to an in vitro method for enhancing immune cells activity, comprising the step of contacting activated immune cells with a lipid-based nanoparticle according to the present invention or with the pharmaceutical composition according to the present invention.
  • antibody describes a type of immunoglobulin molecule and is used in its broadest sense.
  • antibodies include immunoglobulin molecules and immunologically active fragments of immunoglobulin molecules, i.e., molecules that contain an antigen binding site.
  • Immunoglobulin molecules can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2) or subclass.
  • the heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively.
  • antibody particularly refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding domain that specifically binds an antigen.
  • antibody encompasses whole antibody molecules such as four-chain antibodies comprising 2 heavy chains and 2 light chains, such as polyclonal antibodies, monoclonal antibodies or recombinant antibodies, but also any antibody fragments thereof that comprise an antigen binding domain.
  • antigen-binding fragment refers to one or more fragments or derivatives of an antibody that retain the ability to specifically bind to an antigen (e.g., PD-1).
  • antigen e.g., PD-1
  • binding fragments encompassed within the term "antigen-binding fragment" of an antibody include a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CHI domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a dAb fragment (Ward et al., 1989 Nature 341 :544-546), which consists of a VH domain, or any fusion proteins comprising such antigen-binding fragments.
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single chain protein in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al., 1988 Science 242:423-426; and Huston et al., 1988 Proc. Natl. Acad. Sci. 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody.
  • isolated indicates that the recited material (e.g., antibody, polypeptide, nucleic acid, etc.) is substantially separated from, or enriched relative to, other materials with which it occurs in nature.
  • an “isolated” antibody is one which has been identified and separated and/or recovered from a component of its natural environment.
  • treatment refers to any act intended to ameliorate the health status of patients such as therapy, prevention, prophylaxis and retardation of the disease.
  • such term refers to the amelioration or eradication of a disease or symptoms associated with a disease, such as according to the present disclosure, the disruption or the delay in the resolution of the inflammation leading to inflammation associated disease.
  • this term refers to minimizing the spread or worsening of the disease resulting from the administration of one or more therapeutic agents to a subject with such a disease.
  • sequence identity between two sequences is described by the parameter “sequence identity”, “sequence similarity” or “sequence homology”.
  • sequence identity the percent identity between two sequences (A) and (B) is determined by comparing the two sequences aligned in an optimal manner, through a window of comparison.
  • Said alignment of sequences can be carried out by well- known methods in the art, for example, using the algorithm for global alignment of Needleman-Wunsch. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions, and other modifications, including conservative amino acid substitutions. Once the total alignment is obtained, the percentage of identity can be obtained by dividing the full number of identical amino acid residues aligned by the full number of residues contained in the longest sequence between the sequence (A) and (B). Sequence identity is typically determined using sequence analysis software. For comparing two amino acid sequences, one can use, for example, the tool “Emboss needle” for pairwise sequence alignment of proteins providing by EMBL-EBI and available on: www.ebi.ac.
  • the percent identity between two amino acid sequences or nucleotide sequences can also be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4:11- 17, 1988) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two amino acid sequences or nucleotide sequences can be determined using the Needleman and Wunsch (J. Mol, Biol.
  • sequence identity can also be typically determined using sequence analysis software Clustal Omega using the HHalign algorithm and its default settings as its core alignment engine.
  • the algorithm is described in Sbding, J. (2005) 'Protein homology detection by HMM-HMM comparison'. Bioinformatics 21, 951-960, with the default settings.
  • Eu numbering also known as Eu index refers to the antibody numbering system (Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda), which is based on the sequential numbering of the first human IgGl sequenced (the Eu antibody; Edelman, et al., 1969, Proc Natl Acad Sci USA 63: 78-85).
  • amino acid change or “amino acid modification” is meant herein a change in the amino acid sequence of a polypeptide.
  • amino acid modifications include substitution, insertion and/or deletion in a polypeptide sequence.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with another amino acid.
  • amino acid insertion or “insertion” is meant the addition of an amino acid at a particular position in a parent polypeptide sequence.
  • amino acid deletion or “deletion” is meant the removal of an amino acid at a particular position in a parent polypeptide sequence. The amino acid substitutions may be conservative.
  • amino acid position or “amino acid position number” are used interchangeably and refer to the position of a particular amino acid in an amino acids sequence, generally specified with the one letter codes for the amino acids.
  • the first amino acid in the amino acids sequence i.e., starting from the N terminus
  • a conservative substitution is the replacement of a given amino acid residue by another residue having a side chain (“R-group”) with similar chemical properties (e.g., charge, bulk and/or hydrophobicity).
  • R-group residue having a side chain
  • a conservative amino acid substitution will not substantially change the functional properties of a protein.
  • Conservative substitutions and the corresponding rules are well-described in the state of the art. For instance, conservative substitutions can be defined by substitutions within the groups of amino acids reflected in the following tables:
  • a or “an” can refer to one of or a plurality of the elements it modifies (e.g., “a reagent” can mean one or more reagents) unless it is contextually clear either one of the elements or more than one of the elements is described.
  • the biological sequence consists of that sequence, but it may also include 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 substitutions, additions, deletions or a mixture thereof, preferably 1, 2, 3, 4, or 5 substitutions, additions, deletions or a mixture thereof, with the proviso that said biological sequence varies from the reference sequence contained in the sequence listing by up to 10% of the biological sequence length.
  • the present disclosure relates to a lipid-based nanoparticle comprising an antigen-binding domain capable of specifically binding to a target expressed on activated immune cells surface and one or several mRNA molecule(s) encoding an activity-enhancing protein of said activated immune cells.
  • the invention particularly concerns a lipid-based nanoparticle comprising an antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface and one or several mRNA molecule(s) encoding an activity-inhibiting protein of said activated immune cells, wherein the activity-inhibiting protein is i) an intracellular protein having an intracellular effect on the activated immune cell and/or ii) a transmembrane protein, wherein the antigen binding domain is an antibody or an antigen binding fragment thereof.
  • the lipid-based nanoparticle according to the invention is particularly formulated either as a liposome or a lipid nanoparticle (LNP), especially a lipid nanoparticle comprising a mixture of lipids.
  • LNP lipid nanoparticle
  • the lipid-based nanoparticle also encompasses similar nanoparticles such as but not limited to micelles and nano-emulsions.
  • Lipid based nanoparticles also include Hybrid nanoparticles comprising polymers-lipids hybrid compounds, such as polamines-polaxamers, in particular as described herein.
  • the lipid-based nanoparticle according to the invention is preferably a t-LNP.
  • t-LNP refers to a targeted lipid nanoparticle, i.e., a lipid nanoparticle comprising an antigen binding domain.
  • nt-LNP refers to a lipid nanoparticle devoid of antigen binding domain.
  • Lipid-based nanoparticles of the invention typically comprise helper lipid, sterol and/or PEG lipid components along with the mRNA of interest.
  • the elements of a LNP may be selected based on a particular application or target, and/or based on the efficacy, toxicity, expense, ease of use, availability, or other feature of one or more elements.
  • the particular formulation of a lipid-based nanoparticle may be selected for a particular application or target according to, for example, the efficacy and toxicity of particular combination of elements.
  • the lipid-based nanoparticles of the disclosure can particularly be generated using components, compositions, and methods as generally known in the art, for example such as disclosed in WO2017049245; WO2017112865; WO2017218704; WO2015164674; WO2017031232; WO2017099823; WO2016118724; WO2016118724; WO2017223135; WO2014152211; WO2015038892; W02017049074; W02013090648; W02017180917; WO2017075531 and WO2017117528 all of which are incorporated by reference herein in their entirety.
  • LNPs The manufacture of LNPs is well described in the art, for example in U.S. Patent Application Publication No. US20120276209, Semple et al., 2010, Nat Biotechnol., 28(2): 172-176; Akinc et al., 2010, Mol Ther., 18(7): 1357-1364; Basha et al., 2011, Mol Ther, 19(12): 2186- 2200; Leung et al., 2012, J Phys Chem C Nanomater Interfaces, 116(34): 18440-18450; Lee et al., 2012, Int J Cancer., 131(5): E781-90; Belliveau et al., 2012, Mol Ther nucleic Acids, 1 : e37; Jayaraman et al., 2012, Angew Chem Int Ed Engl., 51(34): 8529-8533; Mui et al., 2013, Mol Ther Nucleic Acids. 2, el39; Maier
  • the method for obtaining the lipid-based nanoparticles of the invention is as described under the “Examples” section below, in particular in Examples 1-3, or as described in Figure 21J-K.
  • the method for obtaining the lipid-based nanoparticles of the invention is as described in PCT/EP2024/058775.
  • the lipid-based nanoparticle comprises one or more ionizable or cationic lipid(s), one or more helper lipid(s), one or more sterol(s), and/or one or more polyethylene glycol (PEG)-modified lipid(s).
  • the lipid-based nanoparticle comprises one or more ionizable or cationic lipid(s) (also referred as component N° 1), one or more helper lipid(s) (also referred as component N° 2), one or more sterol(s) (also referred as component N° 3), and/or one or more polyethylene glycol (PEG)-modified lipid(s) (also referred as component N° 4).
  • ionizable or cationic lipid(s) also referred as component N° 1
  • helper lipid(s) also referred as component N° 2
  • sterol(s) also referred as component N° 3
  • PEG polyethylene glycol
  • the lipid-based nanoparticle according to the invention comprises one or more ionizable or cationic lipid(s).
  • ionizable or cationic lipid refers to a lipid molecule positively charged in an acidic environment.
  • Ionizable or cationic lipids particularly promotes nucleic acid molecule(s) delivery and transfection efficiency. Their mechanism of action is based on complexing the nucleic acid by electrostatic interactions. Several properties, such as the charge or lipid shape, as well as the protein corona formation, have been described as important factors to consider when understanding structure-activity relationship studies, and thus, the design of new ionizable lipids.
  • the ionizable or cationic lipid comprises a head group which includes at least one nitrogen atom (N) which is positively charged or capable of being protonated.
  • the ionizable or cationic lipid is selected from the group consisting of [(4- hydroxybutyl)azanediyl]di(hexane-6,l-diyl) bis(2 -hexyldecanoate) (ALC-0315), 1,2- dioleoyl-3 -trimethylammonium propane (DOTAP); N,N-dimethyl-2,3- di oleyloxypropylamine (DODMA), l,2-di-O-octadecenyl-3 -trimethylammoniumpropane (DOTMA), 3-(N-(N',N'-dimethylaminoethane)-carbamoyl)cholesterol (DC-Chol), dimethyldioctadecylammonium (DDAB); l,2-dioleoyl-3 -dimethylammonium -propane (DODAP); l,2-diacyloxy
  • Ionizable or cationic lipids can be selected from the group consisting of l,2-dioleoyl-3- trimethylammonium propane (DOTAP); N,N-dimethyl-2,3-dioleyloxypropylamine (DODMA), l,2-di-O-octadecenyl-3 -trimethylammonium propane (DOTMA), 3-(N- (N',N'- dimethylaminoethane)-carbamoyl)cholesterol (DC-Chol), dimethyldioctadecylammonium (DDAB); l,2-dioleoyl-3-dimethylammonium-propane (DODAP); l,2-diacyloxy-3- dimethylammonium propanes; l,2-dialkyloxy-3 -dimethylammonium propanes; dioctadecyldimethyl ammonium chloride (DODAC), l,2-distearyloxy
  • DMOBA l,2-N,N'-dioleylcarbamyl-3 -dimethylaminopropane
  • DOcarbDAP 2,3- Dilinoleoyloxy-N,N-dimethylpropylamine
  • DLincarbDAP 2,3- Dilinoleoyloxy-N,N-dimethylpropylamine
  • DLincarbDAP 2,3- Dilinoleylcarbamyl-3- dimethylaminopropane
  • DLincarbDAP 2,2-Dilinoleoylcarbamyl-3 -dimethylaminopropane
  • DLinCDAP 2,2-dilinoleyl-4-dimethylaminomethyl-[l,3]-dioxolane
  • DLin-K-DMA 2,2- dilinoleyl-4-dimethylaminoethyl-[l,3]-di oxolane
  • ionizable or cationic lipids are described in WO 2016/021683, WO 2015/011633, WO 2011/153493, WO 2013/126803, WO 2010/054401, WO 2010/042877, WO 2016/104580, WO 2015/005253, WO 2014/007398, WO 2017/117528, WO 2017/075531, WO 2017/00414, WO 2015/199952, US 2015/0239834, WO2019/131839 all of which are incorporated by reference herein in their entirety.
  • synthetic ionizable or cationic lipids e.g., K-E12, H-A12, Y-E12, G-O12, K- A12, R-A12, CKK-E12, cPK-E12, PK1K-E12, PK500-E12, cQK-E12, cKK-A12, KK-A12, PK-4K-E12, CWK-E12, PK500-012, PK1K-O12, cYK-E12, cDK-E12, cSK-E12, cEK-E12, CMK-E12, cKK-012, cIK-E12, cKK-ElO, cKK-E14, and cKK-E16, preferably, cKK-E12, cKK-E14) described in Dong et al.
  • the lipid-based nanoparticle according to the invention comprises an ionizable or cationic lipid such as described in WO 2016/021683 or WO 2019/131839 which are incorporated herein by reference in their entirety.
  • the lipid composition of the lipide-based nanoparticle according to the invention comprises an ionizable or cationic lipid selected from the group consisting of [(4- hydroxybutyl)azanediyl]di(hexane-6,l-diyl) bis(2 -hexyldecanoate) (ALC-0315), heptatriaconta-6,9,28,3 l-tetraen-19-yl-4-(dimethylamino)butanoate (DLin-MC3-DMA), 9- Heptadecanyl 8- ⁇ (2-hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino ⁇ octanoate (SM- 102), and bis[2-(4- ⁇ 2-[4-(cis-9-octadecenoyloxy)phenylacetoxy]ethyl ⁇ piperidinyl)ethyl]
  • the ionizable or cationic lipid is selected from the group consisting of ALC- 0315, SM-102, Dlin-MC3-DMA and SS-OP.
  • the ionizable or cationic lipid comprised in the lipid-based nanoparticle is ALC- 0315.
  • the ionizable or cationic lipid comprised in the lipid-based nanoparticle is SS-OP.
  • the ionizable or cationic lipid represents from about 10 mol% to about 100 mol%, about 20 mol% to about 100 mol%, about 30 mol% to about 100 mol%, about 40 mol% to about 100 mol%, or about 50 mol% to about 100 mol% of the total lipids present in the lipid-based nanoparticle according to the invention.
  • the ionizable or cationic lipid preferably ALC-0315 or SS-OP, represents from about 45 mol % to about 55 mol % of the total lipids present in the lipid composition of the lipid-based nanoparticle according to the invention.
  • the ionizable or cationic lipid preferably ALC-0315 or SS-OP, represents from about 48 mol % to about 52 mol % of the total lipids present in the lipid composition of the lipid-based nanoparticle according to the invention.
  • the ionizable or cationic lipid preferably ALC-0315 or SS-OP, represents about 50 mol % of the total lipids present in the lipid composition of the lipid-based nanoparticle according to the invention.
  • the lipid-based nanoparticle according to the invention comprises a helper lipid.
  • helper lipid refers to a class of lipid molecules that increases particle stability, fluidity tolerability and/or biodistribution of lipid-based nanoparticles.
  • Helper lipids are also constituents of LNPs, playing an important role in terms of stability and fusogenicity. These lipids are mainly phospholipids (such as DOPE, DSPC, DEPC, DSPE), which forms the main skeleton of LNPs. Helper lipids modulate nanoparticle fluidity and enhance efficacy by promoting lipid phase transitions that aid membrane fusion with the endosome. Helper lipids are generally saturated phospholipids, which can increase the phase transition temperature of cationic liposomes, support the formation of lamellar lipid bilayers and stabilize their structural arrangement.
  • phospholipids such as DOPE, DSPC, DEPC, DSPE
  • the helper lipid can be selected from the group consisting of 1,2-distearoyl-sn- glycero-3 -phosphocholine (DSPC), l,2-dilinoleoyl-sn-glycero-3 -phosphocholine (DLPC), 1,2-dimyristoyl-sn-glycero-phosphocholine (DMPC), l,2-dioleoyl-sn-glycero-3- phosphocholine (DOPC), l,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2- diundecanoyl-sn-glycero-phosphocholine (DUPC), l-palmitoyl-2-oleoyl-sn-glycero-3- phosphocholine (POPC), l,2-di-0-octadecenyl-sn-glycero-3 -phosphocholine (18:0 Diether PC), l-ole,
  • PA 1.2-dioleoyl-sn-glycero-3 -phosphate
  • PG l,2-dioleoyl-sn-glycero-3-phospho-(l '-rac- glycerol)
  • PG l,2-dioleoyl-sn-glycero-3 -phosphomethanol
  • 1.2-dioleoyl-sn-glycero-3 -phosphoethanol (18 : 1 Phosphatidyethanol), 1,2-dioleoyl-sn- glycero-3 -phosphopropanol (18 : 1 Phosphatidypropanol), l,2-dioleoyl-sn-glycero-3- phospho-L-serine (18: 1 PS, DOPS), l,2-distearoyl-sn-glycero-3-phospho-L-serine (18:0 PS), N-oleoyl-D-erythro-sphingosine (Ceramide), Sphingomyelin (SM), Phosphatidylinositol (PI), 9A1P9, l,2-dioleoyl-3 -trimethylammonium -propane (DOTAP),
  • DOTMA 1.2-di-O-octadecenyl-3 -trimethylammonium propane
  • helper lipid can be selected from the group consisting of 1,2-distearoyl-sn- glycero-3 -phosphocholine (DSPC), l,2-dilinoleoyl-sn-glycero-3 -phosphocholine (DLPC),
  • DMPC 1.2-dimyristoyl-sn-glycero-phosphocholine
  • DOPC l,2-dioleoyl-sn-glycero-3- phosphocholine
  • DPPC 1,2- diundecanoyl-sn-glycero-phosphocholine
  • POPC l-palmitoyl-2-oleoyl-sn-glycero-3- phosphocholine
  • POPC l,2-di-0-octadecenyl-sn-glycero-3 -phosphocholine (18:0 Diether PC), l-oleoyl-2-cholesterylhemisuccinoyl-sn-glycero-3-phosphocholine (OChemsPC), 1- hexadecyl-sn-glycero-3 -phosphocholine (Cl 6 Lyso PC), l,2-dilin
  • N-(7-nitrobenz-2-oxa-l,3-diazol-4-yl)-phosphatidylethanolamine N-(7-nitrobenz-2-oxa-l,3-diazol-4-yl)-phosphatidylethanolamine (NBD-PE), N-(7-nitrobenz-2-oxa-l,3-diazol-4-yl)-phosphatidylethanolamine (NBD-PE), N-
  • PA 1.2-dioleoyl-sn-glycero-3 -phosphate
  • PG l,2-dioleoyl-sn-glycero-3-phospho-(l'-rac- glycerol)
  • PG l,2-dioleoyl-sn-glycero-3 -phosphomethanol
  • 1.2-dioleoyl-sn-glycero-3 -phosphoethanol (18 : 1 Phosphatidyethanol), 1,2-dioleoyl-sn- glycero-3 -phosphopropanol (18 : 1 Phosphatidypropanol), l,2-dioleoyl-sn-glycero-3- phospho-L-serine (18: 1 PS, DOPS), l,2-distearoyl-sn-glycero-3-phospho-L-serine (18:0 PS), N-oleoyl-D-erythro-sphingosine (Ceramide), Sphingomyelin (SM), Phosphatidylinositol (PI), 9A1P9, l,2-dioleoyl-3 -trimethylammonium -propane (DOTAP),
  • DOTMA 1.2-di-O-octadecenyl-3 -trimethylammonium propane
  • Dimethyldioctadecylammonium (18:0 DDAB), l,2-dioleyloxy-3 -dimethylaminopropane (DODMA), l,2-dioleoyl-3-dimethylammonium-propane (DODAP), 1,2-dierucoyl-sn- glycero-3 -phosphocholine (DEPC), l,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) and any mixtures or combinations thereof.
  • DODMA dioleyloxy-3 -dimethylaminopropane
  • DODAP l,2-dioleoyl-3-dimethylammonium-propane
  • DEPC 1,2-dierucoyl-sn- glycero-3 -phosphocholine
  • DSPE 1,2-distearoyl-sn-glycero-3-phosphoethanolamine
  • the helper lipid is selected from the group consisting of DOPE, DOPS, DODMA, DOTAP, DODAP, DDAB, POPE, DSPC, DOPC, DEPC and DSPE, and any combinations thereof.
  • the helper lipid is selected from the group consisting of from the group consisting of DOPE, DOPC, DDAB, POPE and DSPC, and any combinations thereof.
  • the helper lipid is DOPE.
  • the helper lipid is DSPC.
  • the helper lipid represents from about 5 mol% to about 100 mol%, from about 10 mol% to about 100 mol%, about 20 mol% to about 100 mol%, about 30 mol% to about 100 mol%, about 40 mol% to about 100 mol%, or about 50 mol% to about 100 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • helper lipid preferably DOPE or DSPC, represents from about 5 mol% to about 15 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • helper lipid preferably DOPE or DSPC, represents from about 8 mol% to about 12 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • helper lipid preferably DOPE or DSPC, represents about 10 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the lipid of the lipid-based nanoparticle disclosed herein comprises one or more molecules comprising polyethylene glycol (PEG). Accordingly, the lipid-based nanoparticle may comprise PEG or PEG-modified lipids.
  • PEG polyethylene glycol
  • PEG lipid may refer to polyethylene glycol (PEG)-modified lipids.
  • PEG lipid stabilizes lipid nanoparticles, regulates nanoparticle size by limiting lipid fusion and increases nanoparticle half-life by reducing nonspecific interactions with macrophages, improving colloidal stability and preventing the formation of the protein corona.
  • PEG lipid may refer to polyethylene glycol (PEG) -modified lipids.
  • PEG lipids include PEG-modified phosphatidylethanolamine and phosphatidic acid, PEG-ceramide conjugates (e.g., PEG- CerCI4 or PEG-CerC20), PEG- modified dialkylamines and PEG-modified 1,2- diacyloxypropan-3-amines.
  • PEGylated lipids are also referred to as PEGylated lipids.
  • a PEG lipid can be PEG-c-DOMG, PEG-DMG, PEG-DLPE, PEG-DMPE, PEG- DPPC, or a PEG-DSPE lipid.
  • the PEG-modified lipids are a modified form of PEG-DMG.
  • a PEG lipid may particularly be selected from the non-limiting group including PEG-modified phosphatidylethanolamines, PEG-modified phosphatidic acids, PEG-modified ceramides, PEG-modified dialkylamines, PEG-modified diacylglycerols, PEG-modified dialkylglycerols, and mixtures thereof.
  • a PEG lipid is selected from the group consisting of PEG-c-DOMG, PEG-DMG, DMG-PEG-2000, PEG- DLPE, PEG- DMPE, PEG-DPPC, and PEG-DSPE lipid.
  • the PEG-lipid includes, but is not limited to 1,2-dimyristoyl-sn-glycerol methoxypolyethylene glycol (PEG-DMG), l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), PEG-dipalmetoleyl, PEG-dioleyl, PEG-distearyl, PEG-diacylglycamide (PEGDAG), PEG-dipalmitoyl phosphatidylethanolamine (PEG-DPPE), or PEG- 1,2- dimyristyloxlpropyl-3-amine (PEG-c-DMA).
  • PEG-DMG 1,2-dimyristoyl-sn-glycerol methoxypolyethylene glycol
  • PEG-DSPE l,2-distearoyl-s
  • the PEG-lipid includes, but is not limited to 1,2-dimyristoyl-sn-glycerol methoxypolyethylene glycol (PEG-DMG), l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), PEG-dipalmetoleyl, PEG-dioleyl, PEG-distearyl, PEG-diacylglycamide (PEGDAG), PEG-dipalmitoyl phosphatidylethanolamine (PEG-DPPE), or PEG- 1,2- dimyristyloxlpropyl-3-amine (PEG-c-DMA) and ALC-0159 (N,N-dimyristylamide of 2- hydroxyacetic acid, O-pegylated to a PEG), and any mixture thereof.
  • the PEG-lipid is selected from the group consisting of a PEG-modified phosphatidylethanolamine, a PEG-modified phosphatidic acid, a PEG-modified ceramide, a PEG-modified dialkylamine, a PEG-modified diacylglycerol, a PEG-modified dialkylglycerol, and mixtures thereof.
  • such PEG is selected from the group consisting of 1,2-dimyristoyl-sn-glycerol methoxypolyethylene glycol (PEG-DMG), l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), PEG-dipalmetoleyl, PEG-dioleyl, PEG-distearyl, PEG-diacylglycamide (PEGDAG), PEG-dipalmitoyl phosphatidylethanolamine (PEG-DPPE), or PEG- 1,2- dimyristyloxlpropyl-3-amine (PEG-c-DMA), PEG-c-DOMG, PEG-DMG, DMG-PEG- 2000, PEG-DLPE, PEG- DMPE, PEG-DPPC, and PEG-DSPE.
  • the PEG-lipid is selected from the group consisting of PEG-DMG, PEG-DSPE, PEG-c- DOMG, PEG-DLPE, PEG-DMPE, PEG-DPPC, PEG-DPPE, PEG-DAG and PEG- c-DMA, ALC-0159 (N,N-dimyristylamide of 2-hydroxyacetic acid, O-pegylated to a PEG), and any mixture thereof; particularly from the group consisting of PEG-DMG, PEG-DSPE, ALC-0159 and any mixture thereof.
  • the PEG is comprised between 2000 Daltons and 5000 Daltons (i.e., PEG-2000 to PEG-5000), preferably is DSPE-PEG-2000, DMG-PEG-2000, DSPE-PEG-5000, DMG- PEG-5000 or any mixture thereof.
  • the LNP comprises a PEG that is not functionalized, which is a PEG that does not comprises any reactive species at its end, said reactive species being usable to conjugate a target moiety such as an antibody or a fragment thereof to the PEG.
  • the lipid moiety of the PEG-lipids includes those having lengths of from about C14 to about C22, preferably from about C14 to about C16.
  • the PEG-lipid has an impact on the biodistribution of the LNPs.
  • the choice of the PEG-lipid can modify the uptake of the LNPs by the capturing organs (mostly liver, and optionally spleen and/or lungs). For instance, a PEG-C14 lipid seems to favor liver uptake whereas a PEG-C18 lipid decreases liver uptake and may optionally favor spleen targeting.
  • the PEG-lipid and the amount thereof in the LNP can be selected so as favoring free distribution of the LNP throughout the circulation of the body, in particular in direction of an area of interest for a specific targeted therapeutic treatment.
  • the PEG-lipid and the amount thereof in the LNP can be selected so as to be sufficiently free to target immune cells and tumoral cells, especially in the tumor microenvironment.
  • targeted LNP carrying targeting ligand such as antibodies are designed to escape the uptake by organs and target activated immune cells (such as PD1+ T lymphocytes) located within the tumor micro-environment.
  • target activated immune cells such as PD1+ T lymphocytes located within the tumor micro-environment.
  • the RNA carried by the LNP and transfected into said activated immune cells allows to reinforce their anti-tumoral activity locally and specifically.
  • the lipid, in particular the lipid moiety of the PEG- lipids includes those having a length of from about C16 to about C22 (C16, Cl 7, Cl 8, Cl 9, C20, C21 or C22), preferably C16 to C20 (C16, C17, C18, C19 or C20), especially Cl 8.
  • PEG lipid can be selected in the group consisting of l,2-distearoyl-sn-glycero-3- phosphoethanolamine-N-[amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), PEG-dipalmetoleyl, PEG-dioleyl, PEG-distearyl, PEG-diacylglycamide (PEGDAG), and PEG-dipalmitoyl phosphatidylethanolamine (PEG-DPPE), optionally in the group consisting of l,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- [amino(polyethylene glycol)] (PEG-DSPE), PEG-disteryl glycerol (PEG-DSG), and PEG- dioleyl, PEG-distearyl.
  • the PEG-lipid is PEG-d
  • the PEG may present a molecular weight within the range from 0.5 to 50 kD, more preferably from 1 to 20 kD.
  • a PEG moiety for example a mPEG-NH2
  • the PEG has a size of between 2000 Daltons and 5000 Daltons.
  • the PEG is selected from the group consisting of PEG-2000, PEG-2500, PEG-3000, PEG-3500, PEG-4000, PEG-4500 and PEG-5000.
  • the PEG has a size of about 2000.
  • the PEG has a size of about 5000 Daltons.
  • the PEG lipid is selected from the group consisting of DSPE-PEG-2000, DMG-PEG-2000, DSPE-PEG-5000, DMG-PEG-5000 or a mixture thereof.
  • the PEG-lipid is PEG 5000-DMG. In another very particular aspect, the PEG-lipid is PEG 5000-DSG.
  • the PEG-lipid is PEG 2000-DMG. In another very particular aspect, the PEG-lipid is PEG 2000-DSG. In another very particular aspect, the PEG-lipid is ALC-0159. In some aspects, the PEG lipid represents from about 1 mol% to about 100 mol%, from about 2 mol% to about 100 mol%, about 3 mol% to about 100 mol%, about 4 mol% to about 100 mol%, about 5 mol% to about 100 mol%, about 10 mol% to about 100 mol%, or about 15 mol% to about 100 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the PEG lipid in the lipid-based nanoparticle is within the range from about 0.5 mol% to about 2 mol% of the total lipids present in the nanoparticle, for instance about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mol%, especially 1.5 mol%.
  • the inventors have surprisingly observed that the amount of PEG lipid has an impact of the biodistribution of the LNPs. It appears that a lower amount of PEG lipid may decrease the uptake of LNPs by capturing organs, especially liver but also spleen.
  • the PEG lipid in said lipid-based nanoparticle could be less than 1.5 mol% of the total lipids present in the nanoparticle, in particular, less than 1.4, 1.3, 1.2, 1.1, 1.0, 0.9, 0.8, 0.7, 0.6 or 0.5 mol% of the total lipids present in the nanoparticle.
  • the PEG lipid preferably PEG-DMG or PEG DSPE, represents from about 0.5 mol% to about 5 mol% or from about 0.5 mol% to about 2.5 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the PEG lipid preferably PEG-DMG or PEG DSPE, represents from about 1 mol% to about 2 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the PEG lipid preferably PEG-DMG or PEG DSPE, represents about 1.5 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the lipid-based nanoparticle of the invention comprises one or more sterol.
  • the sterol can particularly be selected from the group consisting of cholesterol, fecosterol, sitosterol, ergosterol, campesterol, stigmasterol, brassicasterol, tomatidine, ursolic acid, alpha-tocopherol, and any mixtures thereof.
  • the sterol is cholesterol.
  • Sterol more so cholesterol, enhances the stability of the nanoparticles by filling gaps between lipids, and aids fusion with the endosomal membrane during uptake into the cell.
  • the sterol represents from about 10 mol% to about 100 mol%, from about 20 mol% to about 100 mol%, about 30 mol% to about 100 mol%, about 40 mol% to about 100 mol%, about 50 mol% to about 100 mol%, about 60 mol% to about 100 mol%, or about 70 mol% to about 100 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the sterol preferably cholesterol, represents from about 30 mol% to about 50 mol% or from about 35 mol% to about 45 mol% of the total lipids present in the lipid- based nanoparticle of the invention.
  • the sterol preferably cholesterol
  • the sterol represents from about 35 mol% to about 40 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • Such sterol being preferably cholesterol.
  • the sterol preferably cholesterol
  • the sterol represents about 38.5 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • Such sterol being preferably cholesterol.
  • the lipid-based nanoparticle of the invention comprises an ionizable or cationic lipid, a helper lipid, a sterol and a PEG lipid.
  • the lipid composition of the lipid-based nanoparticle of the invention consists of an ionizable or cationic lipid, a helper lipid, a sterol and a PEG lipid, these lipids being preferably as described above.
  • the ionizable or cationic lipid is from about 10 mol % to about 70 mol % of the total lipids present in the nanoparticle
  • the helper lipid is from about 5 mol% to about 70 mol % of the total lipids present in the nanoparticle
  • the sterol is from about 10 mol% to about 70 mol% of the total lipids present in the nanoparticle
  • the PEG lipid is from about 0.5 mol% to about 4 mol% of the total lipids present in the nanoparticle.
  • the lipid-based nanoparticle comprises or consists of from about 35 mol % to about 55 mol % of a cationic or ionizable lipid, from about 5 mol% to about 20 mol % of a helper lipid, from about 30 mol% to about 60 mol% of a sterol, and from about 0.5 mol% to about 4 mol% of a PEG-lipid.
  • the lipid-based nanoparticle comprises or consists of from about 45 mol % to about 55 mol % of a cationic or ionizable lipid, from about 5 mol% to about 15 mol % of a helper lipid from about 35 mol% to about 45 mol% of a sterol, and from about 0.5 mol% to about 2.5 mol% of a PEG-lipid.
  • the PEG lipid in said lipid-based nanoparticle is within the range from about 0.5 mol% to about 2 mol% of the total lipids present in the nanoparticle, for instance about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mol%, especially 1.5 mol%.
  • the inventors have surprisingly observed that the amount of PEG lipid has an impact of the biodistribution of the LNPs. It appears that a lower amount of PEG lipid may decrease the uptake of LNPs by capturing organs, especially liver but also spleen.
  • the PEG lipid in said lipid-based nanoparticle could be less than 1.5 mol% of the total lipids present in the nanoparticle, in particular, less than 1.4, 1.3, 1.2, 1.1, 1.0, 0.9, 0.8, 0.7, 0.6 or 0.5 mol% of the total lipids present in the nanoparticle.
  • the PEG lipid preferably PEG-DMG or PEG DSPE, represents from about 0.5 mol% to about 5 mol% or from about 0.5 mol% to about 2.5 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the PEG lipid preferably PEG-DMG or PEG DSPE, represents from about 1 mol% to about 2 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • the PEG lipid preferably PEG-DMG or PEG DSPE, represents about 1.5 mol% of the total lipids present in the lipid-based nanoparticle of the invention.
  • a polymer may be included in and/or used to encapsulate or partially encapsulate the lipid-based nanoparticle according to the invention.
  • the polymer may be biodegradable and/or biocompatible.
  • the polymer may be selected from, but is not limited to, polyamines, polyethers, polyamides, polyesters, poly carbamates, polyureas, polycarbonates, polystyrenes, polyimides, polysulfones, polyurethanes, polyacetylenes, polyethylenes, polyethyleneimines, polyisocyanates, polyacrylates, polymethacrylates, polyacrylonitriles, and polyarylates.
  • the polymer may include poly(caprolactone) (PCL), ethylene vinyl acetate polymer (EVA), poly(lactic acid) (PLA), poly(L-lactic acid) (PLLA), poly(gly colic acid) (PGA), poly(lactic acid-co-gly colic acid) (PLGA), poly(L-lactic acid-co-gly colic acid) (PLLGA), poly(D,L-lactide) (PDLA), poly(L- lactide) (PLLA), poly(D,L-lactide-co-caprolactone), poly(D,L-lactide-co-caprolactone-co- glycolide), poly(D,L-lactide-co-PEO-co-D,L-lactide), poly(D,L-lactide-co-PPO-co-D,L- lactide), polyalkyl cyanoacrylate, polyurethane, poly-L-lysine (PLL), hydroxypropyl methacrylate (HPMA), poly(C
  • the lipid based particle comprises a poloxamine and/or a poloxamer.
  • the lipid based particle comprises a plyethyleneimine, rotamine and/or polyaspartamide.
  • a surface altering agent may be disposed within a lipid-based nanoparticle of the invention and/or on the surface of the lipid-based nanoparticle (e.g., by coating, adsorption, covalent linkage, or other process).
  • Surface altering agents include, but are not limited to, anionic proteins (e.g., bovine serum albumin), surfactants (e.g., cationic surfactants such as dimethyldioctadecyl-ammonium bromide), sugars or sugar derivatives (e.g., cyclodextrin), nucleic acids, polymers (e.g., heparin, polyethylene glycol, and poloxamer), mucolytic agents (e.g., acetylcysteine, mugwort, bromelain, papain, clerodendrum, bromhexine, carbocisteine, eprazinone, mesna, ambroxol, sobrerol, domiodol, letosteine, stepronin, tiopronin, gelsolin, thymosin b4, domase alfa, neltenexine, and erdosteine), and DNases (e.g.,
  • the lipid-based nanoparticle comprises a lipid mixture.
  • the lipid mixture comprises or consists of an ionizable or cationic lipid, a helper lipid, a sterol and a PEG lipid, these lipids being preferably as described here above.
  • the lipid-based nanoparticle comprises [(4- hydroxybutyl)azanediyl]di(hexane-6,l-diyl) bis(2-hexyldecanoate) (ALC-0315) as ionizable or cationic lipid, l,2-dioleoyl-sn-glycero-3 -phosphoethanolamine (DOPE) as neutral lipid, cholesterol as sterol and one or more polyethylene glycol (PEG)-modified lipid(s).
  • ALC-0315 ionizable or cationic lipid
  • DOPE l,2-dioleoyl-sn-glycero-3 -phosphoethanolamine
  • cholesterol as sterol
  • PEG polyethylene glycol
  • ALC-0315 is from about 35 mol % to about 55 mol % of the total lipids present in the LNP
  • DOPE is from about 5 mol% to about 20 mol % of the total lipids present in the LNP
  • cholesterol is from about 30 mol% to about 60 mol% of the total lipids present in the LNP
  • said one or more polyethylene glycol (PEG)-modified lipid(s) is from about 0.5 mol% to about 4 mol% of the total lipids present in the LNP.
  • said one or more polyethylene glycol (PEG)-modified lipid(s) is from about 0.5 mol% to about 2 mol%, for instance about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mol%, especially 1.5 mol%.
  • said one or more polyethylene glycol (PEG)- modified lipid(s) is from about 0.5 mol% to about 1.5 mol% of the total lipids present in the LNP.
  • said one or more polyethylene glycol (PEG)-modified lipid(s) is from about 0.5 mol% to about 0.6, 0.7, 0.8, 0.9 or 1.0 mol% of the total lipids present in the LNP.
  • the lipid-based nanoparticle comprises [(4- hydroxybutyl)azanediyl]di(hexane-6,l-diyl) bis(2-hexyldecanoate) (ALC-0315) as ionizable or cationic lipid, l,2-dioleoyl-sn-glycero-3 -phosphoethanolamine (DOPE) as neutral lipid, cholesterol as sterol and 1- monom ethoxypoly ethyleneglycol-2, 3- dimyristylglycerol with polyethylene glycol of average molecular weight 2000 (PEG2000- DMG) as PEG-modified lipid.
  • ALC-0315 ionizable or cationic lipid
  • DOPE l,2-dioleoyl-sn-glycero-3 -phosphoethanolamine
  • cholesterol as sterol
  • 1- monom ethoxypoly ethyleneglycol-2 1- monom ethoxypoly ethyleneg
  • ALC-0315 is from about 35 mol % to about 55 mol % of the total lipids present in the LNP
  • DOPE is from about 5 mol% to about 20 mol % of the total lipids present in the LNP
  • cholesterol is from about 30 mol% to about 60 mol% of the total lipids present in the LNP
  • the PEG2000-DMG is from about 0.5 mol% to about 4 mol% of the total lipids present in the LNP.
  • PEG2000-DMG is from about 0.5 mol% to about 2 mol% of the total lipids present in the LNP, for instance about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mol%, especially 1.5 mol%.
  • PEG2000-DMG is from about 0.5 mol% to about 1.5 mol% of the total lipids present in the LNP.
  • PEG2000-DMG is from about 0.5 mol% to about 0.6, 0.7, 0.8, 0.9 or 1.0 mol% of the total lipids present in the LNP, preferably about 0.5 mol%.
  • the lipid-based nanoparticle comprises [(4- hydroxybutyl)azanediyl]di(hexane-6,l-diyl) bis(2-hexyldecanoate) (ALC-0315) as ionizable or cationic lipid, l,2-dioleoyl-sn-glycero-3 -phosphoethanolamine (DOPE) as neutral lipid, cholesterol as sterol and a disteryl glycerol (DSG) with polyethylene glycol of average molecular weight 2000 (PEG2000-DSG) as PEG-modified lipid.
  • ALC-0315 ionizable or lipid
  • DOPE l,2-dioleoyl-sn-glycero-3 -phosphoethanolamine
  • DSG disteryl glycerol
  • PEG2000-DSG polyethylene glycol of average molecular weight 2000
  • ALC-0315 is from about 35 mol % to about 55 mol % of the total lipids present in the LNP
  • DOPE is from about 5 mol% to about 20 mol % of the total lipids present in the LNP
  • cholesterol is from about 30 mol% to about 60 mol% of the total lipids present in the LNP
  • the PEG2000-DSG is from about 0.5 mol% to about 4 mol% of the total lipids present in the LNP.
  • PEG2000-DSG is from about 0.5 mol% to about 2 mol% of the total lipids present in the LNP, for instance about 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0 mol%, especially 1.5 mol%.
  • PEG2000-DSG is from about 0.5 mol% to about 1.5 mol% of the total lipids present in the LNP.
  • PEG2000-DSG is from about 0.5 mol% to about 0.6, 0.7, 0.8, 0.9 or 1.0 mol% of the total lipids present in the LNP, preferably about 0.5 mol%.
  • the lipid composition of the LNP according to the invention comprises:
  • ALC-0315 from about 35 mol % to about 55 mol % of the total lipids present in the LNP
  • PEG 2000-DSG and/or PEG 2000-DMG preferably PEG 2000-DSG, from about 0.5 mol% to about 4 mol% of the total lipids present in the LNP, preferably from about 0.5 mol% to about 1.5 or 2 mol% of the total lipids present in the LNP, optionally from about 0.5 mol% to about 1.0 mol% of the total lipids present in the LNP.
  • the lipid composition of the LNP according to the invention comprises: ALC-0315 from about 35 mol % to about 55 mol % of the total lipids present in the LNP,
  • PEG 2000-DSG and/or PEG 2000-DMG preferably PEG 2000-DSG, from about 0.5 mol% to about 4 mol% of the total lipids present in the LNP, preferably from about 0.5 mol% to about 1.5 or 2 mol% of the total lipids present in the LNP, optionally from about 0.5 mol% to about 1.0 mol% of the total lipids present in the LNP.
  • the lipid-based composition of the lipid-based nanoparticle comprises or consists of a lipid mixture selected from the group consisting of: a) ALC-0315, DOPE, cholesterol and DMG-PEG; b) ALC-0315, DDAB, cholesterol and DMG-PEG; c) ALC-0315, POPE, cholesterol and DMG-PEG; d) ALC-0315, DOPE, cholesterol and DSPE-PEG; e) ALC-0315, DSPC, cholesterol and DMG-PEG; f) ALC-0315, DSPC, cholesterol and ALC-0159; g) SM-102, DSPC, cholesterol and DMG-PEG; h) Dlin-MC3-DMA, DSPC, cholesterol and DMG-PEG; i) ALC-0315, DOPE, cholesterol, DMG-PEG and DSPE-PEG; j) SS-OP, DOPE, cholesterol and DMG-PEG; k)
  • the lipid-based composition comprises or consists of a lipid mixture selected from the group consisting of: a) SS-OP, POPE, cholesterol and DMG-PEG 2000; b) SS-OP, DEPC, cholesterol and DMG-PEG 2000; c) SS-OP, DOPC, cholesterol and DMG-PEG 2000; d) SS-OP, DOPC, cholesterol and DSG-PEG 2000 or 5000; and e) SS-OP, DSPC, cholesterol and DSPE-PEG 2000.
  • the lipid-based composition comprises or consists of the following lipid mixture SS-OP, DSPC, cholesterol and DSPE-PEG, preferably DSG-PEG 2000 or DSG-PEG 5000.
  • the lipid-based composition comprises or consists of a lipid mixture selected from the group consisting of: a) ALC-0315, DOPE, cholesterol and DMG-PEG; b) ALC-0315, DDAB, cholesterol and DMG-PEG; c) ALC-0315, POPE, cholesterol and DMG-PEG; d) ALC-0315, DOPE, cholesterol and DSPE-PEG; e) ALC-0315, DSPC, cholesterol and DMG-PEG; f) ALC-0315, DSPC, cholesterol and ALC-0159; g) ALC-0315, DOPE, cholesterol, DMG-PEG and DSPE-PEG;
  • the PEG has a size of 2000 Daltons (i.e., PEG-2000).
  • the lipid composition of the lipid-based nanoparticle comprises or consists of from about 35 mol % to about 55 mol % of a cationic or ionizable lipid, from about 5 mol% to about 20 mol % of a helper lipid, from about 30 mol% to about 60 mol% of a sterol, and from about 0.5 mol% to about 4 mol% of a PEG-lipid.
  • the lipid composition of the lipid-based nanoparticle comprises or consists of from about 45 mol % to about 55 mol % of a cationic or ionizable lipid, from about 5 mol% to about 15 mol % of a helper lipid from about 35 mol% to about 45 mol% of a sterol, and from about 0.5 mol% to about 2.5 mol% of a PEG-lipid.
  • the lipid composition of the LNP according to the invention comprises:
  • ALC-00315, SM-102, Dlin-MC3-DMA or SS-OP or any mixture thereof from about 45 mol % to about 55 mol 0%, preferably from about 48 mol % to about 52 mol %, more preferably of about 50 mol% of the total lipids present in the LNP,
  • DOPE, DDAB, DOPC, POPE or DSPC or any mixture thereof from about 5 mol% to about 15 mol %, preferably from about 8 mol% to about 12 mol %, more preferably of about 10 mol% of the total lipids present in the LNP, Cholesterol from about 35 mol% to about 45 mol%, preferably from about 37 mol% to about 40 mol %, more preferably of about 38.5 mol% of the total lipids present in the LNP, and
  • the lipid-based composition comprises or consists of:
  • SS-OP from about 35 mol % to about 55 mol % of the total lipids present in the LNP, preferably from about 48 mol % to about 52 mol %, more preferably of about 50 mol% of the total lipids present in the LNP
  • DSPC from about 5 mol% to about 20 mol % of the total lipids present in the LNP, preferably from about 8 mol% to about 12 mol %, more preferably of about 10 mol% of the total lipids present in the LNP,
  • Cholesterol from about 35 mol% to about 45 mol%, preferably from about 37 mol% to about 40 mol %, more preferably of about 38.5 mol% of the total lipids present in the LNP, and
  • PEG 2000-DSPE from about 0.5 mol% to about 2.5 mol%, preferably from about 1 mol% to about 2 mol%, more preferably of about 1.5 mol% of the total lipids present in the LNP.
  • the lipid-based composition comprises or consists of:
  • ALC-0315 from about 35 mol % to about 55 mol % of the total lipids present in the LNP, preferably from about 48 mol % to about 52 mol %, more preferably of about 50 mol% of the total lipids present in the LNP;
  • DOPE from about 5 mol% to about 20 mol % of the total lipids present in the LNP, preferably from about 8 mol% to about 12 mol %, more preferably of about 10 mol% of the total lipids present in the LNP,
  • Cholesterol from about 30 mol% to about 60 mol% of the total lipids present in the LNP, preferably from about 37 mol% to about 40 mol %, more preferably of about 38.5 mol% of the total lipids present in the LNP, and and PEG 2000-DSG and/or PEG 2000-DMG, preferably PEG 2000-DSG, from about 0.5 mol% to about 4 mol% of the total lipids present in the LNP, preferably from about 0.5 mol% to about 1.5 or 2 mol% of the total lipids present in the LNP, optionally from about 0.5 mol% to about 1.5 mol% of the total lipids present in the LNP.
  • the lipid-based nanoparticle according to the invention may also comprise one or more functionalized lipids.
  • a lipid may be functionalized with an alkyne group that, when exposed to an azide under appropriate reaction conditions, may undergo a cycloaddition reaction.
  • a lipid bilayer may be functionalized in this fashion with one or more groups useful in facilitating membrane permeation, cellular recognition, imaging, or for the conjugation of the antigen binding domain to the LNP.
  • the LNP of the invention may optionally comprise one or more coatings.
  • the LNP may be formulated in a capsule, film, or tablet having a coating.
  • a capsule, film, or tablet including a LNP such as described herein may have any useful size, tensile strength, hardness, or density.
  • Lipid-based nanoparticles or a composition comprising LNPs may be characterized by a variety of methods. For example, microscopy (e.g., transmission electron microscopy or scanning electron microscopy) may be used to examine the morphology and size distribution of a LNP or of a composition comprising LNPs. Dynamic light scattering or potentiometry (e.g., potentiometric titrations) may be used to measure zeta potentials. Dynamic light scattering may also be utilized to determine particle sizes.
  • microscopy e.g., transmission electron microscopy or scanning electron microscopy
  • Dynamic light scattering or potentiometry e.g., potentiometric titrations
  • Dynamic light scattering may also be utilized to determine particle sizes.
  • Instruments such as the Zetasizer Nano ZS (Malvern Instruments Ltd, Malvern, Worcestershire, UK) may also be used to measure multiple characteristics of a LNP or of a composition comprising LNPs, such as particle size, polydispersity index, and zeta potential.
  • Physiochemical properties of lipid-based nanoparticles may be altered in order to increase selectivity for particular bodily targets. For instance, particle sizes may be adjusted based on the fenestration sizes of different organs.
  • the mean size of the lipid-based nanoparticle of the invention may be between 10 of nm and 200 of nm, e.g., measured by dynamic light scattering (DLS).
  • the mean size may be from about 40 nm to about 200 nm, such as about 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, 150 nm, 155 nm, 160 nm, 165 nm, 170 nm, 175 nm, 180 nm, 185 nm, 190 nm, 195 nm or 200nm.
  • the mean size of a LNP is from about 50 nm to about 200 nm, from about 50 nm to about 190 nm, from about 50 nm to about 180 nm, from about 50 nm to about 170 nm, from about 50 nm to about 160 nm, from about 60 nm to about 200 nm, from about 60 nm to about 190 nm, from about 60 nm to about 180 nm, from about 60 nm to about 170 nm, from about 70 nm to about 200 nm, from about 70 nm to about 190 nm, from about 70 nm to about 180 nm, from about 80 nm to about 200 nm, from about 80 nm to about 190 nm, or from about 90nm to about 200 nm.
  • the mean size of a LNP is from about 50 nm to about 200 nm, from about 50 nm to about 190 nm, from about 50 nm to about 180 nm, from about 50 nm to about 170 nm, from about 50 nm to about 160 nm, from about 60 nm to about 190 nm, from about 60 nm to about 180 nm, from about 60 nm to about 170 nm, from about 60 nm to about 160 nm, from about 70 nm to about 180 nm, from about 70 nm to about 170 nm, from about 70 nm to about 160 nm, from about 80 nm to about 170 nm, from about 80 nm to about 160 nm, or from about 90nm to about 160 nm.
  • the mean size of a LNP may be from about 70 nm to about 150 nm. In a particular aspect, the mean size is about 120 nm. In other embodiments, the mean size of the lipid- based nanoparticle is about 150 nm.
  • the mean size of the lipid-based nanoparticle of the invention may be between 10 nm and 400 nm, e.g., measured by dynamic light scattering (DLS).
  • the mean size may be from about 40 nm to about 350 nm, such as about 40 nm, 45 nm, 50 nm, 55 nm, 60 nm, 65 nm, 70 nm, 75 nm, 80 nm, 85 nm, 90 nm, 95 nm, 100 nm, 105 nm, 110 nm, 115 nm, 120 nm, 125 nm, 130 nm, 135 nm, 140 nm, 145 nm, 150 nm, 155 nm, 160 nm, 165 nm, 170 nm, 175 nm, 180 nm, 185 nm, 190 nm, 195 nm, 200 nm, 250 nm, 300
  • the zeta potential of a lipid-based nanoparticle may be used to indicate the electrokinetic potential of a composition comprising said lipid-based nanoparticle.
  • the zeta potential may describe the surface charge of a lipid-based nanoparticle.
  • Lipid-based nanoparticles with relatively low charges, positive or negative, are generally desirable, as more highly charged species may interact undesirably with cells, tissues, and other elements in the body.
  • the zeta potential of a lipid-based nanoparticle may be from about -10 mV to about +20 mV, from about -10 mV to about +15 mV, from about -10 mV to about +10 mV, from about -10 mV to about +5 mV, from about -10 mV to about 0 mV, from about -10 mV to about - 5 mV, from about -5 mV to about +20 mV, from about -5 mV to about +15 mV, from about -5 mV to about +10 mV, from about -5 mV to about +5 mV, from about -5 mV to about 0 mV, from about 0 mV to about +20 mV, from about 0 mV to about +15 mV, from about 0 mV to about +10 mV, from about 0 mV to about +5 mV, from about 0 mV to about +
  • the lipid-based nanoparticle comprises an imaging agent in particular for MRI (magnetic resonance imaging), PET (Positron Emission Tomography), SPECT (Single Photon Emission Computed Tomography), ultrasound, radiography, X-ray tomography and optical imaging (fluorescence, bioluminescence, diffusion).
  • MRI magnetic resonance imaging
  • PET Positron Emission Tomography
  • SPECT Single Photon Emission Computed Tomography
  • ultrasound radiography
  • X-ray tomography X-ray tomography
  • optical imaging fluorescence, bioluminescence, diffusion
  • imaging agents include but are not limited to paramagnetic gadolinium chelates, paramagnetic lanthanide chelate (DOTA, D03A, DTPA, PCTA), especially with a membrane lipophilic part, and, iron gluconates, and iron sulfates or iron oxide comprised in the cavity of the lipid-based nanoparticle, or instance, magnetite (FesCU), maghemite (y- Fe203), wiistite (FeO), hematite (a-Fe203), or combinations thereof, for example such as described in WO2021194672 the disclosure thereof being incorporated herein by reference.
  • the lipid-based nanoparticle of the invention comprises i) an antigen-binding domain and ii) one or several mRNA molecule(s).
  • an antigen-binding domain ii) one or several mRNA molecule(s).
  • the lipid-based nanoparticle according to the invention comprises an antigen binding domain capable of specific binding to a target expressed on activated immune cells surface.
  • an antigen binding domain that specifically (or preferentially) binds to an antigen is an antigen binding domain that binds said antigen for example with greater affinity, avidity, more readily, and/or with greater duration than it binds to other/different antigens.
  • the term “specifically binds to” or “binds specifically” refers to the ability of an antigen receptor to bind to an antigen with an affinity of at least about 1 x IO' 6 M, 1 x IO' 7 M,1 x IO' 8 M, 1 x IO' 9 M, 1 x IO' 10 M, 1 x 10' 11 M, 1 x 10' 12 M, or more, and/or bind to a target with an affinity that is at least two-fold greater than its affinity for a nonspecific antigen.
  • the affinity can be determined by various methods well known from one skilled in the art. These methods include, but are not limited to, Biacore Analysis, Blitz analysis and Scatchard plot.
  • the antigen-binding domain comprised in the lipid-based nanoparticle according to the invention has a KD value inferior or equal to 10' 8 M, preferably inferior or equal to 10' 9 M for the target expressed on activated immune cells, more preferably inferior or equal to 1.10' 10 M, as may be determined by biosensor analysis, particularly by Biacore Analysis.
  • target of the antigen binding domain refers to a carbohydrate, lipid, peptide, polypeptide, protein, antigen or epitope that is specifically recognized or bound by the antigen binding domain according to the invention and expressed on the external surface of activated immune cells.
  • expression of a target on the surface of immune cells the term “expressed” refers to a target, such as carbohydrates, lipids, peptides, polypeptides, proteins, antigens or epitopes that are present or presented at the outer surface of an immune cell.
  • the target is specifically expressed by activated immune cells in a healthy subject or in a subject suffering from a disease, in particular such as a cancer or an infectious disease.
  • a disease in particular such as a cancer or an infectious disease.
  • the target has a higher expression level in activated immune cells than in other cells or that the ratio of activated immune cells expressing the target by the total immune cells is higher than the ratio of other cells expressing the target by the total other cells.
  • the expression level or ratio is higher by a factor 2, 5, 10, 20, 50 or 100.
  • Immuno cell as used herein includes neutrophils, eosinophils, basophils, mast cells, monocytes, macrophages, dendritic cells, natural killer cells, and lymphocytes (B cells and T cells). It preferably refers to T cells, more specifically CD4+ T cells, CD8+ T cells, effector T cells and/or exhausted T cells.
  • activated immune cells it is meant immune cells that are involved or have been activated during an immune response towards the presence of non-self cells such as pathogens or cancer cells. Activated immune cells are particularly recruited in the localization wherein the inflammation, triggered by the presence of non-self cells, occurs. Particular markers of immune cells of activation that can be targeted by the antigen binding domain are particularly described here after. In a particular aspect, immune cells are not T regulatory cells.
  • the activated immune cells are selected from the group consisting of activated T cells, activated B cells, activated myeloid cells including activated macrophages and activated dendritic cells.
  • the target of the antigen binding domain is selected from the group comprising or consisting of PD-1, BCMA/TNFRSF17, BTLA, CD101/IGSF2, CD103, CD119, CD137/4-1BB/TNFRSF9, CD150, CD153, CD154, CD223, CD226, CD25, CD254, CD26, CD27, CD275, CD39/ENTPD1, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, GPR35, FPR2, CD80, CD83, CD86, CD95, CMKLR1, CRTAM, CST7, CTLA4, CXCR3, CXCR4, CXCR5, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, TIM3/HAVCR2, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LAG3, TRAILR, OX40L, LY108 /SlamF6, NKG2D, O
  • TNFRSF 19/TROY TNFRSF21/DR6, TNFRSF25/DR3 /TNFRSF 12, CD301, IL4R, CLEC- 1A, CD21, CLEC-9A, CD 180, CD59, CD54, CD71, CD35, CD218a, CD74, CD 165, 4- 1BBL/CD137L, ICOSL, CD160, CD127 and SIRPa.
  • the target of the antigen binding domain is selected from the group comprising or consisting of BCMA/TNFRSF17, BTLA, CD101/IGSF2, CD103, CD119, CD137/4-1BB/TNFRSF9, CD150, CD153, CD154, CD223, CD226, CD25, CD254, CD26, CD27, CD275, CD39/ENTPD1, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, GPR35, FPR2, CD80, CD83, CD86, CD95, CMKLR1, CRTAM, CST7, CTLA4, CXCR3, CXCR4, CXCR5, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, TIM3/HAVCR2, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LAG3, TRAILR, OX40L, LY108 /SlamF6, NKG2D, OX40/TN
  • TNFRSF 12A/FN14/TWEAKR B AFFR/TNFRSF 13 C/CD268,
  • TNFRSF 19/TROY TNFRSF21/DR6, TNFRSF25/DR3 /TNFRSF 12, CD301, IL4R, CLEC- 1A, CD21, CLEC-9A, CD 180, CD59, CD54, CD71, CD35, CD218a, CD74, CD 165, 4- 1BBL/CD137L, ICOSL and CD160.
  • T cell or “T lymphocytes” as used herein includes CD4 + T cells, CD8 + T cells, T helper 1 type T cells, T helper 2 type T cells, T helper 17 type T cells, effector T cells, effector memory stem like T cells, Tumor Infiltrating Lymphocyte (TIL), anergic T cells and exhausted T cells.
  • TIL Tumor Infiltrating Lymphocyte
  • the T cell is an effector T cell, an exhausted T cell, a Tumor Infiltrating Lymphocyte (TIL) or an effector memory stem like T cell.
  • Activated T cell or “Activated T lymphocytes” are T cells activated by simultaneously receiving signal- 1 from T-cell recognition of antigen via the T cell receptor and signal-2 from costimulatory molecule.
  • Markers expressed by activated T cells include but are not limited to CD137/41BB/TNFRSF9, PD-1, CTLA4, FasL/TNFSF6, ITGAE and OX40/TNFRSF4 .
  • Exhausted T cells can be characterized by the presence of the marker TNFRSF9.
  • T cell does not include regulatory T cells (Treg), inhibitory T cells and/or senescent T cells. Regulatory T cells can be characterized by the presence of the marker CD25. Preferably, the T cell is an activated T cell.
  • Treg regulatory T cells
  • inhibitory T cells inhibitory T cells
  • senescent T cells Regulatory T cells can be characterized by the presence of the marker CD25.
  • the T cell is an activated T cell.
  • T cell does not include regulatory T cells (Treg), inhibitory T cells and/or senescent T cells. Regulatory T cells can be characterized by the presence of the marker CD25.
  • B cells or “B lymphocytes” as used herein include but are not limited to B-l B cells, follicular B cells, and marginal zone (MZ) B cells.
  • Activated B cells or “Activated B lymphocytes” are B cells activated when the B cell binds to an antigen via its B cell Receptor. Activated B cells are particularly able to secrete immunoglobulins. Markers expressed by activated B cells include but are not limited to BCMA/TNFRSF17, CD 150 and CD86..
  • Neutrophils are a type of white blood cells called granulocytes and are produced from stem cells in the bone marrow. They play an important role in innate immunity
  • Activated Neutrophils are neutrophils activated upon their entry into inflammatory or infected tissue site, in response to pro-inflammatory stimuli in the tissue. Neutrophils activation is characterized by the release of granule proteins, acquisition of phagocytic capabilities, and production of NETs, all of which are designed to enhance the cells’ pathogen-destruction capacity. Markers of activated neutrophils include but are not limited to CD 11b, CD 18, CD66b, CD 177 and PRTN3.
  • Eosinophils are a variety of white blood cells and one of the immune system components responsible for combating multicellular parasites and certain infections in vertebrates. They are granulocytes that develop during hematopoiesis in the bone marrow before migrating into blood. “Activated Eosinophils” are eosinophils which were recruited from the blood into the tissues at sites of inflammation or infection, and received activation signal through cytokine mediation. Upon activation, eosinophils can release an array of inflammatory mediators. Markers of activated eosinophils include but are not limited to CD69, L-selectin, ICAM-1, CD44 and PSGL-1.
  • Basophils are a type of polymorphonuclear leukocyte characterized by having a nucleus with two or three lobes, and by the presence of cytoplasmic granules.
  • Activated Basophils are activated by antigen crosslinking of FceRI receptor-bound IgE to undergo rapid degranulation and release their cellular contents.
  • basophils can be activated without IgE crosslinking by inflammatory mediators such as complement factors C5a and C3a, MBP, PAF and chemokines. Markers of activated basophils include but are not limited to CD63, CD203c and CD164.
  • Mast cells also known as mastocytes or labrocytes are resident cells of connective tissue that contains many granules rich in histamine and heparin.
  • Activated Mast cells are mast cells stimulated by allergens through cross-linking with immunoglobulin E receptors (e.g., FcsRI), physical injury through pattern recognition receptors for damage-associated molecular patterns (DAMPs), microbial pathogens through pattern recognition receptors for pathogen-associated molecular patterns (PAMPs), and various compounds through their associated G-protein coupled or ligand-gated ion channels.
  • immunoglobulin E receptors e.g., FcsRI
  • DAMPs damage-associated molecular patterns
  • PAMPs pathogen-associated molecular patterns
  • various compounds through their associated G-protein coupled or ligand-gated ion channels.
  • activated mast cells Upon activation, they can selectively or quickly release mediators or inflammation-inducing components, such as histamine, heparin, cytokines, and growth factors.
  • mediators or inflammation-inducing components such as histamine, heparin, cytokines, and growth factors.
  • Markers of activated mast cells include but are not limited to CD63, CD203, c-Kit, IL-3Ra, and FceRI.
  • Macrophages are a type of white blood cells that help eliminate foreign substances by engulfing foreign materials and initiating an immune response. As used herein, this term includes for example Adipose tissue macrophages, Monocytes, Kupffer cells, Sinus histiocytes, Alveolar macrophages (dust cells), Tissue macrophages (histiocytes) leading to giant cells, Microglia, Hofbauer cells, Intraglomerular mesangial cells, Osteoclasts, Langerhans cells, Epithelioid cells, Red pulp macrophages (sinusoidal lining cells), Peritoneal macrophages, LysoMac.
  • Adipose tissue macrophages Monocytes, Kupffer cells, Sinus histiocytes, Alveolar macrophages (dust cells), Tissue macrophages (histiocytes) leading to giant cells, Microglia, Hofbauer cells, Intraglomerular mesangial cells, Osteoclasts,
  • Activated Macrophages are macrophages activated either by a priming signal through IFNg followed by encountering an appropriate stimulus, such as bacterial LPS, or by direct stimulation by IL4 and/or IL13. Activated macrophages are able to kill non-self cells through phagocytosis. Markers of activated macrophages include but are not limited to TNFRSF12A/FN14/TWEAKR.
  • Dendritic cells or “DCs” are antigen-presenting cells of the immune system. Their main function is to process antigen material and present it on the cell surface to the T cells of the immune system. As used herein, this term includes for example plasmacytoid dendritic cells (pDC) and myeloid dendritic cells (mDC). “Activated Dendritic cells” are dendritic cells directly activated by conserved pathogen molecules and indirectly by inflammatory mediators produced by other cell types that recognize such molecules. Markers of activated dendritic cells include but are not limited to TRAF4.
  • Natural killer cell Natural killer cell
  • Activated Natural killer cell are natural killer cells activated by a prevalence of activating receptor stimulation over inhibitory receptor stimulation. Markers of activated natural killer cells include but are not limited to NKG2A and TRAF3IP1.
  • the antigen binding domain of the invention particularly targets a marker of activation of an immune cell, in particular such as described here above.
  • the target expressed on activated immune cells can particularly be selected among the target described in Table D below.
  • Table D Examples of targets of interest.
  • the target expressed on activated immune cells surface is in addition specifically expressed on activated immune cells surface.
  • the target of the antigen binding domain is expressed at the outer surface of immune cells, but is not substantially expressed by other cell types, for example such as tumor cells. It particularly means that the expression of the target is higher in immune cells than in other cells (i.e., non- immune cells).
  • the target specifically expressed on activated immune cells surface is selected from the group consisting of BCMA/TNFRSF17, BTLA, CD101/IGSF2, CD103, CD119, CD137/4- 1BB/TNFRSF9, CD150, CD153, CD154, CD223, CD226, CD25, CD254, CD26, CD27, CD275, CD30, CD39/ENTPD1, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, GPR35, FPR2, CD80, CD83, CD86, CD95, CMKLR1, CRTAM, CST7, CTLA4, CXCR3, CXCR4, CXCR5, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, TIM3/HAVCR2, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LAG3, TRAILR, OX40L, LY108 /SlamF6, NKG2D, OX40/TN
  • TNFRSF 12A/FN14/TWEAKR B AFFR/TNFRSF 13 C/CD268,
  • TNFRSF 19/TROY TNFRSF21/DR6, TNFRSF25/DR3 /TNFRSF 12, CD301, IL4R, CLEC- 1A, CD21, CLEC-9A, CD 180, CD59, CD54, CD71, CD35, CD218a, CD74, CD 165, 4- 1BBL/CD137L, ICOSL and CD160.
  • the target expressed on activated B cells surface is selected from the group consisting of BCMA/TNFRSF17, CD 150, CD86, OX40L, LOX1, TACI/TNFRSF13B, CD138 (SDC1), FCRL4, CD78, FRAF3/CD40BP, TRAP1, BAFFR/TNFRSF13C/CD268, CD21, CLEC-9A, CD 180, CD59, CD54, CD71, CD35, CD218a, CD74, CD 165.
  • the target expressed on activated myeloid cells surface is selected from the group consisting of CD 163, CD206, SIGLEC 6, TRAF1, TRAF4, TRAF7, TRAP100/MED24, TNFRSF12A/FN14/TWEAKR, CD301, IL4R, CLEC-1A.
  • the target expressed on activated natural killer cells surface is selected from the group consisting of CST7, CXCR4, NKG2A, TRAF3IP1, CMKLR1.
  • the target specifically expressed on activated immune cells surface is not an antigen of the TCR pathway (interaction between antigen presenting cells and T cells).
  • the target expressed on activated T cells surface is selected from the group consisting of CD101/IGSF2, CD103, CD119, CD137/4-1BB/TNFRSF9, CD154, CD183, CD25, CD254, CD26, CD275, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, CD80, CD95, CTLA4, CXCR3, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LY108 /SlamF6, OX40/TNFRSF4, RGS1, LTBR/CD70, TNFSF14, CD112R, CD28H, CD164, TRAF2, CDCR3/TNFRSF6B, GITR/TNFRSF8/CD357, RELT/TNFRSF19L, TNFRSF19/TROY, TNFRSF21/DR6, TNFRSF25/DR3/TNFRSF
  • the target expressed on TILs surface is selected from the group consisting of CD101, CD137 (Tnfrsf9/4-lBBL), CRTAM, CST7, CTLA4, CXCR3, FAS, IL18R1/CXCR1/CD218A, LAG-3 PTPN22, RGS1, TNFSF14 and PD1.
  • the target expressed on activated immune cells surface is selected from the group consisting of PD-1, CD127, SIRPa and CLEC-1A.
  • the lipid-based nanoparticle of the invention aims to target activated immune cells that are present in a tumoral environment (e.g., that are recruited on tumoral site), for example PD-1 positive immune cells.
  • the target is PD-1.
  • the target specifically expressed on activated immune cells surface is a target that allows the interaction between an immune cell and a tumor cell (for example: PD-1 on T cells and PD-L1 on tumoral cells).
  • the target specifically expressed on activated immune cells surface is not a target that allows the interaction between a T cell and an antigen presenting cell (APC) via the TCR pathway.
  • APC antigen presenting cell
  • the immune cell is a T cell so that the lipid-based nanoparticle according to the invention comprises an antigen-binding domain capable of specifically binding to a target expressed on activated T cells surface.
  • the target expressed on activated T cells surface is selected from the group consisting of CD137/41BB/TNFRSF9, PD-1, CRTAM, CD39, CXCR5, CD70, CTLA-4, TIM-3/HAVCR2, ITGAE, LAG-3, OX40/TNFRSF4,and TIGIT.
  • the target specifically expressed on activated immune cells surface is not a pan- T-cell marker, i.e., a marker that is expressed on multiple sub-types of T cells.
  • the pan-T antigen is CD2, CD3, CD5 or CD7. Then, when the lipid-based nanoparticle is aimed at targeting immune cells such as T cells, the target of the antigen binding domain is not CD2, CD3, CD5 and/or CD7.
  • the activated T cell is an effector memory stem like T cell and the target is a factor expressed on the surface of effector memory stem like T cells, preferably specifically expressed on the surface of effector memory stem like T cells.
  • the target expressed on the surface of effector memory stem like T cells is selected from the group consisting of CXCR5, SLAMF7 and CRTAM.
  • the activated T cell is a Tumor Infiltrating lymphocytes (TILs) and the target is a factor expressed on the surface of Tumor Infiltrating lymphocytes, preferably specifically expressed on the surface of TILs.
  • TILs Tumor Infiltrating lymphocytes
  • the target expressed on the surface of TILs is selected from the group consisting of CD101, CD137 (Tnfrsf9/4-lBBL), CRTAM, CST7, CTLA4, CXCR3, FAS, IL18R1/CXCR1/CD218A, LAG-3 PTPN22, RGS1, TNFSF14 and PD1.
  • the activated T cell is a cytotoxic T cell and the target is a factor expressed on the surface of cytotoxic T cell, preferably specifically expressed on the surface of cytotoxic T cells.
  • the target expressed on the surface of cytotoxic T cells is selected from the group consisting of CD25, CD38, CD69, PD1, TIM-3, LAG-3 and TIGIT.
  • the present invention relates to an approach that is very different from lipid-based nanoparticles that would target tumoral cells (instead of immune cells) thanks to at least a targeting agent specific of tumor cells, such as an antibody at the membrane of the LNP (said antibody targeting a tumor associated antigen such as HER2 or VEGFR).
  • Such tumor targeted LNP may comprise at least an RNA encoding a protein activating i) the expression by the tumor cell of a gene inducing the apoptosis or death of the tumor cell, and/or ii) the expression by the tumor cell of a gene encoding the production by the tumor cell of a checkpoint inhibitor (such as PD-L1) between an immune cell (notably a T cell) and the tumor cell.
  • a checkpoint inhibitor such as PD-L1
  • the tumor cell modified by the RNA of the LNP produces less PD-L1 at its surface, and there is thus less inhibition of T cells (expressing PD-1, the ligand of PD- Ll).
  • the RNA of the LNP interferes directly with the activity of the activated immune cell (notably T cell), not of a tumor cell.
  • the antigen binding domain has an antagonist activity on the target when the target is a receptor having an inhibitory effect on the cell, in particular for checkpoint inhibitors between tumor cells and T cells (for example PD-1).
  • antagonist refers to a substance that blocks or reduces the activity or functionality of another substance. Particularly, this term refers to a binding domain that binds to a cellular receptor (e.g., PD-1) as a reference substance (e.g., PD-L1 and/or PD-L2), preventing it from producing all or part of its usual biological effects (e.g., the creation of an immune suppressive microenvironment).
  • a cellular receptor e.g., PD-1
  • PD-L1 and/or PD-L2 a cellular receptor
  • the antagonist activity may be assessed by competitive ELISA.
  • the antigen binding domain does not interfere nor compete with the binding between its target and its natural ligand. In a preferred aspect, the antigen binding domain of the lipid-based nanoparticle does not compete with an antigen binding domain which binds to the same target.
  • the antigen binding domain has an agonist activity on the target when the target is a receptor having an activator effect on the cell (e.g CD137).
  • agonist refers to a substance that activates or increases the activity or functionality of another substance. Particularly, this term refers to a binding domain that binds to a cellular receptor as a reference substance, causing it to produce all or part of its usual biological effects (e.g., the creation of an immune suppressive microenvironment).
  • the antigen binding domain has no activity on the target, in particular for checkpoint inhibitors between tumor cells and T cells (for example PD-1).
  • the antigen binding domain does not interfere with the binding between its target and its natural ligand.
  • the antigen binding domain according to the invention is not an agonist and/or not an antagonist of the interaction between its target and its natural ligand. The absence of such an agonist and/or antagonist capability may be assessed with methods usually known by the person skilled in the art.
  • the antigen binding domain of the lipid-based nanoparticle does not compete with the natural ligand for the binding to the target specifically expressed on activated immune cells.
  • the absence of competition between the antigen binding domain of the invention and the natural ligand of the target specifically expressed on the activated immune cells may be determined when, in presence of the antigen binding domain of the invention, the binding of the natural ligand to the target specifically expressed on the activated immune cells is at least 50%, more preferably at least 80%, still more preferably at least 90% and most preferably similar, to the binding of the natural ligand to the target specifically expressed on the activated immune cells, under the same experimental conditions but without the presence of the antigen binding domain of the invention.
  • the antigen binding domain is an antibody, a fragment thereof or a derivative thereof.
  • the antigen binding domain is preferably derived from a format selected from the group consisting of IgA, IgM, IgE, IgD and IgG, or a variant thereof.
  • the antigen binding domain is preferably a monoclonal antibody, preferably a human, humanized, chimeric or recombinant antibody or antigen binding fragment thereof.
  • the antigen binding domain is a monoclonal antibody or an antigen binding fragment thereof. In some aspects, the antigen binding domain is a monoclonal antibody or an antigen binding fragment thereof.
  • the term "monoclonal antibody” as used herein refers to a preparation of antibody molecules of single specificity. A monoclonal antibody displays a single binding specificity and affinity for a particular epitope. Accordingly, the term “human monoclonal antibody” refers to an antibody displaying a single binding specificity which has variable and constant regions derived from or based on human germline immunoglobulin sequences or derived from completely synthetic sequences. The method of preparing the monoclonal antibody is not relevant for the binding specificity. In an aspect, the antibodies of the disclosure are monoclonal antibodies.
  • the antigen binding domain is a recombinant antibody or an antigen binding fragment thereof.
  • the term "recombinant antibody” refers to antibodies which are produced, expressed, generated or isolated by recombinant means, such as antibodies which are expressed using a recombinant expression vector transfected into a host cell; antibodies isolated from a recombinant combinatorial antibody library; antibodies isolated from an animal (e.g. a mouse) which is transgenic due to human immunoglobulin genes; or antibodies which are produced, expressed, generated or isolated in any other way in which particular immunoglobulin gene sequences (such as human immunoglobulin gene sequences) are assembled with other DNA sequences.
  • Recombinant antibodies include, for example, chimeric and humanized antibodies.
  • the antigen binding domain is a chimeric antibody or an antigen binding fragment thereof.
  • a “chimeric antibody” refers to an antibody in which the sequence of the variable domain derived from the germline of a mammalian species, such as a mouse, have been grafted onto the sequence of the constant domain derived from the germline of another mammalian species, such as a human.
  • Chimeric antibodies generally comprise constant domains from human and variable domains from another mammalian species, reducing the risk of a reaction to foreign antibodies from a non-human animal when they are used in therapeutic treatments.
  • the antigen binding domain is a humanized antibody or an antigen binding fragment thereof.
  • humanized antibody refers to an antibody in which CDR sequences derived from the germline of another mammalian species, such as mouse, have been grafted onto human framework sequences”.
  • a "humanized form" of an antibody e.g., a non-human antibody, also refers to an antibody that has undergone humanization.
  • a humanized antibody is generally a human immunoglobulin (recipient antibody) in which residues from one or more CDRs are replaced by residues from at least one CDR of a nonhuman antibody (donor antibody) while maintaining the desired specificity, affinity, and capacity of the original antibody. Additional framework region modifications may be made within the human framework sequences.
  • humanized antibody has a T20 humanness score greater than 80%, 85% or 90%.
  • “Humanness” of an antibody can for example be measured using the T20 score analyzer to quantify the humanness of the variable region of antibodies as described in Gao S H, Huang K, Tu H, Adler A S. BMC Biotechnology. 2013: 13:55 or via a web-based tool to calculate the T20 score of antibody sequences using the T20 Cutoff Human Databases: https://abAnalyzer.lakepharma.com.
  • the antigen binding domain according to the present disclosure is a modified antibody or an antigen binding fragment thereof.
  • a “modified antibody” corresponds to a molecule comprising an antibody or an antigen-binding fragment thereof, wherein said antibody or functional fragment thereof is associated with a functionally different molecule.
  • a modified antibody of the invention may be either a fusion chimeric protein or a conjugate resulting from any suitable form of attachment including covalent attachment, grafting, chemical bonding or coupling with a chemical or biological group or with a molecule, such as a PEG polymer or another protective group or molecule suitable for protection against proteases cleavage in vivo, for improvement of stability and/or half-life of the antibody or functional fragment.
  • PEGylation of the antibody or functional fragments thereof is a particular interesting embodiment as it improves the delivery conditions of the active substance to the host, especially for a therapeutic application.
  • PEGylation can be site specific to prevent interference with the recognition sites of the antibodies or functional fragments and can be performed with high molecular weight PEG.
  • PEGylation can be achieved through free cysteine residues present in the sequence of the antibody or functional fragment or through added free Cysteine residues in the amino sequence of the antibody or functional fragment.
  • the antigen binding domain comprises a hydrophobic domain.
  • the hydrophobic domain may comprise or consist of one or more alpha-helical regions.
  • the hydrophobic region is particularly configured to interact with the hydrophobic lipid of the lipid-based nanoparticle of the invention.
  • the antigen binding domain comprises or is modified to comprise a group such as a thiol group, capable of reacting with a group carried by a lipid of the nanoparticle, such as a PEG-maleimide, so as to conjugate the antigen binding domain to a lipid of the lipid-based nanoparticle.
  • a group such as a thiol group
  • a group carried by a lipid of the nanoparticle such as a PEG-maleimide
  • the antigen binding domain is conjugated to a lipid of the lipid-based nanoparticle, for instance through a maleimide moiety which is conjugated, attached or linked to a PEG-derivative.
  • the antigen binding domain may comprise a thiol group capable of reacting with the PEG-maleimide to conjugate the antigen binding domain to the lipid-based nanoparticle.
  • Other methods known to the person skilled in the art may be used to conjugate the antigen binding domain to a lipid of the lipid-based nanoparticle, optionally through a linker, such as disclosed in Kedmi, Ranit et al. “A modular platform for targeted RNAi therapeutics.” Nature nanotechnology vol. 13,3 (2016): 214-219. doi: 10.1038/s41565-017-0043-5, the disclosure thereof being incorporated herein by reference.
  • the antigen binding domain is not covalently bound to any of the lipids of the LNP or does not comprise any modification for coupling or grafting the antigen binding domain to a lipid.
  • the antigen binding domain does not comprise a lipophilic moiety or a grafting moiety such as free cysteine(s) or a thiol group.
  • the LNP does not include any antigen binding domain specific for an antigen present on the antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface, in particular an antigen binding domain directed against the Fc domain of an antibody.
  • the antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface is not bound to the LNP by a type of interaction “antigen-antibody”. More specifically, the antigen binding domain is not bound by an antigen binding domain specific for an antigen present on the antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface, in particular an antigen binding domain directed against the Fc domain of an antibody.
  • the lipid-based nanoparticle does not comprise a secondary antibody that allows the attachment of the antigen binding domain to the lipid-based nanoparticle (e.g., such as an anti-Fc antibody or antigen binding fragment or derivative thereof).
  • the lipid-based nanoparticle does not comprise an antibody or any fragment or derivative thereof that is lipidated or that is covalently bound to a peptide or motif that is lipidated.
  • the lipid-based nanoparticle does not comprise a lipidated secondary antibody that allows the attachment of the antigen binding domain to the lipid-based nanoparticle.
  • the lipid-based nanoparticle does not comprise a moiety comprising a lipidation peptide or motif.
  • the antigen binding domain does not comprise or is not covalently bound to an anchoring moiety comprising a lipidation peptide or motif.
  • anchoring moiety or “anchoring molecule” or “anchoring entity” refers to a component that anchors or attaches the antigen binding domain into the lipid- based nanoparticle.
  • anchoring moiety is a protein.
  • lipidation peptide or motif refers to a specific sequence pattern in proteins or proteic entity (such as antibodies or fragment thereof) that is associated with the attachment or anchoring of lipid moi eties.
  • an entity that comprises a lipidation peptide, motif or pattern is an entity (e.g., an antigen binding domain, scFv or antibody) that will be anchored or attachment to lipid molecules, in particular to lipids of the lipid-based nanoparticle.
  • entity e.g., an antigen binding domain, scFv or antibody
  • Lipidation peptide or motif can involve different types of lipid modifications, including: cysteine prenylation (e.g., the attachment of hydrophobic isoprene polymers such as farnesyl or geranylgeranyl to cysteine residues of proteins), N-terminal Glycine Myristoylation, Cysteine Palmitoyl ati on, Serine and Lysine Fatty Acylation (e.g., the addition of fatty acyl groups to serine and lysine residues of proteins), Palmitoylation, GPI-Anchor Addition, or peptides that derive from part of an inner membrane bacterial lipoprotein.
  • cysteine prenylation e.g., the attachment of hydrophobic isoprene polymers such as farnesyl or geranylgeranyl to cysteine residues of proteins
  • N-terminal Glycine Myristoylation Cysteine Palmitoyl ati on
  • Serine and Lysine Fatty Acylation
  • the lipid based nanoparticle does not comprise a bacterial anchor polypeptide, a lipoprotein, such as a bacterial lipoprotein, or a recombinant membrane- anchored lipoprotein.
  • the antigen binding domain does not comprise or is not covalently bound to a bacterial anchor polypeptide, a lipoprotein, such as a bacterial lipoprotein, or a recombinant membrane-anchored lipoprotein.
  • the lipid based nanoparticle of the invention does not comprise a NipA lipoprotein or any fragment thereof.
  • the antigen binding domain does not comprise or is not covalently bound to a NipA lipoprotein or any fragment thereof.
  • the lipid based nanoparticle or antigen binding domain does not comprise a moiety comprising a lipidation peptide or motif that comprises or consists of the amino acid sequence : CDNSSS (SEQ ID NO: 41) or CDQSSS (SEQ ID NO: 42).
  • the antigen-binding domain to be comprised in the lipid-based nanoparticle according to the invention has a binding activity which is similar to the same antigen-binding domain in a free form.
  • a “free form antigen binding domain” refers to an antigen-binding domain which is not linked, grafted or conjugated to an LNP.
  • the antigen-binding domain to be comprised in the lipid-based nanoparticle according to the invention has a binding activity equal to about 70% of the binding activity of the free form antigen-binding domain, even preferably to about 75%, even more preferably to about 80%.
  • the antigen binding domain according to the present disclosure is an antigen-binding antibody mimetic.
  • antigen-binding antibody mimetic refers to artificial proteins, peptides and any chemical compounds with the capacity to bind antigens mimicking that of antibodies. Such mimetics comprise affitins and anticalins as well as aptamers (peptide aptamers and oligonucleotide aptamers).
  • the antigen binding domain according to the present disclosure is a targeting peptide.
  • the peptide is not covalently linked to a lipid or to a lipidation motif.
  • the peptide is the RGD peptide such as described in Qin J, Xue L, Gong N, Zhang H, Shepherd SJ, Haley RM, Swingle KL, Mitchell MJ. RGD peptide-based lipids for targeted mRNA delivery and gene editing applications.
  • PMID 36199352; PMCID: PMC9450108.
  • the antigen binding domain is an aptamer.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence.
  • Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods.
  • the antigen binding domain of the lipid-based nanoparticle can be of any format known in the art.
  • the antigen binding domain is an antibody, a fragment or a derivative thereof such as a Fab, a F(ab)2, a Fab’, a F(ab')2, a Fd, a Fv, a crossMAb or a single-chain variable fragment (scFV) a VHH or a single-chain Fab fragment.
  • antigen binding fragments typically include a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consisting of the VH and CHI domains; a Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a dAb fragment (Ward et al., 1989 Nature 341 :544-546), which consists of a VH domain, or any fusion proteins comprising such antigen-binding fragments.
  • a Fab fragment a monovalent fragment consisting of the VL, VH, CL and CHI domains
  • F(ab)2 fragment a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region
  • a Fd fragment consisting of the VH and CHI domains
  • a Fv fragment consisting of
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single chain protein in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al., 1988 Science 242:423-426; and Huston et al., 1988 Proc. Natl. Acad. Sci. 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment" of an antibody.
  • CrossMAb refers to antigen binding domains with an inversion of CL and CHI domains, in particular in one binding arm of antibodies.
  • binding domain comprises a VH domain linked to a CL domain and a VL domain linked to a CHI domain.
  • Such format reduces the byproduct formation caused by a mismatch of a light chain of a first binding domain that specifically binds to a first antigen with the wrong heavy chain of a second biding domain that specifically binds to a second antigen (when compared to approaches without such CL-CH1 domain exchanges).
  • CrossMAb are for example described in WO 2009/080253 and Schaefer, W.
  • the antigen binding domain is an antibody or comprises or consists of a Fab, a Fv, a Fab’, a scFV, a CrossMAb or a VHH covalently linked to a Fc domain, preferably an IgG Fc domain such as described herein.
  • the antigen binding domain is an aptamer or a short peptide sequence such as RGD peptide.
  • RNA ribonucleic acid
  • DNA deoxyribonucleic acid
  • the antigen binding domain is an antibody, a fragment or a derivative thereof such as a F(ab')2, a Fab, a Fab’, a F(ab)2, Fd, Fv, a crossMAb, a single-chain variable fragment (scFV), a VHH or a single chain Fab fragment that is specific to PD-1, CTLA-4, BTLA, TIGIT, CD 160, LAG-3 or TIM-3.
  • scFV single-chain variable fragment
  • VHH single chain Fab fragment that is specific to PD-1, CTLA-4, BTLA, TIGIT, CD 160, LAG-3 or TIM-3.
  • Numerous antibodies directed against PD-1, TIM- 3, CTLA-4, LAG-3, BTLA, TIGIT and CD160 have already been described in the art.
  • the antigen binding domain is designed to be specifically activable in the TME.
  • the antigen binding domain may be a pH sensitive antigen binding domain, having a lower affinity for its ligand at a physiological pH, and a higher affinity at a pH of the TME, typically more acidic than the physiological pH.
  • the antigen binding domain may comprise a masking group designed to bar the antigen binding domain from binding with its ligand, or at least limit its binding to its ligand, when out of the TME, while exposing the antigen binding domain once in the TME.
  • the exposure of the antigen binding domain may be caused by the switch of pH, the presence of a protease, or any other mechanism known to the person skilled in the art.
  • the terms “Programmed Death 1”, “Programmed Cell Death 1”, “PD-1”, “PDCD1”, “PD-1 antigen”, “human PD-1”, “hPD-1” and “hPD-1” are used interchangeably and refer to the Programmed Death- 1 receptor, also known as CD279, and include variants and isoforms of human PD-1, and analogs having at least one common epitope with PD-1.
  • PD-1 is a key regulator of the threshold of immune response and peripheral immune tolerance. It is expressed on activated T cells, B cells, monocytes, and dendritic cells and binds to its ligands PD-L1 and PD-L2. Human PD-1 is encoded by the PDCD1 gene.
  • PD-1 has four splice variants expressed on human Peripheral blood mononuclear cells (PBMC).
  • PD-1 proteins include full-length PD-1, as well as alternative splice variants of PD-1, such as PD-lAex2, PD-lAex3, PD-lAex2,3 and PD- lAex2,3,4.
  • the terms include any variant and, isoform of human PD-1 that are naturally expressed by PBMC, or that are expressed by cells transfected with a PD-1 gene.
  • the anti -PD-1 antibody can be selected from the group consisting of Pembrolizumab (also known as Keytruda lambrolizumab, MK-3475), Nivolumab (Opdivo, MDX-1106, BMS-936558, ONO-4538), OSE-279 (see WO2020/127366), Pidilizumab (CT-011), Cemiplimab (Libtayo), Camrelizumab, AUNP12, AMP-224, AGEN-2034, BGB-A317 (Tisleizumab), PDR001 (spartalizumab), MK-3477, SCH-900475, PF-06801591, JNJ-63723283, Genolimzumab (CBT-501), LZM-009, BCD- 100, SHR-1201, BAT-1306, AK-103 (HX-008), MEDI-
  • Antibodies directed against TIM-3 targeting TIM-3 are also known such as Sym023, TSR- 022, MBG453, LY3321367, INCAGN02390, BGTB-A425, LY3321367.
  • the TFM-3 antibody is as disclosed in International Patent Application Publication Nos. W02013006490, W02016/161270, WO 2018/085469, or WO 2018/129553, WO 2011/155607, U.S. 8,552,156, EP 2581113 and U.S 2014/044728, the disclosure thereof being incorporated herein by reference.
  • Antibodies directed against CTLA-4 targeting CTLA-4 are also known such as Ipilimumab, Tremelimumab, MK-1308, AGEN-1884, XmAb20717 (Xencor), MEDI5752
  • Anti-CTLA-4 antibodies are also disclosed in WO 18025178, WO 19179388, WO19179391, WO19174603, WO19148444, WO19120232, WO19056281, WO19023482, W018209701, WO18165895, WO18160536, WO18156250, WO18106862, WO18106864, WO18068182, W018035710, WO18025178, WO17194265, WO17106372, W017084078, WO17087588, WO16196237, WO16130898, WO16015675, WO12120125, W009100140 and W007008463, the disclosure thereof being incorporated herein by reference.
  • Antibodies directed against LAG-3 targeting LAG-3 are also known such as BMS- 986016, IMP701 or MGD012.
  • Anti-LAG-3 antibodies are also disclosed in W02008132601, EP2320940, WO19152574, the disclosure thereof being incorporated herein by reference.
  • Antibodies directed against BTLA are also known in the art such as hu Mab8D5, hu Mab8A3, hu Mab21H6, hu Mabl9A7, or hu Mab4C7.
  • the antibody TAB004 against BTLA are currently under clinical trial in subjects with advanced malignancies.
  • Anti -BTLA antibodies are also disclosed in W008076560, W010106051 (e.g., BTLA8.2), WO1 1014438 (e.g., 4C7), W017096017 and WO17144668 (e.g., 629.3), the disclosure thereof being incorporated herein by reference.
  • Antibodies directed against TIGIT are also known in the art, such as BMS-986207 or AB154, BMS-986207 CPA.9.086, CHA.9.547.18, CPA.9.018, CPA.9.027, CPA.9.049, CPA.9.057, CPA.9.059, CPA.9.083, CPA.9.089, CPA.9.093, CPA.9.101, CPA.9.103,
  • the target is CD127 or IL-7R and the antigen binding domain is specific to CD 127, preferably human CD 127.
  • the antigen binding domain is an antagonist of CD127.
  • IL-7R refers to any form of IL-7R and variants thereof that retain at least part of the activity of IL-7R.
  • IL-7R is found as Uniprot Accession Number P16871.
  • Antagonist IL-7R antibodies encompass antibodies that block, antagonize, suppress or reduce (to any degree including significantly) IL-7R biological activity, including downstream pathways mediated by IL-7R signaling, such interaction with IL-7 and/or elicitation of a cellular response to IL-7.
  • Antibodies directed against CD127 or IL7-R are also known in the art, such as GSK2618960, RN168, AbD11590, MAB306-100, R34.34, A019D5, eBioRDR5, 40131, 1A12, M21, 47H4, HIL-7R-M21, eBioYL8, RDR5.
  • Anti-CD127 antibodies are also disclosed in W014102430, W020077190, W004000238, WO11104687, WO16059512 and WO 17062748, the disclosure thereof being incorporated herein by reference.
  • the target is SIRPa and the antigen binding domain is specific to SIRPa, preferably human SIRPa.
  • the antigen binding domain is an antagonist of SIRPa.
  • SIRPa Simulatory protein alpha
  • SIRPa a receptor-type transmembrane glycoprotein that is mammalian Immunoglobulin-like cell surface receptor for CD47.
  • anti-SIRP refer to an antibody of the disclosure which is intended for use as a therapeutic or diagnostic agent, and specifically binds to SIRPa, in particular to a human SIRPa, to one or both of two common variants identified, SIRPaVl and SIRPaV2.
  • the human SIRPa amino acid sequence is about 504 amino acids and has a Genbank accession number of NP_001035111.1, NP_001035112.1, NP_001317657.1, or NP_542970.1.
  • Antibodies directed against SIRPa are also known in the art, such as OSE-172, CC-95251, BI 765063, HPA054437, Magrolimab, TTI-621, TTL622 and Evorpacept (ALX148).
  • Anti- SIRPa antibodies are also disclosed in WO17178653, W019073080, WO22254379, W020102422, WO23202672, WO21222746, W018008470, W016205042, WO22121980, WO221 10922, WO19226973, WO22254379, WO23020459 and W018107058 the disclosure thereof being incorporated herein by reference.
  • the antigen binding domain comprises or consists of an anti-SIRPa antibody such as disclosed in W019073080.
  • the antigen binding domain is an anti-SIRPa domain, comprising or consisting of a VH domain comprising or consisting of a sequence as set forth in SEQ ID NO: 41 and a VL domain comprising or consisting of a sequence as set forth in SEQ ID NO: 42.
  • said antigen binding domain further comprises or is covalently linked to a Fc domain, preferably an IgG Fc domain such as described herein.
  • the antigen binding domain is an anti-SIRPa antibody, comprising or consisting of : an heavy chain comprising or consisting of a sequence as set forth in SEQ ID NO: 54 and a light chain comprising or consisting of a sequence as set forth in SEQ ID NO: 55.
  • the target is CLEC-1 A and the antigen binding domain is specific to CLEC- 1 A, preferably human CLEC-1 A.
  • the antigen binding domain is an antagonist of CLEC-1 A.
  • CLEC-1 A relates to a C- type lectin-like receptor- 1 A from a mammal species, preferably a human CLEC-1 A.
  • a reference sequence of the human CLEC- 1 A corresponds to the sequence associated to the Accession number Q8NC01 Uniprot.
  • CLEC-1 antagonist has its general meaning in the art and refers to any compound, such as an antibody or a fragment thereof, that blocks, suppresses, or reduces the biological activity of CLEC-1. In particular, the CLEC-1 antagonist inhibits the interactions between the CLEC-1 and at least one of its ligands.
  • Antibodies directed against CLEC-1 A are also known in the art, such as MAB1704, ABIN526589, AF1704 and ABIN526590.
  • the target is PD-1 and the antigen binding domain is specific to PD-1.
  • the antigen binding domain is an antagonist of PD-1.
  • the anti-PD-1 antibody is Pembrolizumab (also known as Keytruda lambrolizumab, MK-3475), Nivolumab (Opdivo, MDX-1106, BMS-936558, ONO-4538) or OSE-279 (such as described in WO2020/127366, the disclosure thereof being incorporated herein by reference).
  • the invention concerns a t-LNP, comprising an anti-PD-1 antigen-binding domain such as disclosed herein, capable of specifically binding to PD-1 expressed on activated T cells surface and one or several mRNA molecule(s) encoding an activity-enhancing protein of said activated T cells.
  • the antigen binding domain comprised in the lipid-based nanoparticle according to the invention is an anti-PD-1 antibody such as described above an or antigen binding fragment thereof, preferably a human, humanized or chimeric anti-PD-1 antibody or antigen binding fragment thereof.
  • the antigen binding domain is a F(ab')2, a Fab, a crossMAb or a scFv that is specific to PD-1.
  • said antigen binding domain further comprises or is covalently linked to a Fc domain, preferably an IgG Fc domain such as described herein.
  • the antigen binding domain targets PD-1 and is derived from the antibody disclosed in WO2020/127366, the disclosure thereof being incorporated herein by reference in its entirety.
  • the antigen binding domain is an anti-PD-1 antigen-binding domain comprising:
  • the heavy chain CDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 1, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof, in particular at any position but position 3 of SEQ ID NO: 1;
  • HCDR2 the heavy chain CDR2 (HCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 2, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof, in particular at any position but positions 13, 14 and 16 of SEQ ID NO: 2;
  • HCDR3 the heavy chain CDR3 (HCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 3; optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof, in particular at any position but positions 2, 3, 7 and 8 of SEQ ID NO: 3;
  • the light chain CDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 4, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof, in particular at any position but positions 5, 6, 10, 11 and 16 of SEQ ID NO: 4;
  • LCDR2 the light chain CDR2 (LCDR2) comprises or consists of an amino acid sequence of SEQ ID NO: 5, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof;
  • LCDR3 the light chain CDR3 (LCDR3) comprises or consists of an amino acid sequence of SEQ ID NO: 6, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof at any position but positions 1, 4 and 6 of SEQ ID NO: 6.
  • the anti-PD-1 antigen-binding domain comprises or consists essentially of: (i) a heavy chain variable region (VH) comprising a CDR1 of SEQ ID NO: 1, a CDR2 of SEQ ID NO: 2 and a CDR3 of SEQ ID NO: 3; and (ii) a light chain variable region (VL) comprising a CDR1 of SEQ ID NO: 4, a CDR2 of SEQ ID NO: 5 and a CDR3 of SEQ ID NO: 6.
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-PDl antibody or antigen binding fragment according to the invention comprises framework regions, in particular heavy chain variable region framework regions (HFR) HFR1, HFR2, HFR3 and HFR4 and light chain variable region framework regions (LFR) LFR1, LFR2, LFR3 and LFR4.
  • HFR heavy chain variable region framework regions
  • LFR light chain variable region framework regions
  • the anti-PD-1 antigen-binding domain comprises or consists essentially of:
  • VH heavy chain variable region
  • HFR1 of SEQ ID NO : 7 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • a HCDR1 of SEQ ID NO: 1 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • a HCDR2 of SEQ ID NO: 2 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • a HCDR3 of SEQ ID NO: 3 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • a HCDR3 of SEQ ID NO: 3 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • a HCDR3 of SEQ ID NO: 3 optionally with one, two or three modification(
  • VL light chain variable region
  • LCDR1 of SEQ ID NO: 11 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • LCDR1 of SEQ ID NO: 4 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • LCDR2 of SEQ ID NO: 5 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof
  • LCDR3 of SEQ ID NO: 6 and a LFR4 of SEQ ID NO : 14 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the anti-PD-1 antigen-binding domain comprises or consists essentially of: (a) a heavy chain variable region (VH) comprising or consisting of an amino acid sequence SEQ ID NO: 15, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof, in particular at any position but positions 7, 16, 17, 20, 33, 38, 43, 46, 62, 63, 65, 69, 73, 76, 78, 80, 84, 85, 88, 93, 95, 96, 97, 98, 100, 101, 105, 106 and 112 of SEQ ID NO: 15;
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-PD-1 antigen-binding domain comprises or consists essentially of:
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-PD-1 antigen-binding domain comprises or consists essentially of:
  • VH heavy chain variable region
  • VL light chain variable region
  • the anti-PD-1 antigen-binding domain comprises VH, VL, CHI and a CL domains, so that the antigen binding domain is a Fab.
  • the heavy chain constant domain (CHI) comprises or consists essentially of SEQ ID NO: 17, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the light chain constant domain (CL) comprises or consists essentially of SEQ ID NO: 18 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the anti-PD-1 antigen-binding domain is a Fab or a Fab’, a Fab or a F(ab’)2 and comprises i) a VH domain and a CHI domain, said VH and CHI domains having the amino acid sequence as set forth in SEQ ID Nos: 15 and 17, respectively, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof; and ii) a VL domain and a CL domain, said domains having the amino acid sequence as set forth in SEQ ID Nos: 16 and 18, respectively, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the antigen binding domain is an anti-PD-1 Fab or F(ab’)2, comprising or consisting of i) a chain comprising or consisting of a VH domain and a CHI domain, said VH and CHI domains having the amino acid sequence as set forth in SEQ ID Nos: 15 and 17 respectively and ii) a chain comprising or consisting of VL and CL domains, said domains having the amino acid sequence as set forth in SEQ ID Nos: 16 and 18, respectively.
  • the antigen binding domain is an anti-PD-1 Fab or F(ab’)2, comprising or consisting of i) a chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 19 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof and of ii) a chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 20 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the antigen binding domain comprises an anti-PD-1 CrossMAb comprising or consisting of i) a chain comprising or consisting of VH and CL domains, said domains having the amino acid sequence as set forth in SEQ ID NOs: 15 and 18, respectively, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof; ii) a chain comprising or consisting of VL and CHI domains, said domains having the amino acid sequence as set forth in SEQ ID NOs: 16 and 17, respectively, optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the antigen binding domain comprises an anti-PD-1 CrossMAb, comprising or consisting of i) a chain comprising or consisting of a VH domain and a CL domain, said domains having the amino acid sequence as set forth in SEQ ID NOs: 15 and 18, respectively, and ii) a chain comprising or consisting of VL and CHI domains, said domains having the amino acid sequence as set forth in SEQ ID NOs: 16 and 17, respectively.
  • the antigen binding domain is an anti-PD-1 CrossMAb, comprising or consisting of i) a chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 21 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof and of ii) a chain comprising or consisting of an amino acid sequence as set forth in SEQ ID NO: 22 optionally with one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof.
  • the antigen binding domain when described as having one, two or three modification(s) selected from substitution(s), addition(s), deletion(s) and any combination thereof, said modifications are outside of the CDRs.
  • the antigen binding domain is an anti-PD-1 antigen-binding domain comprising:
  • HCDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 23,
  • HCDR2 comprises or consists of an amino acid sequence of SEQ ID NO: 24,
  • HCDR3 comprises or consists of an amino acid sequence of SEQ ID NO: 25,
  • LCDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 26,
  • LCDR2 comprises or consists of an amino acid sequence of SEQ ID NO: 27, and
  • LCDR3 comprises or consists of an amino acid sequence of SEQ ID NO: 28,
  • the antigen binding domain is an anti-PD-1 antigen-binding domain comprising: (a) a heavy chain variable region (VH) comprising or consisting of an amino acid sequence of SEQ ID NO: 29; (b) a light chain variable region (VL) comprising or consisting of an amino acid sequence of SEQ ID NO: 30.
  • VH heavy chain variable region
  • VL light chain variable region
  • HCDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 31,
  • HCDR2 comprises or consists of an amino acid sequence of SEQ ID NO: 32,
  • HCDR3 comprises or consists of an amino acid sequence of SEQ ID NO: 33,
  • LCDR1 comprises or consists of an amino acid sequence of SEQ ID NO: 34,
  • LCDR2 comprises or consists of an amino acid sequence of SEQ ID NO: 35, and
  • LCDR3 comprises or consists of an amino acid sequence of SEQ ID NO: 36,
  • the antigen binding domain is an anti-PD-1 antigen-binding domain comprising:
  • VH heavy chain variable region
  • VL light chain variable region
  • the antigen binding domain is an anti-PD-1 antibody, comprising or consisting of : an heavy chain comprising or consisting of a sequence as set forth in SEQ ID NO: 48 and a light chain comprising or consisting of a sequence as set forth in SEQ ID NO: 49; an heavy chain comprising or consisting of a sequence as set forth in SEQ ID NO: 50 and a light chain comprising or consisting of a sequence as set forth in SEQ ID NO: 51; or an heavy chain comprising or consisting of a sequence as set forth in SEQ ID NO: 52 and a light chain comprising or consisting of a sequence as set forth in SEQ ID NO: 53.
  • the antigen binding domain is an antagonist anti-PD-1 antibody comprising or consisting of an heavy chain comprising or consisting of a sequence as set forth in SEQ ID NO: 48 and a light chain comprising or consisting of a sequence as set forth in SEQ ID NO: 49.
  • variable regions of the antigen binding domain as described above may be associated with antibody constant regions, in particular from IgA, IgM, IgE, IgD or IgG such as IgGl, IgG2, IgG3, IgG4, preferably IgGl, IgG2, or IgG4.
  • the antigen binding domain comprises an IgG Fc region, preferably an IgGl, IgG2, or IgG4 Fc region.
  • the Fc domain includes all or a portion of a hinge region.
  • the hinge region can be derived from an immunoglobulin heavy chain, e.g., IgGl, IgG2, IgG3, IgG4, or other classes.
  • the hinge region is derived from a human or humanized IgGl, IgG2, IgG3 or IgG4, preferably from a human or humanized IgGl, IgG2, or IgG4.
  • the antigen binding domain comprises a Fc domain, preferably an IgG Fc domain.
  • the antigen binding domain is an antibody that comprises an IgG Fc domain and/or that the antigen binding domain comprises an antigen binding fragment of an antibody (such as a Fab or scFv) that is covalently linked to an IgG Fc domain.
  • the antigen binding domain comprises or is covalently linked to a Fc domain, preferably an IgG Fc domain.
  • the Fc region is derived from human or humanized IgGl, IgG2 or IgG4.
  • the antigen binding domain comprises a Fc domain and a hinge region derived from a human or humanized IgGl, IgG2 or IgG4.
  • the antigen binding domain comprises or is covalently linked to a Fc domain, preferably an IgG Fc domain.
  • the Fc region is derived from human or humanized IgGl, IgG2 or IgG4.
  • the antigen binding domain comprises a Fc domain and a hinge region derived from a human or humanized IgGl, IgG2 or IgG4.
  • IgGFc region or “IgGFc domain” is used to define the C-terminal region of an immunoglobulin heavy chain, including native sequence Fc region and variant Fc regions.
  • the human IgG heavy chain Fc region is generally defined as comprising the amino acid residue from position C226 or from P230 to the carboxyl-terminus of the IgG antibody.
  • the numbering of residues in the Fc region is that of the EU index of Kabat.
  • the constant regions may be further mutated or modified, by methods known in the art, for modifying their binding capability towards Fc receptor.
  • a Fc domain comprises two heavy chain constant domain, known as CH2 and CH3 domains.
  • the Fc domain envisioned herein also comprises a hinge region.
  • the antigen binding domain comprises a truncated Fc region or a fragment of the Fc region.
  • the Fc region includes a CH2 domain.
  • the Fc region includes CH2 and CH3 domains or includes hinge-CH2- CH3.
  • the Fc region can include all or a portion of the hinge region, a CH2 domain and/or a CH3 domain.
  • Antibodies or antigen-binding fragments thereof with amino acid sequences having at least 90%, for example, at least 95%, 96%, 97%, 98%, or 99% identity to any one of the above defined amino acid sequences are also part of the present disclosure.
  • the amino acid differences are conservative substitutions, i.e., substitutions of one amino acid with another having similar chemical or physical properties (size, charge or polarity), which substitution generally does not adversely affect the biochemical, biophysical and/or biological properties of the antibody or antigen-binding fragment thereof.
  • the substitution does not disrupt the interaction of the antibody or antigenbinding fragment thereof with its target.
  • Said conservative substitution(s) are advantageously chosen within one of the following five groups: Group 1 -small aliphatic, non-polar or slightly polar residues (A, S, T, P, G); Group 2-polar, negatively charged residues and their amides (D, N, E, Q); Group 3 -polar, positively charged residues (H, R, K); Group 4-large aliphatic, nonpolar residues (M, L, I, V, C); and Group 5-large, aromatic residues (F, Y, W).
  • the skilled person is able to determine the location of the various regions/domains of antibodies by reference to the standard definitions in this respect set forth, including a reference numbering system, a reference to the numbering system of KABAT or by application of the IMGT algorithm.
  • the delimitation of the regions/domains may vary from one reference system to another.
  • the regions/domains as defined in the present invention encompass sequences showing variations in length or localization of the concerned sequences within the full-length sequence of the variable domains of the antibodies, of approximately +/- 10%.
  • the lipid-based nanoparticle comprises an additional antigen binding domain which binds to a target specifically expressed on activated immune cell surface (i.e., in addition to the first antigen binding domain).
  • the lipid-based nanoparticle particularly comprises a first antigen binding domain which binds to a first target specifically expressed on activated immune cell surface and a second antigen binding domain which binds to a second target specifically expressed on activated immune cell surface.
  • the additional or second antigen binding domain is i) not covalently bound to any of the lipids of the lipid-based nanoparticle, ii) does not comprise any modification for coupling or grafting the antigen binding domain to a lipid and/or iii) is not covalently bound to a lipidation peptide or motif, in particular a lipidation peptide or motif such as described herein.
  • the additional or second antigen binding domain is not a secondary antibody, typically a secondary antibody that allows the attachment of the first antigen binding domain to the lipid-based nanoparticle (e.g., such as an anti-Fc antibody or antigen binding fragment or derivative thereof).
  • the additional or second antigen binding domain is not a secondary antibody comprising a lipidation peptide or motif such as described herein.
  • the first antigen binding domain and the additional (second) antigen binding domain bind to targets expressed on the surface of the same activated immune cells.
  • Said targets can be the same or different.
  • the first antigen binding domain binds for example to PD-1
  • the second antigen binding domain binds to an antigen selected from the group consisting of CD137/41BB/TNFRSF9, CRT AM, CTLA4, FasL/TNFSF6, TIM-3/HAVCR2, ITGAE, LAG-3, OX40/TNFRSF4, SIRPg and TIGIT.
  • both the first and second antigen binding domains may bind to PD-1.
  • the first and second antigen binding domains are not competing antibodies for PD-1, which means that the first and second antigen binding domains recognize a different (non-overlapping) epitope of PD-1.
  • each of the first and second targets specifically expressed on activated immune cell surface are selected from the group comprising CD4, CD8, BCMA/TNFRSF17, BTLA, CD101/IGSF2, CD103, CD119, CD137/4-1BB/TNFRSF9, CD150, CD153, CD154, CD223, CD226, CD25, CD254, CD26, CD27, CD275, CD39/ENTPD1, CD40L, CD44, CD45RO, CD45RC, LGR6, CD69, GPR18, GPR35, FPR2, CD80, CD83, CD86, CD95, CMKLR1, CRTAM, CST7, CTLA4, CXCR3, CXCR4, CXCR5, CXCR6, FasL/TNFSF6, GITR/TNFRSF18, GPR32, TIM3/HAVCR2, ICOS, IL18Rl/CXCRl/CD218a, ITGAE, LAG
  • Adding the additional antigen binding domain enhances the specificity of the lipid-based nanoparticle for a specific activated immune cell population, based on their nature and/or localization. Enhancing the already high specificity of the lipid-based nanoparticle of the invention limits or completely removes the risk of off-target or toxicity in healthy tissue and/or organs.
  • the first antigen binding domain and the second antigen binding domain are derived from Fab, Fab', F(ab')2, Fv, single chain (scFv), CrossMAb or nanobody (VHH), preferably a F(ab’)2, a Fab, a CrossMAb or a scFV and the first and second antigen binding domains may have the same or different formats.
  • the first antigen binding domain and the second antigen binding domain can be both Fab or scFV.
  • the first antigen binding domain and the second antigen binding domain are not both scFv.
  • the first antigen binding domain and the second antigen binding antigen binding domain comprise or are covalently linked to a Fc domain, preferably an IgG Fc domain such as described herein.
  • the first antigen binding domain and the second antigen binding domain may have the same or different formats.
  • the first antigen binding domain and the second antigen binding domain can be both antibodies, Fab or scFV.
  • the first antigen binding domain and the second antigen binding domain are not both scFv.
  • the lipid-based nanoparticle comprises 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 different antigen binding domains.
  • Such antigen binding domains can be two distinct antigen biding domains that recognize the same target (e.g., such as pembrolizumab and nivolumab that both binds to PD-1) or two distinct antigen biding domains that recognize two different target (e.g., such as PD-1 and CTLA-4).
  • additional targeting molecules may be used in the method of the invention.
  • modified sugars or modified lipids can be used in combination with the antigen binding domain of the invention.
  • the lipid-based nanoparticle of the invention comprises a mRNA molecule encoding an immune cell activity enhancing protein.
  • the lipid-based nanoparticle of the invention comprises one or more isolated mRNA molecule(s).
  • the lipid-based nanoparticle of the invention particularly includes an mRNA encoding a polypeptide of interest capable of being translated by the immune cell to produce the polypeptide of interest.
  • the lipid-based nanoparticle comprises a mRNA polynucleotide or a set of mRNA polynucleotides.
  • mRNA polynucleotide or a set of mRNA polynucleotides.
  • the technology of mRNA polynucleotide is now well-known by the person skilled in the art, as illustrated in WO21159130, the disclosure thereof being incorporated herein by reference.
  • the amount of a mRNA molecule in a lipid-based nanoparticle may depend on the size, composition, desired target and/or application, or other properties of the lipid-based nanoparticle.
  • the amount of mRNA useful in the lipid-based nanoparticle may also depend on the size, sequence, and other characteristics of the mRNA.
  • the relative amounts of a mRNA molecule and other elements (e.g., lipids) in a lipid-based nanoparticle may also vary.
  • the wt/wt ratio of the lipid component to a mRNA molecule in a lipid-based nanoparticle may be from about 5: 1 to about 60: 1, such as 5: 1, 6: 1, 7: 1, 8: 1, 9:1, 10: 1, 11 : 1, 12: 1, 13: 1, 14: 1, 15: 1, 16: 1, 17: 1, 18: 1, 19: 1, 20:1, 25: 1,30: 1,35: 1, 40: 1, 45:1, 50: 1, and 60:1.
  • the wt/wt ratio of the lipid component to a mRNA molecule may be from about 10: 1 to about 40: 1.
  • the wt/wt ratio is about 20:1.
  • the amount of a mRNA molecule in a lipid-based nanoparticle may, for example, be measured using absorption spectroscopy (e.g., ultraviolet-visible spectroscopy).
  • the amount of lipid and mRNA may be selected to provide a specific N:P ratio.
  • the N:P ratio of the composition refers to the molar ratio of nitrogen atoms in one or more lipids to the number of phosphate groups in an mRNA. In general, a lower N:P ratio is preferred.
  • the one or more mRNA, lipids, and amounts thereof may be selected to provide an N:P ratio from about 2: 1 to about 30: 1, such as 2: 1, 3: 1, 4: 1, 5: 1, 6: 1, 7: 1, 8:1, 9: 1, 10: 1, 12:1, 14: 1, 16:1, 18: 1, 20:1, 22: 1, 24:1, 26: 1, 28: 1, or 30: 1.
  • the N:P ratio may be from about 2: 1 to about 8: 1. In other aspects, the N:P ratio is from about 5: 1 to about 8:1.
  • the N:P ratio may be about 5.0: 1, about 5.5: 1, about 5.67: 1, about 6.0: 1, about 6.5: 1, or about 7.0: 1.
  • the N:P ratio may be about 5.67: 1.
  • the N:P ratio is between about 5: 1 and about 7: 1.
  • the N:P ratio is about 6: 1.
  • the efficiency of encapsulation of mRNA molecules describes the amount of mRNA encapsulated or otherwise associated with a LNP after preparation, relative to the initial amount provided.
  • the encapsulation efficiency is preferably desirably high (e.g., close to 100%).
  • the encapsulation efficiency may be measured, for example, by comparing the amount of mRNA in a solution containing the lipid-based nanoparticle before and after breaking up the lipid-based nanoparticle with one or more organic solvents or detergents. Fluorescence may be used to measure the amount of free mRNA in a solution.
  • the encapsulation efficiency may be of at least 50%, for example 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%.
  • the encapsulation efficiency is of at least 70%.
  • the encapsulation efficiency is of at least 80%, preferably 90%.
  • the mRNA molecule of the invention particularly comprises structural elements that allows its encapsulation into the lipid-based nanoparticle and/or its expression into the targeted immune cell.
  • the mRNA molecule contains stabilizing elements, including, but not limited to untranslated regions (UTR) at their 5'-end (5' UTR) and/or at their 3'-end (3' UTR), in addition to other structural features, such as a 5'-cap structure or a 3'-poly(A) tail.
  • mRNA polynucleotides comprise at least one base modification and at least one 5'-terminal cap.
  • mRNA envisioned herein typically include a region of linked nucleosides encoding a polypeptide of interest (e.g., a coding region), a first flanking region located at the 5'- terminus of the region of linked nucleosides (e.g., a 5’-UTR), a second flanking region located at the 3'-terminus of the region of linked nucleosides (e.g., a 3’-UTR), at least one 5'-cap region, and a 3 '-stabilizing region.
  • a region of linked nucleosides encoding a polypeptide of interest e.g., a coding region
  • a first flanking region located at the 5'- terminus of the region of linked nucleosides
  • a second flanking region located at the 3'-terminus of the region of linked nucleosides
  • at least one 5'-cap region e.g., a 3’-UTR
  • the mRNA of the invention includes a poly-A region or a Kozak sequence (e.g., in the 5’-UTR).
  • the mRNA of the invention comprises one or more intronic nucleotide sequences capable of being excised from the polynucleotide.
  • the mRNA of the invention includes a flanking region, a 5'-cap structure, a chain terminating nucleotide, a stem loop, a poly-A sequence and/or a polyadenylation signal.
  • the mRNA of the invention comprises a flanking region.
  • a 5'-UTR or a 3’- UTR may be provided as a flanking region to the mRNA of the invention.
  • a 5’-UTR may be homologous or heterologous to the coding region of the mRNA.
  • Multiple 5'- UTRs or 3’- UTRs may be included in the flanking region and may be of the same or of different sequences. Any portion of the flanking regions, including none, may be codon optimized and any may independently contain one or more different structural or chemical alterations, before and/or after codon optimization.
  • 5'-UTRs or 3 ’-UTRs which are heterologous to the coding region of an mRNA may be engineered.
  • the mRNA may then be administered to cells, tissue or organisms and outcomes such as protein level, localization, and/or halflife may be measured to evaluate the beneficial effects the heterologous 5'-UTR and/or 3’- UTR may have on the mRNA.
  • Variants of the 5'-UTRs and/or 3 ’-UTRs may be utilized wherein one or more nucleotides are added or removed to the termini, including A, T, C or G.
  • 5'-UTRs and/or 3’ -UTR may also be codon-optimized, or altered in any manner described herein.
  • the mRNA comprises an Internal ribosome entry site (IRES) or a Kozak sequence in the 5 ’-UTR region.
  • the Kozak consensus sequence (Kozak consensus or Kozak sequence) is a nucleic acid motif that functions as the protein translation initiation site.
  • An internal ribosome entry site (IRES) is an RNA element that allows for translation initiation in a cap-independent manner.
  • the mRNA of the invention comprises a 5 ’-capping region or structure.
  • the 5'-cap structure of a polynucleotide is involved in nuclear export and increasing polynucleotide stability and binds the mRNA Cap Binding Protein (CBP), which is responsible for polynucleotide stability in the cell and translation competency through the association of CBP with poly-A binding protein to form the mature cyclic mRNA species.
  • CBP mRNA Cap Binding Protein
  • the cap further assists the removal of 5'-proximal introns during mRNA splicing.
  • Endogenous polynucleotide molecules may be 5'-end capped generating a 5'-ppp-5'- triphosphate linkage between a terminal guanosine cap residue and the 5'-terminal transcribed sense nucleotide of the polynucleotide.
  • This 5'-guanylate cap may then be methylated to generate an N7-methyl-guanylate residue.
  • the ribose sugars of the terminal and/or anteterminal transcribed nucleotides of the 5'-end of the polynucleotide may optionally also be 2'-O-methylated.
  • 5'-decapping through hydrolysis and cleavage of the guanylate cap structure may target a polynucleotide molecule, such as an mRNA molecule, for degradation.
  • Alterations to polynucleotides may generate a non-hydrolyzable cap structure preventing decapping and thus increasing polynucleotide half-life. Because cap structure hydrolysis requires cleavage of 5'-ppp-5' phosphorodiester linkages, alternative nucleotides may be used during the capping reaction. For example, a Vaccinia Capping Enzyme from New England Biolabs (Ipswich, MA) may be used with a-thio-guanosine nucleotides according to the manufacturer's instructions to create a phosphorothioate linkage in the 5'-ppp-5' cap.
  • a Vaccinia Capping Enzyme from New England Biolabs (Ipswich, MA) may be used with a-thio-guanosine nucleotides according to the manufacturer's instructions to create a phosphorothioate linkage in the 5'-ppp-5' cap.
  • Additional alternative guanosine nucleotides may be used such as a-methyl-phosphonate and seleno-phosphate nucleotides. Additional alterations include, but are not limited to, 2'-O- methylation of the ribose sugars of 5'-terminal and/or 5'-anteterminal nucleotides of the polynucleotide (as mentioned above) on the 2'-hydroxy group of the sugar. Multiple distinct 5'-cap structures can be used to generate the 5'-cap of an mRNA molecule. mRNAs may also be capped post-transcriptionally, using enzymes, in order to generate more authentic 5'-cap structures.
  • the phrase "more authentic” refers to a feature that closely mirrors or mimics, either structurally or functionally, an endogenous or wild type feature. That is, a “more authentic” feature is better representative of an endogenous, wild-type, natural or physiological cellular function, and/or structure as compared to synthetic features or analogs of the prior art, or which outperforms the corresponding endogenous, wild-type, natural, or physiological feature in one or more respects.
  • Nonlimiting examples of more authentic 5'-cap structures useful in the polynucleotides of the present disclosure are those which, among other things, have enhanced binding of cap binding proteins, increased half-life, reduced susceptibility to 5'-endonucleases, and/or reduced 5'-decapping, as compared to synthetic 5'-cap structures known in the art (or to a wild-type, natural or physiological 5'-cap structure).
  • recombinant Vaccinia Virus Capping Enzyme or a Faustovirus Capping Enzyme and recombinant 2'-O- methyltransferase enzyme can create a canonical 5'-5'-triphosphate linkage between the 5'- terminal nucleotide of a polynucleotide and a guanosine cap nucleotide wherein the cap guanosine contains an N7-methylation and the 5'-terminal nucleotide of the polynucleotide contains a 2'-O-methyl.
  • Capl structure Such a structure is termed the Capl structure.
  • cap results in a higher translational-competency, cellular stability, and a reduced activation of cellular pro- inflammatory cytokines, as compared, e.g., to other 5'-cap analog structures known in the art.
  • Other exemplary cap structures include 7mG(5')ppp(5')N,pN2p (Cap 0), 7mG(5')ppp(5')NlmpNp (Cap 1), 7mG(5')-ppp(5')NlmpN2mp (Cap 2), and m(7)Gpppm(3)(6,6,2')Apm(2')Apm(2')Cpm(2)(3,2')Up (Cap 4).
  • 5'- terminal caps may include endogenous caps or cap analogs.
  • a 5'-terminal cap may include a guanosine analog.
  • guanosine analogs include inosine, Nl-methyl-guanosine, 2'- fluoro-guanosine, 7-deaza-guanosine, 8-oxo-guanosine, 2-amino-guanosine, LNA- guanosine, and 2-azido-guanosine.
  • a polynucleotide contains a modified 5'- cap. A modification on the 5'-cap may increase the stability of polynucleotide, increase the half-life of the polynucleotide, and could increase the polynucleotide translational efficiency.
  • the modified 5'-cap may include, but is not limited to, one or more of the following modifications: modification at the 2'- and/or 3'-position of a capped guanosine triphosphate (GTP), a replacement of the sugar ring oxygen (that produced the carbocyclic ring) with a methylene moiety (CH2), a modification at the triphosphate bridge moiety of the cap structure, or a modification at the nucleobase (G) moiety.
  • GTP capped guanosine triphosphate
  • CH2 methylene moiety
  • G nucleobase
  • 5'-capping of polynucleotides may be completed concomitantly during the in vitro- transcription reaction using the following chemical RNA cap analogs to generate the 5'- guanosine cap structure according to manufacturer protocols: 3'-O-Me-m7G(5')ppp(5') G [the ARCA cap]; G(5')ppp(5')A; G(5')ppp(5')G; m7G(5')ppp(5')A; m7G(5')ppp(5')G (New England BioLabs, Ipswich, MA).
  • 5'-capping of modified RNA may be completed post- transcriptionally using a Vaccinia Virus Capping Enzyme or a Faustovirus Capping Enzyme to generate the "Cap 0" structure: m7G(5')ppp(5')G (New England BioLabs, Ipswich, MA).
  • Cap 1 structure may be generated using both Vaccinia Vims Capping Enzyme or a Faustovirus Capping Enzyme and a 2'-O- methyl-transferase to generate: m7G(5')ppp(5')G- 2'-0-methyl.
  • Cap 2 structure may be generated from the Cap 1 structure followed by the 2'- O-methylation of the 5'-antepenultimate nucleotide using a 2'-Omethyl-transferase.
  • Cap 3 structure may be generated from the Cap 2 structure followed by the 2'-O-methylation of the 5'-preantepenultimate nucleotide using a 2'-O-methyl-transferase.
  • Enzymes may be derived from a recombinant source.
  • other caps may be used, such as CleanCap structures (Trilink). Cleancap is a trinucleotide with a 5'-m7Gjoined by a 5'-5' triphosphate linkage to an AG sequence.
  • the mRNA of the invention may particularly comprise a 5’-cap analog.
  • Cap analogs which herein are also referred to as synthetic cap analogs, chemical caps, chemical cap analogs, or structural or functional cap analogs, differ from natural (i.e., endogenous, wild-type, or physiological) 5'-caps in their chemical structure, while retaining cap function.
  • Cap analogs may be chemically (i.e., non-enzymatically) or enzymatically synthesized and/linked to a polynucleotide.
  • the Anti-Reverse Cap Analog (ARCA) cap contains two guanosines linked by a 5'-5'-triphosphate group, wherein one guanosine contains an N7- methyl group as well as a 3'-O-methyl group (i.e., N7, 3'-O-dimethyl-guanosine-5'- triphosphate-5'-guanosine, m7G-3'mppp-G, which may equivalently be designated 3'0-Me- m7G(5')ppp(5')G).
  • N7, 3'-O-dimethyl-guanosine-5'- triphosphate-5'-guanosine, m7G-3'mppp-G which may equivalently be designated 3'0-Me- m7G(5')ppp(5')G).
  • the 3'-0 atom of the other, unaltered, guanosine becomes linked to the 5'-terminal nucleotide of the capped polynucleotide (e.g., an mRNA).
  • the N7- and 3'-O- methylated guanosine provides the terminal moiety of the capped polynucleotide (e.g., mRNA).
  • Another exemplary cap is mCAP, which is similar to ARCA but has a 2'-O-methyl group on guanosine (i.e., N7,2'-O-dimethyl-guanosine-5'-triphosphate-5'-guanosine, m7Gm-ppp-G).
  • the 5 ’-cap of the mRNA may be a dinucleotide cap analog.
  • the dinucleotide cap analog may be modified at different phosphate positions with a boranophosphate group or a phophoroselenoate group such as the dinucleotide cap analogs described in US Patent No. 8,519,110, the cap structures of which are herein incorporated by reference.
  • a cap analog may be a N7-(4-chlorophenoxy-ethyl) substituted dinucleotide cap analog known in the art and/or described herein.
  • Non-limiting examples of N7-(4-chlorophenoxy-ethyl) substituted dinucleotide cap analogs include a N7-(4- chlorophenoxy-ethyl)-G(5)ppp(5’)G and a N7-(4-chlorophenoxy-ethyl)-m3'- OG(5)ppp(5')G cap analog (see, e.g., the various cap analogs and the methods of synthesizing cap analogs described in Kore et al. Bioorganic & Medicinal Chemistry 20132: 4570-4574; the cap structures of which are herein incorporated by reference).
  • a cap analog useful in the polynucleotides of the present disclosure is a 4- chloro/bromophenoxy-ethyl analog.
  • the mRNA of the invention includes a stem loop such as, but not limited to, a histone stem loop.
  • the histone stem loop may be before and/or after the poly-A region.
  • the mRNA including the histone stem loop and a poly-A region sequence may include a chain terminating nucleoside described herein.
  • the mRNA includes a histone stem loop and a 5'-cap structure such as described herein and/or known in the art.
  • the conserved stem loop region may include a miR sequence.
  • the stem loop region may include the seed sequence of a miR sequence.
  • the stem loop region may include a miR-122 seed sequence.
  • the stem loop is a nucleotide sequence of about 25 or about 26 nucleotides in length.
  • the histone stem loop may be located 3'-relative to the coding region (e.g., at the 3'- terminus of the coding region).
  • the stem loop may be located at the 3 '-end of a mRNA described herein.
  • an mRNA includes more than one stem loop (e.g., two stem loops).
  • a stem loop may be located in a second terminal region of a polynucleotide.
  • the stem loop may be located within an untranslated region (e.g., 3'-UTR) in a second terminal region.
  • a mRNA which includes the histone stem loop may be stabilized by the addition of a 3 ’-stabilizing region (e.g., a 3 ’-stabilizing region including at least one chain terminating nucleoside).
  • a 3 ’-stabilizing region e.g., a 3 ’-stabilizing region including at least one chain terminating nucleoside.
  • the addition of at least one chain terminating nucleoside may slow the degradation of a polynucleotide and thus can increase the half-life of the polynucleotide.
  • the mRNA, which includes the histone stem loop is stabilized by an alteration to the 3'-region of the polynucleotide that can prevent and/or inhibit the addition of oligo(U).
  • the mRNA, which includes the histone stem loop is stabilized by the addition of an oligonucleotide that terminates in a 3 '-deoxynucleoside, 2', 3'- dideoxynucleoside 3'-O-methylnucleosides, 3’-O- ethylnucleosides, 3 '-arabinosides, and other alternative nucleosides known in the art and/or described herein.
  • the mRNA may particularly include at least one histone stem-loop and a poly-A region or polyadenylation signal.
  • the mRNA comprised in the lipid-based nanoparticle of the invention includes a poly-A sequence and/or polyadenylation signal.
  • a poly-A sequence may be comprised entirely or mostly of adenine nucleotides or analogs or derivatives thereof.
  • a poly-A sequence may be a tail located adjacent to a 3 '-untranslated region of a nucleic acid.
  • mRNA messenger RNA
  • poly-A polymerase adds a chain of adenosine nucleotides to the RNA.
  • the process called polyadenylation, adds a poly-A region that is between 100 and 250 residues long.
  • Unique poly-A region lengths may provide certain advantages to the mRNAs of the present disclosure.
  • the length of a poly-A region of the present disclosure is at least 30 nucleotides in length.
  • the poly-A region is at least 35 nucleotides in length.
  • the length is at least 40 nucleotides.
  • the length is at least 45 nucleotides.
  • the length is at least 55 nucleotides.
  • the length is at least 60 nucleotides. In another embodiment, the length is at least 70 nucleotides. In another embodiment, the length is at least 80 nucleotides. In another embodiment, the length is at least 90 nucleotides. In another embodiment, the length is at least 100 nucleotides. In another embodiment, the length is at least 120 nucleotides. In another embodiment, the length is at least 140 nucleotides. In another embodiment, the length is at least 160 nucleotides. In another embodiment, the length is at least 180 nucleotides. In another embodiment, the length is at least 200 nucleotides. In another embodiment, the length is at least 250 nucleotides. In another embodiment, the length is at least 300 nucleotides.
  • the length is at least 350 nucleotides. In another embodiment, the length is at least 400 nucleotides. In another embodiment, the length is at least 450 nucleotides. In another embodiment, the length is at least 500 nucleotides. In another embodiment, the length is at least 600 nucleotides. In another embodiment, the length is at least 700 nucleotides. In another embodiment, the length is at least 800 nucleotides. In another embodiment, the length is at least 900 nucleotides. In another embodiment, the length is at least 1000 nucleotides. In another embodiment, the length is at least 1100 nucleotides. In another embodiment, the length is at least 1200 nucleotides.
  • the length is at least 1300 nucleotides. In another embodiment, the length is at least 1400 nucleotides. In another embodiment, the length is at least 1500 nucleotides. In another embodiment, the length is at least 1600 nucleotides. In another embodiment, the length is at least 1700 nucleotides. In another embodiment, the length is at least 1800 nucleotides. In another embodiment, the length is at least 1900 nucleotides. In another embodiment, the length is at least 2000 nucleotides. In another embodiment, the length is at least 2500 nucleotides. In another embodiment, the length is at least 3000 nucleotides.
  • the poly-A region may be 80 nucleotides, 120 nucleotides, 160 nucleotides in length on an mRNA molecule described herein. In other instances, the poly-A region is of 20, 40, 80, 100, 120, 140 or 160 nucleotides in length on an mRNA molecule described herein. In some cases, the poly-A region is designed relative to the length of the overall mRNA. This design may be based on the length of the coding region, the length of a particular feature or region of the mRNA or based on the length of the ultimate product expressed from the mRNA.
  • the poly-A region When relative to any feature of the mRNA (e.g., other than the mRNA portion which includes the poly-A region) the poly-A region may be 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% greater in length than the additional feature.
  • the poly-A region may also be designed as a fraction of the mRNA to which it belongs. In this context, the poly-A region may be 10, 20, 30, 40, 50, 60, 70, 80, or 90% or more of the total length of the construct or the total length of the construct minus the poly-A region.
  • the mRNA includes a poly-A-G quartet.
  • the G-quartet is a cyclic hydrogen bonded array of four guanosine nucleotides that can be formed by G-rich sequences in both DNA and RNA.
  • the G-quartet is incorporated at the end of the poly-A region.
  • the resultant mRNA may be assayed for stability, protein production and other parameters including half-life at various time points. It has been discovered that the poly-A-G quartet results in protein production equivalent to at least 75% of that seen using a poly-A region of 120 nucleotides alone.
  • the mRNA with a poly-A-G quartet may further include a 5'-cap structure.
  • the 3 '-stabilizing region which may be used to stabilize mRNA includes a poly-A region or poly-A-G quartet.
  • the 3 '-stabilizing region which may be used with the present disclosure include a chain termination nucleoside such as 3'-deoxyadenosine (cordycepin), 3 '-deoxyuridine, 3'- deoxycytosine, 3 '-deoxy guanosine, 3 '-deoxythymine, 2',3'-dideoxynucleosides, such as 2', 3'- dideoxyadenosine, 2', 3 '-dideoxyuridine, 2', 3 '-dideoxy cytosine, 2', 3'- dideoxyguanosine, 2', 3 '-dideoxythymine, a 2'-deoxynucleoside, or an O-methylnucleoside.
  • a chain termination nucleoside such as 3'-deoxyadenosine (cord
  • mRNA which includes a poly-A region or a poly-A-G quartet may be stabilized by an alteration to the 3'-region of the polynucleotide that can prevent and/or inhibit the addition of oligo(U).
  • mRNA which includes a poly-A region or a poly-A-G quartet may be stabilized by the addition of an oligonucleotide that terminates in a 3'- deoxynucleoside, 2',3'-dideoxynucleoside 3 -O-m ethylnucleosides, 3'-O-ethylnucleosides, 3 '-arabinosides, and other alternative nucleosides known in the art and/or described herein.
  • engineered binding sites and/or the conjugation of mRNA for poly-A binding protein is used to enhance expression.
  • the engineered binding sites may be sensor sequences which can operate as binding sites for ligands of the local microenvironment of the mRNA.
  • the mRNA may include at least one engineered binding site to alter the binding affinity of poly-A binding protein (PABP) and analogs thereof.
  • PABP poly-A binding protein
  • multiple distinct mRNA molecules may be linked together to the PABP (poly-A binding protein) through the 3 '-end using alternative nucleotides at the 3'- terminus of the poly-A region. While not wishing to be bound by theory, the poly-A region recruits PABP which in turn can interact with translation initiation complex and thus may be useful for protein synthesis.
  • the mRNA molecule of the invention may include one or more naturally occurring components, including any of the canonical nucleotides A (adenosine), G (guanosine), C (cytosine), U (uridine), or T (thymidine).
  • Any one of the regions of a nucleic acid may include one or more alternative components (e.g., an alternative nucleoside) which impart useful properties including increased stability and/or the lack of a substantial induction of the innate immune response of a cell into which the polynucleotide is introduced.
  • a modRNA may exhibit reduced degradation in a cell into which the modRNA is introduced, relative to a corresponding unaltered mRNA.
  • Non-limiting examples of such non-naturally occurring modified nucleotides and nucleosides can be found, inter alia, in published patent application Nos. WO2013052523; WO2014093924; W02015051173; W02015051169; W02015089511; W02015196130; WO2015196118; WO2015196128; or WO2017153936 all of which are incorporated by reference herein.
  • nucleotide analogs or other alteration(s) may be located at any position(s) of a polynucleotide such that the function of the polynucleotide is not substantially decreased.
  • An alteration may also be a 5'- or 3'-terminal alteration.
  • the polynucleotide includes an alteration at the 3 '-terminus.
  • the mRNA may contain from about 1% to about 100% alternative nucleotides (either in relation to overall nucleotide content, or in relation to one or more types of nucleotide, i.e., any one or more of A, G, U or C) or any intervening percentage (e.g., from 1% to 20%, from 1% to 25%, from 1% to 50%, from 1% to 60%, from 1% to 70%, from 1% to 80%, from 1% to 90%, from 1% to 95%, from 10% to 20%, from 10% to 25%, from 10% to 50%, from 10% to 60%, from 10% to 70%, from 10% to 80%, from 10% to 90%, from 10% to 95%, from 10% to 100%, from 20% to 25%, from 20% to 50%, from 20% to 60%, from 20% to 70%, from 20% to 80%, from 20% to 90%, from 20% to 95%, from 20% to 100%, from 50% to 60%, from 50% to 70%, from 50% to 80%, from 50% to 90%, from 50% to 95%, from 50% to 100%, from 70% to 80%
  • the mRNA may contain at a minimum zero and at maximum 100% alternative nucleotides, or any intervening percentage, such as at least 5% alternative nucleotides, at least 10% alternative nucleotides, at least 25% alternative nucleotides, at least 50% alternative nucleotides, at least 80% alternative nucleotides, or at least 90% alternative nucleotides.
  • polynucleotides may contain an alternative pyrimidine such as an alternative uracil or cytosine.
  • At least 5%, at least 10%, at least 25%, at least 50%, at least 80%, at least 90% or 100% of the uracil in a polynucleotide is replaced with an alternative uracil (e.g., a 5-substituted uracil).
  • the alternative uracil can be replaced by a compound having a single unique structure or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • cytosine in the polynucleotide is replaced with an alternative cytosine (e.g., a 5-substituted cytosine).
  • the alternative cytosine can be replaced by a compound having a single unique structure or can be replaced by a plurality of compounds having different structures (e.g., 2, 3, 4 or more unique structures).
  • the nucleobase is an alternative uracil.
  • Exemplary nucleobases and nucleosides having an alternative uracil include pseudouridine (y), pyridin-4-one ribonucleoside, 5-aza-uracil, 6-aza-uracil, 2-thio-5-aza-uracil, 2-thio-uracil (s2U), 4-thio- uracil (s4U), 4-thio-pseudouridine, 2-thio-pseudouridine, 5 -hydroxy -uracil (ho5U), 5- aminoallyl-uracil, 5-halo-uracil (e.g., 5-iodo-uracil or 5 -bromo-uracil), 3-methyl-uracil (mU), 5 -methoxy -uracil (mo5U), uracil 5-oxyacetic acid (cmo5U), uracil 5-oxyacetic acid methyl ester (mcmo5U),
  • the nucleobase is an alternative cytosine.
  • Exemplary nucleobases and nucleosides having an alternative cytosine include 5 -aza-cytosine, 6-aza-cytosine, pseudoisocytidine, 3-methyl-cytosine (m3C), N4-acetyl-cytosine (ac4C), 5-formyl- cytosine (f5C), N4-methyl-cytosine (m4C), 5-methyl-cytosine (m5C), 5-halo- cytosine (e.g., 5- iodocytosine), 5-hydroxymethyl-cytosine (hm5C), 1-methyl-pseudoisocytidine, pyrrolo- cytosine, pyrrolo-pseudoisocytidine, 2-thio-cytosine (s2C), 2-thio-5-methyl-cytosine, 4- thio-pseudoisocytidine, 4-thio- 1 -methyl-
  • the nucleobase is an alternative adenine.
  • Exemplary nucleobases and nucleosides having an alternative 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-adenine, 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-methy 1 -adenine (ml A), 2-methyl-adenine (m2A), N6-methyl- adenine (m6A), 2-methyl
  • the nucleobase is an alternative guanine.
  • Exemplary nucleobases and nucleosides having an alternative guanine include inosine (I), 1-methyl-inosine (mil), wyosine (imG), methylwyosine (mimG), 4-demethyl-wyosine (imG- 14), isowyosine (imG2), wybutosine (yW), peroxywybutosine (o2yW), hydroxywybutosine (OHyW), undermodified hydroxywybutosine (OHyW*), 7-deaza-guanine, queuosine (Q), epoxy queuosine (oQ), galactosyl-queuosine (galQ), mannosyl- queuosine (manQ), 7-cyano- 7-deaza-guanine (preQO), 7-aminomethyl-7-deaza- guanine (preQi), archa
  • the alternative nucleobase of a nucleotide can be independently a purine, a pyrimidine, a purine or pyrimidine analog.
  • the nucleobase can be an alternative to adenine, cytosine, guanine, uracil, or hypoxanthine.
  • the nucleobase can also include, for example, naturally-occurring and synthetic derivatives of a base, including pyrazolo[3,4- d]pyrimidines, 5-methylcytosine (5-me-C), 5 -hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2 -propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2 -thiocytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo (e.g., 8-bromo), 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxy and other 8-substitute
  • each letter refers to the representative base and/or derivatives thereof, e.g., A includes adenine or adenine analogs, e.g., 7-deaza adenine).
  • the mRNA can be a circular RNA, in particular a circular mRNA, especially as described in WO2014/186334 and WO2022/261490.
  • the lipid-based nanoparticle of the invention comprises one or more different mRNA molecule encoding an immune cell activating protein.
  • immune cell activating protein or “immune cell enhancing protein” refer to a protein that induces, increases, enhances or boosts the activity of immune cells or activates immune cells. These proteins typically play critical roles in augmenting the immune response, improving the ability of the immune system to detect and eliminate pathogens, infected cells, or abnormal cells, such as cancer cells. Immune cell enhancing proteins can act by promoting the proliferation, activation, or function of various types of immune cells, including T cells, B cells, natural killer cells, macrophages, dendritic cells, and others. They are known to be crucial for mounting effective immune responses against infections and tumors.
  • the lipid-based nanoparticle may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mRNA molecule(s) encoding 1, 2, 3, 4, 5, 6,7 8, 9 or 10 different immune cell activating protein(s), respectively.
  • the lipid-based nanoparticle comprises mRNA molecules encoding 2 different immune cell activating proteins such as disclosed herein.
  • the immune cell activating protein can be selected from a single or from different cell types.
  • the mRNA molecules may encode different T cells activating proteins.
  • the mRNA molecules may encode different immune cell activating protein, for example for a T cell activating protein and for another immune cell activating protein such as a Natural killer activating protein.
  • the immune cell enhancing protein encoded by a mRNA molecule included in the lipid- based nanoparticle of the invention may particularly be selected based on the type of immune cell targeted by the antigen binding domain comprised in the lipid-based nanoparticle.
  • the immune cell targeted is an activated T-cell (i.e., so that the lipid-based nanoparticle comprises an antigen binding domain that binds to a target expressed on activated T cells such as PD-1)
  • the mRNA encodes an activated T cell enhancing protein.
  • the immune cell enhancing protein encoded by a mRNA molecule included in the lipid-based nanoparticle of the invention may particularly be selected based on the desired effect.
  • the immune cell enhancing protein may be selected for a particular indication, condition, disease, or disorder.
  • the immune cell enhancing protein exhibits an effect on activated immune cells selected from the group consisting of:
  • TAE tumor microenvironment
  • the immune cell enhancing protein inducing or increasing the sternness of immune cells is selected from the group comprising or consisting of TCF1, LEF1, WNT, FRIZZLED and Beta catenin.
  • the immune cell enhancing protein inducing or increasing the proliferation or renewal of immune cells is selected from the group comprising or consisting of LRP6, CYCLIN, TOP2A, MUCL1 and MDM2.
  • the immune cell enhancing protein inhibiting or decreasing the apoptosis of immune cells is selected from the group comprising or consisting of BCL2, BCLXL, BIRC3 and MCLl .
  • the immune cell enhancing protein increasing the production or concentration of mitochondrial enzymes and/or transporters of immune cells is PGCla.
  • the immune cell enhancing protein increasing the production or concentration of transcription factors of immune cells is selected from the group comprising or consisting of TCF7, NF AT, NFKB, RORgt, TRAM, TBK1, TBET, EOMES, RUNX3, GATA3, JUNB, POU2AF1, OCTI, BLIMP- 1, XBP-1 and FOXO1.
  • the immune cell enhancing protein inhibiting or decreasing hypoxia of the microenvironment or tissue is PTGS2.
  • the immune cell enhancing protein increasing the production or concentration of metabolism enzymes of immune cells is selected from the group comprising or consisting of CSE, Glutl, Glut3, HK2, FOXO1, arginine resynthesis enzymes, argininosuccinate synthase (ASS), Ornithine transcarb amylase (OTC), GYS.
  • the immune cell enhancing protein increasing the production or concentration of metabolism enzymes of immune cells is selected from the group comprising or consisting of CSE, Glutl, Glut3, HK2, FOXO1, arginine resynthesis enzymes, argininosuccinate synthase (ASS), Ornithine transcarbamylase (OTC), GYS, OXPHOS.
  • the immune cell enhancing protein increasing the efficiency of signaling pathways of immune cells is selected from the group comprising or consisting of AKT, PLC, SMAD, Blys, BTK and BLK.
  • the immune cell enhancing protein inducing or increasing the production of cytotoxic proteins by immune cells is selected from the group comprising or consisting of CD 107a, Lymphotoxin (LT) aip2, granzyme B, perforin.
  • the immune cell enhancing protein inducing or increasing internalization of endosome is POU2F1.
  • the immune cell enhancing protein increasing the production or concentration of chaperone protein of immune cells is selected from the group comprising or consisting of BBS10, BBS12, TCP1 and HSP.
  • the immune cell enhancing protein inducing or increasing the production of cytoskeleton regulation protein, the migration and/or motility of immune cells is selected from the group comprising or consisting of Integrin alpha 1, Integrin alpha 2, Integrin alpha 2b, Integrin alpha 11, Integrin alpha 3, Integrin alpha 6, Integrin alpha7, Integrin alpha E, Integrin beta 2, Integrin beta 4, LFA-1, LFA-2, LFA-3, Integrin beta 1, Integrin beta 7, CD103, Integrin alpha V, ITGAE, CD11C, CRTAM, CXCR5, CXCR3, CCR7, SELL, GAL3, Laminin, Actin, Vimentin, DEFI, Dynein, Kinesin, Rab protein.
  • the immune cell enhancing protein inducing or increasing the degradation of protein through immuno-proteasome and ubiquitination, as well as the antigen presentation of immune cells is selected from the group comprising or consisting of NLRP3, TAP, LAMP, Ubiquitin ligase, CD74, Peptidase, Calreticulin and Aurora.
  • the immune cell enhancing protein inducing or increasing the inflammation of TME is LGR6.
  • the immune cell enhancing protein modifying the epigenetic of immune cells is selected from the group comprising or consisting of HAT, KDM1, TGD, TET1.
  • the immune cell enhancing protein increasing the anergy resistance of immune cells is selected from the group comprising or consisting of C-FOS, JUN, EGR-2, EGR-3.
  • the mRNa molecule encodes for a protein selected in Table E.
  • the effect of an immune cell enhancing protein on the activated immune cells may be determined when, in presence of the immune cell enhancing protein of the invention, activated immune cells exhibits a greater activity compared to activated immune cells under the same experimental conditions but without the presence of the immune cell enhancing protein.
  • Said sample may be an immune cell culture, from a sample of at least one healthy patient or from a sample of at least one patient in need of a treatment as detailed hereunder. Immune cell activity can be measured by any method known to the person skilled in the art.
  • the enhancement of immune cell activity may be measured by comparing the immune cell activity of a population of immune cells obtained from a sample, without the immune cell enhancing protein to be assessed, to the immune cell activity of a population of immune cells obtained from a sample and treated with the immune cell enhancing protein to be assessed.
  • the immune cell enhancing protein is an intracellular protein having an intracellular effect on the activated immune cell or a transmembrane protein, preferably an intracellular protein.
  • intracellular proteins in combination with the targeting of a specific subset of immune cells, preferentially T cells, even more preferably TILs, results in a very specific and efficient enhancement of said immune cells.
  • Potent intracellular proteins may be used, with an elevated activation and/or proliferation of immune cells in a specific environment.
  • the activated immune cell is a regulatory T cell
  • the immune cell activity enhancing protein is selected to induce a phenotype switch from the regulatory T cell to a T cell type having an anti-tumoral effect, such as a cytotoxic or effector T cell.
  • a “protein that have an intracellular effect on the activated immune cell” or an “intracellular protein” refers to a protein which is produced/expressed inside a cell and which does not enter the extracellular medium, either alone or in a vesicle, nor that is expressed in the cellular membrane.
  • the intracellular protein is thus contained in the boundaries of the cellular membrane, and acts in one of the cellular compartments (e.g. cytosol, endoplasmic reticulum, mitochondria, nucleus, etc).
  • cytosol cytosol, endoplasmic reticulum, mitochondria, nucleus, etc.
  • Such protein can be present in any of the cellular compartments, such as nucleus, interci sternal space, organelles or cytosol.
  • the protein can be a cytoplasmic protein, a nuclear protein or a mitochondrial protein, or an interci sternal protein, preferably nuclear or cytoplasmic protein.
  • the immune cell intracellular protein can be an enzyme, an intracellular signaling protein or a transcription factor, preferably a transcription factor.
  • the transmembrane protein is selected from the group comprising FRIZZLED, BCLXL, CCR4, CCR10, CXCR3, CCR10, CCR5, CCR2, CX3CR1, CCR7, CXCR4, CXCR3, CXCR5, CRTAM, CCR7, CXCR5, GPR35, GPR37 and TAP.
  • the immune cell enhancing protein is a transcription factor.
  • a “transcription factor” refers to a DNA-binding protein that regulates the transcription of a gene.
  • the transcription factor is selected from the group consisting of RORgt, SOCS, NFKB, TRAM, RIPK1, and TBK1 and a variant thereof having at least 80% of identity with the wildtype protein or having 1 to 10 modifications selected from the group consisting of addition, deletion, substitution and combinations thereof.
  • the transcription factor is selected from the group consisting of RORgt, NFKB, TRAM and TBK1 and a variant thereof having at least 80% of identity with the wildtype protein or having 1 to 10 modifications selected from the group consisting of addition, deletion, substitution and combinations thereof.
  • the transcription factor is selected from the group consisting of RORgt, NFKB, TRAM and TBK1 and a variant thereof having at least 80% of identity with the wildtype protein or having 1 to 10 modifications selected from the group consisting of addition, deletion, substitution and combinations thereof.
  • the transcription factor is selected from the group consisting TCF7, NF AT, NFKB, RORgt, TRAM, TBK1, TBET, EOMES, RUNX3, GATA3, JUNB, POU2AF1, OCTI, BLIMP- 1, XBP-1 and FOXO1 and any combinations thereof.
  • the immune cell enhancing protein is an enzyme.
  • such enzyme can be phosphatidylinositol-3 -kinase (PI3K).
  • the immune cell enhancing protein is an intracellular signaling protein.
  • intracellular signaling molecules can be D3 -phosphoinositides and derivatives of phosphatidylinositol such as phosphorylated at the D-3 position of the inositol ring and encompasses the compounds phosphatidylinositol-(3)-monophosphate (PtdIns(3)P), phosphatidylinositol(3,4)-bisphosphate (PtdIns(3,4)P2), and phosphatidylinositol(3,4,5)-trisphosphate (PtdIns(3,4,5)P3). Therefore, the intracellular signaling protein can be those involved in the synthesis of these molecules. I l l
  • the immune cell activating protein is not a chimeric antigen receptor (CAR).
  • the immune cell activating protein is not a CAR, a T cell receptor (TcR) (i.e., including TCR alpha, TCR beta, CD3 and CD247) or a B cell receptor (BcR).
  • TcR T cell receptor
  • BcR B cell receptor
  • the one or more mRNA molecule(s) does not encode a cytokine and/or a chemokine.
  • the immune cell enhancing protein is a cytokine receptor.
  • the cytokine receptor is selected from the group consisting of IL7R, CD122, CD132, CD25, CD215, IL12R, IL17R, IL8R, IL21R, IL11R, IL18R, IL10R, IL1R and IL6R.
  • the immune cell enhancing protein is a chemokine receptor.
  • the chemokine receptor is selected from the group consisting of CXCR3, CXCR5, CXCR4, CXCR1, CXCR2, CXCR6, CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, CCR11, CX3CR1 and XCR1.
  • the immune cell enhancing protein is a lectin receptor.
  • the lectin receptor is selected from the group consisting of DECTIN-1, CLEC-9A, CLEC-2, DECTIN-2, MCL, MINCLE and BDCA-2.
  • the immune cell enhancing protein is an engineered surface receptor or anchored membrane cytokine.
  • the engineered surface receptor or anchored membrane cytokine is selected from the group consisting of IL12A, IL12B, IFNG, IFNa, IL21, IL7, IL2, IL15 and IL18.
  • the immune cell enhancing protein is a costimulation receptor or ligand.
  • the costimulation receptor or ligand is selected from the group consisting of ICOS, ICOSL, CD28, CD80, CD86, CD70, CD40L, CD226, GITR, GITRL, 4-1BB, 4- 1BBL, 0X40, OX40L, CD155, LIGHT, HVEM, CD30, CD30L, SLAM CD2 family, CD27, TL1A, DR3, TM1, TIM4, CD150, CD48, CD58, CD112, BAFFR, BCMA, TACI, KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5, DAP12, KIR3DS1 and CD160.
  • the activity-enhancing protein is selected from the group consisting of TCF1, LEF1, WNT, FRIZZLED, Beta catenin, LRP6, CYCLIN, TOP2A, MUCL1, MDM2, BCL2, BCLXL, BIRC3, MCL1, PGCla, TCF7, NF AT, NFKB, RORgt, TBET, EOMES, RUNX3, GATA3, JUNB, POU2AF1, OCTI, BLIMP-1, XBP-1, FOXO1, PTGS2, CSE, Glutl, Glut3, HK2, FOXO1, arginine resynthesis enzymes, argininosuccinate synthase (ASS), Ornithine transcarbamylase (OTC), GYS, AKT, PLC, STAT, SMAD, Blys, BTK, BLK, CD 107a, Lymphotoxin (LT) aip2, granzyme B, perforin, POU2F
  • the activity-enhancing protein is selected from the group consisting of: TCF1, WNT, BCL2, BCLXL, TBET, Glutl, LGR6, ICOS, CD28, CD40L, 4- IBB, Perforin, CXCL9, CXCL10, GrB, OXPHOS, Integrin alpha 1, Integrin alpha 2, Integrin alpha 2b, Integrin alpha 11, Integrin alpha 3, Integrin alpha 6, Integrin alpha?, Integrin alpha E, Integrin beta 2, Integrin beta 4, Integrin beta 1, Integrin beta 7 and Integrin alpha V.
  • the activity-enhancing protein is selected from the group consisting of: TCF1, BCL2, IL7, IL7R, CXCL9 and CXCL10, preferably from BCL2, IL7, IL7R, CXCL9 and CXCL10.
  • the mRNA molecule encoding for BLC2 comprises or consists of a nucleic acid sequence as set forth in SEQ ID NO: 43 or of a nucleic acid sequence having at least 80%, 85%, 90%, 95% or 97% sequence identity thereto.
  • the mRNA molecule encoding for IL7 comprises or consists of a nucleic acid sequence as set forth in SEQ ID NO: 44 or of a nucleic acid sequence having at least 80%, 85%, 90%, 95% or 97% sequence identity thereto.
  • the mRNA molecule encoding for IL7R comprises or consists of a nucleic acid sequence as set forth in SEQ ID NO: 45 or of a nucleic acid sequence having at least 80%, 85%, 90%, 95% or 97% sequence identity thereto.
  • the mRNA molecule encoding for CXCL9 comprises or consists of a nucleic acid sequence as set forth in SEQ ID NO: 46 or of a nucleic acid sequence having at least 80%, 85%, 90%, 95% or 97% sequence identity thereto.
  • the mRNA molecule encoding for CXCL10 comprises or consists of a nucleic acid sequence as set forth in SEQ ID NO: 47 or of a nucleic acid sequence having at least 80%, 85%, 90%, 95% or 97% sequence identity thereto.
  • the mRNA molecule encodes for a protein selected in Table F. Table F.
  • the immune cell enhancing protein is a protein inhibiting a compound, molecule or protein that inhibits the proliferation of immune cells, the migration of immune cells, the motility of immune cells, the active membrane transport of immune cells, the production or concentration of carrier protein of immune cells, the production or concentration of metabolism enzymes of immune cells, the production or concentration of transcription factors of immune cells, the polarization of immune cells, the production or concentration of membrane receptor of immune cells, the antigen presentation of immune cells or the production of cytotoxic compounds by immune cells (granzyme, perforin and the like).
  • the immune cell enhancing protein is specific of T cells.
  • the T cell enhancing compound is selected from the group comprising T cell transcription factor, T cell growth factors, in particular growth factors to increase number and repertoire of naive immune cells, agonists to activate and stimulate immune cells, inhibitors of T cell checkpoint blockade, T cell growth factors to increase the growth and survival of immune T cells and T cell stimulating membrane protein.
  • the invention concerns a lipid-based nanoparticle comprising an antigenbinding domain capable of specifically binding to a target expressed on activated T cells surface and one or several mRNA molecule(s) encoding an activity-enhancing protein of said T cells.
  • the invention concerns a lipid-based nanoparticle comprising an anti-PD-1 antigen-binding domain and one or several mRNA molecule(s) encoding an activityenhancing protein of said T cells.
  • the invention concerns a lipid-based nanoparticle comprising an anti- PD-1 antigen-binding domain and one or several mRNA molecule(s) encoding T cells transcription factors, T cell growth factors and T cell stimulating membrane protein.
  • the lipid-based nanoparticle of the invention may comprise several mRNA molecules encoding different immune cell activity enhancing proteins.
  • the lipid-based nanoparticle comprises a first mRNA molecule encoding a T cell activity enhancing protein and a second mRNA molecule encoding a second immune cell activity enhancing protein.
  • the second immune cell activity enhancing protein may be another T cell activity enhancing protein (i.e., different form the first T cell activity enhancing component) or can be an activity enhancing component of another type of immune cell, such as a NK or a macrophage.
  • the lipid-based nanoparticle comprises at least two mRNA molecule(s), wherein one of said at least two mRNA molecules encodes a transmembrane protein that is a receptor and another of said at least two mRNA molecules encodes a secreted protein that is a ligand of said receptor.
  • the lipid-based nanoparticle comprises a mRNA encoding an interleukin receptor (e.g., IL7-R) and a mRNA encoding the associated interleukin (e.g., IL- 7). This allows the activation at the level of the same/single immune cell (i.e., cis-activation).
  • IL7-R interleukin receptor
  • IL- 7 mRNA encoding the associated interleukin
  • IL2Ralpha IL2Ralpha
  • IL2Rbeta IL2Rgamma
  • IL2Rgamma IL2Ralpha
  • the lipid-based nanoparticle of the invention comprises a mRNA encoding IL7-R and a mRNA encoding IL-7.
  • Interleukin receptors and ligands are detailed in Table G hereunder.
  • the mRNa molecule encodes for a protein selected in Table G.
  • the mRNa molecule encodes for a protein as described in Tables E, F and/or G.
  • Table G Interleukin receptors
  • the lipid-based nanoparticle comprises at least two mRNA molecule(s), wherein one of said at least two mRNA molecules encodes a costimulatory molecule, preferably selected from the group consisting of CD28, CD80, CD86, ICOS, ICOSL, 0X40, OX40L, CD40, CD40L, GITRL, CD137 and CD137L and another of said at least two mRNA molecules encodes another immune cell enhancing compound, such as an intracellular protein or another transmembrane protein.
  • a costimulatory molecule preferably selected from the group consisting of CD28, CD80, CD86, ICOS, ICOSL, 0X40, OX40L, CD40, CD40L, GITRL, CD137 and CD137L
  • another immune cell enhancing compound such as an intracellular protein or another transmembrane protein.
  • the lipid-based nanoparticle comprises one or more mRNA molecule selected from the group consisting of: a mRNA encoding for BCL2, preferably a mRNA molecule comprising a nucleic acid sequence such as described in SEQ ID NO: 43 or having at least 80%, 90%, 95% or 99% sequence identity thereto; a mRNA encoding for IL7, preferably a mRNA molecule comprising a nucleic acid sequence such as described in SEQ ID NO: 44 or having at least 80%, 90%, 95% or 99% sequence identity thereto; a mRNA encoding for IL7R, preferably a mRNA molecule comprising a nucleic acid sequence such as described in SEQ ID NO: 45 or having at least 80%, 90%, 95% or 99% sequence identity thereto; a mRNA encoding for CXCL9, preferably a mRNA molecule comprising a nucleic acid sequence such as described in SEQ ID NO: 46 or having at least 80%,
  • the effect of an immune cell enhancing protein on the immune cells may be determined when, in presence of the immune cell enhancing protein of the invention, immune cells exhibits a greater activity compared to immune cells under the same experimental conditions but without the presence of the immune cell enhancing protein.
  • Immune cell activity can be measured by any method known to the person skilled in the art.
  • the enhancement of immune cell activity may be measured by comparing the immune cell activity of a population of immune cells obtained from a sample, without the immune cell enhancing protein to be assessed, to the immune cell activity of a population of immune cells obtained from a sample and treated with the immune cell enhancing compound to be assessed.
  • the invention particularly concerns a lipid-based nanoparticle comprising: a) a lipid based composition comprising:
  • Cholesterol from about 35 mol% to about 45 mol%, preferably from about 37 mol% to about 40 mol %, more preferably of about 38.5 mol% of the total lipids present in the LNP, and
  • the invention particularly concerns a lipid-based nanoparticle comprising: a lipid based composition comprising:
  • an antigen binding domain that specifically binds to PD-1, CTLA4, 4- IBB, ICOS, LAG3, TIM3, TIGIT, BTLA or CLEC-1, preferably to PD-1; one or more mRNA molecule that encodes for a protein selected from the group consisting of TCF1, LEF1, WNT, FRIZZLED, Beta catenin, LRP6, CYCLIN, TOP2A, MUCL1, MDM2, BCL2, BCLXL, BIRC3, MCL1, PGCla, TCF7, NF AT, NFKB, RORgt, TBET, EOMES, RUNX3, GATA3, JUNB, POU2AF1, OCTI, BLIMP-1, XBP-1, FOXO1, PTGS2, CSE, Glutl, Glut3, HK2, FOXO1, arginine resynthesis enzymes, argin
  • the invention particularly concerns a lipid-based nanoparticle comprising: a lipid based composition comprising:
  • an antigen binding domain that specifically binds to PD-1, CTLA4, 4- IBB, ICOS, LAG3, TIM3, TIGIT, BTLA or CLEC-1, preferably to PD-1; one or more mRNA molecule that encodes for a protein selected from the group consisting of TCF1, LEF1, WNT, FRIZZLED, Beta catenin, LRP6, CYCLIN, TOP2A, MUCL1, MDM2, BCL2, BCLXL, BIRC3, MCL1, PGCla, TCF7, NF AT, NFKB, RORgt, TBET, EOMES, RUNX3, GATA3, JUNB, POU2AF1, OCTI, BLIMP-1, XBP-1, FOXO1, PTGS2, CSE, Glutl, Glut3, HK2, FOXO1, arginine resynthesis enzymes, argin
  • the invention particularly concerns a lipid-based nanoparticle comprising: a lipid based composition comprising:
  • an antigen binding domain that specifically binds to preferably to PD-1; one or more mRNA molecule that encodes for a protein selected from the group consisting of BCL2, IL-7, IL7-R, CXCL9 and CXCL10.
  • the mRNA molecule(s) encodes for an antigen or an antigen fragment.
  • the antigen or fragment thereof is preferably selected among any molecule that is expressed by any viral, bacterial, or parasitic pathogen prior to or during entry into, colonization of, or replication in their host. These pathogens can be infectious in humans, domestic animals or wild animal hosts.
  • the antigen or fragment thereof encoded by the nucleic acid molecule(s) of the invention promote protective immunity.
  • the antigen is a protective antigen or a fragment thereof.
  • any viral, bacterial, or parasitic molecule that elicits an immunological response that results in long-term acquired immune resistance in an host is called a "protective antigen”.
  • the nucleic acid molecule encodes for a viral antigen or a fragment thereof.
  • the viral pathogens, from which the viral antigens are derived include, but are not limited to: Orthomyxoviruses, such as influenza virus; Retroviruses, such as RSV, HTLV-1, and HTLV-II, Herpesviruses such as EBV; CMV or herpes simplex virus; Lentiviruses, such as HIV-1 and HIV-2; Rhabdoviruses, such as rabies virus; Picornaviruses, such as Poliovirus; Poxviruses, such as vaccinia virus; Rotavirus; and Parvoviruses, such as Adeno- Associated Viruses (AAV); Betacoronaviruses, such as SARS-CoV, SARS-CoV 2 and MERS-CoV.
  • Orthomyxoviruses such as influenza virus
  • Retroviruses such as RSV, HTLV-1, and HTLV-II
  • protective antigens of viral pathogens include the Human Immunodeficiency Virus (HIV) antigens Rev, Pol, Nef, Gag, Env, Tat, mutant derivatives of Tat, such as Tat- A31-45, T- and B-cell epitopes of gpl20, chimeric derivatives of HIV-1 Env and gpl20, such as a fusion between gpl20 and CD4, a truncated or modified HIV-1 Env, such as gpl40 or derivatives of HIV-1 Env and/or gpl40.
  • HIV Human Immunodeficiency Virus
  • hepatitis B surface antigen examples include hepatitis B surface antigen, rotavirus antigens, such as VP4 and VP7, influenza virus antigens such as hemagglutinin, neuraminidase, or nucleoprotein, and herpes simplex virus antigens such as thymidine kinase.
  • rotavirus antigens such as VP4 and VP7
  • influenza virus antigens such as hemagglutinin, neuraminidase, or nucleoprotein
  • herpes simplex virus antigens such as thymidine kinase.
  • the nucleic acid molecule encodes for a bacterial antigen or a fragment thereof.
  • bacterial pathogens from which the bacterial antigens may be derived, include but are not limited to, Mycobacterium spp., Helicobacter pylori, Salmonella spp., Shigella spp., E. coli, Rickettsia spp., Listeria spp., Legionella pneumoniae, Fansicella spp., Pseudomonas spp., Vibrio spp., and Boreilia burgdorferi.
  • protective antigens of bacterial pathogens include the somatic antigens of enterotoxigenic E. coli, such as the CFA/I fimbrial antigen and the nontoxic B-subunit of the heat-labile toxin; pertactin of Bordetella pertussis, adenylate cyclase-hemolysin of B.
  • pertussis fragment C of tetanus toxin of Clostridium tetani, OspA of Boreilia burgdorferi, protective paracrystalline-surface-layer proteins of Rickettsia prowazekii and Rickettsia typhi, the listeriolysin (also known as “Lio” and “Hly”) and/or the superoxide dismutase (also known as "SOD" and "p60”) of Listeria monocytogenes, urease of Helicobacter pylori, and the receptor-binding domain of lethal toxin and/or the protective antigen of Bacillus anthrax.
  • the nucleic acid molecule encodes for a parasitic antigen or a fragment thereof.
  • the parasitic pathogens, from which the parasitic antigens are derived include but are not limited to: Plasmodium spp. such as Plasmodium falciparum, Trypanosome spp. such as Trypanosoma cruzi, Giardia spp. such as Giardia intestinalis, Boophilus spp., Babesia spp. such as Babesia microti, Entamoeba spp. such as Entamoeba histolytica, Eimeria spp.
  • Eimeria maxima Leishmania spp., Schistosome spp., Brugia spp., Fascida spp., Dirofilaria spp., Wuchereria spp., and Onchocerea spp.
  • protective antigens of parasitic pathogens include the circumsporozoite (CS) or Liver Stage Specific (LSA) antigens LSA-1 and LSA-3 of Plasmodium spp. such as those of P. bergerii or P. falciparum, or immunogenic mutants thereof; the merozoite surface antigen of Plasmodium spp., the galactose specific lectin of Entamoeba histolytica, gp63 of Leishmania spp., gp46 of Leishmania major, paramyosin of Brugia malayi, the triosephosphate isomerase of Schistosoma mansoni, the secreted globin-like protein of Trichostrongylus colubriformis, the glutathione-S-transferase of Frasciola hepatica, Schistosoma bovis and S. japonicum, and KLH of Schistosoma bovis and S. japonicum.
  • CS circumspor
  • the antigen or fragment thereof may be encoded by a codon-optimized, synthetic gene and may be constructed using conventional recombinant DNA methods.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a lipid-based nanoparticle as described hereabove, preferably as the active ingredient or compound and optionally a pharmaceutically acceptable carrier or excipient.
  • a “pharmaceutical composition” refers to a preparation of one or more of the active agents, such as comprising a LNP according to the invention, with optional other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of the active agent to an organism.
  • Compositions of the present invention can be in a form suitable for any conventional route of administration or use.
  • a “composition” typically intends a combination of the active agent, e.g., compound or composition, and a naturally-occurring or non-naturally-occurring carrier, inert or active, such as an adjuvant, diluent, binder, stabilizer, buffers, preservative or the like and include pharmaceutically acceptable carriers.
  • An "acceptable vehicle” or “acceptable carrier” as referred to herein, is any known compound or combination of compounds that are known to those skilled in the art to be useful in formulating pharmaceutical compositions.
  • compositions can be sterilized and, if desired, mixed with auxiliary agents such as pharmaceutically acceptable carriers, excipients, salts, antioxidant and/or stabilizers which do not deleteriously interact with the lipid-based nanoparticle of the invention and does not impart any undesired toxicological effects.
  • auxiliary agents such as pharmaceutically acceptable carriers, excipients, salts, antioxidant and/or stabilizers which do not deleteriously interact with the lipid-based nanoparticle of the invention and does not impart any undesired toxicological effects.
  • the pharmaceutical composition according to the invention can be formulated for any conventional route of administration including a topical, enteral, oral, parenteral, intranasal, intravenous, intramuscular, subcutaneous or intraocular administration and the like.
  • the lipid-based nanoparticle as described herein can particularly be made into a pharmaceutical composition for in vivo administration.
  • the means of making such a composition have been described in the art (see, for instance, Remington: The Science and Practice of Pharmacy, Lippincott Williams & Wilkins, 21st edition (2005)).
  • the pharmaceutical composition is administered intranodally or intratum orally.
  • the pharmaceutical composition may be prepared by mixing a lipid-based nanoparticle having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients, antioxidant, and/or stabilizers in the form of lyophilized formulations or aqueous solutions.
  • suitable carriers, excipients, antioxidants, and/or stabilizers are well known in the art and have been for example described in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • the pharmaceutical composition comprising the lipid-based nanoparticle is relatively homogenous.
  • a poly dispersity index may be used to indicate the homogeneity of the composition, e.g., the particle size distribution of the lipid-based nanoparticles comprised in the composition.
  • a small (e.g., less than 0.3) poly dispersity index generally indicates a narrow particle size distribution.
  • the pharmaceutical composition has a poly dispersity index from about 0 to about 0.25, such as 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.20, 0.21, 0.22, 0.23, 0.24, or 0.25.
  • the poly dispersity index of the pharmaceutical composition is from about 0.10 to about 0.25.
  • compositions according to the invention may be formulated to release the active ingredients (e.g. the lipid-based nanoparticle of the invention) substantially immediately upon administration or at any predetermined time or time period after administration.
  • the pharmaceutical composition in some aspects can employ time-released, delayed release, and sustained release delivery systems such that the delivery of the composition occurs prior to, and with sufficient time to cause, sensitization of the site to be treated. Means known in the art can be used to prevent or minimize release and absorption of the composition until it reaches the target tissue or organ, or to ensure timed-release of the composition. Such systems can avoid repeated administrations of the composition, thereby increasing convenience to the subject and the physician.
  • the formulations of the invention may be isotonic with human blood that is the formulations of the invention have essentially the same osmotic pressure as human blood.
  • Such isotonic formulations generally have an osmotic pressure from about 250 mOSm to about 350 mOSm. Isotonicity can be measured by, for example, a vapor pressure or ice-freezing type osmometer.
  • composition typically must be sterile and stable under the conditions of manufacture and storage. Prevention of presence of microorganisms may be ensured both by sterilization procedures (for example by microfiltration), and/or by the inclusion of various antibacterial and antifungal agents.
  • the pharmaceutical composition includes one or more pharmaceutically acceptable excipients or accessory ingredients such as, but not limited to, one or more solvents, dispersion media, diluents, dispersion aids, suspension aids, surface active agents, buffering agents and/or preservatives.
  • one or more solvents such as, but not limited to, one or more solvents, dispersion media, diluents, dispersion aids, suspension aids, surface active agents, buffering agents and/or preservatives.
  • Surface active agents and/or emulsifiers may include, but are not limited to, natural emulsifiers (e.g., acacia, alginic acid, sodium alginate, cholesterol, and lecithin), sorbitan fatty acid esters (e.g., polyoxyethylene sorbitan monolaurate [TWEEN®20], polyoxyethylene sorbitan [TWEEN® 60], polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], sorbitan monostearate [SPAN®60], sorbitan tristearate [SPAN®65], glyceryl monooleate, sorbitan monooleate [SPAN®80]), polyoxyethylene esters (e.g., polyoxyethylene monostearate [MYRJ® 45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and SOLUTOL®), sucrose fatty
  • preservatives may include, but are not limited to, antioxidants, chelating agents, free radical scavengers, antimicrobial preservatives, antifungal preservatives, alcohol preservatives and/or acidic preservatives.
  • antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxy toluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite and/or sodium sulfite.
  • chelating agents include, but are not limited to, ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid and/or trisodium edetate.
  • EDTA ethylenediaminetetraacetic acid
  • citric acid monohydrate disodium edetate
  • dipotassium edetate dipotassium edetate
  • edetic acid fumaric acid, malic acid
  • phosphoric acid sodium edetate
  • tartaric acid tartaric acid and/or trisodium edetate.
  • antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and/or thimerosal.
  • antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate and/or sorbic acid.
  • alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, benzyl alcohol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol.
  • acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroascorbic acid, ascorbic acid, sorbic acid, and/or phytic acid.
  • preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisole (BHA), butylated hydroxy toluene (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, GLYDANT PLUS®, PHENONIP®, methylparaben, GERMALL® 115, GERMAB EN®II, NEOLONETM, KATHONTM and/or EUXYL®.
  • An exemplary free radical scavenger includes butylated hydroxytoluene (BHT or butylhydroxytoluene) or deferoxamine.
  • buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, d-gluconic acid, calcium glycerophosphate, calcium lactate, calcium lactobionate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, amino-sulfonate buffers (e.g., HEPES), magnesium
  • the pharmaceutical composition including a lipid-based nanoparticle according to the invention further includes a salt, such as a chloride salt.
  • the pharmaceutical composition including a lipid-based nanoparticle further includes a sugar such as a disaccharide.
  • the pharmaceutical composition further includes a sugar but not a salt, such as a chloride salt.
  • a pharmaceutical composition further includes one or more small hydrophobic molecules such as a vitamin (e.g., vitamin A or vitamin E) or a sterol.
  • Carbohydrates may include simple sugars (e.g., glucose) and polysaccharides (e.g., glycogen and derivatives and analogs thereof).
  • Formulations comprising amphiphilic polymers and lipid-based nanoparticles may be formulated in whole or in part as pharmaceutical compositions.
  • Pharmaceutical compositions may particularly include one or more amphiphilic polymers and one or more lipid-based nanoparticles.
  • a pharmaceutical composition may include one or more amphiphilic polymers and one or more lipid-based nanoparticles including one or more different mRNA therapeutics and/or prophylactics.
  • excipients and accessory ingredients may be used in said pharmaceutical composition, except insofar as any conventional excipient or accessory ingredient may be incompatible with one or more components of the lipid-based nanoparticle(s) or amphiphilic polymer(s).
  • the pharmaceutical composition comprises between 0.1% and 15% (wt/vol) of one or more amphiphilic polymers (e.g., 0.5%, 1%, 2.5%, 5%, 10%, or 12.5% w/vol).
  • one or more amphiphilic polymers e.g. 0.5%, 1%, 2.5%, 5%, 10%, or 12.5% w/vol.
  • Relative amounts of the lipid-based nanoparticles, pharmaceutically acceptable excipients and/or any additional ingredients in a pharmaceutical composition in accordance with the present disclosure may vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the pharmaceutical composition comprises between 0.1% and 100% (wt/wt) of one or more lipid-based nanoparticles such as disclosed herein.
  • the amount of lipid-based nanoparticles which can be combined with a carrier material to produce a single dosage form will generally be that amount of the lipid-based nanoparticles which produces a therapeutic effect.
  • one or more excipients or accessory ingredients may make up greater than 50% of the total mass or volume of the pharmaceutical composition including a lipid- based nanoparticle such as disclosed herein.
  • the one or more excipients or accessory ingredients may make up 50%, 60%, 70%, 80%, 90%, or more of a pharmaceutical composition.
  • a pharmaceutically acceptable excipient is at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% pure.
  • an excipient is approved for use in humans and/or for veterinary use.
  • an excipient is approved by United States Food and Drug Administration.
  • an excipient is pharmaceutical grade.
  • an excipient meets the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
  • the pharmaceutical composition comprises 2, 3, 4, 5, 6, 7, 8, 9 or 10 different lipid-based nanoparticles comprising an antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface and comprising one or several mRNA molecule(s) encoding an immune cell activity enhancing component.
  • different lipid-based nanoparticles it is meant lipid-based nanoparticles that differ from i) the lipid composition of the nanoparticle, ii) the antigen binding domain (e.g., different sequences and/or target) and/or iii) the mRNA molecule(s).
  • the pharmaceutical composition comprises at least two different lipid-based nanoparticles comprising an antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface and comprising one or several mRNA molecule(s) encoding an immune cell activity enhancing component.
  • the pharmaceutical composition comprises at least two different lipid-based nanoparticles comprising an antigen binding domain capable of specifically binding to a target expressed on activated immune cells surface and comprising one or several mRNA molecule(s) encoding an immune cell activity inhibiting component, wherein the antigen binding domain is an antibody or an antigen binding fragment thereof and preferably comprises a Fc domain , preferably an IgG Fc domain.
  • the pharmaceutical composition comprises at least two different lipid-based nanoparticles as described above, wherein none of the lipid-based nanoparticles comprises one or more antigen binding domain(s) that i) is/are not covalently bound to any of the lipids of the lipid-based nanoparticle, ii) does/do not comprise any modification for coupling or grafting the antigen binding domain to a lipid and/or iii) is/are not covalently bound to a lipidation peptide or motif.
  • composition according to the invention may comprise:
  • a first lipid-based nanoparticle comprising a first antigen binding domain capable of specifically binding to a first target expressed on activated immune cells surface and one or several mRNA molecule(s) encoding an immune cell activity enhancing component;
  • a second lipid-based nanoparticle comprising a second antigen binding domain capable of specifically binding to a second (different) target expressed on activated immune cells surface and one or several mRNA molecule(s) encoding an immune cell activity enhancing component.
  • the one or several mRNA molecule(s) encoding an immune cell activity enhancing component may be the same or different in the first and second lipid-based nanoparticles.
  • the antigen binding domain of the lipid-based nanoparticle and the antigen binding domain of the additional lipid-based nanoparticle are capable of specifically binding to the same target (e.g., PD-1) while each having mRNA molecule(s) encoding for a different immune cell activity enhancing component, respectively.
  • the antigen binding domain of the first lipid-based nanoparticle and the antigen binding domain of the additional (second) lipid-based nanoparticle are capable of specifically binding to a different/non overlapping epitope of the same target (e.g., different epitopes of PD-1).
  • the antigen binding domain of the first lipid-based nanoparticle and the antigen binding domain of the additional (second) lipid-based nanoparticle are capable of specifically binding to the same epitope of the target (e.g., the same epitope on PD-1).
  • the lipid-based nanoparticle and the pharmaceutical composition comprising such as defined above have numerous in vitro and in vivo utilities and applications. Particularly, the lipid-based nanoparticles and the pharmaceutical compositions provided herein may be used in therapeutic methods and/or for therapeutic purposes.
  • the present invention relates to a lipid-based nanoparticle or a pharmaceutical composition comprising such for use as a medicament or vaccine and/or for use in the treatment of a disorder or disease, such as a cancer or an infection.
  • lipid-based nanoparticle or a pharmaceutical composition comprising such for treating a disease or disorder, such as a cancer or an infection, in a subject. It also concerns the use a lipid-based nanoparticle or pharmaceutical composition as disclosed herein in the manufacture of a medicament for treating a disease or disorder, such as a cancer or an infection, in a subject.
  • the present invention relates to the use of one or more lipid-based nanoparticle(s) according to the invention or of the pharmaceutical composition according to the invention, for the manufacture of a medicament for the treatment of a cancer or an infectious disease.
  • the present invention relates to a method for treating a a cancer or an infectious disease in a subject, wherein the method comprises administering to said subject one or more lipid-based nanoparticle(s) according to the invention or the pharmaceutical composition according to the invention.
  • a method for treating a disease or a disorder, such as a cancer or an infection, in a subject comprising administering a therapeutically effective amount of a pharmaceutical composition or a lipid-based nanoparticle such as disclosed herein.
  • the invention relates to a method of treatment of a disease and/or disorder selected from the group consisting of a cancer, an infectious disease and a chronic viral infection in a subject in need thereof, comprising administering to said subject an effective amount of the lipid-based nanoparticle or pharmaceutical composition as defined above.
  • a disease and/or disorder selected from the group consisting of a cancer, an infectious disease and a chronic viral infection in a subject in need thereof, comprising administering to said subject an effective amount of the lipid-based nanoparticle or pharmaceutical composition as defined above. Examples of such diseases and disorders are more particularly described hereafter.
  • the invention concerns a treatment method that comprises: (a) identifying a patient in need of treatment; and (b) administering to the patient a therapeutically effective amount of the lipid-based nanoparticle or pharmaceutical composition described herein.
  • an effective amount” or a “therapeutic effective amount” as used herein refers to the amount of active agent (i.e., the lipid-based nanoparticle disclosed herein) required to confer therapeutic effect on the subject, either alone or in combination with one or more other active agents, e.g., the amount of active agent that is needed to treat the targeted disease or disorder, or to produce the desired effect.
  • the “effective amount” will vary depending on the agent(s), the disease and its severity, the characteristics of the subject to be treated including age, physical condition, size, gender and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art.
  • the invention provides a lipid-based nanoparticle or a pharmaceutical composition for use in the treatment of a subject having a cancer.
  • cancer as used herein is defined as disease characterized by the rapid and uncontrolled growth of aberrant cells. Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body.
  • the invention provides the use a lipid-based nanoparticle or pharmaceutical composition as disclosed herein in the manufacture of a medicament for treating a cancer.
  • the invention provides a method of treating a cancer in a subject, comprising administering to the subject a therapeutically effective amount of the lipid-based nanoparticle or pharmaceutical composition of the invention, preferably such that the subject is treated from cancer.
  • the present invention relates to the treatment of a subject using a lipid-based nanoparticle or pharmaceutical composition of the invention such that immune response, especially the CTL response, is enhanced and growth of cancerous cells is inhibited.
  • the lipid-based nanoparticle or composition disclosed herein are for use in the treatment of a subject suffering from cancer with a poor prognosis.
  • the invention also provides the use of a lipid-based nanoparticle or pharmaceutical composition as disclosed herein in the manufacture of a medicament for treating a subject suffering from cancer with a poor prognosis. It also concerns a method of treating a cancer in a subject with a poor prognosis comprising administering to the subject a therapeutically effective amount of the lipid-based nanoparticle or pharmaceutical composition of the invention.
  • the term “poor prognosis” refers to a decreased subject survival and/or an early cancer progression and/or an increased or early cancer recurrence and/or an increased risk or occurrence of metastasis. Particularly, the poor prognosis is correlated with a cancer in which a population of Treg cells is present in the tumor or wherein the Treg/Teff ratio is high in the tumor (Chraa et al., 2018 J Leukoc Biol. 2018; 1—13).
  • the cancer to be treated is associated with exhausted T cells.
  • Preferred cancers for treatment include cancers typically responsive to immunotherapy.
  • preferred cancers for treatment are cancers non-responsive to immunotherapy.
  • any suitable cancer may be treated with the lipid-based nanoparticle provided herein, such as hematopoietic cancer or solid cancer.
  • Such cancers include carcinoma, cervical cancer, colorectal cancer, esophageal cancer, gastric cancer, gastrointestinal cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, lymphoma, glioma, mesothelioma, melanoma, stomach cancer, urethral cancer environmentally induced cancers and any combinations of said cancers.
  • the present invention is also useful for the treatment of metastatic cancers.
  • the present invention is also useful for the treatment of refractory or recurrent malignancies.
  • the cancer to be treated is selected from the group consisting of metastatic or not metastatic, Melanoma, malignant mesothelioma, Non-Small Cell Lung Cancer, Renal Cell Carcinoma, Hodgkin's Lymphoma, Head and Neck Cancer, Urothelial Carcinoma, Colorectal Cancer, Hepatocellular Carcinoma, Small Cell Lung Cancer, Metastatic Merkel Cell Carcinoma, Gastric or Gastroesophageal cancers and Cervical Cancer.
  • metastatic or not metastatic Melanoma
  • Melanoma malignant mesothelioma
  • Non-Small Cell Lung Cancer Renal Cell Carcinoma
  • Hodgkin's Lymphoma Head and Neck Cancer
  • Urothelial Carcinoma Colorectal Cancer
  • Hepatocellular Carcinoma Small Cell Lung Cancer
  • Metastatic Merkel Cell Carcinoma Gastric or Gastroesophageal cancers and Cervical Cancer.
  • the cancer is a hematologic malignancy or a solid tumor with high expression of PD-1 and/or PD-L1.
  • a cancer can preferably be selected from the group consisting of hematolymphoid neoplasms, angioimmunoblastic T cell lymphoma, myelodysplastic syndrome, acute myeloid leukemia.
  • the cancer is a cancer induced by a virus or associated with immunodeficiency.
  • a cancer can be selected from the group consisting of Kaposi sarcoma (e.g., associated with Kaposi sarcoma herpes virus); cervical, anal, penile and vulvar squamous cell cancer and oropharyngeal cancers (e.g., associated with human papilloma virus); B cell non-Hodgkin lymphomas (NHL) including diffuse large B cell lymphoma, Burkitt lymphoma, plasmablastic lymphoma, primary central nervous system lymphoma, HHV-8 primary effusion lymphoma, classic Hodgkin lymphoma, and lymphoproliferative disorders (e.g., associated with Epstein-Barr virus (EBV) and/or Kaposi sarcoma herpes virus); hepatocellular carcinoma (e.g., associated with hepatitis B and/or C viruses); Merkel cell carcinoma (e.g.,
  • the cancer is a PD-L1 negative cancer.
  • the lipid-based nanoparticles or pharmaceutical compositions are for use in the treatment of an infectious disease or for use in the treatment of patients that have been exposed to toxins or pathogens.
  • the invention also provides the use of a lipid-based nanoparticle or pharmaceutical composition as disclosed herein in the manufacture of a medicament for treating an infectious disease.
  • an aspect of the invention provides a method of treating an infectious disease in a subject comprising administering to the subject a therapeutically effective amount of a lipid-based nanoparticle according to the present invention, or a pharmaceutical composition comprising such, preferably such that the subject is treated for the infectious disease.
  • Any suitable infection may be treated with a lipid-based nanoparticle or pharmaceutical composition according to the present invention.
  • the infectious disease is a chronic viral infection.
  • pathogenic viruses causing infections treatable by methods of the invention include HIV, hepatitis (A, B, or C), herpes virus (e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr virus), adenovirus, influenza virus, flaviviruses, echovirus, rhinovirus, coxsackie virus, coronavirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus and arboviral encephalitis virus.
  • herpes virus e.g., VZV, HSV-1, HAV-6, HSV-II, and CMV, Epstein Barr virus
  • adenovirus e.g., influenza virus, flaviviruses, echovirus, rhinovirus, coxsacki
  • the lipid-based nanoparticles or pharmaceutical compositions of the invention are used to treat patients that have chronic viral infection, such infection being caused by viruses selected from the group consisting of Retroviruses, Anellovirus, Circovirus, Herpesvirus, Varicella zoster virus (VZV), Cytomegalovirus (CMV), Epstein-Barr virus (EBV), Polyomavirus BK, Polyomavirus, Adeno-associated virus (AAV), Herpes simplex type 1 (HSV-1), Adenovirus, Herpes simplex type 2 (HSV-2), Kaposi's sarcoma herpesvirus (KSHV), Hepatitis B virus (HBV), GB virus C, Papilloma virus, Hepatitis C virus (HCV), Human immunodeficiency virus (HIV), Hepatitis D virus (HDV), Human T cell leukemia virus type 1 (HTLV1), Xenotropic murine leukemia virus-related virus (XMLV), Rubella virus
  • pathogenic bacteria causing infections treatable by methods of the invention include chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumonococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, legionella, diphtheria, salmonella, bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lymes disease bacteria.
  • pathogenic fungi causing infections treatable by methods of the invention include Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum.
  • pathogenic parasites causing infections treatable by methods of the invention include Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi, and Nippostrongylus brasiliensis.
  • the lipid-based nanoparticle or the pharmaceutical composition according to the invention may be used in combination with another therapeutic agent or therapy, in particular for the treatment of cancer, of an infectious disease or of a chronic viral infection.
  • the present invention also relates to a method for treating a disease, such as a cancer or an infectious disease, in a subject, comprising administering to said subject a therapeutically effective amount of the lipid-based nanoparticle or the pharmaceutical composition described herein and a therapeutically effective amount of an additional or second therapeutic agent or therapy.
  • a disease such as a cancer or an infectious disease
  • the lipid-based nanoparticle according to the invention can particularly be combined with some other potential strategies for overcoming immune evasion mechanisms with agents in clinical development or already on the market (see for example Table 1 from Antonia et al. Immuno-oncology combinations: a review of clinical experience and future prospects. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 20, 6258-6268, 2014). Such combination with the lipid-based nanoparticle according to the invention may be useful notably for:
  • lipid-based nanoparticle or pharmaceutical composition comprising such, as described herein and a suitable second agent, for the treatment of a disease or disorder.
  • the lipid-based nanoparticle and the second agent can be present in a unique pharmaceutical composition.
  • the terms “combination therapy” or “combined therapy”, as used herein embrace administration of these two agents (e.g., a lipid-based nanoparticle as described herein and an additional or second suitable therapeutic agent) in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the agents, in a substantially simultaneous manner. Sequential or substantially simultaneous administration of each agent can be affected by any appropriate route.
  • the agents can be administered by the same route or by different routes.
  • a first agent e.g., a lipid-based nanoparticle
  • an additional therapeutic agent e.g., an anti-cancer agent, an anti-infection agent; or an immune modulator
  • an agent of the combination selected may be administered by intravenous injection while the other agents of the combination may be administered intramuscularly.
  • the additional therapeutic agent can be selected in the non-exhaustive list comprising alkylating agents, angiogenesis inhibitors, antibodies, antimetabolites, antimitotic, antiproliferative, antivirals, aurora kinase inhibitors, apoptosis promoters (for example, Bcl-2 family inhibitors), activators of death receptor pathway, Bcr-Abl kinase inhibitors, BiTE (Bi-Specific T cell Engager) antibodies, antibody drug conjugates, biologic response modifiers, Bruton's tyrosine kinase (BTK) inhibitors, cyclin-dependent kinase inhibitors, cell cycle inhibitors, cyclooxygenase-2 inhibitors, leukemia viral oncogene homolog (ErbB2) receptor inhibitors, growth factor inhibitors, heat shock protein (HSP)-90 inhibitors, histone deacetylase (HDAC) inhibitors, hormonal therapies, inhibitors of inhibitors of apoptosis proteins (IAPS), intercal
  • the additional agent is an anti-cancer agent.
  • treatment with an anti-cancer antibody or an anti-cancer immunoconjugate or other current anti-cancer therapy that lead to cancer cell death would potentiate an immune response against tumor cells.
  • an anti-cancer treatment may include a lipid-based nanoparticle combined with an anti-cancer treatment, concurrently or sequentially or any combination thereof, which may potentiate an anti-tumor immune response by the host.
  • a lipid-based nanoparticle may be used in combination with other immunogenic agents, standard cancer treatments, or other antibodies.
  • the additional therapeutic agent can be selected in the group consisting of chemotherapy, radiotherapy, targeted therapy, anti angiogenic agents, hypomethylating agents, cancer vaccines, epitopes or neoepitopes from tumor antigens, myeloid checkpoints inhibitors, other immunotherapies, and HD AC inhibitors.
  • the invention relates to a combined therapy as defined above, wherein the second therapeutic agent is particularly selected from the group consisting of therapeutic vaccines, immune checkpoint blockers or activators, in particular of adaptive immune cells (T and B lymphocytes) and antibody-drug conjugates.
  • the second therapeutic agent is particularly selected from the group consisting of therapeutic vaccines, immune checkpoint blockers or activators, in particular of adaptive immune cells (T and B lymphocytes) and antibody-drug conjugates.
  • suitable agents for co-use with any of the lipid-based nanoparticle or with the pharmaceutical composition according to the invention include an antibody binding to a co-stimulatory receptor (e.g., 0X40, CD40, ICOS, CD27 or HVEM), an agent that induces immunogenic cell death (e.g., a chemotherapeutic agent, a radio-therapeutic agent, an anti -angiogenic agent, or an agent for targeted therapies), an agent that inhibits a checkpoint molecule (e.g., LAG-3, TIM-3, BTLA, or TIGIT), a cancer vaccine, an agent that modifies an immunosuppressive enzyme (e.g., IDO1 or iNOS), an agent that targets Treg cells, an agent for adoptive cell therapy, or an agent that modulates myeloid cells.
  • a co-stimulatory receptor e.g., 0X40, CD40, ICOS, CD27 or HVEM
  • an agent that induces immunogenic cell death e.g.
  • the invention relates to a combined therapy as defined above, wherein the second therapeutic agent is an immune checkpoint blocker or activator of adaptive immune cells (T and B lymphocytes) selected from the group consisting of anti-CD2, anti-CD40, anti-HVEM, anti-BTLA, anti-CD160, anti-TIGIT, anti-TIM-1/3, anti-LAG-3, anti-2B4, and anti-OX40, anti-CD40 agonist, CD40-L, TLR agonists, anti-ICOS, ICOS-L and B-cell receptor agonists.
  • T and B lymphocytes adaptive immune cells
  • the second therapeutic agent is selected from the group consisting of chemotherapeutic agents, radiotherapy agents, immunotherapeutic agents, cell therapy agents (such as CAR-T cells), antibiotics and probiotics.
  • the lipid-based nanoparticle or the pharmaceutical composition according to the invention is combined with other forms of immunotherapy such as cytokine treatment (e.g., interferons, GM-CSF, G-CSF, IL-2), or any therapy which provides for enhanced presentation of tumor antigens.
  • cytokine treatment e.g., interferons, GM-CSF, G-CSF, IL-2
  • any therapy which provides for enhanced presentation of tumor antigens e.g., interferons, GM-CSF, G-CSF, IL-2
  • the lipid-based nanoparticle in particular obtained or obtainable by the methods of the invention, or the pharmaceutical composition comprising said lipid-based nanoparticle may be used in combination with IL-7, in particular a human wild-type IL-7, typically such as described in Uniprot accession number P13232.
  • the IL-7 and the LNP are administered sequentially, IL-7 being administered before the administration of the LNP or the pharmaceutical composition according to the invention.
  • Combination therapy could also rely on the combination of the administration of the lipid- based nanoparticle according to the invention or the pharmaceutical composition comprising such with surgery.
  • a subject in need of a treatment may be a human having, at risk for, or suspected of having a disease such as a cancer or an infectious disease. Such a patient can be identified by routine medical examination.
  • the term “subject”, “host”, “individual,” or “patient” refers to human, including adult and child.
  • the subject to treat may particularly be a human, particularly a human at the prenatal stage, a new-born, a child, an infant, an adolescent or an adult, in particular an adult of at least 30 years old, 40 years old, preferably an adult of at least 50 years old, still more preferably an adult of at least 60 years old, even more preferably an adult of at least 70 years old.
  • a subject who needs a treatment is a patient having, suspected of having, or at risk for a cancer.
  • a human patient suitable for the treatment can be identified by examining whether such a patient carries PD-L1 and/or PD-L2 positive cancer cells.
  • the subject can be immunosuppressed or immunocompromised.
  • An immunocompromised subject is a subject who is incapable of developing or unlikely to develop a robust immune response, usually as a result of disease, malnutrition, or immunosuppressive therapy.
  • Those who can be considered to be immunosuppressed or immunocompromised include, but are not limited to, subjects who have been treated with or is a candidate for treatment with an immunosuppressant, subjects with AIDS (or HIV positive), subjects with severe combined immunodeficiency, diabetics, subjects who have had transplants and who are taking immunosuppressants, and those who are receiving chemotherapy for cancer.
  • compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired.
  • the lipid-based nanoparticle, the pharmaceutical composition or the combined therapy is administered via subcutaneous, intra-cutaneous, intravenous, intramuscular, intra-articular, intra-arterial, intra-synovial, intra-tumoral, intra-sternal, intra-thecal, intra-lesion, and intracranial injection or infusion techniques.
  • the lipid-based nanoparticle, the pharmaceutical composition or the combined therapy is administered intravenously.
  • the lipid-based nanoparticle, the pharmaceutical composition or the combined therapy is administered intratum orally.
  • the subject has already received at least one line of treatment, preferably several lines of treatment, prior to the administration of the lipid-based nanoparticle, the pharmaceutical composition or the combined therapy of the invention.
  • the subject suffers from cancer and has already received at least one line of treatment with an immune checkpoint inhibitor.
  • the subject suffers from cancer has a primary or secondary resistance to an immune checkpoint inhibitor, preferably against an anti-programmed cell death 1 (PD-1) inhibitor, an anti-programmed cell death 1 ligand 1 (PD-L1) inhibitor, or a combination of an anti-PDl inhibitor and an anti CTLA-4 inhibitor.
  • the immune cell enhancing compound is selected among inhibitors of the secretion of inhibitory checkpoint molecules.
  • the inhibitory checkpoint molecule is different from PD-1, such as TIM-3 or LAG-3.
  • the use of lipid-based nanoparticle targeting specifically T cells of the TME will allow to inactivate these inhibiting pathways of T cells: one or several mRNA inactivating said pathways are delivered specifically towards and within said T cells of the TME.
  • the invention provides new targeted-LNP (t-LNP) carrying at least a mRNA encoding an inhibitor of the expression of checkpoint inhibitor other than PD-1 on T cells, in particular of at least LAG-3 and/or TIM-3, and their use for the prevention or treatment of a secondary resistance against anti-PD-1 treatment.
  • the t-LNP comprises at its surface a targeting anti-PD-1 antibody of activated T cells, and inside the t-LNP i) at least one mRNA encoding an inhibitor of the expression of checkpoint inhibitor other than PD-1 by T cells, and ii) a mRNA encoding proteins for inducing effector functions of activated T cells.
  • the invention also relates to a method of enhancing immune cell activity, comprising the step of contacting immune cells with a lipid-based nanoparticle or a pharmaceutical composition according to the invention.
  • Such methods aim to enhance the immune potential of T cells and provide powerful tools to amplify the immune response, in particular for the treatment of a disease such as a cancer or an infectious disease.
  • the lipid-based nanoparticle disclosed herein can be administered to a subject, e.g., in vivo, to enhance immunity or to enhance the immune response, especially the cytotoxic T-lymphocyte (CTL) response, preferably in order to treat a disorder and/or disease.
  • the invention provides a method of enhancing an immune response in a subject comprising administering to the subject a lipid-based nanoparticle or pharmaceutical composition of the invention such that the immune response in the subject is enhanced.
  • the lipid-based nanoparticle or pharmaceutical composition is preferably used to enhance immune responses such as immune cell activation in a subject in need of a treatment.
  • the lipid-based nanoparticle or pharmaceutical composition according to the invention is used to reduce T cells exhaustion or to reactivate exhausted T cells.
  • the invention particularly provides a method of enhancing an immune response in a subject, comprising administering to the subject a therapeutic effective amount of any of the lipid- based nanoparticles or pharmaceutical compositions comprising such described herein, such that an immune response in the subject is enhanced.
  • the lipid- based nanoparticle or pharmaceutical composition can be used to promote the cytotoxic T- lymphocyte (CTL) response, to reduce T cells exhaustion or to reactivate exhausted T cells.
  • CTL cytotoxic T- lymphocyte
  • the method is an ex vivo or an in vitro method.
  • the lipid-based nanoparticle or pharmaceutical composition according to the invention may be contacted with immune cells taken from a biological sample of a subject, such as a blood sample.
  • the method for promoting immune cell activation may comprise a first step of providing a biological sample from a subject.
  • the method of enhancing immune cells activation comprises a step of purification/isolation of immune cells from a biological sample, in particular from a blood sample.
  • a biological sample in particular from a blood sample.
  • General techniques for purifying immune cells by cell sorting or antibody and complement treatment are routinely performed and are well-known to those skilled in the art (see for example Leo, O, Sachs, DH, Samelson, L, et al. J. Immunol. 137:3874 (1986)).
  • the method of immune cells activation comprises a step of immune cell culture, expansion or amplification in particular in an immune cell culture medium.
  • Culture of immune cells are well described in the art (see for example Samuelson, LE, Germain, RN, Schwartz, RH, PNAS(USA) 80:6972 (1983)). Immune cells may particularly be cultured/amplified prior to their contact with the lipid-based nanoparticle or pharmaceutical composition of the invention.
  • immune cells culture medium refers to a medium that enables immune cell growth and/or survival and includes all conventional media used in the field appropriate for immune cell culture, in particular an aqueous or liquid medium.
  • the basic medium used for immune cell culture is preferably a cell culture minimum medium (CCMM), and generally includes a carbon source, a nitrogen source, and a trace element component.
  • CCMM cell culture minimum medium
  • the medium for immune cell culture may also be selected from the group consisting of DMEM (Dulbecco's Modified Eagle's Medium ®), MEM (Minimal Essential Medium Fix), BME (Basal Animal Medium National Eagle ®), RPMI1640 ®, F -10 MS, F -12 MS, (Minimal Essential Medium Fix), GMEM SCF (Glaggo's Animal Essential Medium Fix), Iscove's Modified Deldulbecco's Medium, Lonza X-VIVO15 TM medium, Coming 88-581-CM DC CIK cell serum-free medium and Lonza X-VIVO15 TM phenol-free red medium..
  • the lipid-based nanoparticles according to the invention are comprised in or added to the immune cell culture medium and to contact with immune cells in such medium.
  • the method further comprises a step of measuring the activation of immune cells, in particular after the step of contacting the lipid-based nanoparticles or pharmaceutical compositions of the invention with the immune cells.
  • Immune cell activity can be measured by any method known to the person skilled in the art. Activities which may be monitored to indicate degree of activation include, but are not limited to, cell proliferation, cytokine secretion, target cell killing, helper cell function (inducing other cell types to activity), cell migration, cell differentiation, or secretion of other cell products such as hormones or proteins.
  • the immune cell activity is monitored by assessing the presence or amount of the immune cell enhancing component encoded by the mRNA comprised in the lipid-based nanoparticle among the immune cell population and/or in the immune cell culture medium.
  • the activated immune cells are administered to a patient to enhance his immune system, in particular to treat various forms of cancer, infectious diseases,
  • the method of enhancing immune cells activation may comprise: a. Providing a biological sample from a subject, in particular a blood sample; b. Isolating immune cells from the biological sample; c. Culturing/amplifying the isolated immune cells, preferably in an immune cell culture medium; d. Contacting the amplified immune cells with the lipid-based nanoparticles or pharmaceutical compositions of the invention, preferably in the immune cell culture medium; e. Optionally, assessing the activity of immune cells.
  • Such method may further comprise a step f. of selecting the immune cells that are activated.
  • Such method may further comprise a step f. or g of administering the activated immune cells into the subject, in particular for promoting his immune response or for the treatment of a disease such as cancer or infectious disease.
  • kits for use in treating diseases or disorders (e.g., cancer and/or infection) or for promoting immune cells activation.
  • kit means two or more components (one of which corresponding to the lipid-based nanoparticle) packaged in a container, recipient or otherwise.
  • a kit can hence be described as a set of products and/or utensils that are sufficient to achieve a certain goal, which can be marketed as a single unit.
  • the kits of this invention are in suitable packaging.
  • the kit may include, in suitable container means, the pharmaceutical composition or lipid- based nanoparticle of the present invention.
  • the components comprised in the kit according to the invention may particularly be formulated into a syringe compatible composition.
  • the kit further includes an additional agent for treating cancer or an infectious disease, and the additional agent may be combined with the pharmaceutical composition and/or lipid-based nanoparticle of the present invention, or other components of the kit of the present invention or may be provided separately in the kit.
  • the kit described herein may include one or more additional therapeutic agents such as those described in the “Combined Therapy” described hereabove.
  • the kit(s) may be tailored to a particular cancer for an individual and comprise respective second cancer therapies for the individual as described hereabove.
  • the kit may further comprise an immune cell culture medium.
  • the lipid- based nanoparticles according to the invention may be comprised in such immune cell culture medium.
  • the instructions related to the use of the lipid-based nanoparticle or pharmaceutical composition described herein generally include information as to dosage, dosing schedule, route of administration for the intended treatment, means for reconstituting the lipid-based nanoparticle and/or means for diluting the lipid-based nanoparticle of the invention.
  • Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit in the form of a leaflet or instruction manual).
  • FIG. 1 Flow cytometry analyses of U937 (WT vs transduced), Jurkat (WT vs transduced) and HPB-ALL. Flow cytometry was used to measure the percentage of PD- 1+ cells. Dashed line: cells were stained with a PE-Cy7 labelled control isotype mouse IgGl (#557646 batch: 8155598, BD Biosciences). Dark grey: cells were stained with a PE-Cy7 labelled anti-human PD-1 antibody (#561272 batch: 1319137, BD Biosciences). Dead cells were stained and excluded from analysis with LIVE/DEADTM Fixable Yellow Dead Cell Stain Kit (#L34968A batch: 2438368, Life Technologies).
  • Figure 2 Addition of the anti-PD-1 mAb OSE-279 before the formation of LNPs allows for the preparation of targeted-LNPs with improved transfection potency in PD- 1+ cells.
  • Different modalities for mAb OSE-279 addition during the LNPs production process were tested.
  • Transfection efficiencies of LNPs encapsulating FLuc-mRNA were assessed by comparing OSE-279 addition modalities #1A, #1B and #1C in WT and PD-1+ U937 cells (Fig. 2A), and in WT and PD-1+ Jurkat cells (Fig. 2B).
  • Figure 3 Increasing the dose of mAb in targeted-LNPs induces improved transfection.
  • Targeted-LNPs were prepared using increasing amounts of mAb OSE-279 (1, 10 and 50 pL) added during LNPs preparation.
  • Transfection efficiencies of t-LNPs encapsulating FLuc- mRNA were assessed in WT and PD-1+ U937 cells (Fig. 3 A), in WT and PD-1+ Jurkat cells (Fig. 3B) and in HPB-ALL cells (Fig. 3C).
  • Non-targeted-LNPs were used as negative control.
  • FIG. 4 Different ratios of lipids can be used for the preparation of efficient targeted- LNPs.
  • OSE-279 t-LNPs were prepared with varying the relative ratios of lipids constituting the LNPs (ratio 1 and ratio 2, as described in Table 2).
  • Comparative transfection efficiency of targeted-LNPs encapsulating FLuc-mRNA were performed in WT and PD-1+ U937 cells (Fig. 4A), in WT and PD-1+ Jurkat cells (Fig. 4B) and in HPB-ALL cells (Fig. 4C).
  • Non- targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • FIG. 5 Blocking the PD-1 receptors on PD-1+ cells reduced the potency of targeted- LNPs for improved transfection.
  • Targeted-LNPs were prepared using increasing amounts of mAb OSE-279 (1, 10 and 50 pL) added in the course of LNPs preparation. Pre-incubation experiments were performed with the addition of mAb OSE-279 - in a concentration allowing for the full occupancy of the cells PD-1 receptors - prior to the addition of targeted- LNPs.
  • Transfection efficiency of LNPs encapsulating FLuc-mRNA were compared in the presence or the absence of the pre-incubation step in WT and PD-1+ U937 cells (Fig. 5A), in WT and PD-1+ Jurkat cells (Fig. 5B) and in HPB-ALL cells (Fig. 5C). Non-targeted- LNPs were used as negative control.
  • Figure 6 Targeted-LNPs display efficient binding to PD-1 receptors.
  • ELISA binding assays were performed to characterize the binding efficiency to PD-1 receptors of OSE-279 targeted-LNPs compared to OSE-279 mAb “alone” (meaning mAb not complexed in an LNP).
  • FIG. 7 The nature of the anti-PD-1 mAb used in targeted-LNPs can be modified while maintaining improved transfection in PD-1 positive cells.
  • Targeted-LNPs were prepared using either Pembrolizumab, Nivolumab or OSE-279 anti-PD-1 mAbs. Transfection efficiency of LNPs encapsulating FLuc-mRNA were compared for Pembrolizumab t-LNPs, Nivolumab t-LNPs and OSE-279 t-LNPs (in the presence or the absence of the anti-PD-1 pre-incubation step), in WT and PD-1+ U937 cells (Fig. 7A), in WT and PD-1+ Jurkat cells (Fig. 7B) and in HPB-ALL cells (Fig. 7C). Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 8 - Targeted-LNPs are able to more efficiently transfect activated T-cells expressing PD-1 compared to non-targeted-LNPs.
  • Activated T-cells isolated from PBMC and stimulated via agonistic CD3/CD28 mAbs
  • flow cytometry determine the percentage of PD-1 positive cells (Fig. 8A).
  • Dashed line cells were stained with a PE-Cy7 labelled control isotype mouse IgGl (#557646 batch: 8155598, BD Biosciences). Dark grey: cells were stained with a PE-Cy7 labelled anti-human PD-1 antibody (#561272 batch: 1319137, BD Biosciences).
  • Figure 9 The receptors targeted by the t-LNPs can be modified by varying the mAb introduced in the t-LNPs.
  • U937 cells were transduced to express both PD-1 and target 2 receptors, while HPB-ALL cells naturally express PD-1 and target 2.
  • Flow cytometry was used to measure the percentage of PD-1+ and cells expressing target 2 (Fig. 9A). Dashed line: cells were stained with a PE-Cy7 labelled control isotype mouse IgGl (#557646 batch: 8155598, BD Biosciences).
  • Targeted-LNPs were prepared using either an anti-target 2 mAb or an anti- PD-1 mAb and their relative transfection efficiency was assessed in U937 (Fig. 9B-C) and in HPB-ALL (Fig. 9D-E) cells expressing both target 2 and PD-1 receptors. Non-targeted- LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Target 2 is CD127.
  • Figure 10 In vivo and ex-vivo biodistribution of LNPs.
  • Figure 10A in vivo distribution of LNP#1 and LNP#2;
  • Figure 10B ex vivo distribution of LNP#1 and LNP#2;
  • Figure 10C in vivo distribution of LNP#3 and LNP#4;
  • Figure 10D ex vivo distribution of LNP#3 and LNP#4.
  • Figure 12 In vivo and ex-vivo biodistribution of non-targeted and targeted-LNPs.
  • Figure 13 - LNPs targeted with an anti-CD127 antagonist mAb are able to target CD- 127 expressing cells.
  • U937 and Jurkat cells were transduced to express CD-127 receptors.
  • Flow cytometry was used to measure the percentage of CD127+ cells (Fig. 13A).
  • Dashed line cells were stained with a PE-Cy7 labelled control isotype mouse IgGl (#557646 batch: 8155598, BD Biosciences). Dark grey: cells were stained with a PE-Cy7 labelled anti-human CD-127 antibody (#351320 batch: B251081, BioLegend) to measure CD-127 positive cells.
  • Figure 14 - LNPs targeted with an anti-CLEC antagonist mAb are able to target CLEC-1 expressing cells.
  • U937 and THP-1 cells were transduced to express CLEC-1 receptors.
  • Flow cytometry was used to measure the percentage of CLEC-1 positive cells (Fig. 14A).
  • Dashed line cells were stained with a purified control isotype human IgGl (OSE Immunotherapeutics) at lOpg/mL.
  • Dark grey cells were stained with a purified anti-human CLEC-1 antibody (OSE Immunotherapeutics) at 10 pg/mL to measure CLEC-1 positive cells.
  • a PE-labelled anti-human IgG antibody (clone: QA19A42 #366904 batch: B359783, BioLegend) was used for detection of primary purified antibodies. Dead cells were stained and excluded from analysis with LIVE/DEADTM Fixable Yellow Dead Cell Stain Kit (#L34968Abatch: 2438368, Life Technologies) and human Fc Receptor were saturated with Human FcBlock (#564220 batch: 2122225, BD Biosciences). Transfection efficiency of anti-CLEC targeted-LNPs was assessed in U937 (Fig. 14B) and in THP-1 (Fig. 14C) cells expressing CLEC-1 receptors. Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 15 - LNPs targeted with an anti-SIRPa antagonist mAb are able to target SIRPa expressing cells.
  • U937 cells were transduced to express SIRPa receptors.
  • Flow cytometry was used to measure the percentage of SIRPa positive cells (Fig. 15 A).
  • Dashed line cells were stained with a purified control isotype human IgG4m (MOTA hIgG4m #PI08898, EVITRIA) at 10 pg/mL.
  • Dark grey cells were stained with a purified anti -human SIRPa antibody at 10 pg/mL to measure SIRPa positive cells.
  • a PE-labelled anti -human IgG antibody (clone: QA19A42 #366904 batch: B359783, BioLegend) was used for detection of primary purified antibodies. Dead cells were stained and excluded from analysis with LIVE/DEADTM Fixable Yellow Dead Cell Stain Kit (#L34968A batch: 2622316, Life Technologies) and human Fc Receptor were saturated with Human FcBlock (#564220, 2122225, BD Biosciences). Transfection efficiency of anti-human SIRPa targeted-LNPs was assessed in U937 cells (Fig. 15B) cells expressing SIRPa receptors. Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 16 - LNPs targeted with both an anti-PD-1 and an anti-CD-127 antagonist mAb are able to target PD-1 and CD-127 expressing cells.
  • U937 cells were transduced to express both PD-1 and CD- 127 receptors.
  • Flow cytometry was used to measure the percentage of PD-1 and CD127 positive cells (Fig. 16A).
  • Dashed line cells were stained with a PE-Cy7 labelled control isotype mouse IgGl (#557646 batch: 8155598, BD Biosciences).
  • Figure 17 Different ratios of lipids can be used for the preparation of efficient targeted-LNPs.
  • OSE-279 targeted-LNPs were prepared while varying the relative ratios of DOPE and Cholesterol constituting the LNPs. Comparative transfection efficiency of targeted-LNPs were performed in PD-1 positive cells: U937 cells (Fig. 17A), and in HPB- ALL cells (Fig. 17B).
  • Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • FIG 18 Different nature of helper lipids can be used for the preparation of efficient targeted-LNPs.
  • OSE-279 targeted-LNPs were prepared while varying the nature of the helper lipid constituting the LNPs (phospholipids DOPE and POPE or cationic lipid DDAB). Comparative transfection efficiency of targeted-LNPs were performed in U937 cells PD-1 positive cells. Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 19 Different lipid length of the PEG-lipid can be used for the preparation of efficient targeted-LNPs.
  • OSE-279 targeted-LNPs were prepared while varying the length of the lipid tail of the PEG-lipid constituting the LNPs (C14-long DMG-PEG2000 and CIS- long DSG-PEG2000). Comparative transfection efficiency of targeted-LNPs were performed in PD-1 positive cells: U937 cells (Fig. 19A) and Jurkat cells (Fig. 19B).
  • Non- targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 20 Different lipidic composition can be used for the preparation of efficient tar eted-LNPs.
  • OSE-279 targeted-LNPs were prepared using the same lipid constituents as the several FDA and EMA-approved mRNA-based LNPs (Cominarty®, Spike Vax® and Onpattro®). Comparative transfection efficiency of non-targeted vs targeted-LNPs were performed in PD-1 positive cells: U937 cells (Fig. 20A-D), Jurkat cells (Fig. 20E-H) and HPB-ALL (Fig. 20I-K).
  • FIG. 21 Comparison of transfection efficiency and binding of two types of OSE-279 targeted LNPs prepared either via the method of the invention or via Thiol-Michael addition.
  • experiments comparing this new process with the classical method used for grafting mAbs on LNP surface were conducted.
  • different experimental conditions were tested to optimize the process of Thiol -Mai eimide reaction by modifying the amount of DSPE-PEG- Maleimide in the LNP (Fig. 21 A-C) and the amount of OSE-279 mAb added to the reaction (Fig. 21D-F).
  • Fig. 21G the transfection capacities of OSE-279 targeted LNP prepared either via the Thiol-Maleimide route or via the method of the invention were then compared in Jurkat (Fig. 21G) and HPB-ALL (Fig. 21H) cells. Binding studies of the two types of targeted LNPs were also assessed (Fig. 211).
  • Fig21J illustrates a process of synthesis of targeted LNPs of the prior art
  • Fig. 2 IK presents a process of synthesis of targeted LNPs according to one of the embodiments of the invention.
  • OSE-279 targeted-LNPs were prepared with various formats of anti-PD-1 mAb OSE-279 (IgG vs Monovalent IgG formats) and their transfection efficiency assessed in different cell lines expressing the PD-1 receptors: U937 (Fig. 22A), Jurkat (Fig. 22B) and HPB-ALL (Fig. 22C).
  • Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 23 Different formats of OSE-279 mAbs can be used to target PD-1 positive cells with targeted LNPs.
  • OSE-279 targeted-LNPs were prepared with various formats of anti-PD-1 mAb OSE-279 (Monovalent IgG vs ScFv-Fc formats) and their transfection efficiency assessed in different cell lines expressing the PD-1 receptors: U937 (Fig. 23A), Jurkat (Fig. 23B) and HPB-ALL (Fig. 23C).
  • Non-targeted-LNPs and targeted-LNPs prepared with a control isotype mAb were used as negative control.
  • Figure 24 Improved expression of the anti-apoptotic BCL-2 protein is observed in PD-1 expressing cells transfected with targeted-LNPs encapsulating mRNA encoding for the BCL-2 protein.
  • mRNA encoding for the anti-apoptotic protein BCL-2 (B-Cell Lymphoma-2) was produced by IVT and encapsulated in non-targeted-LNPs and in OSE- 279 targeted-LNPs.
  • Transfection experiments were conducted in WT and in PD-1 expressing cell lines U937 (Fig 24A-B) and Jurkat (Fig. 24C-D).
  • FIG. 25 Improved expression of the CXCL-9 chemokine is observed in PD-1 expressing cells transfected with targeted-LNPs encapsulating mRNA encoding for CXCL-9.
  • mRNA encoding for the Chemokine Ligand-9 (CXCL-9) was produced by IVT and encapsulated in non-targeted-LNPs and in OSE-279 targeted-LNPs. Transfection experiments were conducted in WT and in PD-1 expressing Jurkat cells (Fig. 25A-B).
  • FIG. 26 Improved expression of the CXCL-10 chemokine is observed in PD-1 expressing cells transfected with targeted-LNPs encapsulating mRNA encoding for CXCL-10.
  • mRNA encoding for the Chemokine Ligand- 10 (CXCL-10) was produced by IVT and encapsulated in non-targeted-LNPs and in OSE-279 targeted-LNPs. Transfection experiments were conducted in WT and in PD-1 expressing U937 cells (Fig. 26A-B).
  • Figure 27 Improved luciferase expression is observed ex-vivo for mice treated with PD-1 targeted-LNPs in an EL-4 tumor model. Mice were treated with EL-4 cells transduced with human PD-1 and treated at D16 with OSE-279 targeted-LNPs and nontargeted LNPs. Bioluminescence was measured in tumors ex-vivo (Fig 27A-D).
  • Figure 28 Sequential transfection with LNPs encapsulating IL-7Ra mRNA and IL-7 mRNA induce stronger pSTAT5 signal for OSE-279 targeted-LNPs in PD-1 positive cells, compared to non-targeted LNPs or WT cells.
  • Cell phenotype for natural CD- 127 and CD-132 expression was assessed at the beginning of the experiment: U937 naturally express CD-132 but not CD-127 while Jurkat cells naturally express low CD-132 but not CD- 127.
  • WT or PD-1 positive cells were first transfected with OSE-279 targeted-LNPs or non-targeted-LNPS encapsulating IL-7Ra mRNA.
  • Dashed line live cells were unstained Dark grey: cells were stained with a PE labelled anti-human PD-1 antibody (#130-117-384 batch: 5230902507, Miltenyi)). Dead cells were stained and excluded from analysis with LIVE/DEADTM Fixable Yellow Dead Cell Stain Kit (#L34968A batch: 2622316, Life Technologies). The transfection efficiency of OSE-279 targeted-LNPs on activated T-cells was compared to non-targeted-LNPs (Fig. 29).
  • Lipids ALC-0315 (890900), DDAB (890810), DSPC (850365), DMG-PEG-2000 (880151), DSG-PEG2000 (880152), DSPE-PEG2000 (880120) were purchased from Avanti Lipids (8909000), Cholesterol (1046720001) and POPE (01991) were purchased from Merck, DOPE was purchased from Corden Pharma (LP-R4-069), ALC-0159 (BP-25711) and DSPE-PEG2000-Maleimide (BP-23307) were purchased from Broadpharm, Coatsome SS-OP was purchased from NOF Corporation, MC3-DLin-DMA (N-1282) was purchased from Echelon Biosciences, SM-102 (LP001-2) was purchased from ABP Biosciences.
  • mRNA CleanCap FLuc mRNA (5moU) was purchased from Trilink Biotechnologies (L- 7202).
  • mRNA sequences are the following :
  • mRNA encoding for BCL2 comprises a nucleic acid sequence as described in SEQ ID NO: 43;
  • the mRNA encoding for IL7 comprises a nucleic acid sequence as described in SEQ ID NO: 44;
  • the mRNA encoding for IL7R comprises a nucleic acid sequence as described in SEQ ID NO: 45;
  • the mRNA encoding for CXCL9 comprises a nucleic acid sequence as described in SEQ ID NO: 46;
  • the mRNA encoding for CXCL10 comprises a nucleic acid sequence as described in SEQ ID NO: 47.
  • Cells were cultured in T75 flasks using RPMI 1640 medium (Gibco) supplemented with 10% heat inactivated fetal bovine serum (FBS) (Jurkat and U937) or 20% FBS (HPB-ALL), 100 U/mL of penicillin, 0,1 mg/mL of streptomycin, and 2mM of L-Glutamine.
  • FBS heat inactivated fetal bovine serum
  • HPB-ALL 20% FBS
  • the Jurkat and U937 cell lines stably expressing human PD-1 were obtained by lentiviral transduction. Briefly, an optimized cDNA sequence (Genscript) of human PD-1 Uniprot (QI 5116) was inserted in the BamHI / Sbfl sites of the lentiviral T54-pHRSIN plasmid. The construct was transfected along with packaging plasmids psPAX2 (Trono Lab) and pLTRG (Reiser Lab) into HEK cells to produce lentiviral particles. Wild-type U937 and Jurkat cells were transduced by the lentiviral particles, cultured for two weeks before being sorted for PD-1 expression.
  • PBMCs Human Peripheral Blood Mononuclear Cells
  • T-cells were then plated in a 6-well plate previously coated with agonistic anti-human CD3 (clone: OKT3) and CD28 (clone: CD28.2) monoclonal antibodies at 3 pg/mL in PBS for 2 hours at 37°C, 5% CO2. T cells were incubated overnight at 37°C, 5% CO2 before use.
  • agonistic anti-human CD3 clone: OKT3
  • CD28 clone: CD28.2
  • Flow cytometry was used to measure the percentage of PD-1+ and cells expressing target 2.
  • a Cytoflex flow cytometer (Beckman) was used for reading and analysis were performed with FlowJo software.
  • Anti-human PD-1 OSE-279 was produced by LFB Biomanufacturing - BMG167 batch: 21M00297 at 50 mg/mL.
  • the OSE-279 antibody comprises a heavy chain having an amino-acid sequence as disclosed in WO2020/127366.
  • OSE-279 particularly comprises a VH sequence as set forth in SEQ ID NO: 15 and a VL sequence as set forth in SEQ ID NO: 16.
  • OSE-279 particularly comprises a heavy chain sequence as set forth in SEQ ID NO: 48 and a light chain sequence as set forth in SEQ ID NO: 49.
  • Pembrolizumab - batch OFR229207LA at 25 mg/mL.
  • Pembrolizumab typically comprises a VH sequence as set forth in SEQ ID NO: 37 and a VL sequence as set forth in SEQ ID NO: 38.
  • Pembrolizumab particularly comprises a heavy chain sequence as set forth in SEQ ID NO: 50 and a light chain sequence as set forth in SEQ ID NO: 51.
  • Nivolumab - batch AXY5480 at 10 mg/mL.
  • Nivolumab particularly comprises a VH sequence as set forth in SEQ ID NO: 29 and a VL sequence as set forth in SEQ ID NO: 30.
  • Nivolumab particularly comprises a heavy chain sequence as set forth in SEQ ID NO: 52 and a light chain sequence as set forth in SEQ ID NO: 53.
  • Anti-human target 2 was produced by OSE Immunotherapeutics - batch #140521 at 1.6 mg/mL.
  • Target 2 is CD 127.
  • Anti -human CLEC was produced by OSE Immunotherapeutics.
  • Anti-human SIRPa was produced by LFB Biomanufacturing - batch: LCP21-CP04.
  • the anti-SIRPa antibody particularly comprises a VH sequence as set forth in SEQ ID NO: 39 and a VL sequence as set forth in SEQ ID NO: 40.
  • the anti-SIRPa antibody particularly comprises a heavy chain sequence as set forth in SEQ ID NO: 54 and a light chain sequence as set forth in SEQ ID NO: 55.
  • Control isotype was produced by EVITRIA at 7,4mg/mL.
  • the mAbs are not covalently bound to any of the lipids of the lipid-based nanoparticle or do not comprise any modification for coupling or grafting the antigen binding domain to a lipid.
  • Example 20 features mAbs that are covalently bound to the lipids of the lipid-based nanoparticle, and mAbs that are not covalently bound to any of the lipids of the lipid-based nanoparticle or do not comprise any modification for coupling or grafting the antigen binding domain to a lipid.
  • LNPs were 4-fold diluted into RPMI medium supplemented with 10 % FBS, 10 % heat inactivated fetal bovine serum (FBS), 100 U/mL of penicillin, 0.1 mg/mL of streptomycin, and 2 mM of L-Glutamine. 2.5 pL, 10 pL or 25 pL of the obtained solution were added on cells previously prepared in a final volume of 60 pL per well in their culture medium: 0.1 M cells/well (U937 PD1+ and Jurkat PD1+) or 0.2 M cells/well (HPB-ALL) in a 96-well transparent flat bottom culture plate.
  • FBS fetal bovine serum
  • cells were harvested in a 96-well conical bottom plate and cell viability was evaluated by cell death staining for 30 minutes at 4°C with LIVE/DEADTM Fixable Yellow Dead Cell Stain Kit (#L34959, Invitrogen) following manufacturer’s recommendations.
  • Cell death was quantified by using a flow cytometer CytoFLEX (Beckman Coulter) and analyzed with Flow Jo software. Fluorescent threshold was obtained by staining on control conditions (untreated cells and cells treated with LNP containing noncoding mRNA).
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 38% / DOPE: 12% / Cholesterol: 48 % / DMG- PEG-2000: 2 % (examples 1-3) or ALC-0315: 50% / DOPE: 10% / Cholesterol: 38.5 % / DMG-PEG-2000: 1.5 % (examples 4-9), at a total lipid concentration of 9.1 mM (unless otherwise specified).
  • FLuc-mRNA (1 mg/mL) was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of 0.083 mg/mL.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 kDa MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 mL in PBS at 4°C.
  • Targeted-LNPs were prepared as described previously, with one additional step consisting of the addition of a monoclonal antibody (mAb) used as a targeting agent.
  • mAb monoclonal antibody
  • the mean hydrodynamic diameter of the mRNA-LNPs was measured by dynamic light scattering using a Malvern NanoZS (Malvern Instruments, UK). mRNA encapsulation efficiency was determined by mRNA accessibility to Ribogreen using the Quant-iT Ribogreen RNA assay (Thermo Fisher).
  • LNPs were 5-fold diluted into PBS IX and 25 pL were added on cells previously prepared in a final volume of 100 pL per well in their culture medium.
  • the luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 48 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • mAb pre-incub ati on experiments 5 pL of mAb were added on the cells for a final concentration of 100 pg/mL (saturating conditions) and incubated at room temperature for 30 minutes before treatment with LNPs.
  • ELISA binding assays For activity ELISA assay, recombinant hPD-1 (Sino Biologicals, Beijing, China; reference 10377-H08H) was immobilized on plastic at 1 pg/mL in carbonate buffer (pH 9.2) and t- LNPs or OSE-279 mAb were added to measure binding. After incubation and washing, peroxidase-labeled donkey anti-human IgG (Jackson Immunoresearch; USA; reference 709- 035-149) was added and revealed by conventional methods.
  • Flow cytometry was used to measure the percentage of PD-1+ cells. Analyses confirmed that all 3 cell types effectively express the PD-1 receptors (Fig. 1). Wild type U937 cells do not express PD-1 receptors, while wild type Jurkat cells express PD-1 receptors at a low level. Addition of anti-PD-1 monoclonal antibody OSE-279 during the preparation of LNPs allows for improved transfection in PD-1 positive cells compared to WT cells and to LNPs without targeting agent.
  • anti-PD-1 mAb OSE-279 The ability of anti-PD-1 mAb OSE-279 to be incorporated into the core of the LNPs for effective targeting of PD-1 expressing cells was evaluated by adding OSE-279 (10 pL) during the process of LNP formation.
  • Results show that OSE-279 can be added before the microfluidic mixing step via 2 different modalities to produce targeted-LNPs able to transfect PD-1 expressing cells with improved efficacy compared to WT cells.
  • o Channel 1 mRNA (50 pL, 1 mg/mL in sodium citrate) + mAb (15 pL, 0.95 mg/mL in PBS) + acetate buffer pH 4.3 (535 pL, 25 mM)
  • o Channel 2 lipid mix (200 pL, 9.1 mM in EtOH)
  • Modality #1B o Channel 1 mRNA (50 pL, 1 mg/mL in sodium citrate) + acetate buffer pH
  • t-LNPs targeted-LNPS
  • nt-LNPs non-transfected-LNPs
  • targeted-LNPs can be produced by adding OSE-279 mAb either in the mRNA solution (mixing modality #1 A) or in the lipid solution (mixing modality #1B).
  • targeted-LNPs displayed improved transfection efficacy in PD-1 -expressing U937 (Fig. 2A) and Jurkat (Fig. 2B) cells, compared to WT cells and compared to non- targeted-LNPs.
  • Example 3 Preparation and characterization of non-targeted LNPs and targeted- LNPs encapsulating FLuc-mRNA.
  • the non-targeted-LNPs and targeted-LNPs were prepared as detailed above using microfluidic mixing system (Inside Tx).
  • the mixing modality #1 A was used in the following examples.
  • targeted-LNPs To confirm the potency of targeted-LNPs to preferentially deliver mRNA in cells expressing the PD-1 receptors, 3 different targeted-LNPs were prepared with increasing amount of OSE-279 mAh (1, 10 and 50 pL).
  • Non-targeted-LNPs and targeted-LNPs were then compared for their transfection potency, on different cell lines expressing or not-expressing PD-1 : U937 WT vs U937 PD-1+ (Fig. 3 A), Jurkat WT vs Jurkat PD-1+ (Fig. 3B) and HPB-ALL (cell line naturally expressing PD- l, Fig. 3C).
  • Results indicate that improved transfection compared to non-targeted-LNPs was observed in conditions combining PD-1 -expressing cells and targeted-LNPs, in a dose-dependent manner (with the exception of the t-LNP with 50 pL of OSE-279 in WT Jurkat cells, where an increase in transfection was observed compared to nt-LNP).
  • Example 5 OSE-279 targeted-LNPs prepared using different lipid ratios are efficient in preferentially transfecting PD-1 positive cells.
  • Results show that targeted-LNPs prepared with 10 pL of OSE-279 (mixing modality #1A) were able to preferably transfect PD-1 positive cells, compared i) to WT cells, ii) to non- targeted-LNPs and iii) to targeted-LNPs prepared with a control isotype mAb.
  • Both lipid ratios 1 and 2 allow to generate targeted-LNPs capable of inducing improved transfection on PD-1 expressing cells U937 (Fig. 4A), Jurkat (Fig. 4B) and HPB-ALL (Fig. 4C).
  • Incubation of PD-1 positive cells with the PD-1 inhibitor OSE-279 before the transfection reduced the efficacy of targeted-LNPs, confirming a PD-l-mediated endocytosis pathway for targeted-LNPs.
  • the lipid ratio 2 was used in the following examples.
  • ELISA binding assays were performed to compare the binding of mAb OSE-279 with OSE-279 targeted-LNPs.
  • Pembrolizumab, Nivolumab and OSE-279 were used to produce targeted-LNPs (addition of 10 pL of mAb during the formation of the t-LNP).
  • Results show that targeted-LNPs prepared from Pembrolizumab, Nivolumab or OSE-279 all display improved transfection in PD-1 expressing-cells compared to WT-cells and to non- targeted-LNPs (Fig. 7A for U937, Fig. 7B for Jurkat, Fig. 7C for HPB-ALL).
  • pre-incub ati on experiments were conducted using the same mAb than the one used for preparing the targeted-LNPs (i.e., Pembrolizumab was used to saturate the PD- 1 receptors in the Pembrolizumab t-LNP pre-incubation).
  • the pre-incubation step suppressed the beneficial effect of targeted-LNPs on the transfection of PD-1 expressing cells.
  • Targeted-LNPs display improved transfection on hard-to-transfect activated T cells expressing PD-1 receptor, in a dose-dependent manner, compared to non-targeted-LNPs.
  • T cells were isolated from human PBMCs and activated via CD3/CD28 stimulation to induce expression of PD-1 receptor.
  • Flow cytometry was used to measure the percentage of PD-1 + and cells expressing target 2.
  • a Cytoflex flow cytometer (Beckman) was used for reading and analysis were performed with FlowJo software. Analysis show that activated T cells express PD-1 receptors (46.9 % for donor 1, 37.2 % for donor 2, Fig. 8A).
  • the ability of OSE-279 targeted-LNPs to transfect activated T cells was evaluated compared to non-targeted-LNPs, either in Relative Light Units (Fig. 8B) or illustrated in fold changes (Fig. 8C).
  • Targeted-LNPs are able to target different cell receptors depending on the nature of the mAh used in their preparation, for an improved transfection in the cells expressing these receptors.
  • targeted-LNPs their ability to efficiently target and transfect cells expressing various receptors was investigated.
  • a model of U937 cells transduced to express both PD-1 and target 2 receptors was used.
  • Flow cytometry was used to measure the percentage of PD-1+ and cells expressing target 2 (Fig. 9A).
  • Targeted-LNPs were prepared using 15 pL of mAb, being either an anti -target 2 antagonist mAb or an anti -PD-1 mAb (OSE-279, as described previously). Results show that targeted- LNPs were able to induce an improved transfection in cells expressing the target 2 and the PD-1 receptors (U937 cells in Fig. 9A-B and HPB-ALL cells in Fig. 9C-D):
  • nt-LNP non-targeted LNP
  • nt-LNPs Preparation and characterization of nt-LNP s with various PEG lipids length and ratio.4 nt- LNPs were prepared with either DMG-PEG lipid (Cl 4 saturated chains, LNP #1 and LNP #2) or DSG-PEG lipid (C18 saturated chains, LNP #3 and LNP #4) and with either 38.5 % of Cholesterol + 1.5% of PEG-lipid (LNP #1 and LNP #3) or 39.5 % of Cholesterol + 0.5% of PEG-lipid (LNP #2 and LNP #4).
  • LNPs were formulated with FLuc-mRNA as previously described. Physico-chemical characterization of the LNPs showed similar encapsulation efficiency (> 85%) and low poly dispersity (PDI ⁇ 0.1) for all formulations. DLS measurements showed that 0.5%-PEG- lipid LNPs #2 and #4 were larger than their 1.5% PEG-lipid counterpart.
  • mice Male, 6-8 weeks old, ⁇ 20 g were intravenously injected via the lateral tail vein with 100 pL of various LNP formulations at a fixed mRNA dose of 10 pg/mL. After 6 h, mice were injected intraperitoneally with D-luciferin solution. 15 minutes after luciferin injection, a whole-body bioluminescence was performed then the mice were sacrificed, and the 3 organs of interest (liver, spleen, lung) were collected. The organs’ luminescence were analysed using an IVIS imaging system (Perkin Elmer, Waltham, MA) and quantified using Livingimage software (Perkin Elmer) to measure the radiance of each organ in photons/ sec.
  • IVIS imaging system Perkin Elmer, Waltham, MA
  • Livingimage software Perkin Elmer
  • Results Figure 10 and Figure 11 show that the biodistribution of the LNP can be controlled depending on the choice of PEG-lipid compound and of PEG-lipid ratio. More particularly, LNP1 targets liver mostly whereas LNP2, LNP3 and LNP4 are able to escape liver and lungs.
  • non-targeted and targeted (with an illustrative example of anti- PD1 antibody OSE-279) LNPs based on the lipid compositions used for LNP3 as detailed above were prepared and compared.
  • In-vivo experiments ( Figure 12) showed a lower luciferase expression for the targeted-LNP at least in the liver or spleen compared to the non- targeted-LNP. Accordingly, it can be hypothesized that the targeted LNP allows for a higher potential circulation of the LNP outside of the liver and/or spleen.
  • Example 12 Targeted-LNPs are able to target different cell receptors depending on the nature of the mAh used in their preparation, for an improved transfection in the cells expressing these receptors: example of CD-127 expressing cells targeted with anti-CD- 127 targeted LNPs.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 50% /DOPE: 10% / Cholesterol: 38.5 % /DMG- PEG-2000: 1.5 %, at a total lipid concentration of 4.55 mM. FLuc-mRNA(25 pL, 1 mg/mL) was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of 0.041 mg/mL.
  • the anti-human CD-127 mAh (0.95 pg/pL in PBS, 1 ,3xl0' 6 pmol mAb/pg RNA) was added to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was then immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 to 1.5 mL in PBS at 4°C for characterization.
  • LNPs were then diluted to 3.9 ng/pL with PBS IX and added on cells previously prepared in a final volume of 60 pL per well in their culture medium (addition of 25 pL LNP/well corresponding to 97 ng RNA/well for U937 cells and 2.5 pL LNP/well corresponding to 9.7 ng RNA/well for Jurkat cells).
  • the luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 48 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • Results show that anti-CD-127-targeted LNPs were able to induce an improved transfection in cells expressing the CD-127 receptors (U937 cells in Fig. 13B and Jurkat cells in Fig. 13C).
  • Example 13 Targeted-LNPs are able to target different cell receptors depending on the nature of the mAh used in their preparation, for an improved transfection in the cells expressing these receptors: example of CLEC-1 expressing cells targeted with anti- CLEC-1 targeted LNPs.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 50% /DOPE: 10% / Cholesterol: 38.5 % /DMG- PEG-2000: 1.5 %, at a total lipid concentration of 9.1 mM. FLuc-mRNA (50 pL, 1 mg/mL) was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of 0.083 mg/mL.
  • the anti-CLECl mAb (0.95 pg/pL in PBS, 1.3xl0' 6 pmol mAb/pg RNA) was added to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was then immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 to 1.5 mL in PBS at 4°C for characterization.
  • LNPs were diluted to 5 ng/pL with PBS IX and added (5 pL of LNP/well corresponding to 25 ng RNAfor U937 cells and 2.5 pL of LNP/well corresponding to 12.5 ng RNAfor THP1 cells) on cells previously prepared in a final volume of 60 pL per well in their culture medium and pre-treated with 5 pL of pure serum albumin bovine (SAB pre-treatment 15 min before LNP addition).
  • the luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 48 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • Results show that anti-CLECl -targeted LNPs were able to induce an improved transfection in cells expressing the CLEC-1 receptors (U937 cells in Fig. 14B and THP-1 cells in Fig. 14C).
  • Example 14 Targeted-LNPs are able to target different cell receptors depending on the nature of the mAh used in their preparation, for an improved transfection in the cells expressing these receptors: example of SIRPa expressing cells targeted with anti- SIRPa targeted LNPs.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 50% /DOPE: 10% / Cholesterol: 38.5 % /DMG- PEG-2000: 1.5 %, at a total lipid concentration of 9.1 mM. FLuc-mRNA (50 pL, 1 mg/mL) was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of 0.083 mg/mL.
  • the anti-human SIRPa mAb (0.95 pg/pL in PBS, 1 ,3xl0' 6 pmol mAb/pg RNA) was added to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was then immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 to 1.5 mL in PBS at 4°C for characterization.
  • LNPs were diluted to 1.2 ng/pL with PBS IX and added on cells previously prepared in a final volume of 60 pL per well in their culture medium (addition of 1 pL LNP/well corresponding to 1.2 ng RNA/well).
  • the luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 24 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • Results show that anti-human SIRPa targeted LNPs were able to induce an improved transfection in cells expressing the SIRPa receptors (U937 cells in Fig. 15B).
  • Targeted-LNPs are able to target different cell receptors depending on the nature of the mAb used in their preparation, for an improved transfection in the cells expressing these receptors: example of PD-1 and CD-127 expressing cells targeted with anti-PD-1 (OSE-279) and anti-CD-127 dual-targeted LNPs.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 50% /DOPE: 10% / Cholesterol: 38.5 % /DMG- PEG-2000: 1.5 %, at a total lipid concentration of 9.1 mM. FLuc-mRNA (50 pL, 1 mg/mL) was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of 0.083 mg/mL.
  • the 2 mAbs anti-CD-127 and OSE-279 (0.95 pg/pL in PBS, each 0.65xl0' 6 pmol mAb/pg RNA) were successively added in equimolar amount to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was then immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 to 1.5 mL in PBS at 4°C for characterization.
  • LNPs were diluted to 3.9 ng/pL with PBS IX and added on cells previously prepared in a final volume of 60 pL per well in their culture medium (addition of 25 pL LNP/well corresponding to 97 ng RNA/well).
  • luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 48 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • mAb pre-incubation experiments 5 pL of each mAb (9 mg/mL) were added on the cells for a final concentration of 500 pg/mL (saturating conditions) and incubated at room temperature for 30 minutes before treatment with LNPs.
  • results show that (anti-human CD-127 antibody + OSE-279) dual-targeted LNPs were able to induce an improved transfection in cells expressing the PD-1 and CD-127 receptors (U937 cells in Fig. 16B).
  • the improvements in transfection potency were suppressed when a pre-incubation step with a saturating concentration of an both anti -PD-1 and anti-CD-127 mAbs was performed before the transfection protocols, confirming that the binding of the targeted-LNP with its receptor is a prerequisite for an improved transfection.
  • the targeting efficacy was still observed when only a single mAb was used for the pre-incubation step (either anti -PD-1 alone or anti-CD-127 alone), showing that both types of receptors were involved in the improvement of transfection observed with the dualtargeted LNPs.
  • Example 16 OSE-279 targeted-LNPs prepared using 2 different DOPE/Cholesterol ratios are both efficient in preferentially transfecting PD-1 positive cells.
  • the influence of the ratio between the phospholipid DOPE and Cholesterol in the LNPs was assessed by preparing OSE-279 targeted-LNP with the lipid ratios described below.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios, at a total lipid concentration of 9.1 mM:
  • Ratio 1 ALC-0315: 50% / DOPE: 10% / Cholesterol: 38.5 % / DMG-PEG-2000: 1.5 %,
  • Ratio 2 ALC-0315: 50% / DOPE: 38.5% / Cholesterol: 10 % / DMG-PEG-2000: 1.5 %.
  • FLuc-mRNA 50 pL, 1 mg/mL was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of 0.083 mg/mL.
  • the mAb 0.95 pg/pL in PBS, 1 ,9xl0' 6 pmol mAb/pg RNA was added to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was then immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 to 1.5 mL in PBS at 4°C for characterization.
  • LNPs were diluted to 5.0 ng/pL with PBS IX and added on cells previously prepared in a final volume of 60 pL per well in their culture medium (addition of 25 pL LNP/well corresponding to 125 ng RNA/well).
  • the luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 48 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • mAb pre-incubation experiments 5 pL of mAb (9 mg/mL) were added on the cells for a final concentration of 500 pg/mL (saturating conditions) and incubated at room temperature for 30 minutes before treatment with LNPs.
  • Results show that, independently of the DOPE/Cholesterol ratio used, OSE-279 targeted LNPs were able to induce an improved transfection in cells expressing the PD-1 receptors (U937 cells in Fig. 17A, and HPB-ALL cells in Fig. 17B).
  • Example 17 OSE-279 targeted-LNPs prepared using 3 different helper lipids are efficient in preferentially transfecting PD-1 positive cells.
  • the influence of the nature of the helper lipid in the LNPs was assessed by preparing OSE-279 targeted-LNP with either DOPE, POPE or DDAB as helper lipid.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 50% /DOPE: 10% / Cholesterol: 38.5 % /DMG- PEG-2000: 1.5 % (Ratio 1) or ALC-0315: 50% / DOPE or DDAB or POPE : 38.5% / Cholesterol: 10 % / DMG-PEG-2000: 1.5 % (Ratio 2), at a total lipid concentration of 9.1 mM.
  • FLuc-mRNA 50 pL, 1 mg/mL
  • the mAb (0.95 pg/pL in PBS, 1.9xl0' 6 pmol mAb/pg RNA) was added to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.
  • the lipids (200 pL) and the mRNA solutions (600 pL) were then injected into a microfluidic mixer (LNP Pack, Inside Therapeutics, France) at a flow rate ratio of 1 :3 and a combined flow rate of 4 mL/min.
  • the resultant formulation was then immediately dialyzed against PBS IM (pH 7.4) for at least 3.5 h using 3.5 MWCO dialysis cassettes (Pur-A-Lyzer, Sigma Aldrich). After dialysis, the LNPs were stored in a final volume of 1 to 1.5 mL in PBS at 4°C for characterization.
  • LNPs were diluted to 5.0 ng/pL with PBS IX and added on cells previously prepared in a final volume of 60 pL per well in their culture medium (addition of 25 pL LNP/well corresponding to 125 ng RNA/well).
  • luciferase assay (ONE-GloTM Luciferase Assay System, Promega) was performed 48 h post-treatment in a 96-wells white cell culture plate and TECAN Spark plate reader were used to quantify the luminescence.
  • mAb pre-incubation experiments 5 pL of each mAb (9 mg/mL) were added on the cells for a final concentration of 500 pg/mL (saturating conditions) and incubated at room temperature for 30 minutes before treatment with LNPs.
  • Results show that, irrespective of the nature of the helper lipid, OSE-279 targeted LNPs were able to induce an improved transfection in U937 cells expressing the PD-1 receptors (Fig. 18). As observed in the previous experiments, the improvements in transfection potency were suppressed when a pre-incubation step with a saturating concentration of an anti -PD-1 mAbs was performed before the transfection protocols.
  • Example 18 OSE-279 targeted-LNPs prepared using 2 different PEG-lipid are efficient in preferentially transfecting PD-1 positive cells.
  • the influence of the chainlength of the PEG-lipid in the LNPs was assessed by preparing OSE-279 targeted-LNP with either DMG-PEG2000 or DSG-PEG2000 as PEG lipid.
  • LNPs were prepared using microfluidic mixing. Briefly, lipids were dissolved in ethanol with the following molar ratios ALC-0315: 50% /DOPE: 10% / Cholesterol: 38.5 % /DMG- PEG2000 or DSG-PEG2000 : 1.5 %, at a total lipid concentration of 9.1 mM. FLuc-mRNA (25 pL, 1 mg/mL) was diluted with 25 mM acetate buffer (pH 4.3) to a final concentration of0.083 mg/mL.
  • the mAb (0.95 pg/pLinPBS, 1.3x10" 6 pmol mAb/pg RNA) was added to the mRNA solution prior to microfluidic mixing.
  • the selected experimental conditions allow for a N/P ratio of 6.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nanotechnology (AREA)
  • Cell Biology (AREA)
  • Medicinal Preparation (AREA)

Abstract

L'invention concerne une nanoparticule à base de lipide comprenant un domaine de liaison à l'antigène capable de se lier spécifiquement à une cible exprimée à la surface de cellules immunitaires activées et une ou plusieurs molécule(s) d'ARNm codant pour une protéine améliorant l'activité desdites cellules immunitaires activées, et leurs utilisations.
PCT/EP2024/058778 2023-03-30 2024-03-29 Nanoparticule à base de lipide ciblant des cellules immunitaires activées pour l'expression d'une molécule d'amélioration de cellule immunitaire et son utilisation WO2024200823A1 (fr)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP23305452 2023-03-30
EP23305452.7 2023-03-30
EP23306239.7 2023-07-18
EP23306239 2023-07-18
EP23306891.5 2023-10-31
EP23306891 2023-10-31

Publications (1)

Publication Number Publication Date
WO2024200823A1 true WO2024200823A1 (fr) 2024-10-03

Family

ID=90468478

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2024/058778 WO2024200823A1 (fr) 2023-03-30 2024-03-29 Nanoparticule à base de lipide ciblant des cellules immunitaires activées pour l'expression d'une molécule d'amélioration de cellule immunitaire et son utilisation

Country Status (1)

Country Link
WO (1) WO2024200823A1 (fr)

Citations (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US806972A (en) 1904-08-17 1905-12-12 George Lyon Harvey Draft-rigging.
WO2004000238A2 (fr) 2002-03-22 2003-12-31 Tittle Thomas V Utilisation de motifs de liaison au cmh de classe ii dans le domaine de l'immunisation pour produire un immun-serum, des anticorps monoclonaux et des vaccins
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007008463A2 (fr) 2005-07-07 2007-01-18 Coley Pharmaceutical Group, Inc. Polytherapie associant un anticorps anti-ctla-4 et un oligodesoxynucleotide synthetique a motif cpg destinee au traitement du cancer
WO2008076560A2 (fr) 2006-11-15 2008-06-26 Medarex, Inc. Anticorps monoclonaux humains contre le btla et procédés d'utilisation
WO2008132601A1 (fr) 2007-04-30 2008-11-06 Immutep Anticorps monoclonal anti-lag-3 cytotoxique et son utilisation dans le traitement ou la prévention d'un rejet du greffon d'organe et de maladies auto-immunes
WO2009080253A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009100140A1 (fr) 2008-02-04 2009-08-13 Medarex, Inc. Anticorps anti-clta-4 avec blocage réduit de la liaison de ctla-4 à b7 et leurs utilisations
WO2010042877A1 (fr) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Lipides aminés améliorés et procédés d'administration d'acides nucléiques
WO2010054401A1 (fr) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Nouveaux lipides et nouvelles compositions pour l’administration d’agents thérapeutiques
WO2010106051A1 (fr) 2009-03-17 2010-09-23 Universite De La Mediterranee Anticorps anti-btla et leurs utilisations
WO2011014438A1 (fr) 2009-07-31 2011-02-03 N.V. Organon Anticorps totalement humains dirigés contre le btla
EP2320940A2 (fr) 2008-08-11 2011-05-18 Medarex, Inc. Anticorps humains qui se lient au gène 3 d'activation des lymphocytes (lag-3), et leurs utilisations
WO2011104687A1 (fr) 2010-02-24 2011-09-01 Rinat Neuroscience Corporation Anticorps récepteurs anti-il-7 antagonistes et procédés
WO2011153493A2 (fr) 2010-06-03 2011-12-08 Alnylam Pharmaceuticals, Inc. Lipides biodégradables pour l'administration de principes actifs
WO2011155607A1 (fr) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anticorps anti-tim-3
WO2012120125A1 (fr) 2011-03-09 2012-09-13 Antitope Ltd Anticorps anti-ctla4 humanisés
US20120276209A1 (en) 2009-11-04 2012-11-01 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013052523A1 (fr) 2011-10-03 2013-04-11 modeRNA Therapeutics Nucléosides, nucléotides et acides nucléiques modifiés, et leurs utilisations
WO2013090648A1 (fr) 2011-12-16 2013-06-20 modeRNA Therapeutics Nucléoside, nucléotide, et compositions d'acide nucléique modifiés
US8519110B2 (en) 2008-06-06 2013-08-27 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College mRNA cap analogs
WO2013126803A1 (fr) 2012-02-24 2013-08-29 Protiva Biotherapeutics Inc. Lipides cationiques trialkylés et leurs procédés d'utilisation
WO2014007398A1 (fr) 2012-07-06 2014-01-09 協和発酵キリン株式会社 Lipide cationique
WO2014093924A1 (fr) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Molécules d'acide nucléique modifiées et leurs utilisations
WO2014102430A1 (fr) 2012-12-27 2014-07-03 Consejo Superior De Investigaciones Científicas (Csic) Traitement thérapeutique de leucémies lymphoblastiques aiguës t et b et de lymphomes humains par inhibition du récepteur de l'interleukine-7 (il-7r)
WO2014152211A1 (fr) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation et administration de compositions de nucléosides, de nucléotides, et d'acides nucléiques modifiés
WO2014186334A1 (fr) 2013-05-15 2014-11-20 Robert Kruse Traduction intracellulaire d'arn circulaire
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2015005253A1 (fr) 2013-07-08 2015-01-15 第一三共株式会社 Nouveau lipide
WO2015011633A1 (fr) 2013-07-23 2015-01-29 Protiva Biotherapeutics, Inc. Compositions et procédés pour l'administration d'arn messager
WO2015038892A1 (fr) 2013-09-13 2015-03-19 Moderna Therapeutics, Inc. Compositions polynucléotididiques contenant des acides aminés
WO2015051169A2 (fr) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Molécules de polynucléotides et leurs utilisations
WO2015051173A2 (fr) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc Molécules polynucléotidiques et leurs utilisations
WO2015089511A2 (fr) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Molécules d'acides nucléiques modifiés et leurs utilisations
US20150239834A1 (en) 2013-11-18 2015-08-27 Arcturus Therapeutics, Inc. Symmetric ionizable cationic lipid for rna delivery
WO2015164674A1 (fr) 2014-04-23 2015-10-29 Moderna Therapeutics, Inc. Vaccins à base d'acide nucléique
WO2015196118A1 (fr) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Molécules d'acide nucléique différentes et utilisations
WO2015196128A2 (fr) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Molécules d'acide nucléique alternatives et leurs utilisations
WO2015196130A2 (fr) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Molécules d'acide nucléique alternatives et leurs utilisations
WO2015199952A1 (fr) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Nouveaux lipides et formulations nanoparticulaires lipidiques pour l'administration d'acides nucléiques
WO2016015675A1 (fr) 2014-08-01 2016-02-04 中山康方生物医药有限公司 Anticorps monoclonal anti-ctla4 ou fragment de celui-ci se liant à l'antigène, composition médicinale et son utilisation
WO2016021683A1 (fr) 2014-08-07 2016-02-11 武田薬品工業株式会社 Lipide cationique
WO2016028656A1 (fr) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anticorps anti-tigit
WO2016059512A1 (fr) 2014-10-18 2016-04-21 Pfizer Inc. Compositions d'anticorps anti-il-7r
WO2016106302A1 (fr) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Anticorps contre tigit
WO2016104580A1 (fr) 2014-12-26 2016-06-30 エーザイ・アール・アンド・ディー・マネジメント株式会社 Lipide cationique
WO2016118724A1 (fr) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Compositions de nanoparticules lipidiques
WO2016130898A2 (fr) 2015-02-13 2016-08-18 Sorrento Therapeutics, Inc. Agents thérapeutiques de type anticorps se liant à ctla4
WO2016161270A1 (fr) 2015-04-01 2016-10-06 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de cellule t et protéine 3 de mucine (tim-3)
WO2016191643A2 (fr) 2015-05-28 2016-12-01 Oncomed Pharmaceuticals, Inc. Agents de liaison à tigit et leurs utilisations
WO2016196237A1 (fr) 2015-05-29 2016-12-08 Agenus Inc. Anticorps anti-ctla-4 et méthodes d'utilisation de ceux-ci
WO2016205042A1 (fr) 2015-06-16 2016-12-22 The Board Of Trustees Of The Leland Stanford Junior University Anticorps agonistes de sirpα
WO2017000414A1 (fr) 2015-06-30 2017-01-05 宇龙计算机通信科技(深圳)有限公司 Procédé de détection de service, système de détection de service, terminal et station de base
WO2017019846A1 (fr) 2015-07-30 2017-02-02 Macrogenics, Inc. Molécules se liant à pd-1 et méthodes d'utilisation correspondantes
WO2017024465A1 (fr) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017030823A2 (fr) 2015-08-14 2017-02-23 Merck Sharp & Dohme Corp. Anticorps anti-tigit
WO2017031232A1 (fr) 2015-08-17 2017-02-23 Modernatx, Inc. Procédés de préparation de particules et compositions associées
WO2017040790A1 (fr) 2015-09-01 2017-03-09 Agenus Inc. Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
WO2017037707A1 (fr) 2015-09-02 2017-03-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Anticorps spécifiques contre le domaine d'immunoglobuline de lymphocyte t et itim (tigit)
WO2017049074A1 (fr) 2015-09-18 2017-03-23 Moderna Therapeutics, Inc. Formulations de polynucléotides à utiliser dans le traitement de néphropathies
WO2017049245A2 (fr) 2015-09-17 2017-03-23 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017053748A2 (fr) 2015-09-25 2017-03-30 Genentech, Inc. Anticorps anti-tigit et méthodes d'utilisation
WO2017062748A1 (fr) 2015-10-07 2017-04-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anticorps spécifiques d'il-7r-alpha pour le traitement la leucémie lymphoblastique aiguë
WO2017075531A1 (fr) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Nouveaux lipides et nouvelles formulations de nanoparticules de lipides pour l'administration d'acides nucléiques
WO2017087588A1 (fr) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Liants ctla4
WO2017084078A1 (fr) 2015-11-19 2017-05-26 Zeling Cai Anticorps ctla-4 et leurs utilisations
WO2017096017A1 (fr) 2015-12-02 2017-06-08 Stsciences, Inc. Anticorps spécifiques au btla glycosylé (atténuateur de lymphocytes b et t)
WO2017099823A1 (fr) 2015-12-10 2017-06-15 Modernatx, Inc. Compositions et procédés permettant d'administrer des agents thérapeutiques
WO2017106372A1 (fr) 2015-12-15 2017-06-22 Oncoimmune, Inc. Anticorps monoclonaux chimériques et humanisés anti-ctla4 humaine, et leurs utilisations
WO2017112865A1 (fr) 2015-12-22 2017-06-29 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques et/ou prophylactiques
WO2017117528A1 (fr) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipides et formulations de nanoparticules de lipides pour la libération d'acides nucléiques
WO2017133540A1 (fr) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017132825A1 (fr) 2016-02-02 2017-08-10 华为技术有限公司 Procédé de vérification de puissance d'émission, équipement utilisateur et station de base
WO2017144668A1 (fr) 2016-02-26 2017-08-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour btla et leurs utilisations
WO2017152088A1 (fr) 2016-03-04 2017-09-08 JN Biosciences, LLC Anticorps anti-tigit
WO2017153936A1 (fr) 2016-03-10 2017-09-14 Novartis Ag Arn messager chimiquement modifié
WO2017180917A2 (fr) 2016-04-13 2017-10-19 Modernatx, Inc. Compositions lipidiques et leurs utilisations pour l'administration intratumorale de polynucléotides
WO2017178653A2 (fr) 2016-04-14 2017-10-19 Ose Immunotherapeutics Nouveaux anticorps anti-sirpa et leurs applications thérapeutiques
WO2017194265A1 (fr) 2016-05-10 2017-11-16 Agency For Science, Technology And Research Anticorps anti-ctla -4
WO2017218704A1 (fr) 2016-06-14 2017-12-21 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
WO2017223135A1 (fr) 2016-06-24 2017-12-28 Modernatx, Inc. Nanoparticules lipidiques
WO2018008470A1 (fr) 2016-07-05 2018-01-11 国立大学法人神戸大学 Agent antitumoral
WO2018025178A1 (fr) 2016-08-02 2018-02-08 Aduro Biotech Holdings, Europe B.V. Anticorps contre ctla-4 humain
WO2018033798A1 (fr) 2016-08-17 2018-02-22 Compugen Ltd. Anticorps anti-tigit, anticorps anti-pvrig et combinaisons associées
WO2018035710A1 (fr) 2016-08-23 2018-03-01 Akeso Biopharma, Inc. Anticorps anti-ctla4
WO2018053106A1 (fr) 2016-09-14 2018-03-22 Abbvie Biotherapeutics Inc. Anticorps anti-pd-1 (cd279)
WO2018068182A1 (fr) 2016-10-10 2018-04-19 Crown Bioscience (Taicang) Inc. Nouveaux anticorps anti-ctla4
WO2018085469A2 (fr) 2016-11-01 2018-05-11 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de lymphocyte t et la protéine 3 de mucine (tim-3)
WO2018102746A1 (fr) 2016-12-02 2018-06-07 Rigel Pharmaceuticals, Inc. Molécules de liaison à l'antigène destinées à la tigit
WO2018102536A1 (fr) 2016-11-30 2018-06-07 Oncomed Pharmaceuticals, Inc. Méthodes de traitement du cancer comprenant des agents de liaison à tigit
WO2018106862A1 (fr) 2016-12-07 2018-06-14 Agenus Inc. Anticorps anti-ctla-4 et leurs procédés d'utilisation
WO2018106864A1 (fr) 2016-12-07 2018-06-14 Agenus Inc. Anticorps et procédés d'utilisation de ceux-ci
WO2018107058A1 (fr) 2016-12-09 2018-06-14 Alector Llc Anticorps anti-sirp-alpha et leurs procédés d'utilisation
WO2018129553A1 (fr) 2017-01-09 2018-07-12 Tesaro, Inc. Méthodes de traitement du cancer au moyen d'anticorps anti-tim-3
WO2018156250A1 (fr) 2017-02-21 2018-08-30 Remd Biotherapeutics, Inc. Traitement du cancer à l'aide d'anticorps qui se lient à l'antigène-4 des lymphocytes t cytotoxiques (ctla-4)
WO2018160536A1 (fr) 2017-02-28 2018-09-07 Bristol-Myers Squibb Company Utilisation d'anticorps anti-ctla-4 avec adcc améliorée pour renforcer la réponse immunitaire d'un vaccin
WO2018160704A1 (fr) 2017-02-28 2018-09-07 Adimab Llc Anticorps anti-tigit
WO2018165895A1 (fr) 2017-03-15 2018-09-20 苏州银河生物医药有限公司 Anticorps ctla4, composition pharmaceutique et leur utilisation
WO2018200430A1 (fr) 2017-04-26 2018-11-01 Bristol-Myers Squibb Company Procédés de production d'anticorps qui réduisent au minimum la réduction de liaison disulfure
WO2018204363A1 (fr) 2017-05-01 2018-11-08 Agenus Inc. Anticorps anti-tigit et leurs méthodes d'utilisation
WO2018209701A1 (fr) 2017-05-19 2018-11-22 Wuxi Biologics (Shanghai) Co., Ltd. Nouveaux anticorps monoclonaux dirigés contre la protéine 4 associée aux lymphocytes t cytotoxiques (ctla-4)
WO2019023504A1 (fr) 2017-07-27 2019-01-31 Iteos Therapeutics Sa Anticorps anti-tigit
WO2019023482A1 (fr) 2017-07-27 2019-01-31 Regeneron Pharmaceuticals, Inc. Anticorps anti-ctla-4 et leurs utilisations
WO2019056281A1 (fr) 2017-09-21 2019-03-28 Eucure (Beijing) Biopharma Co., Ltd Anticorps anti-ctla4 et utilisations associées
WO2019062832A1 (fr) 2017-09-29 2019-04-04 江苏恒瑞医药股份有限公司 Anticorps tigit, fragment de liaison à l'antigène de celui-ci, et son utilisation médicale
WO2019073080A1 (fr) 2017-10-13 2019-04-18 Ose Immunotherapeutics Anticorps anti-sirpa modifiés et leurs utilisations
WO2019120232A1 (fr) 2017-12-20 2019-06-27 Harbour Biomed (Shanghai) Co., Ltd Anticorps de liaison à ctla-4 et leurs utilisations
WO2019131839A1 (fr) 2017-12-28 2019-07-04 武田薬品工業株式会社 Lipides cationiques
WO2019129261A1 (fr) 2017-12-30 2019-07-04 Beigene, Ltd. Anticorps anti-tigit et leur utilisation comme agents thérapeutiques et diagnostiques
WO2019129221A1 (fr) 2017-12-28 2019-07-04 Nanjing Legend Biotech Co., Ltd. Anticorps à domaine unique et leurs variants dirigés contre tigit
WO2019137548A1 (fr) 2018-01-15 2019-07-18 Nanjing Legend Biotech Co., Ltd. Anticorps et variants associés dirigés contre tigit
WO2019152574A1 (fr) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Procédés de traitement du cancer ou d'une infection à l'aide d'une combinaison d'un anticorps anti-pd -1, d'un anticorps anti-lag3 et d'un anticorps anti-tigit
WO2019148444A1 (fr) 2018-02-02 2019-08-08 Adagene Inc. Anticorps anti-ctla4 et leurs procédés de fabrication et d'utilisation
WO2019154415A1 (fr) 2018-02-06 2019-08-15 I-Mab Anticorps dirigés contre l'immunorécepteur des lymphocytes t avec des domaines ig et itim (tigit) et leurs utilisations
WO2019168382A1 (fr) 2018-02-28 2019-09-06 Yuhan Corporation Anticorps anti-tigit et leurs utilisations
WO2019174603A1 (fr) 2018-03-14 2019-09-19 上海开拓者生物医药有限公司 Anticorps ciblant ctla-4, son procédé de préparation et son utilisation
WO2019179388A1 (fr) 2018-03-19 2019-09-26 Wuxi Biologics (Shanghai) Co., Ltd. Nouveau polypeptide anticorps anti-ctla-4
WO2019179391A1 (fr) 2018-03-19 2019-09-26 Wuxi Biologics (Shanghai) Co., Ltd. Nouvelles molécules d'anticorps pd-1/ctla-4 bispécifiques
WO2019215728A1 (fr) 2018-05-09 2019-11-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Anticorps spécifiques de la nectine-4 humaine
WO2019226973A1 (fr) 2018-05-25 2019-11-28 Alector Llc Anticorps anti-sirpa et leurs procédés d'utilisation
WO2019232484A1 (fr) 2018-06-01 2019-12-05 Compugen Ltd Anticorps bispécifiques anti-pvrig/anti-tigit et procédés d'utilisation
WO2020077190A1 (fr) 2018-10-12 2020-04-16 Jumaa Weinacht Hassan Anticorps monoclonal pour le traitement de la leucémie lymphoblastique aiguë
WO2020102422A1 (fr) 2018-11-14 2020-05-22 Arch Oncology, Inc. ANTICORPS SIRPα THÉRAPEUTIQUES
WO2020127366A1 (fr) 2018-12-21 2020-06-25 Ose Immunotherapeutics Anticorps anti-pd-1 anti-humain humanisé
WO2021159130A2 (fr) 2020-05-15 2021-08-12 Modernatx, Inc. Vaccins a arn de coronavirus et procédés d'utilisation
WO2021194672A1 (fr) 2020-03-23 2021-09-30 Hdt Bio Corp. Compositions et procédés pour l'administration d'arn
WO2021222746A2 (fr) 2020-04-30 2021-11-04 Arch Oncology, Inc. ANTICORPS SIRPα THÉRAPEUTIQUES
WO2022110922A1 (fr) 2020-11-30 2022-06-02 启愈生物技术(上海)有限公司 ANTICORPS ANTI-SIRPα OU FRAGMENT DE LIAISON À L'ANTIGÈNE ASSOCIÉ, ET SON UTILISATION
WO2022120388A2 (fr) * 2020-12-04 2022-06-09 Tidal Therapeutics, Inc. Nanoparticules lipidiques et lipides cationiques ionisables, et leurs procédés de synthèse et d'utilisation
WO2022121980A1 (fr) 2020-12-11 2022-06-16 浙江博锐生物制药有限公司 ANTICORPS ANTI-SIRPα ET SON APPLICATION
WO2022254379A1 (fr) 2021-06-04 2022-12-08 Boehringer Ingelheim International Gmbh Anticorps anti-sirp-alpha
WO2022261101A1 (fr) 2021-06-07 2022-12-15 Generation Bio Co. Compositions de nanoparticules lipidiques modifiées par apolipoprotéine e et apolipoprotéine b et utilisations associées
WO2022261490A2 (fr) 2021-06-10 2022-12-15 Orna Therapeutics, Inc. Méthodes et compositions d'arn circulaire
WO2023020459A1 (fr) 2021-08-17 2023-02-23 杭州九源基因工程有限公司 ANTICORPS MONOCLONAL CIBLANT SIRPα ET SON UTILISATION
WO2023202672A1 (fr) 2022-04-20 2023-10-26 Biosion Inc. Anticorps ciblant sirp-alpha et leurs utilisations

Patent Citations (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US806972A (en) 1904-08-17 1905-12-12 George Lyon Harvey Draft-rigging.
WO2004000238A2 (fr) 2002-03-22 2003-12-31 Tittle Thomas V Utilisation de motifs de liaison au cmh de classe ii dans le domaine de l'immunisation pour produire un immun-serum, des anticorps monoclonaux et des vaccins
WO2006121168A1 (fr) 2005-05-09 2006-11-16 Ono Pharmaceutical Co., Ltd. Anticorps monoclonaux humains pour mort programmee 1 (mp-1) et procedes pour traiter le cancer en utilisant des anticorps anti-mp-1 seuls ou associes a d’autres immunotherapies
WO2007008463A2 (fr) 2005-07-07 2007-01-18 Coley Pharmaceutical Group, Inc. Polytherapie associant un anticorps anti-ctla-4 et un oligodesoxynucleotide synthetique a motif cpg destinee au traitement du cancer
WO2008076560A2 (fr) 2006-11-15 2008-06-26 Medarex, Inc. Anticorps monoclonaux humains contre le btla et procédés d'utilisation
WO2008132601A1 (fr) 2007-04-30 2008-11-06 Immutep Anticorps monoclonal anti-lag-3 cytotoxique et son utilisation dans le traitement ou la prévention d'un rejet du greffon d'organe et de maladies auto-immunes
WO2009080253A1 (fr) 2007-12-21 2009-07-02 F. Hoffmann-La Roche Ag Anticorps bivalents bispécifiques
WO2009100140A1 (fr) 2008-02-04 2009-08-13 Medarex, Inc. Anticorps anti-clta-4 avec blocage réduit de la liaison de ctla-4 à b7 et leurs utilisations
US8519110B2 (en) 2008-06-06 2013-08-27 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College mRNA cap analogs
EP2320940A2 (fr) 2008-08-11 2011-05-18 Medarex, Inc. Anticorps humains qui se lient au gène 3 d'activation des lymphocytes (lag-3), et leurs utilisations
WO2010042877A1 (fr) 2008-10-09 2010-04-15 Tekmira Pharmaceuticals Corporation Lipides aminés améliorés et procédés d'administration d'acides nucléiques
WO2010054401A1 (fr) 2008-11-10 2010-05-14 Alnylam Pharmaceuticals, Inc. Nouveaux lipides et nouvelles compositions pour l’administration d’agents thérapeutiques
WO2010106051A1 (fr) 2009-03-17 2010-09-23 Universite De La Mediterranee Anticorps anti-btla et leurs utilisations
WO2011014438A1 (fr) 2009-07-31 2011-02-03 N.V. Organon Anticorps totalement humains dirigés contre le btla
US20120276209A1 (en) 2009-11-04 2012-11-01 The University Of British Columbia Nucleic acid-containing lipid particles and related methods
WO2011104687A1 (fr) 2010-02-24 2011-09-01 Rinat Neuroscience Corporation Anticorps récepteurs anti-il-7 antagonistes et procédés
WO2011153493A2 (fr) 2010-06-03 2011-12-08 Alnylam Pharmaceuticals, Inc. Lipides biodégradables pour l'administration de principes actifs
WO2011155607A1 (fr) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anticorps anti-tim-3
EP2581113A1 (fr) 2010-06-11 2013-04-17 Kyowa Hakko Kirin Co., Ltd. Anticorps anti-tim-3
US8552156B2 (en) 2010-06-11 2013-10-08 Kyowa Hakko Kirin Co., Ltd Anti-TIM-3 antibody
WO2012120125A1 (fr) 2011-03-09 2012-09-13 Antitope Ltd Anticorps anti-ctla4 humanisés
WO2013006490A2 (fr) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Anticorps se liant spécifiquement à tim3
WO2013052523A1 (fr) 2011-10-03 2013-04-11 modeRNA Therapeutics Nucléosides, nucléotides et acides nucléiques modifiés, et leurs utilisations
WO2013090648A1 (fr) 2011-12-16 2013-06-20 modeRNA Therapeutics Nucléoside, nucléotide, et compositions d'acide nucléique modifiés
WO2013126803A1 (fr) 2012-02-24 2013-08-29 Protiva Biotherapeutics Inc. Lipides cationiques trialkylés et leurs procédés d'utilisation
WO2014007398A1 (fr) 2012-07-06 2014-01-09 協和発酵キリン株式会社 Lipide cationique
WO2014093924A1 (fr) 2012-12-13 2014-06-19 Moderna Therapeutics, Inc. Molécules d'acide nucléique modifiées et leurs utilisations
WO2014102430A1 (fr) 2012-12-27 2014-07-03 Consejo Superior De Investigaciones Científicas (Csic) Traitement thérapeutique de leucémies lymphoblastiques aiguës t et b et de lymphomes humains par inhibition du récepteur de l'interleukine-7 (il-7r)
WO2014152211A1 (fr) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation et administration de compositions de nucléosides, de nucléotides, et d'acides nucléiques modifiés
WO2014186334A1 (fr) 2013-05-15 2014-11-20 Robert Kruse Traduction intracellulaire d'arn circulaire
WO2014194302A2 (fr) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Protéines de liaison à l'antigène qui se lient à pd-1
WO2015005253A1 (fr) 2013-07-08 2015-01-15 第一三共株式会社 Nouveau lipide
WO2015011633A1 (fr) 2013-07-23 2015-01-29 Protiva Biotherapeutics, Inc. Compositions et procédés pour l'administration d'arn messager
WO2015038892A1 (fr) 2013-09-13 2015-03-19 Moderna Therapeutics, Inc. Compositions polynucléotididiques contenant des acides aminés
WO2015051169A2 (fr) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc. Molécules de polynucléotides et leurs utilisations
WO2015051173A2 (fr) 2013-10-02 2015-04-09 Moderna Therapeutics, Inc Molécules polynucléotidiques et leurs utilisations
US20150239834A1 (en) 2013-11-18 2015-08-27 Arcturus Therapeutics, Inc. Symmetric ionizable cationic lipid for rna delivery
WO2015089511A2 (fr) 2013-12-13 2015-06-18 Moderna Therapeutics, Inc. Molécules d'acides nucléiques modifiés et leurs utilisations
WO2015164674A1 (fr) 2014-04-23 2015-10-29 Moderna Therapeutics, Inc. Vaccins à base d'acide nucléique
WO2015196118A1 (fr) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Molécules d'acide nucléique différentes et utilisations
WO2015196128A2 (fr) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Molécules d'acide nucléique alternatives et leurs utilisations
WO2015196130A2 (fr) 2014-06-19 2015-12-23 Moderna Therapeutics, Inc. Molécules d'acide nucléique alternatives et leurs utilisations
WO2015199952A1 (fr) 2014-06-25 2015-12-30 Acuitas Therapeutics Inc. Nouveaux lipides et formulations nanoparticulaires lipidiques pour l'administration d'acides nucléiques
WO2016015675A1 (fr) 2014-08-01 2016-02-04 中山康方生物医药有限公司 Anticorps monoclonal anti-ctla4 ou fragment de celui-ci se liant à l'antigène, composition médicinale et son utilisation
WO2016021683A1 (fr) 2014-08-07 2016-02-11 武田薬品工業株式会社 Lipide cationique
WO2016028656A1 (fr) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anticorps anti-tigit
WO2016059512A1 (fr) 2014-10-18 2016-04-21 Pfizer Inc. Compositions d'anticorps anti-il-7r
WO2016106302A1 (fr) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Anticorps contre tigit
WO2016104580A1 (fr) 2014-12-26 2016-06-30 エーザイ・アール・アンド・ディー・マネジメント株式会社 Lipide cationique
WO2016118724A1 (fr) 2015-01-21 2016-07-28 Moderna Therapeutics, Inc. Compositions de nanoparticules lipidiques
WO2016130898A2 (fr) 2015-02-13 2016-08-18 Sorrento Therapeutics, Inc. Agents thérapeutiques de type anticorps se liant à ctla4
WO2016161270A1 (fr) 2015-04-01 2016-10-06 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de cellule t et protéine 3 de mucine (tim-3)
WO2016191643A2 (fr) 2015-05-28 2016-12-01 Oncomed Pharmaceuticals, Inc. Agents de liaison à tigit et leurs utilisations
WO2016196237A1 (fr) 2015-05-29 2016-12-08 Agenus Inc. Anticorps anti-ctla-4 et méthodes d'utilisation de ceux-ci
WO2016205042A1 (fr) 2015-06-16 2016-12-22 The Board Of Trustees Of The Leland Stanford Junior University Anticorps agonistes de sirpα
WO2017000414A1 (fr) 2015-06-30 2017-01-05 宇龙计算机通信科技(深圳)有限公司 Procédé de détection de service, système de détection de service, terminal et station de base
WO2017019846A1 (fr) 2015-07-30 2017-02-02 Macrogenics, Inc. Molécules se liant à pd-1 et méthodes d'utilisation correspondantes
WO2017024465A1 (fr) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017025016A1 (fr) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017030823A2 (fr) 2015-08-14 2017-02-23 Merck Sharp & Dohme Corp. Anticorps anti-tigit
WO2017031232A1 (fr) 2015-08-17 2017-02-23 Modernatx, Inc. Procédés de préparation de particules et compositions associées
WO2017040790A1 (fr) 2015-09-01 2017-03-09 Agenus Inc. Anticorps anti-pd1 et méthodes d'utilisation de ceux-ci
WO2017037707A1 (fr) 2015-09-02 2017-03-09 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Anticorps spécifiques contre le domaine d'immunoglobuline de lymphocyte t et itim (tigit)
WO2017049245A2 (fr) 2015-09-17 2017-03-23 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques
WO2017049074A1 (fr) 2015-09-18 2017-03-23 Moderna Therapeutics, Inc. Formulations de polynucléotides à utiliser dans le traitement de néphropathies
WO2017053748A2 (fr) 2015-09-25 2017-03-30 Genentech, Inc. Anticorps anti-tigit et méthodes d'utilisation
WO2017062748A1 (fr) 2015-10-07 2017-04-13 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anticorps spécifiques d'il-7r-alpha pour le traitement la leucémie lymphoblastique aiguë
WO2017075531A1 (fr) 2015-10-28 2017-05-04 Acuitas Therapeutics, Inc. Nouveaux lipides et nouvelles formulations de nanoparticules de lipides pour l'administration d'acides nucléiques
WO2017087588A1 (fr) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Liants ctla4
WO2017084078A1 (fr) 2015-11-19 2017-05-26 Zeling Cai Anticorps ctla-4 et leurs utilisations
WO2017096017A1 (fr) 2015-12-02 2017-06-08 Stsciences, Inc. Anticorps spécifiques au btla glycosylé (atténuateur de lymphocytes b et t)
WO2017099823A1 (fr) 2015-12-10 2017-06-15 Modernatx, Inc. Compositions et procédés permettant d'administrer des agents thérapeutiques
WO2017106372A1 (fr) 2015-12-15 2017-06-22 Oncoimmune, Inc. Anticorps monoclonaux chimériques et humanisés anti-ctla4 humaine, et leurs utilisations
WO2017112865A1 (fr) 2015-12-22 2017-06-29 Modernatx, Inc. Composés et compositions pour l'administration intracellulaire d'agents thérapeutiques et/ou prophylactiques
WO2017117528A1 (fr) 2015-12-30 2017-07-06 Acuitas Therapeutics, Inc. Lipides et formulations de nanoparticules de lipides pour la libération d'acides nucléiques
WO2017133540A1 (fr) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Anticorps anti-pd-1
WO2017132825A1 (fr) 2016-02-02 2017-08-10 华为技术有限公司 Procédé de vérification de puissance d'émission, équipement utilisateur et station de base
WO2017144668A1 (fr) 2016-02-26 2017-08-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps présentant une spécificité pour btla et leurs utilisations
WO2017152088A1 (fr) 2016-03-04 2017-09-08 JN Biosciences, LLC Anticorps anti-tigit
WO2017153936A1 (fr) 2016-03-10 2017-09-14 Novartis Ag Arn messager chimiquement modifié
WO2017180917A2 (fr) 2016-04-13 2017-10-19 Modernatx, Inc. Compositions lipidiques et leurs utilisations pour l'administration intratumorale de polynucléotides
WO2017178653A2 (fr) 2016-04-14 2017-10-19 Ose Immunotherapeutics Nouveaux anticorps anti-sirpa et leurs applications thérapeutiques
WO2017194265A1 (fr) 2016-05-10 2017-11-16 Agency For Science, Technology And Research Anticorps anti-ctla -4
WO2017218704A1 (fr) 2016-06-14 2017-12-21 Modernatx, Inc. Formulations stabilisées de nanoparticules lipidiques
WO2017223135A1 (fr) 2016-06-24 2017-12-28 Modernatx, Inc. Nanoparticules lipidiques
WO2018008470A1 (fr) 2016-07-05 2018-01-11 国立大学法人神戸大学 Agent antitumoral
WO2018025178A1 (fr) 2016-08-02 2018-02-08 Aduro Biotech Holdings, Europe B.V. Anticorps contre ctla-4 humain
WO2018033798A1 (fr) 2016-08-17 2018-02-22 Compugen Ltd. Anticorps anti-tigit, anticorps anti-pvrig et combinaisons associées
WO2018035710A1 (fr) 2016-08-23 2018-03-01 Akeso Biopharma, Inc. Anticorps anti-ctla4
WO2018053106A1 (fr) 2016-09-14 2018-03-22 Abbvie Biotherapeutics Inc. Anticorps anti-pd-1 (cd279)
WO2018068182A1 (fr) 2016-10-10 2018-04-19 Crown Bioscience (Taicang) Inc. Nouveaux anticorps anti-ctla4
WO2018085469A2 (fr) 2016-11-01 2018-05-11 Anaptysbio, Inc. Anticorps dirigés contre l'immunoglobuline de lymphocyte t et la protéine 3 de mucine (tim-3)
WO2018102536A1 (fr) 2016-11-30 2018-06-07 Oncomed Pharmaceuticals, Inc. Méthodes de traitement du cancer comprenant des agents de liaison à tigit
WO2018102746A1 (fr) 2016-12-02 2018-06-07 Rigel Pharmaceuticals, Inc. Molécules de liaison à l'antigène destinées à la tigit
WO2018106862A1 (fr) 2016-12-07 2018-06-14 Agenus Inc. Anticorps anti-ctla-4 et leurs procédés d'utilisation
WO2018106864A1 (fr) 2016-12-07 2018-06-14 Agenus Inc. Anticorps et procédés d'utilisation de ceux-ci
WO2018107058A1 (fr) 2016-12-09 2018-06-14 Alector Llc Anticorps anti-sirp-alpha et leurs procédés d'utilisation
WO2018129553A1 (fr) 2017-01-09 2018-07-12 Tesaro, Inc. Méthodes de traitement du cancer au moyen d'anticorps anti-tim-3
WO2018156250A1 (fr) 2017-02-21 2018-08-30 Remd Biotherapeutics, Inc. Traitement du cancer à l'aide d'anticorps qui se lient à l'antigène-4 des lymphocytes t cytotoxiques (ctla-4)
WO2018160536A1 (fr) 2017-02-28 2018-09-07 Bristol-Myers Squibb Company Utilisation d'anticorps anti-ctla-4 avec adcc améliorée pour renforcer la réponse immunitaire d'un vaccin
WO2018160704A1 (fr) 2017-02-28 2018-09-07 Adimab Llc Anticorps anti-tigit
WO2018165895A1 (fr) 2017-03-15 2018-09-20 苏州银河生物医药有限公司 Anticorps ctla4, composition pharmaceutique et leur utilisation
WO2018200430A1 (fr) 2017-04-26 2018-11-01 Bristol-Myers Squibb Company Procédés de production d'anticorps qui réduisent au minimum la réduction de liaison disulfure
WO2018204363A1 (fr) 2017-05-01 2018-11-08 Agenus Inc. Anticorps anti-tigit et leurs méthodes d'utilisation
WO2018209701A1 (fr) 2017-05-19 2018-11-22 Wuxi Biologics (Shanghai) Co., Ltd. Nouveaux anticorps monoclonaux dirigés contre la protéine 4 associée aux lymphocytes t cytotoxiques (ctla-4)
WO2019023504A1 (fr) 2017-07-27 2019-01-31 Iteos Therapeutics Sa Anticorps anti-tigit
WO2019023482A1 (fr) 2017-07-27 2019-01-31 Regeneron Pharmaceuticals, Inc. Anticorps anti-ctla-4 et leurs utilisations
WO2019056281A1 (fr) 2017-09-21 2019-03-28 Eucure (Beijing) Biopharma Co., Ltd Anticorps anti-ctla4 et utilisations associées
WO2019062832A1 (fr) 2017-09-29 2019-04-04 江苏恒瑞医药股份有限公司 Anticorps tigit, fragment de liaison à l'antigène de celui-ci, et son utilisation médicale
WO2019073080A1 (fr) 2017-10-13 2019-04-18 Ose Immunotherapeutics Anticorps anti-sirpa modifiés et leurs utilisations
WO2019120232A1 (fr) 2017-12-20 2019-06-27 Harbour Biomed (Shanghai) Co., Ltd Anticorps de liaison à ctla-4 et leurs utilisations
WO2019131839A1 (fr) 2017-12-28 2019-07-04 武田薬品工業株式会社 Lipides cationiques
WO2019129221A1 (fr) 2017-12-28 2019-07-04 Nanjing Legend Biotech Co., Ltd. Anticorps à domaine unique et leurs variants dirigés contre tigit
WO2019129261A1 (fr) 2017-12-30 2019-07-04 Beigene, Ltd. Anticorps anti-tigit et leur utilisation comme agents thérapeutiques et diagnostiques
WO2019137548A1 (fr) 2018-01-15 2019-07-18 Nanjing Legend Biotech Co., Ltd. Anticorps et variants associés dirigés contre tigit
WO2019152574A1 (fr) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Procédés de traitement du cancer ou d'une infection à l'aide d'une combinaison d'un anticorps anti-pd -1, d'un anticorps anti-lag3 et d'un anticorps anti-tigit
WO2019148444A1 (fr) 2018-02-02 2019-08-08 Adagene Inc. Anticorps anti-ctla4 et leurs procédés de fabrication et d'utilisation
WO2019154415A1 (fr) 2018-02-06 2019-08-15 I-Mab Anticorps dirigés contre l'immunorécepteur des lymphocytes t avec des domaines ig et itim (tigit) et leurs utilisations
WO2019168382A1 (fr) 2018-02-28 2019-09-06 Yuhan Corporation Anticorps anti-tigit et leurs utilisations
WO2019174603A1 (fr) 2018-03-14 2019-09-19 上海开拓者生物医药有限公司 Anticorps ciblant ctla-4, son procédé de préparation et son utilisation
WO2019179388A1 (fr) 2018-03-19 2019-09-26 Wuxi Biologics (Shanghai) Co., Ltd. Nouveau polypeptide anticorps anti-ctla-4
WO2019179391A1 (fr) 2018-03-19 2019-09-26 Wuxi Biologics (Shanghai) Co., Ltd. Nouvelles molécules d'anticorps pd-1/ctla-4 bispécifiques
WO2019215728A1 (fr) 2018-05-09 2019-11-14 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Anticorps spécifiques de la nectine-4 humaine
WO2019226973A1 (fr) 2018-05-25 2019-11-28 Alector Llc Anticorps anti-sirpa et leurs procédés d'utilisation
WO2019232484A1 (fr) 2018-06-01 2019-12-05 Compugen Ltd Anticorps bispécifiques anti-pvrig/anti-tigit et procédés d'utilisation
WO2020077190A1 (fr) 2018-10-12 2020-04-16 Jumaa Weinacht Hassan Anticorps monoclonal pour le traitement de la leucémie lymphoblastique aiguë
WO2020102422A1 (fr) 2018-11-14 2020-05-22 Arch Oncology, Inc. ANTICORPS SIRPα THÉRAPEUTIQUES
WO2020127366A1 (fr) 2018-12-21 2020-06-25 Ose Immunotherapeutics Anticorps anti-pd-1 anti-humain humanisé
WO2021194672A1 (fr) 2020-03-23 2021-09-30 Hdt Bio Corp. Compositions et procédés pour l'administration d'arn
WO2021222746A2 (fr) 2020-04-30 2021-11-04 Arch Oncology, Inc. ANTICORPS SIRPα THÉRAPEUTIQUES
WO2021159130A2 (fr) 2020-05-15 2021-08-12 Modernatx, Inc. Vaccins a arn de coronavirus et procédés d'utilisation
WO2022110922A1 (fr) 2020-11-30 2022-06-02 启愈生物技术(上海)有限公司 ANTICORPS ANTI-SIRPα OU FRAGMENT DE LIAISON À L'ANTIGÈNE ASSOCIÉ, ET SON UTILISATION
WO2022120388A2 (fr) * 2020-12-04 2022-06-09 Tidal Therapeutics, Inc. Nanoparticules lipidiques et lipides cationiques ionisables, et leurs procédés de synthèse et d'utilisation
WO2022121980A1 (fr) 2020-12-11 2022-06-16 浙江博锐生物制药有限公司 ANTICORPS ANTI-SIRPα ET SON APPLICATION
WO2022254379A1 (fr) 2021-06-04 2022-12-08 Boehringer Ingelheim International Gmbh Anticorps anti-sirp-alpha
WO2022261101A1 (fr) 2021-06-07 2022-12-15 Generation Bio Co. Compositions de nanoparticules lipidiques modifiées par apolipoprotéine e et apolipoprotéine b et utilisations associées
WO2022261490A2 (fr) 2021-06-10 2022-12-15 Orna Therapeutics, Inc. Méthodes et compositions d'arn circulaire
WO2023020459A1 (fr) 2021-08-17 2023-02-23 杭州九源基因工程有限公司 ANTICORPS MONOCLONAL CIBLANT SIRPα ET SON UTILISATION
WO2023202672A1 (fr) 2022-04-20 2023-10-26 Biosion Inc. Anticorps ciblant sirp-alpha et leurs utilisations

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1980
AKINC ET AL., MOL THER., vol. 18, no. 7, 2010, pages 1357 - 1364
ANTONIA ET AL.: "Immuno-oncology combinations: a review of clinical experience and future prospects", CLIN. CANCER RES. OFF. J. AM. ASSOC. CANCER RES., vol. 20, 2014, pages 6258 - 6268, XP055567196, DOI: 10.1158/1078-0432.CCR-14-1457
BASHA ET AL., MOL THER, vol. 19, no. 12, 2011, pages 2186 - 2200
BELLIVEAU ET AL., MOL THER NUCLEIC ACIDS, vol. 1, 2012, pages e37
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
CHRAA ET AL., J LEUKOC BIOL., 2018, pages 1 - 13
DONG ET AL., PROC NATL ACAD SCI USA., vol. 111, no. 15, 15 April 2014 (2014-04-15), pages 5753
E. MEYERSW. MILLER, COMPUT. APPL. BIOSCI., vol. 4, 1988, pages 11 - 17
EDELMAN ET AL., PROC NATL ACAD SCI USA, vol. 63, 1969, pages 78 - 85
HUSTON ET AL., PROC. NATL. ACAD. SCI., vol. 85, 1988, pages 5879 - 5883
JAYARAMAN ET AL., ANGEW CHEM INT, vol. 51, no. 34, 2012, pages 8529 - 8533
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH
KEDMI, RANIT ET AL.: "A modular platform for targeted RNAi therapeutics", NATURE NANOTECHNOLOGY, vol. 13, no. 3, 2018, pages 214 - 219, XP093140904, DOI: 10.1038/s41565-017-0043-5
KHEIROLOMOOM AZADEH ET AL: "In situ T-cell transfection by anti-CD3-conjugated lipid nanoparticles leads to T-cell activation, migration, and phenotypic shift", BIOMATERIALS, ELSEVIER, AMSTERDAM, NL, vol. 281, 29 December 2021 (2021-12-29), XP086945695, ISSN: 0142-9612, [retrieved on 20211229], DOI: 10.1016/J.BIOMATERIALS.2021.121339 *
KORE ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 20132, pages 4570 - 4574
LEE ET AL., INT J CANCER., vol. 131, no. 5, 2012, pages E781 - 90
LEO, OSACHS, DHSAMELSON, L ET AL., J. IMMUNOL., vol. 137, 1986, pages 3874
LEUNG ET AL., J PHYS CHEM C NANOMATER INTERFACES, vol. 116, no. 34, 2012, pages 18440 - 18450
LOVE KT ET AL., PROC NATL ACAD SCI USA., vol. 107, no. 21, 25 May 2010 (2010-05-25), pages 9915
MAIER ET AL., MOL THER., vol. 21, no. 8, 2013, pages 1570 - 1578
MUI ET AL., MOL THER NUCLEIC ACIDS, vol. 2, 2013, pages el39
NEEDLEMANWUNSCH, J. MOL, BIOL., vol. 48, 1970, pages 444 - 453
NUPHAR VEIGA ET AL: "Cell specific delivery of modified mRNA expressing therapeutic proteins to leukocytes", NATURE COMMUNICATIONS, vol. 9, no. 1, 29 October 2018 (2018-10-29), XP055759710, DOI: 10.1038/s41467-018-06936-1 *
QIN JXUE LGONG NZHANG HSHEPHERD SJHALEY RMSWINGLE KLMITCHELL MJ.: "RGD peptide-based lipids for targeted mRNA delivery and gene editing applications", RSC ADV., vol. 12, no. 39, 7 September 2022 (2022-09-07), pages 25397 - 25404
ROBINSON ELISE R ET AL: "Abstract 5372: CD3 and CD8 targeting of ionizable lipid nanoparticles for in vivo mRNA delivery to T cells", vol. 82, no. 12_Supplement, 15 June 2022 (2022-06-15), XP093078452, Retrieved from the Internet <URL:https://aacrjournals.org/cancerres/article/82/12_Supplement/5372/703490/Abstract-5372-CD3-and-CD8-targeting-of-ionizable> *
SBDING, J.: "Protein homology detection by HMM-HMM comparison", BIOINFORMATICS, vol. 21, 2005, pages 951 - 960
SCHAEFER, W. ET AL., PNAS, vol. 108, 2011, pages 11187 - 1191
SEMPLE ET AL., NAT BIOTECHNOL., vol. 28, no. 2, 2010, pages 172 - 176
SI-YANG LIU ET AL., J. HEMATOL. ONCOL., vol. 10, 2017, pages 136
TAM ET AL., NANOMEDICINE, vol. 9, no. 5, 2013, pages 665 - 74
TOMBÁCZ ISTVÁN ET AL: "Highly efficient CD4+ T cell targeting and genetic recombination using engineered CD4+ cell-homing mRNA-LNPs", MOLECULAR THERAPY, vol. 29, no. 11, 1 November 2021 (2021-11-01), US, pages 3293 - 3304, XP055933699, ISSN: 1525-0016, DOI: 10.1016/j.ymthe.2021.06.004 *
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546

Similar Documents

Publication Publication Date Title
US10973917B2 (en) MRNA combination therapy for the treatment of cancer
AU2019253221B2 (en) PD-L1 binding affimers, and uses related thereto
US20230277652A1 (en) Nucleic acid vaccines for coronavirus
CN116113419A (zh) 环状rna组合物和方法
AU2023201482A1 (en) Cytokine conjugates for the treatment of autoimmune diseases
KR20200127207A (ko) Il-15 접합체 및 이의 용도
TW202023629A (zh) 用於調節單核球及巨噬細胞發炎表型之組合物及方法以及其免疫療法用途
KR20190020299A (ko) 면역 조정 폴리펩타이드를 암호화하는 mRNA의 조합물 및 이의 용도
KR20220061158A (ko) 자가면역질환을 치료하기 위한 il-2 접합체 및 사용 방법
TW202342753A (zh) 狂犬病核酸疫苗
CN114555632A (zh) Il-15缀合物及其用途
KR20220097445A (ko) 인터류킨 10 접합체 및 이의 용도
WO2023056915A1 (fr) Polynucléotides codant pour l&#39;interleukine-12 (il-12) et composition associée et procédés associés
WO2024200823A1 (fr) Nanoparticule à base de lipide ciblant des cellules immunitaires activées pour l&#39;expression d&#39;une molécule d&#39;amélioration de cellule immunitaire et son utilisation
WO2024200826A1 (fr) Nanoparticule à base de lipides ciblée au niveau de cellules immunitaires activées pour l&#39;expression d&#39;une molécule inhibitrice de cellules immunitaires et son utilisation
TW202332694A (zh) 血清半衰期延長之pd-l1結合多肽
RU2829811C1 (ru) Конъюгаты интерлейкина 10 и варианты их применения
JP2024160243A (ja) がんの治療のためのmRNA併用療法
EP4413038A1 (fr) Affimers de liaison à pd-l1
EP4408886A1 (fr) Traitement impliquant un arn non immunogène pour vaccination antigénique et antagonistes liant l&#39;axe pd-1
WO2024178413A1 (fr) Compositions de nanoparticules et leurs utilisations pour la réactivation du vih latent
WO2023218243A1 (fr) Protéines de fusion de liaison lag-3/pd-l1