专利摘要:
the present invention relates to extracellular vesicles comprising an immunomodulatory component. methods for producing extracellular vesicles and methods for using extracellular vesicles to treat cancer, gvhd and autoimmune diseases are also provided.
公开号:BR112020013131A2
申请号:R112020013131-8
申请日:2018-12-28
公开日:2020-12-08
发明作者:Nuruddeen D. Lewis;Yu Zhou;Sriram Sathyanarayanan;John D. Kulman;Douglas E. Williams;Leonid A. Gaydukov;Ke Xu;Shelly Martin
申请人:Codiak Biosciences, Inc.;
IPC主号:
专利说明:

[001] [001] This request claims the benefit of Provisional Application No. 62/723. 267, presented on August 27, 2018; and 62 / 611. 140, filed on December 28, 2017, each of which is incorporated by reference in its entirety. FIELD OF THE INVENTION
[002] [002] The invention relates to compositions for interacting and modulating the human immune system, methods for making the compositions and methods for using the compositions to treat cancer, GvHD and autoimmune diseases. FUNDAMENTALS
[003] [003] Immunotherapy is the treatment of the disease by inducing, improving or suppressing the immune response. Immunotherapy can stimulate the patient's own immune system to attack cancer cells. Cancer immunotherapy generally has fewer side effects than traditional cancer therapies, such as chemotherapy and radiation therapy. Anti-inflammatory immunotherapy can downwardly regulate the patient's immune system for the treatment of autoimmune diseases and graft versus host disease (GvHD). What is needed are improved methods for delivering immunomodulatory molecules to cells and tissues in the body. SUMMARY
[004] [004] As drug delivery vehicles, extracellular vesicles offer many advantages over traditional drug delivery methods, especially for gene therapy. The systemic delivery of extracellular vesicles results in the distribution of these lipid nanoparticles to various tissues. Studies have shown that extracellular vesicles can interact with several cells involved in modulating the human immune system. Extracellular vesicles that are selected, enriched or genetically modified to deliver therapeutic molecules to activate, suppress or influence the human immune system can be potent therapies for cancer and other diseases related to the immune system.
[005] [005] Compositions are provided here that comprise selected extracellular vesicles, enriched or genetically modified with immunomodulatory components that can regulate the human immune system in an increasing or decreasing way, stimulating the patient's immune system to fight cancer or suppressing the patient's immune system to relieve symptoms of GvHD and autoimmune diseases.
[006] [006] Methods of producing and using extracellular vesicles to modulate the human immune system are also provided.
[007] [007] Therefore, in a first aspect, a composition is provided here comprising: an extracellular vesicle comprising a cell membrane that connects a closed volume, the cell membrane having an inner surface and an outer surface; and a first immunomodulatory component associated with the cell membrane or closed within the closed volume.
[008] [008] In several modalities, the first immunomodulating component is an inhibitor for a negative checkpoint regulator or an inhibitor for a negative checkpoint regulator binding partner. In some of these modalities, the negative checkpoint regulator is selected from the group consisting of: protein 4 associated with cytotoxic T lymphocytes (CTLA-4), programmed cell death protein 1 (PD-1), gene 3 activated by lin-
[009] [009] In several modalities, the first immunomodulating component is an activator for a positive co-stimulating molecule or an activator for a binding partner for a positive co-stimulating molecule. In some embodiments, the positive co-stimulating molecule is a member of the TNF receptor superfamily. In some of these modalities, the member of the TNF receptor superfamily is selected from the group consisting of: CD120a, CD120b, CD18, OX40, CD40, Fas receptor, M68, CD27, CD30, 4-1BB, TRAILR1I, TRAILR2, TRAILR3, TRAILR4, RANK, OCIF, TWEAK receiver, TACI, BAFF receiver, ATAR, CD271, CD269, AITR, TROY, CD358, TRAMP and XEDAR. In some embodiments, the activator for a positive co-stimulating molecule is a member of the TNF superfamily. In some of these modalities, the member of the TNF superfamily is selected from the group consisting of: TNFa, TNF-C, OX40L, CD40L, FasL, LIGHT, TLIA, CD27L, Siva, CD153, ligand 4-1BB, TRAIL, RANKL, TWEAK , APRIL, BAFF, CAMLG, NGF, BDNF, NT-3, NT-A4, GITR and EDA-2 ligand. In certain embodiments, the member of the TNF superfamily is CD40L. In certain embodiments, the member of the TNF subfamily is CD27L. In certain embodiments, the member of the TNF superfamily is OX40L.
[0010] [0010] In some embodiments, the positive co-stimulating molecule is a co-stimulating molecule of the CD28 superfamily. In some of these modalities, the co-stimulating molecule of the CD28 superfamily is ICOS or CD28. In some embodiments, the activator of a positive co-stimulating molecule is ICOSL, CD80 or CD86. In certain embodiments, the activator of a positive costimulatory molecule is CD80.
[0011] [0011] In some embodiments, the first immunomodulating component is a cytokine or a cytokine binding partner. In some modalities, the cytokine is selected from the group consisting of: I1L-2, IL-7, 11-10, 11-12 and I1L-15. In certain embodiments, cytokine is IL-7. In a certain embodiment, the cytokine is I1L-12. In certain modalities, the cytokine is IL-15.
[0012] [0012] In some embodiments, the first immunomodulating component is a T cell receptor (TCR), a T cell coreceptor, a major histocompatibility complex (MHC), a human leukocyte antigen (HLA) or a derivative of it.
[0013] [0013] In some embodiments, the first immunomodulating component is an activator of a T cell receptor or coreceptor. In certain embodiments, the activator of a T cell receptor or coreceptor is a CD3 activator, optionally an agonist antibody to CD3.
[0014] [0014] In some modalities, the first immunomodulating component is a tumor antigen. In some modalities, the tumor antigen is selected from the group consisting of: alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), tumor epithelial antigen (ETA), mucin 1 (MUC1), Th-MUC1, mucin 16 (MUC16), tyrosinase, melanoma-associated antigen (MAGE), tumor protein p53 (p53), CD4, CD8, CD45, CD80, CD86, programmed death ligand 1 (PD-L1), programmed death ligand 2 (PD-L2), NY-ESO-1, PSMA, TAG-72, HER2, GD2, cMET, EGFR, Mesothelin, VEGFR, alpha-folate receptor, CE7R, IL-3, cancerous testis antigen, MART-1 gp100 and TNF-related apoptosis-inducing ligand. In certain embodiments, the tumor antigen is derived from a reference genome sequence. In certain embodiments, the tumor antigen is derived from an individual's genome sequence.
[0015] [0015] In some modalities, the first immunomodulating component is an agonist or antagonist of a selected target or activity.
[0016] [0016] In some embodiments, the first immunomodulating component is an antibody or antigen-binding fragment.
[0017] [0017] In some embodiments, the first immunomodulating component is a polynucleotide. In some of these modalities, the polynucleotide is selected from the group consisting of: an mRNA, a miRNA, a siRNA, an antisense RNA, a ShRNA, an IncRNA and a dsDNA.
[0018] [0018] In some embodiments, the first immunomodulating component is a protein, a peptide, a glycolipid or a glycoprotein.
[0019] [0019] In some embodiments, the first immunomodulating component is expressed as a fusion protein displayed on the external surface of the said extracellular vesicle. In some embodiments, the fusion protein comprises PTGFRN or a fragment or variant thereof. In some embodiments, the fusion protein sequence is SEQ ID NO: 3.
[0020] [0020] In some embodiments, the extracellular vesicle is an exosome. In some other modalities, the extracellular vesicle is a nanovesicle.
[0021] [0021] In certain embodiments, the composition further comprises a pharmaceutically acceptable carrier.
[0022] [0022] In some embodiments, the extracellular vesicle additionally comprises a second immunomodulatory component.
[0023] [0023] In several modalities, the second immunomotor component
[0024] [0024] In several modalities, the second immunomodulating component is an activator for a positive co-stimulating molecule or an activator for a binding partner for a positive co-stimulating molecule. In some embodiments, the positive co-stimulating molecule is a member of the TNF receptor superfamily. In some of these modalities, the member of the TNF receptor superfamily is selected from the group consisting of: CD120a, CD120b, CD18, OX40, CD40, Fas receptor, M68, CD27, CD30, 4-1BB, TRAILR1I, TRAILR2, TRAILR3, TRAILR4, RANK, OCIF, TWEAK receiver, TACI, BAFF receiver, ATAR, CD271, CD269, AITR, TROY, CD358, TRAMP and XEDAR. In some embodiments, the activator for a positive co-stimulating molecule is a member of the TNF superfamily. In some of these modalities, the member of the TNF superfamily is selected from the group consisting of: TNFa, TNF-C, OX40L, CD40L, FasL, LIGHT, TLIA, CD27L, Siva, CD153, ligand 4-1BB, TRAIL, RANKL, TWEAK , APRIL, BAFF, CAMLG, NGF, BDNF, NT-3, NT-A4, GITR and EDA-2 ligand. In certain embodiments, the member of the TNF superfamily is CD40L. In certain modalities, the member of the
[0025] [0025] In some modalities, the positive co-stimulating molecule is a co-stimulating molecule of the CD28 superfamily. In some of these modalities, the co-stimulating molecule of the CD28 superfamily is ICOS or CD28. In some embodiments, the activator of a positive co-stimulating molecule is ICOSL, CD80 or CD86. In certain embodiments, the activator of a positive co-stimulating molecule is CDB80.
[0026] [0026] In some embodiments, the second immunomodulating component is a cytokine or a cytokine binding partner. In some modalities, the cytokine is selected from the group consisting of: IL-2, IL-7, II-10, 11-12 and 11-15. In certain embodiments, cytokine is IL-7. In a certain embodiment, the cytokine is I1L-12. In some fashion, the cytokine is IL-15.
[0027] [0027] In some embodiments, the second immunomodulating component is a T cell receptor (TCR), a T cell coreceptor, a major histocompatibility complex (MHC), a human leukocyte antigen (HLA) or a derivative of it.
[0028] [0028] In some embodiments, the second immunomodulating component is an activator of a T cell receptor or coreceptor. In certain embodiments, the activator of a T cell receptor or coreceptor is a CD3 activator, optionally an agonist antibody to CD3.
[0029] [0029] In some embodiments, the second immunomodulating component is a tumor antigen. In some modalities, the tumor antigen is selected from the group consisting of: alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), tumor epithelial antigen (ETA), mucin 1 (MUC1), Th-MUC1, mucin 16 (MUC16), tyrosinase, melanoma-associated antigen (MAGE), pro-
[0030] [0030] In some embodiments, the second immunomodulating component is an agonist or antagonist of a selected target or activity.
[0031] [0031] In some embodiments, the second immunomodulating component is an antibody or antigen-binding fragment.
[0032] [0032] In some embodiments, the second immunomodulating component is a polynucleotide. In some of these modalities, the polynucleotide is selected from the group consisting of: an mRNA, a miRNA, a siRNA, an antisense RNA, a sShRNA, an IncRNA and a dsDNA.
[0033] [0033] In some embodiments, the second immunomodulating component is a protein, a peptide, a glycolipid or a glycoprotein.
[0034] [0034] In some embodiments, the second immunomodulating component is expressed as a fusion protein displayed on the outer surface of said extracellular vesicle. In some embodiments, the fusion protein comprises PTGFRN or a fragment or variant thereof. In some embodiments, the sequence of said fusion protein is SEQ ID NO: 3.
[0035] [0035] In some embodiments, the second immunomodulating component is different from the first immunomodulatory component
[0036] [0036] In some embodiments, the extracellular vesicle additionally comprises a third immunomodulatory component. In some embodiments, the third immunomodulatory component is different from the first and second immunomodulatory components.
[0037] [0037] In another aspect, a method of producing the composition is provided here. In some modalities, the method comprises modifying a producer cell with the first, second and / or third immunomodulatory components; obtain the extracellular vesicle of the producing cell; and optionally isolating the obtained extracellular vesicles. In some other modalities, the method comprises obtaining the extracellular vesicle from a producer cell; isolating the extracellular vesicles obtained; and modifying the isolated extracellular vesicle with the first, second and / or third immunomodulatory components. In certain embodiments, the method further comprises formulating the isolated extracellular vesicles in a pharmaceutical composition.
[0038] [0038] In another aspect, a method of treating cancer in an individual is provided here. The method comprises administering to the individual a therapeutically effective amount of the composition, in which the composition is able to increasingly regulate an immune response in the individual, thus improving the targeting of the tumor of the individual's immune system.
[0039] [0039] In another aspect, a method of treating graft versus host disease (GvHD) in an individual is provided here. The method comprises administering to the individual a therapeutically effective amount of the composition, in which the composition is able to downwardly regulate an immune response in the individual, thus alleviating the symptoms of GvHD.
[0040] [0040] In another aspect, a method of treating cancer in an individual is provided here. The method comprises administering to the individual a therapeutically effective amount of the composition, in which the composition is able to downwardly regulate an immune response in the individual, thereby suppressing the individual's immune activity.
[0041] [0041] In another aspect, a method of treating or preventing cancer in an individual is provided herein comprising administering to the individual a therapeutically effective amount of the composition comprising a tumor antigen, wherein the composition is capable of potentiating an immune response to the tumor antigen, thereby improving the individual's immune response to cancer.
[0042] [0042] In some modalities, the tumor antigen is selected from the group consisting of: alpha-fetoprotein (AFP), carcinoid embryo antigen (CEA), tumor epithelial antigen (ETA), mucin 1 (MUC1), Th -MUC1, mucin 16 (MUC16), tyrosinase, melanoma-associated antigen (MAGE), tumor protein p53 (p53), CD4, CD8, CD45, CD80, CD86, programmed death ligand 1 (PD-L1), ligand programmed death 2 (PD-L2), NY-ESO-1, PSMA, TAG-72, HER2, GD2, cMET, EGFR, Mesothelin, VEGFR, alpha-folate receptor, CE7R, IL-3, cancerous testis antigen , MART-1 gp100 and TNF-related apoptosis-inducing ligand.
[0043] [0043] In certain embodiments, the tumor antigen is derived from a reference genome sequence. In certain embodiments, the tumor antigen is derived from an individual's genome sequence. BRIEF DESCRIPTION OF THE FIGURES
[0044] [0044] Figures 1A and 1B show a time course of mice injected with radiolabeled exosomes. Figure 1A shows the intravenous route of administration. Figure 1B shows the intraperitoneal route of administration.
[0045] [0045] Figure 2 is a quantification of the distribution of exosomes in different mouse tissues after intravenous and intraperitoneal administration of radiolabeled exosomes.
[0046] [0046] Figures 3A and 3B show the effects of B cell activation in peripheral blood mononuclear cells (PBMCs) from two human donors after incubation with exosomes that express CD40L.
[0047] [0047] Figures 4A and 4B show the effects of B cell activation of purified B cells from two human donors after incubation with exosomes that express CD40L.
[0048] [0048] Figure 5A is a schematic of a CD40 reporter cell line. Figure 5B shows the concentration-dependent activation of a CD40 reporter cell line treated with a recombinant human anti-CD40 or CD40L agonistic antibody. Figure 5C shows the effects of exosomes expressing CD40L on a CD40 reporter cell image.
[0049] [0049] Figures 6A and 6B show the effects of T cell activation in peripheral blood mononuclear cells (PBMCs) with exosomes that express CD80. Figure 6A shows the effect of exosomes that express CD80 on the number of CD8 * T cells. Figure 6B shows the effect of exosomes that express CD80 on the number of CD4 T cells *.
[0050] [0050] Figures 7A and 7B show the effects of exosomes that express CD80 on the expression of IFNy in human PBMCs.
[0051] [0051] Figures 8A and 8B show the effects of exosomes expressing CD27L on the expression of IFNy in human PBMCs from two donors.
[0052] [0052] Figures 9A and 9B show the effects of exosomes expressing CD27L on IL-2 expression in human PBMCs from two donors.
[0053] [0053] Figures 10A and 10B show the effects of exosomes that express OX40L on the expression of IFNy in human PBMCs from two donors.
[0054] [0054] Figures 11A and 11B show the effects of exosomes that express OX40L on IL-2 expression in human PBMCs from two donors.
[0055] [0055] Figure 12A is a schematic of an OX40 reporter cell line. Figure 12B shows the concentration-dependent activation of an OX40 reporter cell line treated with a recombinant human anti-OX40 or OX40L agonistic antibody. Figure 12C shows the effects of exosomes that express OX40L on an OX40 reporter cell line.
[0056] [0056] Figures 13A and 13B show the effects of exosomes expressing IL-7 in combination with an anti-CD3 antibody on the expression of IFNy in human PBMCs.
[0057] [0057] Figure 14A is a schematic of an IL-7 receptor reporter cell line. Figure 14B shows the concentration-dependent activation of an IL-7 receptor reporter cell line treated with recombinant human IL-7. Figure 14C shows the effects of exosomes expressing IL-7 on an IL-7 receptor reporter cell line.
[0058] [0058] Figures 15A and 15B show the effects of exosomes that express IL-7 on T cell proliferation in mice in vivo as measured by EdU incorporation. Figure 15A shows the effects of exosomes that express IL-7 on CD8 + T cells. Figure 15B shows the effects of exosomes that express IL-7 on the memory CD8 + T cell.
[0059] [0059] Figures 16A and 16B show the effects of exosomes that express IL-7 on T cell proliferation in mice in vivo as measured by CD71 positivity. Figure 16A shows the effects of exosomes that express IL-7 on CD8 + T cells. Figure 16B shows the effects of exosomes that express IL-7 on the memory CD8 + T cell.
[0060] [0060] Figure 17A shows a schematic of a PTGFRN / IL-7 fusion protein expressed in high density on the surface of an exosome and variants of the fusion protein. Figure 17B is the sequence of the optimized PTGFRN / IL-7 fusion protein.
[0061] [0061] Figure 18A is a Western blot showing the relative expression of different IL-7 fusion proteins on the surface of purified exosomes. Figure 18B shows the effects of exosomes that express IL-7 on the down-regulation of the IL-7 receptor as a model of IL-7 mediated T cell activation.
[0062] [0062] Figure 19A shows the effects of anti-CD3 scFv exosomes on T cell activation in PBMCs. Figure 19B shows the effects of exosomes of anti-CD3 scFv on B cell activation in PBMCs.
[0063] [0063] Figure 20A shows the effects of anti-CD3 scFab exosomes on T cell activation in PBMCs. Figure 20B shows the effects of exosomes of anti-CD3 scFv on B cell activation in PBMCs.
[0064] [0064] Figure 21A is a histogram showing the extent of T cell activation after treatment with anti-CD3 scFv exosomes. Figure 21B is a histogram showing the extent of B cell activation after treatment with anti-CDB3 scFv exosomes.
[0065] [0065] Figure 22A shows the effects of anti-CD3 scFab exosomes on T cell activation in a coated plate activation assay, compared to soluble anti-CD3 antibody or anti-
[0066] [0066] Figure 23A shows a schematic of a full-length fusion protein PTGFRN / IL-12. Figure 23B shows a schematic of a shortened PTGFRN / IL-12 fusion protein.
[0067] [0067] Figure 24A shows the effects of recombinant human IL-12 or exosomes that overexpress the full-length or short PTGFRN-IL-12 inducing IFNy in human PBMCs. Figure 24B is a table that summarizes the potency of exosomes containing IL-12 and recombinant IL-12.
[0068] [0068] Figure 25 shows the effects of exosomes of IL-12-PTGFRN and recombinant IL-12 on reducing tumor growth in a murine model of melanoma.
[0069] [0069] Figure 26A shows the tumor growth curves for each of the tumor-bearing mice shown in Figure 25 treated with PBS. Figure 26B shows the tumor growth curves for each of the tumor-bearing mice shown in Figure 25 treated with recombinant IL-12. Figure 26C shows the tumor growth curves for each of the tumor-bearing mice shown in Figure 25 treated with | L-12-PTGFRN exosomes.
[0070] [0070] Figure 27 shows images of all mice bearing a B16F10 tumor in the efficacy study shown in Figure
[0071] [0071] Figure 28 shows the survival curves of mice with B16F10 tumor shown in Figure 25.
[0072] [0072] Figure 29A shows the levels of expression of the IFNy gene in mouse tumors treated with exosomes of | L-12-PTGFRN, PBS or rllL-12. Figure 29B shows the levels of expression
[0073] [0073] Figure 30 shows the percentage of splenic CD8 + T cells positive for IFNy in tumor-bearing mice treated with exosomes of IL-12-PTGFRN, PBS or rlL-12.
[0074] [0074] Figure 31A shows a schematic of a full-length PTGFRN fused to an IFNy monomer. Figure 31B shows a schematic of a full-length PTGFRN fused to an IFNy tandem dimer.
[0075] [0075] Figure 32 shows the results of the PAGE analysis of PTGFRN IFNy exosomes of tandem dimer (td) and monomeric (m) and purified humans.
[0076] [0076] Figure 33 shows the expression of monocyte PD-L1 after addition of native exosomes (WT), monomeric IFNy exosomes PTGFRN (m-IFNy-PTGFRN) and tandem dimer IFNy PTGFRN exosomes (td- IFNy-PTGFRN), respectively. LPS-induced PD-L1 activation was used as a positive control.
[0077] [0077] Figure 34 shows the schemes of the fusion / IL-15Ra proteins fused to the transmembrane domain of PDGFR.
[0078] [0078] Figure 35 shows NK cell activation measured by the percentage of CD69 positive NK cells after adding IL-15 exosomes from pDisplay.
[0079] [0079] Figure 36A shows the schemes of IL-15 fused to full-length PTGFRN and IL-15 N72D fused to full-length PTGFRN. Figure 36B shows Western blotting of IL-15 fused to full-length PTGFRN and IL-15 N72D fused to
[0080] [0080] Figure 37 shows the activation of NK cells measured by the percentage of NK CD69 positive cells after the addition of I | L-15 fused to full-length PTGFRN and IL-15 N72D fused to full-length PTGFRN.
[0081] [0081] Figure 38 shows the schematics of the anti-CD3 antibody fragment fused to the transmembrane region of PDGFR (exoCD3-PD), a full-length PTGFRN (long exoCD3) and a fragment of PTGFRN (short exoCD3), respectively.
[0082] [0082] Figure 39 shows the results of bio-layer interferometry (BLI) after adding native exosomes (WT), exosomes with an anti-CD3 antibody fragment fused to the PDGFR transmembrane region (pDisplay), exosomes with fragment of full-length fused anti-CD3 antibody PTGFRN (FL PTGFRN) and exosters with a fragment of anti-CD3 antibody fused to a fragment of PTGFRN (short PTGFRN), respectively.
[0083] [0083] Figure 40A shows CD4 + T cell activation measured by the percentage of CD4 + CD69 positive T cells after the addition of the anti-CD3 antibody fragment. Figure 40B shows the activation of CD4 + T cells measured by the percentage of CD4 + CD69 positive T cells after the addition of native exosomes (exoNative) and exosomes with anti-CD3 antibody fragment fused to a fragment of PTGFRN (exoCD3 Short ), respectively.
[0084] [0084] Figure 41 shows the schemes of the CD40L-GFP PTGFRN fusion proteins and the EC for each construct in the B cell activation assay measured by CD69 positivity in B cells.
[0085] [0085] Figure 42A shows B cell activation measured by the percentage of CD69 positive B cells after adding native exosomes, exosomes with quarterly CD40L-PTGFRN constructs pOB-527 and exosomes with quarterly CDCL-PTGFRN constructs pCB- 766, respectively. Figure 42B shows B cell activation measured by the percentage of CD69 positive B cells after the addition of exosomes with quarterly CD40L-PTGFRN constructs pCB-527 and pCB-766, respectively, compared to CD40L corresponding to the concentration.
[0086] [0086] Figure 43A shows the activation of B cells in donor 1 measured by the percentage of CD69 positive B cells after adding exosomes with trimester CD40L-PTGFRN constructs, pCB-527. Figure 43B shows B cell activation in donor 2 as measured by the percentage of CD69 positive B cells after adding exosomes with trimester CD40L-PTGFRN constructs, pCB-527.
[0087] [0087] Figure 44A shows the FACS analysis of native exosomes isolated with microspheres decorated with anti-CD40L and stained with fluorescent antibodies against IL-12 and CD40L. Figure 44B shows the FACS analysis of native exosomes isolated with microspheres decorated with anti-CD40L and labeled with fluorescent antibodies against CD81 and CD40L.
[0088] [0088] Figure 45A shows the FACS analysis of genetically modified double exosomes of PTGFRN-CD40L / IL-12 isolated with microspheres decorated with anti-CD40L and labeled with fluorescent antibody against CD81. Figure 45B shows the FACS analysis of genetically modified exosomes of PTGFRN-CD40L / IL-12 isolated with microspheres decorated with anti-CD40L and labeled with fluorescent antibodies against I | L-12 and CDA40L.
[0089] [0089] Figure 46A shows the FACS analysis of genetically modified exosomes of PTGFRN-CD40L / 11-12 isolated with microspheres decorated with anti-IL-12 and marked with fluorescent antibodies against IL-12 and CD40L . Figure 46B shows the FACS analysis of double genetically modified double exosomes of PTGFRN-CD40L / IL-12 isolated with microspheres decorated with anti-IL-12 and labeled with fluorescent antibody against CD81.
[0090] [0090] Figure 47A shows the IFNy response in human PBMCs from donor 1 after the addition of recombinant IL-12, recombinant I1L-12 mixed with recombinant exosomes of CD40L, PTGFRN-IL-12, exosomes PTGFRN- CD40L / IL-12 doubly positive and a mixture of exosomes PTGFRN-IL-12 and exosomes PTGFRN-CD40L, respectively. Figure 47B shows the IFNy response in human PBMCs from donor 2 after adding recombinant IL-12, recombinant IL-12 mixed with recombinant exosomes of CD40L, PTGFRN-IL-12, exosomes PTGFRN-CDA40L / IL - 12 doubly positive and a mixture of exosomes PTGFRN-IL-12 and exosomes PTGFRN-CD40L, respectively.
[0091] [0091] Figure 48 shows the IFNy response in human PBMCs from donor 1 and donor 2 after the addition of recombinant IL-12, recombinant 11-12 mixed with the recombinant exosomes of CD40L, PTGFRN-IL- 12, double positive PTGFRN-CDA40L / IL-12 exosomes and a mixture of PTGFRN-IL-12 exosomes and PTGFRN-CDA40L exosomes, respectively.
[0092] [0092] Figure 49A shows activation of B cells in human PBMCs from donor 1 after addition of recombinant CD40L, | L-12 mixed with recombinant CD40L, exosomes PTGFRN-CD40L, exosomes PTGFRN-CD40L / IL-12 doubly positive and a mixture of exosomes PTGFRN-IL-12 and exosomes PTGFRN-CD40L, respectively. Figure 49B shows the activation of B cells in human PBMCs from donor 2 after the addition of recombinant CD40L, recombinant IL-12 mixed with recombinant CD40L, exosomes PTGFRN-CD40L, exosomes PTGFRN-CDA40L / IL-12 doubly
[0093] [0093] Figure 50 shows the IFNy response in human PBMCs from donor 1 and donor 2 after the addition of recombinant CD40L, 11-12 recombinant mixed with recombinant CD40L, exosomes PTGFRN-CD40L, exosomes PTGFRN-CD40L / 11-12 doubly positive and a mixture of PTGFRN-IL-12 exosomes and PTGFRN-CDA40L exosomes, respectively.
[0094] [0094] Figure 51A shows the FACS analysis of genetically modified exosomes of PTGFRN-CD40L / IL-12 / FLT3L isolated with microspheres decorated with anti-IL-12 and labeled with fluorescent antibodies against IL-12 and CD40L. Figure 51B shows the FACS analysis of genetically modified triple exosomes PTGFRN-CDA40L / IL-12 / FLT3L isolated with microspheres decorated with anti-IL-12 and labeled with fluorescent antibodies against IL-12 and FLT3L. Figure 51C shows the FACS analysis of genetically modified triple exosomes PTGFRN-CDA40L / IL-12 / FLT3L isolated with microspheres decorated with anti-IL-12 and labeled with fluorescent antibodies against CD40L and FLT3L.
[0095] [0095] Figure 52A shows the FACS analysis of genetically modified triple exosomes of PTGFRN-CD40L / IL-12 / FLT3L isolated with microspheres decorated with anti-CD40L and marked with fluorescent antibodies against | L-12 and CD40L. Figure 52B shows the FACS analysis of genetically modified triple exosomes PTG-FRN-CD40L / IL-12 / FLT3L isolated with microspheres decorated with anti-CD40L and labeled with fluorescent antibodies against I | L-12 and FLT3L. Figure 52C shows the FACS analysis of genetically modified triple exosomes PTGFRN-CD40L / IL-12 / FLT3L isolated with microspheres decorated with anti-CD40L and labeled with fluorescent antibodies against CD40L and FLT3L. DETAILED DESCRIPTION
[0096] [0096] Extracellular vesicles capable of modulating the human immune system are disclosed here. Methods are also provided to produce extracellular vesicles and methods for using these extracellular vesicles to treat cancer and other diseases related to the immune system.
[0097] [0097] Before the present invention is described in more detail, it should be understood that this invention is not limited to certain described modalities, as such, of course, may vary. It should also be understood that the terminology used in this document is intended to describe particular modalities only and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
[0098] [0098] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit, unless the context clearly dictates otherwise, between the upper and lower limits of the range and any other value established or intervening in this established range is included in the invention. The upper and lower limits of these smaller ranges can be independently included in the smaller ranges and are also included in the invention, subject to any limit specifically excluded in the established range. Where the indicated range includes one or both limits, ranges excluding either or both of the included limits are also included in the invention.
[0099] [0099] Unless otherwise defined, all technical and scientific terms used in this document have the same meaning as commonly understood by one versed in the technique to which this invention belongs. While any methods and materials similar or equivalent to those described in this document can also be used in the practice or testing of the present invention, representative illustrative materials and methods are now described.
[00100] [00100] All publications and patents cited in this specification in this document are incorporated by reference, as if each individual publication or patent has been specifically and individually indicated to be incorporated by reference and the addendum by reference to be disclosed is incorporated and describe the methods and / or materials in relation to which the publications are cited.
[00101] [00101] This should be noted that as used in this document and in the added claim, the singular forms "one", "one" and "o" include plural reference, unless the context clearly dictates otherwise. It is further noted that claims can be drafted to exclude any optional elements. As such, this statement is intended to serve as an antecedent basis for the use of such exclusive terminology as "only," "only" and the like in connection with the recitation of claim elements, or use of a "negative" limitation .
[00102] [00102] Just as it may be apparent to those skilled in the art when reading this disclosure, each of the individual modalities described and illustrated in this document has discrete components and features that can be readily separated or combined with the characteristics of any of the other several modalities without departing from the scope or meaning of the present invention. Any method recited may be carried out in the order of the events recited or in any other order that is logically possible.
[00103] [00103] In the further description of the invention in question, the systems in question for use in the practice of the methods in question will be discussed in more detail, followed by a review of the associated methods.
[00104] [00104] As used here, the term "extracellular vesicle" refers to a vesicle derived from a cell that comprises a membrane surrounding an internal space. Extracellular vesicles comprise all vesicles attached to the membrane that have a smaller diameter than the cell from which they are derived. Generally, extracellular vesicles vary in diameter from 20 nm to 1000 nm and can comprise several macromolecular charges within the internal space, displayed on the external surface of the extracellular vesicle and / or covering the membrane. The charge may comprise nucleic acids, proteins, carbohydrates, lipids, small molecules and / or combinations thereof. As an example and without limitation, extracellular vesicles include apoptotic bodies, cell fragments, vesicles derived from cells by direct or indirect manipulation, (for example, by serial extrusion or treatment with alkaline solutions), vesiculated organelles and vesicles produced by living cells (for example, by direct budding of the plasma membrane or fusion of the late endosome with the plasma membrane). Extracellular vesicles can be derived from a living or dead organism, explanted tissues or organs and / or cultured cells.
[00105] [00105] As used herein, the term "exosome" refers to a small cell-derived vesicle (between 20 and 300 nm in diameter, more preferably 40 to 200 nm in diameter) that comprises a membrane that surrounds an internal space and that it is generated from the cell by direct budding of the plasma membrane or by the fusion of the late endosome with the plasma membrane. The exosome is a kind of extracellular vesicle. The exosome comprises lipid or fatty acid and polypeptide and optionally comprises a payload, (for example, a therapeutic agent), a receptor, (for example, a chemical targeting portion), a polynucleotide, (for example, a nucleic acid, RNA or DNA), a sugar, (for example, a simple sugar, polysaccharide or glycan) or other molecules. The exosome can be derived from a producer cell and isolated
[00106] [00106] As used here, the term "nanovesicle" refers to a small cell-derived vesicle (between 20 and 250 nm in diameter, more preferably 30 to 150 nm in diameter) comprising a membrane that surrounds an internal space and that is generated from the cell by direct or indirect manipulation, so that the nanovesicle is not produced by the producing cell without manipulation. Appropriate manipulations of the producer cell include, but are not limited to, serial extrusion, treatment with alkaline solutions, sonication or combinations thereof. The production of nanovesicles can, in some cases, result in the destruction of the producing cell. Preferably, nanovesicle populations are substantially free of vesicles that are derived from producer cells through direct budding of the plasma membrane or fusion of the late endosome with the plasma membrane. The nanovesicle is a kind of extracellular vesicle. The nanovesicle comprises lipid or fatty acid and polypeptide and optionally comprises a payload, (for example, a therapeutic agent), a receptor, (for example, a chemical targeting portion), a polynucleotide, (for example, an acid nucleic acid, RNA or DNA), a sugar, (for example, a simple sugar, polysaccharide or glycan) or other molecules. The nanovesicle, once derived from a producer cell according to manipulation, can be isolated from the producer cell based on its size, density, biochemical parameters or a combination of them.
[00107] [00107] The term "extracellular vesicle delivery" or "extracellular vesicle delivery" refers to the administration and location of extracellular vesicles to target the individual's tissues, cells and / or organs. In some embodiments, the immunomodulatory component can be delivered to the cytoplasm of a target cell. In other ways, the immunomodulatory component is delivered to the membrane of the target cell. In some embodiments, the extracellular vesicle membrane fuses with a membrane of a target cell.
[00108] [00108] “As used here, the term" producing cell "refers to any cell from which an extracellular vesicle can be isolated. A producer cell is a cell that serves as a source for the extracellular vesicle. A producer cell can share a protein, lipid, sugar or nucleic acid component with the extracellular vesicle. In some embodiments, the producing cell is a modified or synthetic cell. In some embodiments, the producer cell is a cultured or isolated cell. In certain embodiments, the producing cell is a cell line. In certain other modalities, the producing cell is a primary cell. In some particular modalities, the producing cell is an immune cell.
[00109] [00109] "Membrane", as used here, is a boundary layer that separates an internal space from an external space comprising one or more biological compounds, typically lipids and optionally polypeptides and / or carbohydrates. In some embodiments, the membrane comprises lipids and fatty acids. In some modalities, the membrane comprises phospholipids, glycolipids, fatty acids, sphingolipids, phosphoglycerides, sterols, cholesterols and phosphatidyl serines. In some of these embodiments, the membrane further comprises one or more polypeptides and / or one or more polysaccharides, such as glycan. The extracellular vesicle comprises a membrane as defined herein.
[00110] [00110] As used in this document, the term "immunomodulation component" refers to a therapeutic agent that acts on a target, (for example, a target cell) that is contacted with the extracellular vesicle and regulates the system immune. The immunomodulatory component that can be introduced into an extracellular vesicle and / or a producing cell includes therapeutic agents, such as modulators of checkpoint inhibitors or ligand of checkpoint inhibitors, surface antigens and their derivatives , cytokines and their derivatives. The immunomodulatory component can also include an agonist, an antagonist, an antibody and an antigen-binding fragment or a polynucleotide, such as siRNA, MIRNA, IncRNA and DNA.
[00111] [00111] The term "receptor" refers to a molecule that directs the extracellular vesicle to a target and / or promotes the interaction of the extracellular vesicle with the target in the individual. In some embodiments, the receptor is a polypeptide. In some embodiments, the receptor is able to increase the concentration of the immunomodulatory component in the individual's tissue. Examples of receivers include, but are not limited to, examples listed in Table 3.
[00112] [00112] The term "target" refers to a cell, a pathogen, a metabolite, a polypeptide complex or any molecule or structure that resides in a tissue or circulates in the individual's circulatory system or lymphatic system, such as a immune cell or a cancer cell. Examples of targets include, but are not limited to, examples listed in Table 4.
[00113] [00113] A "therapeutic agent" or "therapeutic molecule" includes a compound or molecule that, when present in an effective amount, produces a desired therapeutic effect, pharmacological and / or physiological effect on an individual in need thereof. It includes any compound, for example, a small molecule drug or a biological agent, (for example, a polypeptide drug or a nucleic acid drug) that, when administered to an individual, has a measurable effect or disease on the individual, for example, relieves or lessens a symptom of a disease, disorder or condition.
[00114] [00114] “As used here, the term" antibody "encompasses an immunoglobulin, whether natural or partially or totally synthetic, and its fragments. The term also encompasses any protein having a binding domain that is homologous to an immunoglobulin binding domain. "Antibody" further includes a polypeptide comprising an immunoglobulin gene framework region or fragments thereof that specifically bind and recognize an antigen. The use of the term antibody should include whole antibodies, polyclonal, monoclonal and recombinant antibodies and their fragments, in addition to including single chain antibodies, humanized antibodies, murine, chimeric antibodies, mouse-human monoclonal antibodies, mouse primates, pre-humans, anti-idiotype antibodies, antibody fragments, such as, for example, scFv, (scFv) 2, Fab, Fab 'and F (ab') 2a, F (ab1) 2 , Fv, dAb and Fd fragments, antibodies and antibody-related polypeptides. The antibody includes bispecific antibodies and multispecific antibodies, as long as they exhibit the desired biological activity or function.
[00115] [00115] The term "antigen binding fragment" used herein refers to fragments of an intact immunoglobulin and to any part of a polypeptide including antigen binding regions with the ability to specifically bind the antigen. For example, the antigen-binding fragment may be an F (ab ') 2 fragment, a Fab' fragment, a Fab fragment, an Fv fragment or an scFv fragment, but is not limited to them. A Fab fragment has an antigen-binding site and contains the variable regions of a light chain and a heavy chain, the light chain constant region and the first heavy chain CH1 constant region. A Fab 'fragment differs from a Fab fragment in that the Fab' fragment additionally includes the hinge region of the heavy chain, including at least
[00116] [00116] The phrase "nucleic acid molecule" refers to a single- or double-stranded polymer of deoxyribonucleotide or ribonucleotide bases. It includes chromosomal DNA and self-replicating plasmids, vectors, MRNA, tRNA, siRNA, miRNA, etc. The nucleic acid molecule can be recombinant and exogenous polypeptides can be expressed when the nucleic acid is introduced into a cell.
[00117] [00117] The term "agonist" refers to a molecule that binds to a receptor and activates the receptor to produce a biological response. The receptors can be activated by an endogenous or exogenous agonist. Non-limiting examples of endogenous agonists include hormones and neurotransmitters. Non-limiting examples of exogenous agonists include drugs. The agonist can be a complete, partial or reverse agonist.
[00118] [00118] The term "antagonist" refers to a molecule that blocks or dampens an agonist-mediated response, instead of provoking the biological response itself by binding to a receptor. Many antagonists reach their potency by competing with endogenous ligands or substrates at structurally defined binding sites on the receptors. Non-limiting examples of antagonists include alpha blockers, beta blockers and calcium channel blockers. The antagonist can be a competitive, non-competitive or non-competitive antagonist.
[00119] [00119] “As used herein, the term" a fragment "of a protein refers to a protein that is deleted at the Ne / or C terminal in comparison to the naturally occurring protein. Preferably, a carrier fragment PTGFRN, BSG, IGSF2, IGSF3, IGSF8, ITGB1, ITGA4, SLC3A2 or ATP maintains the ability to be targeted specifically for exosomes. This fragment is also called a "functional fragment". The possibility of a fragment being a functional fragment in this sense can be evaluated by any method known in the art to determine the protein content of exosomes, including Western Blots, FACS analysis and fragment fusion with autofluorescent proteins such as, for example, GFP. In a specific embodiment, the PTGFRN, BSG, IGSF2, IGSF3, IGSF8, ITGB1, ITGAA4, SLC3A 2, ATP carrier fragment retains at least 50%, 60%, 70%, 80%, 90% or 100%
[00120] [00120] As used here, the term "variant" of a protein refers to a protein that shares a certain identity of amino acid sequence with another protein after alignment by a method known in the art. A variant of a protein can include a substitution, insertion, exclusion, change of frame or rearrangement in another protein. In a specific embodiment, the variant is a variant with at least 70% identity for the PTGFRN, BSG, IGSF2, IGSF3, IGSF8, ITGB1, ITGA4, SLC3A 2, ATP or a fragment of PTGFRN, BSG , IGSF2, IGSF3, IGSF8, ITGB1, Carrier ITGA4, SLC3A2 or ATP. In some variant or variant modalities of PTGFRN fragments they share at least 70%, 80%, 85%, 90%, 95% or 99% of sequence identity with PTGFRN according to SEQ ID NO: 1 or with a functional fragment of it. In some variant or variant embodiments of BSG fragments they share at least 70%, 80%, 85%, 90%, 95% or 99% of sequence identity with BSG according to SEQ ID NO: 9 or with a functional fragment the same. In some variant or variant embodiments of IGSF2 fragments they share at least 70%, 80%, 85%, 90%, 95% or 99% of sequence identity with IGSF2 according to SEQ ID NO: 34 or with a functional fragment the same. In some variant or variant modes of IGSF3 fragments they share at least 70%, 80%, 85%, 90%, 95% or 99% of sequence identity with IGSF3 according to SEQ ID NO: 20 or with a functional fragment of it. In some variant or variant embodiments of IGSF8 fragments they share at least 70%, 80%, 85%, 90%, 95% or 99% of sequence identity with IGSF8 according to SEQ ID NO: 14 or with a functional fragment the same.
[00121] [00121] The sequence alignment methods for comparison are well known in the art. Various alignment programs and algorithms are described in: Smith and Waterman, Adv. Appl. Math. 2: 482 (1981); Needleman and Wunsch, J. Mol. Bio. 48: 443 (1970); Pearson and Lipman, Methods in Mol. Biol. 24: 307-31 (1988); Higgins and Sharp, gene 73:15 237-44 (1988); Higgins and Sharp, CABIOS 5: 151-3 (1989) Corpet et al., Nuc. Acids Res. 16: 10881-90 (1988); Huang et al., Comp. Appl. Biosci. 8: 155-65 (1992); and Pearson et al., Meth. Mol. Biol. 24: 307-31 (1994). The NCBI Basic Local Alignment Search Tool (BLAST) [Altschul 20 et al., J !. Mol. Biol. 215: 403-10 (1990) J is available from several sources, including the National Information Center
[00122] [00122] The recitation of any protein provided here covers a functional variant of the protein. The term "functional variant" of a protein refers to a variant of the protein that retains the ability to be targeted specifically for exosomes.
[00123] [00123] “As used herein, the term" pharmaceutical composition "refers to one or more of the compounds described herein, such as, for example, an extracellular vesicle mixed or mixed with, or suspended in one or more other components chemicals, such as pharmaceutically acceptable carriers and excipients. An objective of a pharmaceutical composition is to facilitate the administration of extracellular vesicle preparations to an individual. The term "pharmaceutically acceptable" and grammatical variations thereof refer to compositions, carriers, diluents and reagents capable of administering to or over an individual without producing undesirable physiological effects to a degree that prohibits administration of the composition. The term "excipient" or "carrier" refers to an inert substance added to a pharmaceutical composition to further facilitate the administration of a compound. The term "pharmaceutically acceptable carrier" or "pharmaceutically acceptable excipient" encompasses any of the agents approved by a US federal government regulatory agency or listed in the US Pharmacopoeia for use in animals, including humans, as well as any carrier or thinner that does not cause significant irritation to an individual and does not
[00124] [00124] “As used here, the terms" isolate, "" isolated "and" isolation "or" purify "" purified "and" purification ", as well as" extracted "and" extraction "are used in a interchangeable and refer to the state of a preparation, for example, a plurality of quantity and / or concentration known or unknown) of desired extracellular vesicles, which have undergone one or more purification processes, for example, a selection or an enrichment of the desired extracellular vesicle preparation. In some modalities, isolating or purifying as used herein is the process of removing, partially removing (for example, a fraction) the extracellular vesicles from a sample containing producer cells. In some modalities, an isolated extracellular vesicle composition has no detectable undesirable activity or, alternatively, the level or amount of undesired activity is equal to or less than an acceptable level or amount. In other embodiments, an isolated extracellular vesicle composition has a desired amount and / or concentration of the extracellular vesicles at or above an acceptable amount and / or concentration. In other embodiments, the isolated extracellular vesicle composition is enriched compared to the starting material (for example, preparations of producer cells) from which the composition is obtained. This enrichment can be 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, 99.9 %, 99.99%, 99.999%, 99.9999% or greater than 99.9999% compared to the starting material. In some embodiments, preparations isolated from extracellular vesicles are substantially free of residual biological products. And bad-
[00125] [00125] The terms "administration", "administration"; and its variants refer to the introduction of a composition, such as an extracellular vesicle or agent in an individual and include the simultaneous and sequential introduction of a composition or agent. The introduction of a composition or agent in an individual is by any suitable route, including oral, pulmonary, intranasal, parenteral (intravenous, intra-arterial, intramuscular, intraperitoneal or subcutaneous), rectal, intra-lymphatic, intrathecal, intratumoral, periocular or topical. Management includes self-administration and administration by third parties. A suitable route of administration allows the composition or agent to perform its intended function. For example, if a suitable route is intravenous, the composition is administered by introducing the composition or agent into the subject's vein.
[00126] [00126] As used here, the term "modular", "modulation", "modify" and / or "modulator" generally refers to the ability to alter, increase or decrease, for example, promoting / stimulating / regulating so increasing or promoting directly or indirectly interfering / inhibiting / regulating in a decreasing way a specific concentration, level, expression, function or behavior, such as, for example, acting as an antagonist or agonist. In some cases, a modulator may increase and / or decrease a certain con-
[00127] [00127] The term "sufficient quantity"; means an amount sufficient to produce the desired effect, for example, an amount sufficient to modulate a condition in the subject.
[00128] [00128] The term "therapeutically effective amount" is an amount that is effective in improving a symptom of a disease. A therapeutically effective amount can be a "prophylactically effective amount", as prophylaxis can be considered therapy.
[00129] [00129] As used herein, the term "substantially" or "substantial" refers, for example, to the presence, level or concentration of an entity in a specific space, the effect of an entity in another entity or the effect of a treatment. For example, an activity, level, or concentration of an entity increases substantially if the increase is 2 times, 3 times, 4 times, 5 times, 10 times, 50 times, 100 times, or 1000 times double in relation to a line. base. An activity, level or concentration of an entity is also increased substantially if the increase is 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% ,, 90%, 100 %, 200% or 500% in relation to a baseline.
[00130] [00130] The term "in vivo" refers to processes that occur in a living organism.
[00131] [00131] The term "mammal", as used herein, includes humans and non-human mammals.
[00132] [00132] The abbreviations used in this application include the following: "MRNA" refers to messenger RNA, "miRNA" refers to microRNA, "siRNA" refers to small interfering RNA, "antisense RNA" refers to Single-stranded RNA that is complementary to an mRNA, "ShRNA" refers to small or short RNA, "LncRNA" refers to the
[00133] [00133] Individual aspects of the disclosure include a composition capable of regulating the immune system. The composition comprises an extracellular vesicle comprising a cell membrane and an immunomodulatory component associated with the cell membrane or closed within the closed volume bound to the membrane. THE EXTRACELLULAR VESICLE
[00134] [00134] In several embodiments, the composition comprises an extracellular vesicle. In certain embodiments, the extracellular vesicle is a vesicle derived from cells that comprises a membrane that surrounds an internal space.
[00135] [00135] In several embodiments, the extracellular vesicle may be a membrane-bound vesicle that has a smaller diameter than the cell from which it is derived. In some embodiments, the extracellular vesicle has a longer dimension between about 20-1000 nm, such as between about 20-100 nm, 20-200 nm, 20-300 nm, 20-400 nm, 20- 500 nm, 20-600 nm, 20-700 nm, 20-800 nm, 20-900 nm, 30-100 nm, 30-200 nm, 30-300 nm, 30-400 nm, 30-500 nm, 30- 600 nm, 30-700 nm, 30-800 nm, 30-900 nm, 40-100 nm, 40-200 nm, 40-300 nm, 40- 400 nm, 40-500 nm, 40-600 nm, 40- 700 nm, 40-800 nm, 40-900 nm, 50-150 nm, 50-500 nm, 50-750 nm, 100-200 nm, 100-500 nm or 500- 1000 nm.
[00136] [00136] In some embodiments, the extracellular vesicle is an exosome. In certain embodiments, the extracellular vesicle is a novesicle. In certain embodiments, the extracellular vesicle is an apoptotic body. In certain embodiments, the extracellular vesicle is a cell fragment. In certain embodiments, the extracellular vesicle is a vesicle derived from the cell by direct or indirect manipulation. In certain modalities, the extracellular vesicle is a vesicle organelle
[00137] [00137] In some embodiments, the extracellular vesicle is derived from a living organism. In some embodiments, the extracellular vesicle is derived from a dead organism. In some embodiments, the extracellular vesicle is derived from an explanted tissue. In some modalities, the extracellular vesicle is derived from an implanted organ. In some embodiments, the extracellular vesicle is derived from cultured cells. In some of these modalities, when the extracellular vesicle is generated in a cell culture system, the extracellular vesicle is even more isolated (for example, separating the extracellular vesicle from cultured cells). Separation can be achieved by sedimentation. For example, the extracellular vesicle can have a specific density between 0.5 and 2.0, 0.6 and 1.0, 0.7 and 1.0, 0.8 and 1.0, 0.9 6 1.0, 1.06 1.1, 1.1 and 1.2, 1.26 1.3, 14 and 1.5, 1.0 and 1.5, 1.5 and 2.0 1.0 and 2.0 kg / m . Separation can also be achieved by affinity purification. For example, the extracellular vesicle can be purified by attaching a population comprising extracellular vehicles to a resin, said resin comprising a plurality of ligands that have specific affinity for one or more target proteins on the surface of the extracellular vesicle. The target proteins can be a tetraspanin (for example, CD63, CD81, CD9), a member of the EWI protein / immunoglobulin superfamily (for example, PTGFRN, IGSF8, IGSF3), an integrin (for example, ITGB1, ITGA4 ), an ATP carrier protein (e.g., ATP1A1, ATP1A2, ATP1A3, ATP1A4, ATP1B3, ATP2B1, ATP2B2, ATP2B3, ATP2B4), SLC3A2, BSG or CD98hc. The target protein may additionally be the immunomodulatory component that is displayed on the surface of the exosomes.
[00138] [00138] In several modalities, the extracellular vesicle comprised
[00139] [00139] In several modalities, the membrane of the extracellular vesicle comprises an internal surface and an external surface and involves an internal space. In some embodiments, the extracellular vesicle further comprises a payload. In certain embodiments, the payload is enclosed within the internal space. In certain embodiments, the payload is displayed on the external surface of the extracellular vesicle. In certain embodiments, the payload is spanning the membrane of the extracellular vesicle. In various modalities, the payload comprises nucleic acids, proteins, carbohydrates, lipids, small molecules and / or combinations thereof. In some embodiments, the extracellular vesicle also comprises a receptor. THE EXOSOME
[00140] [00140] In several modalities, the extracellular vesicle is an exosum. In certain embodiments, the exosome is a small vesicle attached to the membrane secreted by the producing cells.
[00141] [00141] In some embodiments, the exosome of the producing cell has a longer dimension between about 20 to 300 nm, such as between about 20 to 290 nm, 20 to 280 nm, 20 to 270 nm,, 20 to 260 nm, 250 nm, 20 to 240 nm, 20 to 230 nm, 20 to 220 nm, 20 to 210 nm, 20 to 200 nm, 20 to 190 nm, 20 to 180 nm, 20 to 170 nm, 20 to 160 nm , 20 to 150 nm, 20 to 140 nm, 20 to 130 nm, 20 to 120 nm, 20 to 110 nm, 20 to 100 nm, 20 to 90 nm, 20 to 80 nm, 20 to 70 nm, 20 to 60 nm , 20 to 50 nm, 20 to 40 nm, 20 to 30 nm, 30 to 300 nm, 30 to 290 nm, 30 to 280 nm, 30 to 270 nm, 30 to 260 nm, 30 to 250 nm, 30 to 240 nm , 30 to 230 nm, 30 to 220 nm, 30 to 210 nm, 30 to 200 nm, 30 to 190 nm, 30 to 180 nm, 30 to 170 nm, 30 to 160 nm, 30 to 150 nm, 30 to 140 nm , 30 to 130 nm, 30 to 120 nm, 30 to 110 nm, 30 to 100 nm, 30 to 90 nm, 30 to 80 nm,
[00142] [00142] In particularly preferred embodiments, the exosome of the producer cell described here has a longer dimension between about 30 to 100 nm. In another preferred embodiment, the exosome of the producing cell has a longer dimension between about 20 to 300 nm. In another preferred embodiment, the exosome of the producing cell has a longer dimension between about 40 to 200 nm. In another embodiment, a population of the exosomes described here comprises a population in which 90% of the exosomes have a longer dimension 20 to 300 nm. In another embodiment, a population of the exosomes described herein comprises a population in which 95% of the exosomes have a longer dimension 20 to 300 nm. In another embodiment, a population of the exosomes described here comprises a population in which 99% of the exosomes have a longer dimension 20 to 300 nm. In another embodiment, a population of the exosomes described herein comprises a population in which 90% of the exosomes have a longer dimension 40 to 200 nm. In another embodiment, a population of the exosomes described here comprises a population in which 95% of the exosomes have a longer dimension 40 to 200 nm. In another embodiment, a population of the exosomes described herein comprises a population in which 99% of the exosomes have a longer dimension 40 to 200 nm. In other preferred embodiments, the size of the exosome or population of exosomes described herein is measured according to the methods described, below.
[00143] [00143] In some modalities, the exosome is generated by a producer cell. In some embodiments, the membrane of the exosome comprises one or more molecules derived from the producing cell. In some embodiments, the exosome is generated in a cell culture system and isolated (for example, separating the exosome from the producing cell). Separation can be achieved by sedimentation. For example, the exosome can have a specific density between 0.5 to 2.0, 0.6 to 1.0, 0.7 to 1.0, 0.8 to 1.0, 0.9 to 1.0 , 1.0 to 1.1, 1.1 to 1.2, 1.2a 1.3, 1.4 to 1.5, 1.0 to 1.5, 1.5a 2.0 and 1.0 to 2.0 kg / m . Separation can also be achieved by affinity purification. For example, the extracellular vesicle can be purified by binding a population comprising extracellular vesicles to a resin, said resin comprising a plurality of ligands that have specific affinity for one or more target proteins on the surface of the vesicle extracellular. The one or more target proteins may be a tetraspanin (for example, CD63, CD81, CD9), a member of the EWI protein / immunoglobulin superfamily (for example, PTG-FRN, IGSF8 and / or IGSF3), an integrin (for example example, ITGB1 and / or ITGA4), an ATP carrier protein (e.g., ATP1A1, ATP1A2, ATP1A3, ATP1A4, ATP1B3, ATP2B1, ATP2B2, ATP2B3 and / or ATP2B4), SLC3A2, BSG or CD98hc. The target protein may additionally be the immunomodulatory component that is displayed on the surface of the exosomes.
[00144] [00144] In some embodiments, the membrane of the exosome comprises an inner surface and an outer surface. In certain embodiments, the inner surface faces the inner core of the exosome. In certain embodiments, the outer surface may be in contact with the endosome, multivesicular bodies or the membrane | cytoplasm of a producer cell or a target cell.
[00145] [00145] In some modalities, the exosome comprises lipids and fatty acids. In some embodiments, the exosome comprises phospholipids, glycolipids, fatty acids, sphingolipids, phosphoglycerides, sterols, cholesterols and phosphatidyl serines. In some modalities, lipid and fatty acid may be one or more of those listed in Table 1.
[00146] [00146] In certain modalities, the exosome comprises a lipid bilayer composed of an internal leaflet and an external leaflet. The composition of the internal and external pamphlet can be determined by trans-layer distribution tests known in the art, see, for example, Kuypers et al. Biohim Biophys Acta 1985 819: 170. In some embodiments, the composition of the external leaflet is between approximately 70 to 90% choline phospholipids, between approximately 0 to 15% acidic phospholipids and between approximately 5 to 30 % phosphatidylethanolamine. In some embodiments, the composition of the internal leaflet is between approximately 15 to 40% choline phospholipids, between approximately 10 to 50% acidic phospholipids and between approximately 30 to 60% phosphatidylethanolamine.
[00147] [00147] In some embodiments, the membrane of the exosome also comprises one or more polypeptides. In certain embodiments, the exosome comprises one or more polypeptides selected from the list below, including, but not limited to, spectrin, myosin-like polypeptide, band 3, SLC4A1, actin, actin-like polypeptide, 3-P glyceraldehyde dehydrogenase (G3PD), tetraspanins (for example, CD63, CD81 and / or CD9), Alix and TSG101, integrins (for example, ITGB1 and / or ITGA4), selectins, CR1, TNFRI, proteolytic enzymes, proteins or bound histones glycosylphosphatidylinosite! (GPI), EWI protein / immunoglobulin superfamily members (e.g.
[00148] [00148] In some modalities, the exosome comprises polypeptides on its surface. In some embodiments, the exosome is modified to contain the one or more polypeptides. In some
[00149] [00149] In specific modalities, exosomes comprise one or more polypeptides on their surface, in which the said polypeptides
[00150] [00150] In some modalities, the membrane of the exosome also comprises one or more polysaccharides, such as glycan.
[00151] [00151] In some modalities, the exosome delivers the payload (therapeutic agent) to a target. The payload is a therapeutic agent that acts on a target (for example, a target cell) that is contacted with the exosome. Contact can occur in vitro or in an individual. Payloads that can be introduced into an exosome and / or a producing cell include therapeutic agents, such as nucleotides (for example, nucleotides comprising a detectable chemical portion or a toxin or that disrupts transcription), nucleic acids ( for example, DNA or mMRNA molecules that encode a polypeptide, such as an enzyme, or RNA molecules that have a regulatory function, such as mIRNA, dsDNA, IncRKNA or siRNA), amino acids (for example, amino acids comprising a chemical moiety detectable or a toxin that hinders translation), polypeptides (for example, enzymes), lipids, carbohydrates and small molecules (for example, small molecule drugs and toxins).
[00152] [00152] The exosome can interact with the target cell through membrane fusion and deliver payloads (for example,
[00153] [00153] In some embodiments, the exosome comprises a receptor polypeptide. The receptor polypeptide can be synthetic. In some embodiments, the recipient polypeptide is introduced into the producer cell (for example, an exogenous nucleic acid encoding the receptor polypeptide is introduced into the producer cell) or a recombinant receptor polypeptide that is produced outside the producer cell (for example , synthesized by a protein expression system). In some embodiments, the recipient polypeptide (for example, a recombinantly produced polypeptide) is introduced directly into the exosome (for example, after the exosome is isolated from the producing cell). In some embodiments, the receiving polypeptide may be on the surface of exosomes. In some modalities, the receptor polypeptide is able to target the exosome to a specific target (for example, a target such as a pathogen, a metabolite, a polypeptide complex or a cell, such as a non-functional cell or cancer cell) that circulates in the individual's circulatory system, such as blood, or a target that resides in tissue (such as diseased tissue).
[00154] [00154] In some modalities, the exosome is synthetic. For example, the exosome may comprise a payload, such as a therapeutic polypeptide, nucleic acid (such as DNA or RNA) or other polynucleotide, polysaccharide or glycan, lipid or fatty acid, large, small, biological molecule or toxin so that the exosome is not occurring naturally. In some modes, the exosome is modified (for example, by introducing a payload or by modifying the content of the complex, such as altering the protein, lipid or glycan content of the membrane). For example, exosomes are first isolated from a producing cell and then modified as desired, thereby generating synthetic exosomes. In some embodiments, the producing cell is modified. For example, an exogenous nucleic acid, an exogenous polypeptide or small molecule or toxin can be introduced into the producing cell. Alternatively or in addition, the producing cell may otherwise be modified (for example, by modifying the cell or membrane content, as by altering the lipid or glycan content of the cell membrane). The exosomes generated from the modified producer cells comprise one or more of the changes in the producer cell. The process produces synthetic exosomes. In some embodiments, both the producing cell and the exosome isolated from the producing cell are modified as described here. NANOVESICLE
[00155] [00155] In several modalities, the extracellular vesicle is a novesicle. In certain modalities, the nanovesicle is a small vesicle derived from a cell that comprises a membrane that encloses an internal space and that is generated from the cell by direct or indirect manipulation, so that the nanovesicle is not produced by the cell without manipulation. Appropriate cell manipulations include, but are not limited to, serial extrusion, treatment with alkaline solutions, sonication or combinations thereof and may, in some cases, result in the destruction of the producing cell.
[00156] [00156] In various embodiments, the nanovesicle has a longer dimension between about 20 to 250 nm, such as between about 20 to 100 nm, 20 to 150 nm, 20 to 200 nm, 30 to 100 nm, 30 to 150 nm , 30 to 200 nm, 30 to 250 nm, 40 to 100 nm, 40 to 150 nm, 40 to 200 nm, 40 to 250 nm, 50 to 100 nm, 50 to 150 nm, 50 to 200 nm, 50 to 250 nm , 100 to 200 nm or 150 to 250 nm.
[00157] [00157] In several modalities, the nanovesicle is derived from a producing cell. In certain modalities, the nanovesicle is generated from a producing cell by direct or indirect manipulation. Appropriate manipulations include, but are not limited to, serial extrusion, treatment with alkaline solutions, sonication or combinations thereof. In some of these modalities, manipulation can result in the destruction of the producing cell. In some preferred modalities, the nanovesicle population is substantially free of vesicles that are derived from producer cells through direct budding of the plasma membrane or fusion of the late endosome with the plasma membrane.
[00158] [00158] In some modalities, the nanovesicle is isolated from the producing cell based on its size, density, biochemical parameters or a combination of them. In certain embodiments, isolation can be achieved by sedimentation. For example, the particle may have a specific density between 0.5 to 2.0, 0.6 to 1.0, 0.7 to 1.0, 0.8 to 1.0, 0.9 to 1 , 0, 1.0 to 1.1, 1.1 to 1.2, 1.2 to 1.3, 14 to 1.5, 1.0 to 1.5, 1.5 to 2.0 € 1.0 to 2.0 kg / m .
[00159] [00159] In several modalities, the nanovesicle comprises lipids or fatty acids and polypeptides. In certain embodiments, the seedling also comprises sugar. In certain embodiments, the nanovesicle further comprises a polynucleotide. In some modes, the nanovesicle also comprises a receiver. In some modalities, the nanovesicle also comprises a payload. In some of these modalities, the payload comprises nucleic acids, proteins, carbohydrates, lipids, small molecules and / or combinations thereof. THE IMMUNOMODULATIVE COMPONENT
[00160] [00160] In various embodiments, the composition further comprises an immunomodulatory component.
[00161] [00161] In some embodiments, the immunomodulation compound is a protein that is expressed as a translational fusion protein for an exosome surface protein, so that the said protein is retained on the surface of the exosome. In certain modalities, the immunomodulating compound is a membrane protein. In certain embodiments, the immunomodulation compound is a soluble protein. In some embodiments, the exosome surface protein is a tetraspanin (for example, CD63, CD81, CD9), a member of the immunoglobulin / EWI protein superfamily (for example, PTGFRN, IGSF8, IGSF3), an integrin (for example example, ITGB1, ITGA4), an ATP carrier protein (for example, ATP1A1, ATP1A2, ATP1A3, ATP1A4, ATP1B3, ATP2B1, ATP2B2, ATP2B3, ATP2B4), SLC3A2, BSG or CD98hc or a fragment or var.
[00162] [00162] In some embodiments, the immunomodulation compound is a soluble protein that is expressed as a translational fusion protein for an exosome surface protein, so that said soluble protein is retained on the surface of the exosome. In some embodiments, the exosome surface protein is a tetraspanin (for example, CD63, CD81, CD9), a member of the immunoglobulin / EWI protein superfamily (for example, PTGFRN, IGSF8, IGSF3), an integrin (for example, ITGB1, ITGA4), an ATP-carrying protein (for example, ATP1IA1I, ATP1A 2, ATP1A3, ATP1A4, ATP1B3, ATP2B1, ATP2B2, ATP2B3, ATP2B4), SLC3A2, BSG or a CD98hc fragment or a fragment or a different fragment.
[00163] [00163] In certain modalities, the immunomodulatory component has antitumor activity. In some embodiments, the immunomodulatory component regulates the innate immune response. In some of these modalities, the immunomodulatory component targets cells
[00164] [00164] In some modalities, the immunomodulatory component is expressed in the producer cell in its complete form. In other embodiments, the immunomodulatory component is expressed as a translational fusion protein for an exosome surface protein, which results in a higher level of expression of the biologically active portion of the immunomodulation compound on the surface of the exosome . In some embodiments, the immunomodulating compound is a soluble protein that is expressed as a translational fusion protein for an exosome surface protein, so that said soluble protein is retained on the surface of the exosome. In some embodiments, the exosome surface protein is a tetraspanin (for example, CD63, CD81, CD9), a member of the immunoglobulin / EWI protein superfamily (for example, PTGFRN, IGSF8, IGSF3), an integrin (for example example, ITGB1, ITGA4), an ATP carrier protein (for example, ATP1A1, ATP1A2, ATP1A3, ATP1A4, ATP1B3, ATP2B1, ATP2B2, ATP2B3, ATP2B4), SLC3A2, BSG or CD98hc or a fragment or var.
[00165] [00165] In some embodiments, the immunomodulatory component is an inhibitor for a negative checkpoint regulator. In some embodiments, the immunomodulatory component is an inhibitor for a negative checkpoint regulator binding partner.
[00166] [00166] In certain embodiments, the immunomodulatory component is an inhibitor of protein 4 associated with cytotoxic T lymphocytes (CTLA-4). In some of these embodiments, the CTLA-4 inhibitor is a monoclonal antibody to CTLA-4. In certain embodiments, the inhibitor is a fragment of a CTLA-4 monoclonal antibody. In certain modalities, the antibody fragment is a scFv, (scFv) 2, Fab, Fab 'and F (ab', F (ab1) 2, Fv, dAb or Fd of a CTLA4 monoclonal antibody. , the inhibitor is a nanobody, a bispecific antibody or a multispecific antibody against CTLA-4. In some specific modalities, the monoclonal antibody is ipilimumab. In some specific embodiments, the monoclonal antibody is tremelimumab.
[00167] [00167] In certain embodiments, the immunomodulatory component is an inhibitor of programmed cell death protein 1 (PD-1). In certain embodiments, the immunomodulatory component is an inhibitor of the | programmed death ligand 1 (PD-L1). In certain embodiments, the immunomodulatory component is an inhibitor of the programmed death ligand 2 (PD-L2). In some embodiments, the PD-1, PD-L1 or PD-L2 inhibitor is a monoclonal antibody to PD-1, PD-L1 or PD-L2. In certain embodiments, the inhibitor is a fragment of a monoclonal antibody from PD-1, PD-L1 or PD-L2. In certain embodiments, the antibody fragment is a scFv, (scFv) 2, Fab, Fab 'and F (ab') 2, F (ab1)>, Fv, dAb or Fd of a PD-1 monoclonal antibody , PD-L1 or PD-L2. In certain embodiments, the inhibitor is a nanobody, a bispecific antibody or a multispecific antibody against PD-1, PD-L1 or PD-L2. In some specific embodiments, the monoclonal antibody is nivo-lumab. In some specific embodiments, the monoclonal antibody is pembrolizumab. In some specific embodiments, the monoclonal antibody is pidilizumab. In some specific modalities, the monoclonal antibody is atezolizumab. In some specific embodiments, the monoclonal antibody is avelumab.
[00168] [00168] In certain embodiments, the immunomodulatory component is an inhibitor of the lymphocyte-activated gene 3 (LAG3). In some
[00169] [00169] In certain embodiments, the immunomodulatory component is an inhibitor of protein 3 containing mucin from T cell immunoglobulin (TIM-3). In certain embodiments, the immunomodulatory component is an inhibitor of the B and T lymphocyte attenuator (BTLA). In certain modalities, the immunomodulatory component is an inhibitor of the T cell immunoreceptor with Ig and ITIM domains (TIGIT). In certain modalities, the immunomodulatory component is an inhibitor of the Ig suppressor of domain V of T cell activation (VISTA). In certain modalities, the immunomodulatory component is an inhibitor of the A2a adenosine receptor (A2aR). In certain embodiments, the immunomodulating component is an inhibitor of the killer cell immunoglobulin-type receptor (KIR). In certain embodiments, the immunomodulating component is an inhibitor of indoleamine 2,3-dioxigenase (IDO). In certain embodiments, the immunomodulatory component is an inhibitor of CD20, CD39 or CD73.
[00170] [00170] In some embodiments, the immunomodulatory component is an activator for a positive co-stimulating molecule. In some embodiments, the immunomodulatory component is an activator for a positive co-stimulating molecule binding partner.
[00171] [00171] In some embodiments, the immunomodulatory component is an activator of a member of the TNF receptor superfamily. In some of these modalities, the member of the TNF receptor superfamily is selected from the group consisting of: CD120a, CD120b, CD18, OX40, CD40, Fas receptor, M68, CD27, CD30, 4-1BB, TRAILR1I, TRAILR2, TRAILR3, TRAILR4, RANK, OCIF, TWEAK receiver, TACI, BAFF receiver, ATAR, CD271, CD269, AITR, TROY, CD358, TRAMP and XEDAR. In some embodiments, the immunomodulatory component is a member of the TNF superfamily. In certain embodiments, the member of the TNF superfamily is selected from the group consisting of: TNFa, TNF-C, OX40L, CD40L, FasL, LIGHT, TL1A, CD27L, Siva, CD153, ligand 4-1BB, TRAIL, RANKL, TWEAK, APRIL, BAFF, CAMLG, NGF, BDNF, NT-3, NT, GITR and EDA-2 ligand.
[00172] [00172] In some embodiments, the activator of a member of the TNF receptor superfamily is expressed as a monomeric protein. In some embodiments, the activator of a member of the TNF receptor subfamily is expressed as quarterly proteins. In some embodiments, the member of the TNF receptor superfamily is expressed as a monomeric protein. In some modalities, the member of the TNF receptor superfamily is expressed as quarterly proteins.
[00173] [00173] In certain embodiments, the immunomodulatory component is an activator of Member 4 of the TNF Receptor Superfamily (OX40). In some of these modalities, the OX40 activator is an OX40 agonist antibody. In some of these modalities, the OXA40 activator is the OX40 ligand (OX40L).
[00174] [00174] In certain embodiments, the immunomodulatory component is a CD27 activator. In some of these modalities, the CD27 activator is a CD27 agonist antibody. In some of these modalities, the CD27 activator is the CD27 ligand (CD27L).
[00175] [00175] In certain embodiments, the immunomodulatory component is a CD40 activator. In some of these modalities, the CD40 activator is a CD40 agonist antibody. In some of these modalities, the CD40 activator is the CD40 ligand (CD40L). In some embodiments, CD40L is monomeric CD40L. In some modalities, the CD40L is a quarterly CDA40L.
[00176] [00176] In some embodiments, the trimeric CD40L is fused to PTGFRN or a fragment thereof. In some embodiments, the trimeric CDA40L is fused to the N-terminal of PTGFRN or a fragment thereof. In some embodiments, trimeric CD40L is expressed as a fusion protein for PTGFRN, where the polypeptide has the sequence of SEQ ID NO: 19 or SEQ ID NO: 20.
[00177] [00177] In certain modalities, the immunomodulatory component is an activator of the glucocorticoid-induced TNFR-related protein (GITR). In some of these embodiments, the GITR activator is a GITR agonist antibody. In some of these modalities, the GITR activator is a natural GITR ligand.
[00178] [00178] In certain embodiments, the immunomodulatory component is an activator of 4-1BB. In some of these modalities, the 4-1BB activator is a 4-1BB agonist antibody. In some of these modalities, the 4-1BB activator is a natural 4-1BB ligand.
[00179] [00179] In some embodiments, the immunomodulatory component is the Fas receptor (Fas). In some of these modalities, the Fas receptor is displayed on the surface of the extracellular vesicle. In some other modalities, the immunomodulatory component is the Fas ligand (FasL). In some of these modalities, the Fas ligand is displayed on the surface of the extracellular vesicle. In certain embodiments, the immunomodulatory component is an antibody to the Fas receptor. In certain embodiments, the immunomodulatory component is an antibody to the Fas ligand.
[00180] [00180] In some embodiments, the immunomodulatory component is an activator of a co-stimulating molecule of the CD28 superfamily. In certain embodiments, the co-stimulating molecule of the CD28 superfamily is ICOS or CD28. In certain modalities, the immunomodulating component is ICOSL, CD80 or CD86.
[00181] [00181] In certain embodiments, the immunomodulatory component is an activator of the inducible T cell co-stimulator (ICOS). In some of these modalities, the ICOS activator is an ICOS agonist antibody. In some other of these modalities, the ICOS activator is the ICOS ligand (ICOSL).
[00182] [00182] In certain embodiments, the immunomodulatory component is a CD28 activator. In some of these modalities, the CD28 activator is a CD28 agonist antibody. In some of these modalities, the CD28 activator is a natural CD28 ligand. In certain embodiments, the CD28 ligand is CD80.
[00183] [00183] In certain embodiments, the composition comprises an inhibitor for a negative checkpoint regulator or an inhibitor for a binding partner of a negative checkpoint regulator and an activator for a positive co-stimulating molecule or an activator for a positive co-stimulating molecule binding partner.
[00184] [00184] In certain embodiments, the immunomodulatory component is a cytokine. In some embodiments, the cytokine is a soluble cytokine that has been translationally fused to an exosome surface protein or fragment thereof. In some embodiments, the cytokine is interleukin 2 (IL-2). In some embodiments, the cytokine is interleukin 7 (IL-7). In some modalities, the cytokine is interleukin 12 (IL-12). In some modalities, the cytokine is interleukin 15 (IL-15).
[00185] [00185] In certain embodiments, the cytokine is fused to PTGFRN or a fragment thereof. In some embodiments, IL-7 is fused to PTGFRN or a fragment thereof. In some embodiments, IL-7 is fused to the N-terminal of PTGFRN or a fragment thereof. In some embodiments, IL-7 is expressed as a fusion protein for PTGFRN, where the polypeptide has the sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
[00186] [00186] In certain embodiments, the cytokine is fused to PTGFRN or a fragment thereof. In some embodiments, 11-12 is fused to PTGFRN or a fragment thereof. In some modalities, the
[00187] [00187] In certain embodiments, the cytokine is fused to PTGFRN or a fragment thereof. In some embodiments, IL-15 is fused to PTGFRN or a fragment thereof. In some embodiments, IL-15 is fused to the N-terminal of PTGFRN or a fragment thereof. In some embodiments, IL-15 is expressed as a fusion protein for PTGFRN, where the polypeptide has the sequence of SEQ ID NO: 15 or SEQ ID NO: 16.
[00188] [00188] In some embodiments, the cytokine is an interferon (IFN). In certain embodiments, the interferon is fused to PTGFRN or a fragment thereof. In certain embodiments, interferon is interferon y (IFNy). In some embodiments, IL-15 is fused to PTGFRN or a fragment thereof. In some embodiments, IFNy is fused to the N-terminal of PTGFRN or a fragment thereof. In some modalities, IFNy is expressed as a fusion protein for PTG-FRN, where the polypeptide has the sequence of SEQ ID NO: 7 or SEQ ID NO: 8.
[00189] [00189] In some embodiments, the immunomodulatory component is a T cell receptor (TCR) or a derivative thereof. In certain embodiments, the immunomodulatory component is an a TCR chain or a derivative thereof. In certain embodiments, the immunomodulating component is a B chain of TCR or a derivative of the same. In some embodiments, the immunomodulatory component is a T cell coreceptor or a derivative thereof.
[00190] [00190] In some embodiments, the immunomodulatory component is a tumor antigen. In certain modalities, the tumor antigen is selected from the group consisting of: alpha-fetoprotein (AFP),
[00191] [00191] In certain modalities, the tumor antigen is a carcinoembryonic antigen (CEA). In certain embodiments, the tumor antigen is an epithelial tumor antigen (ETA).
[00192] [00192] In certain embodiments, the tumor antigen is a mucin. In some of these modalities, mucin is a secreted mucin. In some other of these modalities, mucin is a transmembrane mucin. In specific modalities, the tumor antigen is mucin 1 (MUC1). In specific modalities, the tumor antigen is Tn-MUC1. In specific modalities, the tumor antigen is mucin 16 (MUC16).
[00193] [00193] In certain modalities, the tumor antigen is an antigen associated with melanoma (MAGE). In some of these modalities, MAGE is a | MAGE type. In some other of these modalities, the MAGE is an MAGE type Il. In specific modalities, the MAGE type | is MAGE-A2. In specific modalities, the MAGE type | is MAGE-AA.
[00194] [00194] In certain embodiments, the tumor antigen is alpha-fetoprotein (AFP). In certain embodiments, the tumor antigen is the tumor protein p53 (p53). In certain modalities, the tumor antigen is tyrosinase. In certain modalities, the tumor antigen is a tyrosinase-related protein (TRP). In some modalities, the tumor antigen is the programmed death ligand 1 (PD-L1) or the
[00195] [00195] In some modalities, the immunomodulatory component is a chimeric antigen receptor (CAR) or a derivative thereof. In some modalities, CAR binds to one or more of alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), tumor epithelial antigen (ETA), mucin 1 (MUC1), Th-MUC1, mucin 16 (MUC16) , tyrosinase, melanoma-associated antigen (MAGE), tumor protein p53 (p53), CD4, CD8, CD45, CD80, CD86, programmed death ligand 1 (PD-L1), programmed death ligand 2 (PD-L2 ), NY-ESO-1, PSMA, TAG-72, HER2, GD2, cMET, EGFR, Mesothelin, VEGFR, alpha-folate receptor, CE7R, IL-3, cancerous testis antigen, MART-1 gp100 and TNF-related apoptosis-inducing ligand.
[00196] [00196] In some embodiments, the immunomodulatory component is an activator for a T cell receptor or coreceptor. In certain embodiments, the immunomodulatory component is a CD3 activator. In certain embodiments, the activator is a fragment of a CD3 monoclonal antibody. In certain embodiments, the antibody fragment is a scFv, (scFv) 2, Fab, Fab 'and F (ab') 2, F (ab1) 2, Fv, dAb or Fd of a monoclonal antibody against CD3. In certain embodiments, the activator is a nanobody, a bispecific antibody or a multispecific antibody against CD3. In some embodiments, the anti-CD3 antibody fragment is fused to PTGFRN or a fragment thereof. In some embodiments, the anti-CD3 antibody fragment is fused to the N-terminus of PTGFRN or a fragment thereof. In some modalities, the anti-CD3 antibody fragment is expressed as a fusion protein for PTGFRN, where the polypeptide has the sequence of SEQ ID NO: 18 or SEQ ID NO: 21. In certain modalities,
[00197] [00197] In some embodiments, the immunomodulatory component is a major histocompatibility complex (MHC) or a derivative of it. In some of these modalities, the immunomodulating component is a class MHC | or a derivative of it. In some of these modalities, the immunomodulatory component is an MHC class II or a derivative thereof. In some of these modalities, the immunomodulatory component is an MHC class Ill or a derivative thereof.
[00198] [00198] In some embodiments, the immunomodulatory component is a human leukocyte antigen (HLA) or a derivative thereof. In some of these modalities, the immunomodulatory component is an HLA-A, HLA-B, HLA-C or a derivative thereof. In some of these modalities, the immunomodulatory component is an HLA-E, HLA-F, HLA-G or a derivative thereof. In some of these modalities, the immunomodulatory component is an HLA-DP, HLA-DQ, HLA-DR or a derivative thereof.
[00199] [00199] In several embodiments, the immunomodulatory component can be a polypeptide, a polynucleotide, a polysaccharide, a lipid, a small molecule or a toxin.
[00200] [00200] In some embodiments, the immunomodulatory component can be a protein, a peptide, a glycolipid or a glycoprotein.
[00201] [00201] In certain modalities, the immunomodulatory component is an agonist. In some of these modalities, the agonist is an endogenous agonist, like a hormone or neurotransmitter. In some other of these modalities, the agonist is an exogenous agonist, like a drug. In some modalities, the agonist is a physical agonist, who can create an agonist response without connecting with the receiver. In some modalities, the agonist is a superagonist, which can produce a maximum response greater than the endogenous agonist. In certain modalities, the agonist is a complete agonist with total efficacy at the recipient. In certain other modalities, the agonist is a partial agonist having only partial efficacy at the receptor in relation to a complete agonist. In some embodiments, the agonist is an inverse agonist that can inhibit the constitutive activity of the receptor. In some embodiments, the agonist is a coagonist who works with other coagonists to produce an effect on the recipient. In certain embodiments, the agonist is an irreversible agonist that permanently binds to a receptor through the formation of a covalent bond. In certain embodiments, the agonist is a selective agonist for a specific type of receptor.
[00202] [00202] In certain embodiments, the immunomodulatory component is an antagonist. In some of these modalities, the antagonist is a competitive antagonist, which reversibly binds to the receptor at the same binding site as the endogenous ligand or agonist without activating the receptor. The competitive antagonist can affect the amount of agonist needed to achieve a maximum response. In some other of these modalities, the antagonist is a non-competitive antagonist, which binds to an active receptor site or an allosteric site of the receptor. The non-competitive antagonist can reduce the magnitude of the maximum response that can be achieved by any amount of agonist. In some other modalities, the antagonist is a non-competitive antagonist, which requires activation of the receptor by an agonist prior to its binding to a separate allosteric binding site.
[00203] [00203] In several embodiments, the immunomodulatory component comprises an antibody or an antigen-binding fragment. The immunomodulatory component can be a full-length protein or a fragment thereof. The antibody or antigen-binding fragment can be derived from natural sources or produced partially or completely synthetically. In some embodiments, the antibody is a monoclonal antibody. In some of these modalities, the monoclonal antibody is an IgG antibody. In certain modalities, the monoclonal antibody is an IgG1, IgG2, IgG3 or IgG4. In some other embodiments, the antibody is a polyclonal antibody. In certain embodiments, the antigen-binding fragment is selected from Fab, Fab 'and F (ab') a, F (ab1) 2, Fv, dAb and Fd. In certain modalities, the antigen-binding fragment is an scFv or (scFv) 2 fragment. In certain other embodiments, the antibody or antigen-binding fragment is a Nanobodyº (single domain antibody). In some embodiments, the antibody or antigen-binding fragment is a bispecific or multispecific antibody.
[00204] [00204] In several modalities, the antibody or antigen-binding fragment is entirely human. In some embodiments, the antibody or antigen-binding fragment is humanized. In some embodiments, the antibody or antigen-binding fragment is chimeric. In some of these modalities, the chimeric antibody has human V region domains and human C region domains. In some embodiments, the antibody or antigen-binding fragment is non-human, such as murine or veterinary.
[00205] [00205] In certain embodiments, the immunomodulatory component is a polynucleotide. In some of these modalities, the polynucleotide includes, but is not limited to, an mMRNA, a miRNA, a siRNA,
[00206] [00206] In some embodiments, the immunomodulatory component is a protein, a peptide, a glycolipid or a glycoprotein.
[00207] [00207] In various embodiments, the composition comprises two or more immunomodulatory components mentioned above, including mixtures, fusions, combinations and conjugates of atoms, molecules, etc. In some embodiments, the composition comprises one, two, three, four, five, six, seven, eight, nine, ten, eleven or twelve different immunomodulatory components associated with the membrane or closed within the closed volume of said extracellular vesicle . In certain embodiments, the composition comprises a nucleic acid combined with a polypeptide. In certain embodiments, the composition comprises two or more polypeptides conjugated to each other. In certain embodiments, the composition comprises a protein conjugated to a biologically active molecule. In some of these modalities, the biologically active molecule is a prodrug.
[00208] [00208] In some embodiments, the composition comprises two different immunomodulatory components associated with the membrane or closed within the closed volume of said extracellular vesicle. In certain embodiments, the two different immunomodulatory components are I1L-12 and CD40L. In some embodiments, CD40L and IL-12 are fused to PTGFRN or a fragment thereof, respectively. In some embodiments, CD40L and IL-12 are fused to the N-terminus of PTGFRN or a fragment thereof, respectively. In some embodiments, CD40L and IL-12 are expressed as fusion proteins for PTGFRN, in which the polypeptides have the sequences of SEQ ID NO: 20 and SEQ ID NO: 3, respectively.
[00209] [00209] In some embodiments, the composition comprises three different immunomodulatory components associated with the membrane or closed within the closed volume of said extracellular vesicle. In certain embodiments, the two different immunomodulatory components are I1L-12, CD40L and tyrosine kinase ligand 3 FMS (FLT3L). In some modalities, CD40L, IL-12 and FLT3L are fused to PTGFRN or a fragment thereof, respectively. In some modalities, the CD40L, I1L-12 and FLT3L are fused to the N terminal of PTGFRN or a fragment thereof, respectively. In some embodiments, CD40L, IL-12 and FLT3L are expressed as fusion proteins for PTGFRN, where the polypeptides have the sequences of SEQ ID NO: 20, SEQ ID NO: 3 and SEQ ID NO: 22, respectively. THE PHARMACEUTICAL COMPOSITION
[00210] [00210] Pharmaceutical compositions generally comprise a plurality of extracellular vesicles and a pharmaceutically acceptable excipient or carrier in a form suitable for administration to an individual. Pharmaceutically acceptable excipients and carriers are determined, in part, by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions comprising a plurality of extracellular vesicles. (See, for example, Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 21st ed. (2005)). Pharmaceutical compositions are generally formulated sterile and in full compliance with all United States Food and Drug Administration (GMP) Good Manufacturing Practice (GMP) regulations.
[00211] [00211] In some embodiments, the pharmaceutical composition comprises one or more therapeutic agents and the extracellular vesicle described herein. In some embodiments, extracellular vesicles are co-administered with one or more separate therapeutic agents, in which coadministration includes the administration of the separate therapeutic agent before, after or concurrent with the administration of extracellular vesicles.
[00212] [00212] Pharmaceutically acceptable excipients include excipients that are generally safe, non-toxic and desirable, including excipients acceptable for veterinary use, as well as for human pharmaceutical use.
[00213] [00213] Examples of carriers or diluents include, but are not limited to, water, saline, Ringer's solutions, dextrose solution and 5% human serum albumin. The use of such media and compounds for pharmaceutically active substances is well known in the art. Except to the extent that any conventional medium or composition is incompatible with the extracellular vesicles described here, its use in the compositions is contemplated. Supplementary therapeutic agents can also be incorporated into the compositions. Typically, a pharmaceutical composition is formulated to be compatible with its intended route of administration. Extracellular vesicles can be administered parenterally, topically, intravenously, orally, subcutaneously, intraarterially, intradermally, transdermally, rectally, intracranially, intraperitoneally, intranasally, intratumorally, intramuscularly or as inhalants. In certain embodiments, the pharmaceutical composition comprising extracellular vesicles is administered intravenously, for example, by injection. Extracellular vesicles can optionally be administered in combination with other therapeutic agents that are at least partially effective in treating the disease, disorder or condition for which the extracellular vesicles are intended.
[00214] [00214] Solutions or suspensions may include the following components: a sterile diluent such as water, saline, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial compounds such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating compounds such as ethylene diaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates and compounds for adjusting tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The preparation can be contained in ampoules, disposable syringes or multi-dose vials made of glass or plastic.
[00215] [00215] Pharmaceutical compositions suitable for injection
[00216] [00216] Sterile injectable solutions can be prepared by incorporating the extracellular vesicles in an effective amount and in an appropriate solvent with one or a combination of ingredients listed here, as desired. Generally, dispersions are prepared by incorporating extracellular vesicles in a sterile vehicle that contains a basic dispersion medium and any other desired ingredients. In the case of sterile powders for the preparation of sterile injectable solutions, the preparation methods are vacuum drying and freeze drying which yield a powder of the active ingredient plus any desired additional ingredient from a previously sterile filtered solution of the same. Extracellular vesicles can be administered in the form of a deposit injection or implant preparation that can be formulated in such a way as to allow a sustained or pulsatile release of extracellular vesicles.
[00217] [00217] Systemic administration of compositions comprising extracellular vesicles can also be by transmucosal means. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art and include, for example, for transmucosal administration, fusidic acid derivatives, detergents, and bile salts. Transmucosal administration can be performed using, for example, nasal sprays.
[00218] [00218] In certain embodiments, the pharmaceutical composition comprising extracellular vesicles is administered intravenously to an individual who would benefit from the pharmaceutical composition. In certain other modalities, the composition is administered to the lymphatic system, for example, by intralymphatic injection or by intravenous injection (see, for example, Senti et al, PNAS 105 (46): 17908 (2008)), or by intramuscular injection, by subcutaneous administration, by intratumoral injection, by direct injection into the thymus or liver.
[00219] [00219] In certain embodiments, the pharmaceutical composition comprising extracellular vesicles is administered as a liquid suspension. In certain embodiments, the pharmaceutical composition is administered as a formulation that is capable of forming a deposit after administration. In certain preferred embodiments, the deposit slowly releases extracellular vesicles into the circulation or remains in the form of a deposit.
[00220] [00220] Typically, pharmaceutically acceptable compositions are highly purified to be free of contaminants, are biocompatible and are non-toxic and are suitable for administration to an individual. If water is a constituent of the carrier, the water is highly purified and processed to be free of contaminants, for example, endotoxins.
[00221] [00221] The pharmaceutically acceptable carrier can be lactose, dextrose, sucrose, sorbitol, mannitol, starch, acacia gum, calcium phosphate, alginates, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methyl hydroxy benzoate, propyl hydroxy benzoate, talc, magnesium stearate and / or mineral oil, but without limitations. The pharmaceutical composition can further include a lubricant, a wetting agent, a sweetener, a flavor enhancer, an emulsifying agent, a suspending agent and / or a preservative.
[00222] [00222] The pharmaceutical compositions described herein comprise the extracellular vesicles described herein and, optionally, a pharmaceutically active or therapeutic agent. The therapeutic agent can be a biological agent, a small molecule agent or a nucleic acid agent.
[00223] [00223] Dosage forms are provided that comprise a pharmaceutical composition comprising the extracellular vesicles described herein. In some modalities, the dosage form is formulated as a liquid suspension for intravenous injection. In some embodiments, the dosage form is formulated as a liquid suspension for intratumor injection.
[00224] [00224] In certain embodiments, the preparation of extracellular vesicles is subject to radiation, for example, X-rays, gamma rays, beta particles, alpha particles, neutrons, protons, elementary nuclei, UV rays, in order to damage nucleic acids competent for residual replication.
[00225] [00225] In certain embodiments, the preparation of extracellular vesicles
[00226] [00226] In certain embodiments, the preparation of extracellular vesicles is subject to X-ray irradiation using an irradiation dose of more than 0.1, 0.5, 1, 5, 10, 15, 20, 25, 30 , 35, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 , 10,000 or more than 10,000 mSv. METHODS
[00227] [00227] Aspects of the present disclosure also include methods of producing the composition comprising the extracellular vesicle and the immunomodulatory component. In some modalities, the method comprises: obtaining the extracellular vesicle of the producing cell, in which the producing cell naturally contains the immunomodulatory component; and optionally isolating the obtained extracellular vesicle. In some modalities, the method comprises: modifying a producer cell with the immunomodulating component; obtaining the extracellular vesicle from the modified producer cell; and optionally isolating the extracellular vesicles obtained. In some other modalities, the method comprises: obtaining the extracellular vesicle of a producing cell; isolating the extracellular vesicles obtained; and modifying the isolated extracellular vesicle with the immunomodulatory component. In certain embodiments, the method further comprises formulating isolated extracellular vesicles in a pharmaceutical composition. METHODS OF PRODUCTION OF EXTRACELLULAR VESICLES METHODS OF MODIFYING THE PRODUCT CELL WITH THE IMMUNOMODULATOR COMPONENT
[00228] [00228] In several modalities, the method comprises the modification of a producer cell with the immunomodulatory component.
[00229] [00229] The producer cell can be a mammalian cell line.
[00230] [00230] In certain preferred embodiments, the producing cell is an immune cell, such as a dendritic cell, a T cell, a B cell, a natural killer cell (NK cell), an antigen presenting cell, a macrophage , an auxiliary T cell or a regulatory T cell (Treg cell).
[00231] [00231] In several modalities, the immunomodulatory component can be expressed in a producing cell from a transgene or mRNA introduced into the producing cell by transfection, viral transduction, electroporation, extrusion, sonication, cell fusion or other methods known to those skilled in the art in the technique.
[00232] [00232] In certain embodiments, the immunomodulatory component is introduced into the producing cell by transfection. In some modalities, the immunomodulatory component can be introduced into suitable producer cells using synthetic macromolecules, such as lipids and cationic polymers (Papapetrou et al, Gene Therapy 12: 8118-S8130 (2005)). In some modalities, cationic lipids form complexes with the immunomodulatory component through charge interactions. In some of these modalities, positively charged complexes bind to the negatively charged cell surface and are absorbed by the cell by endocytosis. In some other embodiments, a cationic polymer can be used to transfect producer cells. In some of these modalities, the cationic polymer is polyethyleneimine (PEI). In certain embodiments, chemical products such as calcium phosphate, cyclodextrin or polybrene, can be used to introduce the immunomodulatory component into the producing cells. The immunomodulatory component can also be introduced into a producer cell using a physical method, such as particle-mediated transfection, "gene weapon", biolistics or particle bombardment technology (Papapetrou et a /. ,, Gene Therapy 12: 8118-S130 (2005)). A reporter gene such as, for example, beta-galactosidase, chloramphenicol acetyltransferase, luciferase, or green fluorescent protein can be used to assess the efficiency of transfection of the producing cell.
[00233] [00233] In certain embodiments, the immunomodulatory component is introduced into the producing cell by viral transduction. Several viruses can be used as gene transfer vehicles, including the moloney murine leukemia virus (MMLV), adenovirus, adeno-associated virus (AAV), herpes simplex virus (HSV), lentivirus and foam virus. Virus-mediated gene transfer vehicles comprise vectors based on DNA viruses, such as adenoviruses, adeno-associated viruses and herpes viruses, as well as vectors based on retrovirals.
[00234] [00234] In certain embodiments, the immunomodulatory component is introduced into the producing cell by electroporation. Electroporation creates transient pores in the cell membrane, allowing the introduction of various molecules into the cell. In some modalities, DNA and RNA, as well as polypeptide and non-polypeptide therapeutic agents, can be introduced into the producing cell by electroporation.
[00235] [00235] In certain embodiments, the immunomodulatory component is introduced into the producing cell by microinjection. In some modalities, a glass micropipette can be used to inject the immunomodulating component into the producing cell at the microscopic level.
[00236] [00236] In certain embodiments, the immunomodulatory component is introduced into the producing cell by extrusion.
[00237] [00237] In certain embodiments, the immunomodulatory component is introduced into the producing cell by sonication. In some modalities, the producing cell is exposed to high intensity sound waves, causing a transient rupture of the cell membrane, allowing the loading of an immunomodulating component.
[00238] [00238] In certain embodiments, the immunomodulatory component is introduced into the producing cell by cell fusion. In some modalities, the immunomodulatory component is introduced by electrical fusion of cells. In some other embodiments, polyethylene glycol (PEG) is used to fuse producing cells. In some other modalities, the sendai virus is used to fuse the producer cells.
[00239] [00239] In some embodiments, the immunomodulatory component is introduced into the producing cell by hypotonic lysis. In some of these modalities, the producing cell is exposed to a low ionic strength buffer, causing it to explode, allowing the loading of an immunomodulating component. In some alternative modalities, controlled dialysis against a hypotonic solution is used to swell the producing cell and create pores in the membrane of the producing cell. The producing cell is subsequently exposed to conditions that allow the membrane to be resealed.
[00240] [00240] In some embodiments, the immunomodulatory component is introduced into the producer cell by treatment with detergent. In certain embodiments, the producer cell is treated with a mild detergent that temporarily compromises the membrane of the producer cell, creating pores that allow the loading of an immunomodulating component. After the producing cells are loaded, the detergent is washed, covering the membrane.
[00241] [00241] In some embodiments, the immunomodulatory component is introduced into the producing cell by receptor-mediated endocytosis. In certain embodiments, the producer cells have a surface receptor that, by binding the immunomodulating component, induces the internalization of the receptor and the associated immunomodulating component.
[00242] [00242] In some embodiments, the immunomodulatory component is introduced into the producing cell by filtration. In certain embodiments, the producing cells and the immunomodulating component can be forced through a filter of pore size smaller than the producing cell, causing transient rupture of the producing cell membrane and allowing the immunomodulating component to enter the producing cell.
[00243] [00243] In some modalities, the producing cell is subjected to several cycles of freezing and thawing, resulting in rupture of the cell membrane, allowing the loading of an immunomodulating component. METHODS OF MODIFICATION OF THE EXTRACELLULAR VESICLE WITH THE IMMUNOMODULATOR COMPONENT
[00244] [00244] In several alternative modalities, the immune component
[00245] [00245] In certain embodiments, the immunomodulatory component is introduced into the extracellular vesicle by transfection. In some modalities, the immunomodulatory component can be introduced into extracellular vesicles using synthetic macromolecules, such as lipids and cationic polymers (Papapetrou et a /., Gene Therapy 12: 8118- S8S130 (2005)). In certain embodiments, chemicals such as calcium phosphate, cyclodextrin or polybrene, can be used to introduce the immunomodulatory component into extracellular vesicles.
[00246] [00246] In certain embodiments, the immunomodulatory component is introduced into the extracellular vesicle by electroporation. In some modalities, the extracellular vesicles are exposed to an electric field that causes transient holes in the membrane of the extracellular vesicle, allowing the loading of an immunomodulating component.
[00247] [00247] In certain embodiments, the immunomodulatory component is introduced into the extracellular vesicle by microinjection. In some embodiments, a glass micropipette can be used to inject the immunomodulatory component directly into the extracellular vesicle at the microscopic level.
[00248] [00248] In certain embodiments, the immunomodulatory component is introduced into the extracellular vesicle by extrusion.
[00249] [00249] In certain embodiments, the immunomodulatory component is introduced into the extracellular vesicle by sonication. In some modalities, extracellular vesicles are exposed to high intensity sound waves, causing transient rupture of the extracellular vesicle membrane, allowing the loading of an immunomodulating component.
[00250] [00250] In some modalities, the immunomodulatory component
[00251] [00251] In some embodiments, the extracellular vesicle comprises an immunomodulatory component that is chemically conjugated. Chemical conjugation can be carried out by covalently bonding the immunomodulatory component to another molecule, with or without the use of a binder. The formation of such conjugates is the competence of the technicians and several techniques are known to perform the conjugation, with the choice of the particular technique being guided by the materials to be conjugated. In certain embodiments, polypeptides are conjugated to the extracellular vesicle. In certain other modalities, non-polypeptides, such as lipids, carbohydrates, nucleic acids and small molecules, are conjugated with the extracellular vesicle.
[00252] [00252] In some embodiments, the immunomodulatory component is introduced into the extracellular vesicle by hypotonic lysis. In some of these modalities, the extracellular vesicles are exposed to a low ionic strength buffer, causing them to explode, allowing the loading of an immunomodulatory component. In some alternative modalities, dialysis controlled against a hypotonic solution is used to swell the extracellular vesicle and create pores in the extracellular vesicle membrane. The extracellular vesicle is subsequently exposed to conditions that allow the membrane to be resealed.
[00253] [00253] In some embodiments, the immunomodulatory component is introduced into the extracellular vesicle by treatment with detergent. In certain embodiments, the extracellular vesicles are treated with a mild detergent that transiently compromises the membrane of the extracellular vesicle, creating pores that allow the loading of an immunomodulatory component. After the extracellular vesicles are loaded, the detergent is washed, covering the membrane.
[00254] [00254] In some embodiments, the immunomodulatory component is introduced into the extracellular vesicle by receptor-mediated endocytosis. In certain embodiments, the extracellular vesicles have a surface receptor that, by binding the immunomodulating component, induces the internalization of the receptor and the associated immunomodulating component.
[00255] [00255] In some embodiments, the immunomodulatory component is introduced into the extracellular vesicle by mechanical firing. In certain embodiments, extracellular vesicles can be bombarded with an immunomodulatory component attached to a heavy or charged particle, such as gold microcarriers. In some of these modalities, the particle can be accelerated mechanically or electrically, so that it crosses the membrane of the extracellular vesicle.
[00256] [00256] In some embodiments, the immunomodulatory component is introduced into the extracellular vesicle by filtration. In certain modalities, the extracellular vesicles and the immunomodulating component can be forced through a filter of pore size smaller than the extracellular vesicle, causing transient rupture of the extracellular vesicle membrane and allowing the immunomodulatory component to enter the extracellular vesicle .
[00257] [00257] In some modalities, extracellular vesicles are subjected to several cycles of freezing and thawing, resulting in rupture of the extracellular vesicle membrane, allowing the loading of an immunomodulating component. INSULATION METHODS OF EXTRACELLULATED VESICLES RES
[00258] [00258] Extracellular vesicles can be isolated from the producer cells. In certain embodiments, the extracellular vesicle is released by the producing cell in the cell culture medium. It is contemplated that all known ways of isolating extracellular vesicles are considered suitable for use in this document. For example, physical properties of extracellular vesicles can be used to separate them from a medium or other source material, including separation based on electrical charge (eg, electrophoretic separation), size (eg, filtration, sieving) molecular, etc.), density (for example, regular or gradient centrifugation), Svedberg constant (for example, sedimentation with or without external force, etc.). Alternatively or additionally, the isolation can be based on one or more biological properties and include methods that can employ surface markers (for example, for precipitation, reversible binding to the solid phase, FACS separation, binding to specific ligand, binding to non-specific binder, affinity purification, etc.).
[00259] [00259] Isolation and enrichment can be done in a general and non-selective way, typically including serial centrifugation. Alternatively, isolation and enrichment can be done in a more specific and selective manner, such as the use of extracellular vesicles or specific surface markers of a producer cell. For example, specific surface markers can be used in immunoprecipitation, FACS classification, affinity purification and magnetic separation with ligands attached to microspheres.
[00260] [00260] In some embodiments, size exclusion chromatography can be used to isolate extracellular vesicles. Size exclusion chromatography techniques are known in the art. Exemplary and non-limiting techniques are provided in this document. In some modalities, a fraction of empty volume is isolated and comprises the extracellular vesicles of interest. In addition, in some embodiments, extracellular vesicles can be further isolated after chromatographic separation
[00261] [00261] In some embodiments, the isolation of extracellular vesicles may involve combinations of methods that include, but are not limited to, differential centrifugation, size-based membrane filtration, immunoprecipitation, FACS classification and magnetic separation. METHODS OF MEASURING THE SIZE OF EXES VESICLES TRACELLULAR
[00262] [00262] In some embodiments, the methods described here include measuring the size of extracellular vesicles and / or populations of extracellular vesicles. Generally, the size of the extracellular vesicle is measured as the largest measurable dimension. Generally, the largest measurable dimension of an extracellular vesicle is also referred to as its diameter.
[00263] [00263] The size of the extracellular vesicle can be measured using dynamic light scattering (DLS) and / or multiangular light scattering (MALS). Methods of using DLS and / or MALS to measure the size of extracellular vesicles are known to those skilled in the art and include the nanoparticle screening assay (NTA, for example, using a screening device).
[00264] [00264] The size of the extracellular vesicle can be measured using adjustable resistive pulse detection (TRPS). In a specific embodiment, the size of the extracellular vesicle as measured by TRPS is determined using an iZON qNANO Gold. In some embodiments, the extracellular vesicles described here have a longer dimension of about 20 to 300 nm, as measured by TRPS (for example, using an iZON qNano Gold). In other ways, the extracellular vesicles described here have a longer dimension of about 40 to 200 nm, as measured by TRPS (for example, an iZON qNano Gold). In other embodiments, the extracellular vesicle populations described here comprise a population, in which 90% of the extracellular vesicles have a longer dimension of about 20 to 300 nm, as measured by TRPS (for example, using an iZON qNano Gold). In other embodiments, the extracellular vesicle populations described herein comprise a population, in which 95% of the extracellular vesicles have a longer dimension of about 20 to 300 nm, as measured by TRPS (for example, using an iZON qNano Gold ). In other embodiments, the extracellular vesicle populations described here comprise a population, in which 99% of the extracellular vesicles have a longer dimension of about 20 to 300 nm, as measured by TRPS (for example, using an iZON qNano Gold ). In other embodiments, the extracellular vesicle populations described herein comprise a population, in which 90% of the extracellular vesicles have a longer dimension of about 40 to 200 nm, as measured by TRPS (for example, using an iZON qNano Gold). In other embodiments, the extracellular vesicle populations described herein comprise a population, in which 95% of the extracellular vesicles have a longer dimension of about 40 to 200 nm, as measured by TRPS (for example, using an iZON qNano Gold ). In other embodiments, the extracellular vesicle populations described here comprise a population, in which 99% of the extracellular vesicles have a longer dimension of about 40 to 200 nm, as measured by TRPS (for example, using an iZON qNano Gold ).
[00265] [00265] The size of the extracellular vesicles can be measured using electron microscopy. In some embodiments, the electron microscopy method used to measure the size of the extracellular vesicle is transmission electron microscopy. In a specific embodiment, the transmission electron microscope used to measure the size of the extracellular vesicle is a Tecnai "MG Spirit Bi-OTWIN. Methods for measuring the size of the extracellular vesicle using an electron microscope are well known to the versa - in the art and any of these methods may be appropriate to measure the size of the extracellular vesicle.In some modalities, the extracellular vesicles described here have a longer dimension of about 20 to 300 nm, as measured by a microscope - scanning electron scope (for example, a Tecnai'y G Spirit BioTWIN scanning electron microscope). In other embodiments, the extracellular vesicles described here have a longer dimension of about 40 to 200 nm as measured by a scanning electron microscope (for example, a Tecnai "mM G Spirit BIOTWIN scanning electron microscope). In other modalities, the populations of extracellular vesicles described here comprise a population, in which 90% of the extracellular vesicles have a size
[00266] [00266] In addition, methods of treating cancer, graft versus host disease (GvHD) and autoimmune disease in an individual are provided in this document.
[00267] [00267] In various modalities, the composition is administered to an individual with cancer. In some of these modalities, the composition can increasingly regulate an immune response and improve the targeting of the tumor of the individual's immune system. In some modalities, the cancer being treated is characterized by the infiltration of leukocytes (T cells, B cells, macrophages, dendritic cells, monocytes) in the tumor microenvironment or the so-called "hot tumors" or "inflammatory tumors". In some modalities, cancer being treated is characterized by low or undetectable levels of leukocyte infiltration into the tumor microenvironment, or so-called "cold tumors" or "non-inflammatory tumors". In some embodiments, the composition is administered in an amount and long enough to convert a "cold tumor" into a "hot tumor", that is, the said administration results in the infiltration of leukocytes (such as T cells) in the tumor microenvironment.
[00268] [00268] In some embodiments, the composition comprising an extracellular vesicle and an immunomodulatory component is administered to an individual as a cancer vaccine. In some of these modalities, the composition is administered to an individual as a personalized cancer vaccine. In some modalities, the immunomodulatory component is a tumor antigen or a peptide derived from a tumor antigen. Suitable examples of tumor antigens include: alpha-fetoprotein (AFP), carcinoma-embryonic antigen (CEA), tumor epithelial antigen (ETA), mucin 1 (MUC1), Th-MUC1, mucin 16 (MUC16), tyrosinase, antigen associated with melanoma (MAGE), tumor protein p53 (p53), CD4, CD8,
[00269] [00269] Cancers that can be treated with the composition include, but are not limited to, the cancers listed in Table 5.
[00270] [00270] In certain embodiments, the composition is administered to an individual with graft versus host disease (GvHD). In some of these modalities, the composition can increasingly regulate an immune response and alleviate the symptoms of GvHD. In some specific modalities, the composition relieves GvHD symptoms by activating apoptotic signaling. In certain modes, the composition for the treatment of GvHD comprises the | Fas ligand (FasL). In some of these modalities, FasL is expressed on the surface of the extracellular vesicle.
[00271] [00271] In various embodiments, the composition is administered to an individual with an autoimmune disease. In some of these modalities, the composition can downwardly regulate an immune response and suppress the individual's immune activity.
[00272] [00272] Autoimmune diseases include, but are not limited to, multiple sclerosis, peripheral neuritis, Sjogren's syndrome, rheumatoid arthritis, alopecia, autoimmune pancreatitis, Behcet's disease, bullous pemphigoid, celiac disease, Devic's disease (neuromyelitis optica) ), glomerulonephritis, IgA nephropathy, varied vasculitis, scleroderma, diabetes, arteritis, vitiligo, ulcerative colitis, irritable bowel syndrome, psoriasis, uveitis and systemic lupus erythematosus.
[00273] [00273] In some embodiments, the composition is administered intravenously to the individual's circulatory system. In some modalities, the composition is infused with an appropriate liquid and administered into an individual's vein.
[00274] [00274] In some modalities, the composition is administered intra-arterially to the individual's circulatory system. In some modalities, the composition is infused with suitable liquid and administered to an individual's artery.
[00275] [00275] In some embodiments, the composition is administered to the individual by intrathecal administration. In some modalities, the composition is administered through an injection in the spinal canal or in the subarachnoid space, so that it reaches the cerebrospinal fluid (CSF).
[00276] [00276] In some embodiments, the composition is administered intratumorally in one or more tumors of the individual.
[00277] [00277] In some embodiments, the composition is administered to the individual by intranasal administration. In some modalities, the composition can be inflated through the nose in a form of topical or systemic administration. In certain modalities, the composition is administered as a nasal spray.
[00278] [00278] In some embodiments, the composition is administered to the individual by intraperitoneal administration. In some modalities, the composition is infused in an appropriate liquid and injected into the individual's peritoneum. In some embodiments, said intraperitoneal administration results in the distribution of the composition (for example, the extracellular vehicles in the composition) to the lymphatics. In some embodiments, said intraperitoneal administration results in the distribution of the composition (for example, the extracellular vesicles in the composition) to the thymus, spleen and / or bone marrow. In some modalities, said intraperitoneal administration results in the distribution
[00279] [00279] In some embodiments, the composition is administered to the individual by periocular administration. In some embodiments, the composition is injected into the periocular tissues. Peri- ocular drug administration includes the subconjunctival, anterior sub Tenon, posterior Sub Tenon and retrobulbar routes of administration.
[00280] [00280] In some embodiments, the composition is administered to the same individual by various routes of administration. In some modes, said multiple routes of administration comprise intravenous administration, intra-arterial administration, intrathecal administration, intranasal administration, intratumoral administration, intraperitoneal and / or periocular administration. In a preferred embodiment, said multiple routes of administration comprise intravenous administration and intraperitoneal administration.
[00281] [00281] In certain embodiments, the dosage of extracellular vesicles is between 1 ng to 10 ng, 10 ng to 100 ng, 100 ng to 1 vo, 1 uga ug, 5 ug to 10 ug, 10 ug to 50 ug, 50 µg to 75 µg, 75 µg to 100 µg, 100 µg to 150 µg, 150 µg to 200 µg, 200 µg to 300 µg, 300 µg to 500 µg, 500 µg to 1 mg or 1 mg to 10 mg.
[00282] [00282] The compositions can be administered once to the individual. Alternatively, multiple administrations can be performed over a period of time. For example, two, three, four,
[00283] [00283] In certain modalities, doses of extracellular vesicles are administered at intervals such as once a day, on alternate days, once a week, twice a week, once a month or twice a month.
[00284] [00284] In some embodiments, the pharmaceutical composition is administered at a frequency sufficient to effectively increase the concentration of the immunomodulatory component in the target cell or tissue above a level that is associated with a symptom of the disease, disorder or condition.
[00285] [00285] In some embodiments, the compositions are administered at least twice during a treatment period, so that the disease, disorder or condition is treated or a symptom of it is improved. In some embodiments, the compositions are administered at least twice during a treatment period, so that the disease, disorder or condition is treated or a symptom thereof is avoided. In some embodiments, the pharmaceutical composition is administered a sufficient number of times during a treatment period, so that a sufficient amount of immunomodulatory component is delivered to the target cell or tissue during the treatment period. In some modalities, the pharmaceutical composition is administered a sufficient number of times during a treatment period, so that a sufficient amount of immunomodulatory component is delivered to the target cell or tissue during the treatment period. so that one or more symptoms of the disease, disorder or condition are avoided, reduced, improved or delayed. In some modalities, increasing the concentration of the immunomodulatory component in the target cell or tissue includes increasing the peak concentration, while in others it includes increasing the average concentration. In some modalities, a substantial increase during the treatment period can be determined by comparing a pre-treatment or post-treatment period in the individual, or by comparing measurements made on a population undergoing treatment with a control population corresponding untreated.
[00286] [00286] In some embodiments, the pharmaceutical composition is administered a sufficient number of times per treatment period, so that the concentration of the immunomodulatory component in the target cell or tissue is increased for at least about a week, two weeks , three weeks, four weeks, one month, two months, three months, four months, five months, six months or more than six months. In some embodiments, the pharmaceutical composition is administered a sufficient number of times per treatment period, so that the concentration of the immunomodulatory component in the target cell or tissue is increased for a period of time at least during the treatment. treatment period.
[00287] [00287] In some modalities, the time interval between repeated administrations within a treatment period is not greater than the period in which the number of circulating extracellular vesicles is reduced to less than about 5%, 10 %, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95% of the number of extracellular vesicles present in the administered pharmaceutical composition.
[00288] [00288] In some embodiments, the methods further comprise one or multiple doses of non-therapeutic extracellular vesicles prior to the injection of an appropriate therapeutic dose of extracellular vesicles that harbor a therapeutic agent. In certain modalities, the non-therapeutic extracellular vesicle is administered separately and in a different dosage than the therapeutic extracellular vesicles. In certain modalities, the dosage of the non-therapeutic extracellular vesicle is greater than the dosage of the therapeutic extracellular vesicle. In certain other modalities, the dosage of the non-therapeutic extracellular vesicle is less than the dosage of the therapeutic extracellular vesicle. In certain embodiments, the dosage of the non-therapeutic extracellular vesicle is the same as the therapeutic extracellular vesicle. In various modalities, non-therapeutic extracellular vesicle methods prior to injecting an adequate dose of therapeutic extracellular vesicles reduce the update of therapeutic extracellular vesicles in the liver, lung and / or spleen.
[00289] [00289] “An effective amount of the composition is provided based, at least in part, on the target tissue, type of target cell, means of administration, physical characteristics of the extracellular vesicle (eg size and, in some cases, cases, the extent of the molecules to be delivered) and other determinants. In general, an effective amount of the composition provides an efficient cellular response of the target cell. The increase in efficiency can be demonstrated by increasing the transfection of cells (that is, the percentage of cells transfected with the constituents of the extracellular vesicle), increasing the cellular response or reducing the innate immune response of the host individual.
[00290] [00290] The dosage and frequency of administration of extracellular vesicles and pharmaceutical compositions thereof can be determined, for example, by the attending physician based on various
[00291] [00291] The following examples are presented in order to provide elements of common skill in the art with a full disclosure and description of how to make and use the present invention and are not intended to limit the scope of what the inventors consider their nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to the numbers used (for example, quantities, temperature, etc.), but some errors and experimental deviations should be considered. Unless otherwise stated, the parts are parts by weight, molecular weight is the average molecular weight, temperature is in degrees Celsius and pressure is atmospheric or close. Standard abbreviations can be used, for example, bp, base pair (s); kb, kilobase (s); pl, picoliters; s or second, second (s); min, minute (s); h or h, hour (s); aa, amino acid (s); nt, nucleotide (s); It's similar.
[00292] [00292] The practice of the present invention will employ, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, recombinant DNA techniques and pharmacology, within the skill of the art. Such techniques are fully explained in the literature. See, for example, T. E. Creighton,
[00293] [00293] The conditioned culture medium was collected and centrifuged at 300 to 800 xg for 5 minutes at room temperature to remove large cells and debris. The medium supernatant was then supplemented with 1000 U / L Benzonaseº and incubated at 37 º C for 1 hour in a water bath. The supernatant was collected and centrifuged at 16,000 xg for 30 minutes at 4 º C to remove residual cell debris and other large contaminants. The supernatant was then ultra-centrifuged at 133,900 xg for 3 hours at 4 º C to pellet the exosomes. The supernatant was discarded and any residual medium was aspirated from the bottom of the tube. The pellet was resuspended in 200 to 1000 µl of PBS (-Ca-Mg).
[00294] [00294] To further enrich the populations of exosomes, the pellet was processed through density gradient purification (sucrose or Optiprep '"). To purify the sucrose gradient, the exosome pellet was layered on a sucrose gradient, as defined in Table 6 below: TABLE 6, SUCHAROSE DENSITY GRADIENT: son (%) touch (mL)
[00295] [00295] The gradient was rotated at 200,000 xg for 16 hours at 4 º C in a 12 mL Ultra-Clear tube (344059) placed in a SW 41 Ti rotor to separate the fraction from the exosome.
[00296] [00296] The exosome layer was gently removed from the top layer and diluted in - 32.5 ml of PBS in a 38.5 ml Ultra-Clear tube (344058) and again ultracentrifuged at 133. 900 xg for 3 hours at 4 º C to pellet the purified exosomes. The resulting pellet was resuspended in a minimum volume of PBS (- 200 uL) and stored at 4º C.
[00297] [00297] For the "Optiprep" gradient, a sterile three-layer gradient was prepared with equal volumes of 10%, 30% and 45% Optiprep in a 12 ml Ultra-Clear tube (344059) for a SW 41 Ti rotor The pellet was added to the Optiprep "" gradient and ultracentrifuged at 200,000 xg for 16 hours at 4 º C to separate the fraction from the exosome. The exosome layer was then carefully collected from the top - 3 mL of the tube.
[00298] [00298] The exosome fraction was diluted in - 32 mL of PBS in a 38.5 mL Ultra-Clear tube (344058) and ultracentrifuged at 133. 900 xg for 3 hours at 4 º C to pellet the purified exosomes. The pelleted exosomes were then resuspended in a minimal volume of PBS (- 200 uL) and stored at 4º C. EXAMPLE 1: GENETICALLY MODIFIED EXOSOMES TO DISPLAY A CHECKING POINT REGULATORY ANTIBODY IMMUNE ACTION
[00299] [00299] “A human embryonic kidney (HEK) cell line is grown to high density, and the resulting exosomes are isolated from the culture medium according to methods known to those skilled in the art (for example, the methods described here). Genetically modified exosomes with protein antibody 4 (CTLA-4) associated with cytotoxic T lymphocytes are prepared by chemical conjugation according to techniques known in the art. Exosomes modified with the CTLA4 antibody are selected by flow cytometry. At the same time, unmodified exosomes are isolated according to the same standard methods.
[00300] [00300] The two populations of exosomes are marked with a radioactive tracker, and 150 µg of each preparation is injected into live mice (for example, mouse melanoma model). Mice that receive the exosomes that exhibit the CTLA-4 antibody or the unmodified exosomes are continuously monitored for 30 minutes and again at four-hour intervals by whole animal PET / CT. Images of whole animals allow real-time, high-resolution tracking of identified exosomes for various tissues.
[00301] [00301] 150 ug of each exosome population are injected into two cohorts of mice intravenously without first identifying with a radioactive tracker. The mice are euthanized five weeks after administration. Tumor samples are collected and analyzed by immunohistochemistry and real-time PCR. EXAMPLE 2: GENETICALLY MODIFIED EXOSOMES RA SHOW LEAGUE FAS
[00302] [00302] Human antigen presenting cells are transfected with a plasmid encoding a selectable marker resistant to puromycin and the Fas ligand. The transfected cells are treated with puromycin and resistant colonies are selected and subjected to assays for expression of the Fas ligand surface by flow cytometry. The cells that express the Fas ligand are
[00303] [00303] The two populations of exosomes are marked with a radioactive tracker, and 150 µg of each preparation is injected into live mice (for example, mouse GvHD model). Mice that receive exosomes derived from unmodified cells or exosomes derived from cells that express the Fas ligand are monitored continuously for 30 minutes and again at four hour intervals by whole animal PET / CT. Images of whole animals allow real-time, high-resolution tracking of identified exosomes for various tissues.
[00304] [00304] The populations of purified exosomes from unmodified producer cells and producer cells genetically modified to express the Fas ligand are purified according to the methods described here. 150 µg of each exosome population is injected into two cohorts of mice without first identifying with a radioactive tracker. Animals from both cohorts are euthanized three to five weeks after administration for immunohistochemical analysis and real-time PCR. EXAMPLE 3: LYMPHATIC ABSORPTION OF EXOSomes AFTER INTRAPERITONEAL ADMINISTRATION
[00305] [00305] To determine the biodistribution of purified exosomes in vivo, the following experiment was carried out:
[00306] [00306] The conditioned culture medium of 293T cells was collected and centrifuged at 300 to 800 xg for 5 minutes at room temperature.
[00307] [00307] To further enrich the populations of exosomes, the pellet was processed through purification by sucrose density gradient, as defined in Table 6.
[00308] [00308] The gradient was rotated at 200,000 xg for 16 hours at 4º C in a 12 mL Ultra-Clear tube (344059) placed in a SW 41 Ti rotor to separate the fraction from the exosome.
[00309] [00309] The exosome layer was gently removed from the top layer and diluted in - 32.5 ml of PBS in a 38.5 ml Ultra-Clear tube (344058) and again ultracentrifuged at 133. 900 xg for 3 hours at 4 º C to pellet the purified exosomes. The resulting pellet was resuspended in a minimum volume of PBS (- 200 uL) and stored at 4º C.
[00310] [00310] To radiolabel the purified exosomes for in vivo imaging, 1xX10 ** purified exosomes in 100 µL were diluted with HEPES (200 µL, 0.1M, pH 8.5) and conjugated to p-SCN-Bn-DFO (5 ug) for one hour at 37 º C followed by overnight incubation at 4 º C, separately. The DFO exosomes were incubated with 897Zr (7.5mCi) diluted in HEPES (100 ul, IM, pH 7.3) for one hour at 37 º C and purified on a qEv column. This resulted in a total yield (0.4 mCi of 89Zr-DFO exosomes in up to 0.8 mL of PBS) at 100 uCi / 1x10 * º exosomes. Quality control (HPLC) was carried out before release to ensure> 95% CPR. STABILITY / N VITRO
[00311] [00311] The exosomes (20 uCi / 2x10º) were incubated at room temperature in: a. Formulation Buffer b. Mouse serum (10% v / v exosome solution in serum, if possible)
[00312] [00312] 2 hours after the start of the incubation, solutions were injected on HPLC to determine the stability of the tracker. IN VIVO IMAGINOLOGY
[00313] [00313] The mice (SKH-1, n = 8, age 5 to 8 weeks) were randomized into two groups, weighed and injected (with the second group injected immediately after the end of the first group dynamic scan) ) with 1x10ºº% / g of exosomes to provide a minimum radioactive dose of 100 uCi / mouse. Group 1 was injected intravenously (IV) while group 2 was injected intraperitoneally (IP).
[00314] [00314] The mice receive a full-body PET / CT in a 4-mouse hotel, using the following schedule: dynamic 1h (5x60, 5x180, 8x300 seconds) and static imaging at 4h (20 min), 24h (Thursday , 20 min) and 48 hours (Friday, 30 min). Each point in the time of imaging was followed by CT for anatomical reference.
[00315] [00315] After the last moment of imaging, the mice were euthanized and the following organs were collected, weighed and counted in the gamma counter: blood, lung (one), liver (lobe), spleen, pancreas, kidney (one) , liver, colon and additional organs of high absorption.
[00316] [00316] The organs were left to decompose for 2 to 3 days if the counts were extremely high and counted again.
[00317] [00317] The two cohorts of treated mice were subjected to imaging 4 hours, 24 hours and 48 hours after treatment. Full-body PET / CT imaging revealed robust delivery to the livers of all mice in group 1 treated IV (Figure 1A) and a distinct non-overlapping distribution for mice in group 2 treated IP (Figure 1B). The organs were dissected and analyzed by gamma radiographic counter, which revealed significant liver and spleen absorption in treated mice | V (Figure 2). In contrast, for IP treated mice, absorption was observed mainly in the pancreas, spleen, thymus and lymph nodes, with additional absorption in the liver and ovaries. These results demonstrate that different routes of administration result in substantially different biodistribution profiles. It is important to note that IP administration leads to significant absorption in the lymphatics, suggesting that IP administration may be an adequate route of administration to reach immune cells. EXAMPLE 4: B-CELL ACTIVATION BY GE-NETICALLY MODIFIED CD40L EXOSOMES
[00318] [00318] “CD40L is a member of the tumor necrosis factor (TNF) superfamily expressed mainly in T cells. The CD40L receptor, CDA40, is expressed in antigen presenting cells, including macrophages, dendritic cells and B cells. Signaling through CD40 activates B cells and induces a specific response to the antigen. Activation of the CD40 pathway, therefore, has implications for the development of antitumor immunity in a wide range of tumor types. To determine whether genetically modified exosomes could be generated to induce a specific immunological effect, exosomes were generated from HEK293SF cells transfected with a plasmid containing full-length human CD40L. The transfected cells were placed under selection of purulicin and resistant cell populations were cultured at high density. The resulting exosomes were collected from conditioned culture medium and purified on an Optiprep "M gradient as described above. Exosomes from unmodified HEK293SF cells were also isolated for use as a control. Human peripheral blood mononuclear cells (PBMCs) ) were plated at 150,000 cells per well of a 96-well plate and incubated with purified CD40L exosomes or native exosomes overnight at 37 º C. A sample of PBMCs was incubated with 1 µg / mL of Soluble recombinant CD40L-Fc as a positive control As shown in Figures 3A and 3B, the CD40L exosomes activated B cells in a dose-dependent manner, as measured by the expression of CD69 in two different donor samples. in inducing B cell activation. It is important to note that the level of B cell activation by CD40L exosomes was comparable to the activation caused by CD40L-Fc.
[00319] [00319] To determine whether the exosome-mediated B cell activation observed was due to direct B cell activation or through
[00320] [00320] To further validate the CD40L exosomes, a reporter system was used to measure the activity of the genetically modified exosomes. Activation of the CD40 pathway results in the activation of NF-KB. Using a genetically modified U2OS cell line to overexpress CD40 on its surface and contain a luciferase reporter downstream of the NF-KB promoter (Promega Corporation), CDA40 activation was confirmed by incubating the cells in the presence of an anti antibody Agonistic CD40 (BioLegend, Inc.) cross-linked with an anti-Fc antibody (Jackson ImmunoResearch, Inc.) or recombinant human CD40L (ACROBiosystems) cross-linked with an anti-IgG antibody (Jackson ImmunoResearch, Inc.) (Figures 5A and 5B). The genetically modified CD40L exosomes were incubated with the genetically modified cells and resulted in a robust increase in luciferase activity comparable to the effects of anti-CD40 + anti-Fc. It is important to note that the genetically modified exosomes did not require a crosslinking antibody, demonstrating that CD40L on the surface of the exosomes can form sufficient functional CD40L trims to activate CD40. EXAMPLE 5: T-CELL ACTIVATION BY GENETICALLY MODIFIED CD80 EXOSOMES
[00321] [00321] CDB80 is expressed in antigen presenting cells and binds to CD28 and CTLAH on the surface of T cells. Stimulation by CD80 (and CD86) through CD28 and CTLA + activates T cells during the initiation of a response immune. To determine whether exosomes could be genetically modified to activate T cells, exosomes containing CD80 were generated by transfection and selection of HEK293SF cells, as described in Example 4. To validate the activity of CD80 exosomes, Human PBMCs were plated in 150,000 cells per well of a 96-well plate and incubated with (i) purified CD80 exosomes and anti-CD3 antibody, (ii) native exosomes and anti-CD3 antibody, (iii) anti-CD3 antibody alone, or (iv) a combination of anti-CD28 and anti-CD3 antibodies. The samples were incubated at 37 ºC for three days and subjected to assays regarding the counting of T cells for CD4 * T cells (Figure 6A) and CD8 * T cells (Figure 6B). The CD80 exosomes activated T cells in a dose-dependent manner and to an extent comparable to the positive control of CD3 and CD28 antibodies. On the other hand, native exosomes had no effect on the proliferation of T cells.
[00322] [00322] To confirm that CD80 exosomes induce functional T cell activation, IFNy levels were measured by AIlphaLISA in PBMCs incubated with native exosomes and CD80 exosomes with additional anti-CD3 antibody. As shown in Figure
[00323] [00323] CD27L (CD70) and OX40L are members of the TNF superfamily and bind to cognate receptors (CD27 and OX40, respectively) in T cells. CD27L is expressed by certain populations of T and B cells, while OX40L is expressed by certain populations of antigen presenting cells. Signaling through CD27 or OXA40, therefore, has implications for immuno-oncology, specifically as a method of activating anergic T cells. To determine whether exosomes could be genetically modified to induce pro-inflammatory cytokine production in PBMCs, exosomes containing CD27L and OX40L were generated by transfection and selection of HEK293SF cells, as described in Example 4. To validate the activity of exosomes CD27L, human PBMCs were plated on a 96-well plate and incubated with purified CD27L exosomes and anti-CD3 antibody, native exosomes and anti-CD3 antibody, anti-CD3 antibody alone or a combination of antibodies anti-CD28 and anti-CD3. The samples were incubated at 37 C for two days and subjected to assays for the production of gamma interferon (IFNy) (Figures 8A and 8B) and production of IL-2 (figures 9A and 9B) in two different donors. The CD27L exosomes induced the production of
[00324] [00324] To further validate the CD40L exosomes, a reporter system was used to measure the activity of the genetically modified exosomes. Activation of the OX40 pathway results in the activation of NF-KB. Using a genetically modified T cell cell line to overexpress OX40 on its surface and contain a luciferase reporter downstream of the NF-KB promoter (Promega Corporation), OXA40 activation was confirmed by incubating the cells in the presence of a agonistic anti-OX40 antibody (BioLegend, Inc.) cross-linked with an anti-Fc antibody (Jackson ImmunoResearch, Inc.) or recombinant human OX40 (ACROBIiosystems) cross-linked with an anti-IgG antibody (Jackson ImmunoResearch, Inc.) (Figures 12A and 12B). The anti-OX40L antibody cross-linked with anti-IgG failed to activate the reporter cells, while the recombinant OX40L cross-linked with anti-Fc led to robust activation of the reporter gene (Figure 12B). Surprisingly, the genetically modified OX40L exosomes induced the expression of the reporter gene to a greater extent than the anti-OX40 antibody or the recombinant OX40L (Figure 12C). It is important to note that the genetically modified exosomes did not require a crosslinking antibody, demonstrating that OX40 on the surface of the exosomes can form sufficient functional OX40L trimers to activate OX40. EXAMPLE 7: T-CELL ACTIVATION BY GE-NETICALLY MODIFIED IL-7 EXOSOMES
[00325] [00325] AIL-7 is a cytokine involved in the proliferation of B cells and T cells and has implications for immunotherapy. Specifically, IL-7 can activate T cells and induce a tumor antigen response in tumors that are poorly infiltrated by leukocytes or in tumor microenvironments that induced T cell anergy. I | L-7 signaling through the heterodimeric receptor IL-7 induces interferon gamma signaling (IFNy), which can improve the tumor-specific antigen response by T cells. To determine whether exosomes could be genetically modified to induce T cell activation , exosomes containing IL-7 were generated by transfection and selection of HEK293SF cells with the plasmid pDisplay "“ (ThermoFisher) which encodes a fusion of IL-7 and PDGF receptor. The genetically modified exosomes were purified as described in the Methods To validate the activity of IL-7 exosomes, human PBMCs were plated on a 96-well plate and incubated with purified IL-7 exosomes and anti-CD3 antibody, native exosomes and antibody ant i-CD3, anti-CD3 antibody alone or a combination of anti-CD28 and anti-CD3 antibodies. The samples were incubated at 37 ºC for two days and subjected to an IFNy test (Figures 13A and 13B). Exosomes of IL-7 in combination with anti-CD3 antibody induced peak IFNy production to a greater extent than anti-CD3 alone (Figure 13A). In addition, IL-7 exosomes induced IFNy in a dose-dependent manner and to an extent comparable to the positive control of CD3 and CD28 antibodies. In contrast, native exosomes had no effect on IFNy production (Figure 13B).
[00326] [00326] The IL-7 receptor is a heterodimeric complex consisting of IL-7R and IL-2RG, which form a ternary complex in the presence of IL-7 and induce signaling downstream through the JAK / STAT pathway, resulting in cell proliferation. A synthetic cell-based assay
[00327] [00327] To determine whether the effects of IL-7 exosomes observed in vitro can be recapitulated in an in vivo model, IL-7 exosomes were administered to C57BL / 6 mice. A cohort of 20 mice was separated in following groups: (1) PBS, (2) recombinant human IL-7 (rhlL-7), (3) exosomes genetically modified by IL-7 and (4) unmodified native exosomes. Five mice in each group were injected intraperitoneally (IP) with 1 mg of EdU and PBS, 1x10 ** native exosomes or IL-7 or 10 µg of rhll-7 once daily for three days. Mice were sacrificed, spleens were isolated and EdU levels were measured in splenic cells by flow cytometry. As shown in Figure 15A, CD8 + T cells with a positive percentage increased significantly in the mice of the | L-7 exosome and in the rhlL-7 mice compared to the control cohorts. Although the T-cell count in the IL-7 exosome mice was lower than the rhlL-7 cohort, it was estimated that there were five times fewer IL-7 molecules administered in the exosome | IL-7 cohort (data not shown) . A similar trend was observed for CD8 + Memory T cells, measuring the levels of the CD45RO memory marker (Figure 15B).
[00328] [00328] “As an orthogonal approach, levels of CD71 (Transferrin receptor) were measured in splenic cells isolated from exosome-treated mice. CD71 is required for proliferation and CD71 levels correlate with the number of T cells. As shown in Figures 15A and 15B, the numbers of CD8 + T cells and CD-8 + Memory T cells followed the same trend seen in Figures 16A and 16B. Together, these data demonstrate that genetically modified exosomes can induce a specific immune cell effect in vivo and that this activation may be more potent per molecule compared to recombinant agonists.
[00329] [00329] To increase the activity of exosomes genetically modified by IL-7, the IL-7 sequence was fused with a truncated portion of PTGFRN, an innovative transmembrane exosome protein that is highly expressed on the surface of the HEK293SF exosomes. IL-7 was expressed as a translation fusion upstream of a short fragment of PTGFRN that spans the region before the most terminal C IgV domain, the transmembrane domain and the intracellular domain of PTGFRN, as well as a marker FLAG. A series of expression constructs was generated by introducing a series of four deletions of amino acids between | L-7 and PTGFRN (Figure 17A). The resulting constructs were numbered from pX-1 to pX-4 (complete sequence of pX-4 shown in Figure 17B). As shown by Western blot analysis using an anti-IL-7 antibody, the pX-3 and pX-4 constructs showed the highest levels of expression. The level of expression of IL-7 in the PTGFRN backbone was dramatically higher than pDisplay-IlL-7, which was used in Example 7 (Figure 18A). The increase in IL-7 expression suggests
[00330] [00330] As shown in the previous examples, exosomes can be genetically modified to overexpress functional endogenous sequences of immunomodulatory proteins. To de-
[00331] [00331] To determine whether T-cell activation mediated by an anti-CD-3 exosome was due to direct T-cell activation or through trans-acting immune cells, purified T-cell activation was measured. 100,000 purified human T cells were plated in a 96-well format in wells pre-coated with a non-bleaching antibody or anti-CD3 exosomes in the presence or absence of anti-CD28 antibody, or in wells that were incubated with anti-CD3 exosomes in the presence or absence of anti-CD3
[00332] [00332] AlL-12 is a potent immunostimulatory cytokine produced by antigen presenting cells in response to infection and other antigenic stimulation. The production of IL-12 by activated dendritic cells, macrophages and neutrophils induces the production of IFNy by both CD8 + and CD4 + T cells and induces cytotoxic effects of natural killer cells (NK). The combined impact of IL-12 secretion on the tumor microenvironment results in the secretion of Th1 cytokines including IFNy, leading to the death of tumor cells, reprogramming of myeloid-derived suppressor cells (MDSCs) and anti-angiogenic effects. The anti-tumor effects mediated by I1L-12 result in a long-lasting T-cell response and anti-tumor immunity in various
[00333] [00333] AIL-12 consists of two domains, p35 and p40. The human IL-12 dimer was encoded as a full-length PTG-FRN fusion protein (Figure 23A, construct 871, SEQ ID NO: 3) or a shortened fragment of PTGFRN that allows the display of high-density surface ( Figure 23B, construct 873, SEQ | ID NO: 5), and the constructs were expressed in a stable manner in HEK293SF cells. Stable cell lines were cultured in chemically defined media and exosomes from the culture supernatant were purified over an Optiprep "'M gradient as described in the Methods. The amount of IL-12 protein on the exosome surface was measured by ELISA and corresponded to the rlL-12 concentration for all functional studies.The purified, full-length, short hlL-12-PTGFRN exosomes, or recombinant hlL-12 (rhlL-12; BioLegend, Catalog No. 573004) were titrated in human PBMCs in the presence of a subideal anti-CD3 antibody to induce IFNy expression. rhIL-12 resulted in robust IFNy expression with an EC 5º of 0.029 ng / ml, which was comparable to IL12-PTGFRN of total length, both of which - 10x more potent than the short IL12-PTGFRN (Figures 24A-B). These results suggest that the IL-12 displayed in the full length PTGFRN framework may be a more potent immunomodulatory reagent than the construct d and short PTGFRN.
[00334] [00334] Human and mouse IL-12 proteins do not cross-react, and the in vitro data shown in Figure 24 suggest that mIL-12 fused to full-length PTGFRN would be more potent than the use of the framework short of PTGFRN. To determine the potency of the mlL-12-PTGFRN exosomes in an in vivo cancer model, the C57BL / 6 mice were implanted subcutaneously with 1x10º B16F10 murine melanoma cells (n = 5 mice per group). On days 5, 6 and 7 after tumor inoculation, animals were injected intratumorally with PBS, 0.2 µg of recombinant murine IL-12 (mIL12; BioLegend, Catalog No. 577004) or 1x10 *! exosomes displaying full-length IL-12-PTGFRN (mIL12-Exosomes; SEQ ID NO: 4). The animals were sacrificed once the tumor volume reached 2,000 mm . As shown in Figures 25 to 27, tumors in the PBS group grew rapidly, while tumors in the rmlL12 and mIL12-Exo groups were drastically reduced (- 65 to 80% reduction in volume). It is important to note that, on day 16, the tumors in the mIL12-Exo group were smaller than those in the rmlL12 group, demonstrating superior efficacy of IL-12 when displayed on the surface of exosomes compared to soluble cytokine. There was also a survival advantage for the groups treated with IL-12 compared to the groups treated with PBS (Figure 28).
[00335] [00335] To understand the mechanistic advantage of the exosomes | L- 12-PTGFRN over rmlL12, the expression of the Th1 gene was profiled in the tumors of the control and treated groups. IFNy (Figure 29A), T cell chemo attractants CXCL9 (Figure 29B) and CXCL10 (Figure 29C) and TGFB (Figure 29D) increased in the groups treated with IL-12 compared to the control group. In most cases, cytokine signs were greater in animals treated with mIL12-Exo compared to rmlL-12. IFNy levels in splenic CD8 + T cells were measured by flow cytometry, and mice treated with Exo-mIL-12 showed significantly greater signal than the PBS group or the rmIiL-12 group (Figure 30). Together, these data demonstrate that IL-12 displayed on the surface of an exosome represents a new and potent immunomodulatory strategy that promotes robust T cell activation in vitro and can be used to obtain potent antitumor effects in an aggressive model of melanoma murino at Vivo. Mechanically, exosomes of IL-12 show superiority over riL-12 and, therefore, represent an innovative differentiated therapeutic modality in immunotherapy against cancer. EXAMPLE 11: EXOSOMES THAT DISPLAY INTERFERON GAMMA ARE POWERFUL IMMUNE CELL ACTIVATORS
[00336] [00336] Gamma interferon (IFNy) is a cytokine involved in the preparation of innate and adaptive immune responses. This is expressed from a variety of cell types in response to numerous signals including I1L-12, and is sufficient to activate NK cells, lead to the presentation of antigens on antigen presenting cells and promote leukocyte activation and invasion. IFNy is naturally expressed as a homodimer and is secreted as a soluble factor. IFNy-expressing exosomes were generated by stable transfection of HEK293SF cells with full-length PTGFRN fused to human and mouse monomeric or dimeric IFNy (Figures 31A and 31B, respectively). Exosomes from suspended cell cultures were purified as described above and analyzed by PAGE. The monomeric PTGFRN IFNy (m) and tandem dimer (td) exosomes were expressed in the predicted molecular weights (arrowheads) at comparable levels (Figure 32). The purified exosomes were analyzed by ELISA and compared to a standard curve using recombinant IFNy (Biolegend, Catalog No. 570206) to calculate the number of IFNy molecules per exosome. The results in Table 9 show the number of IFNy molecules in each of the four types of purified exosomes. Notably, tandem dimer IFNy PTGFRN exosomes contain at least twice as many IFNy molecules as monomeric IFNy exosomes, suggesting that tandem dimer exosomes are adequately expressing dimeric IFNy constructs. TABLE 9
[00337] [00337] The human exosomes of monomeric PTGFRN-IFNy and tandem dimer were incubated with human PBMCs for 24 hours in increasing concentrations. Monocyte activation was measured by the expression of PD-L1, a downstream surface protein induced by IFNy signaling. As shown in Figure 33, native HEK293SF (WT) exosomes failed to induce PD-L1 expression, while IFNy PTGFRN both monomeric and tandem dimer exosomes induced PD-L1 dose-dependent, with greater activation by tandem dimer IFNy PTGFRN exosomes. Exosome-mediated PD-L1 activation was comparable to LPS-induced activation (Figure 33). These data demonstrate that a soluble cytokine, in monomeric or dimeric format, can be functionally expressed on the surface of an exosome and induce immune cell activation. The use of exosomes that express IFNy in immuno-oncology can be useful for inducing NK and T cell responses against tumor cells. EXAMPLE 12: EXOSOMES THAT EXPRESS IL-15 INDUCE THE NK CELL ACTIVATION
[00338] [00338] Interleukin 15 (IL-15) is a cytokine produced by mononuclear cells after pathogenic infection.
[00339] [00339] The results in Example 9 demonstrate that exosomes that exhibit anti-CD3 antibody fragments can activate T cells. To determine whether the PTGFRN framework supports this activity, the anti-CD3 antibody fragments (OKT3 variants) have been fused with the transmembrane region of PDGFR (exoCD3-PD), full-length PTGFRN (long exoCD3) or a fragment of PTGFRN (short exoCD3) and expressed stably in HEK293SF cells (Figure 38). The exosome link was confirmed by bi-layer interferometry (BLI) using an Octet & RED96 (Pall). A fragment of CD3 was attached to the BLI probe (Figure 39, ii), washed (Figure 39, iii) and the exosome constructs were added (Figure 39, iv). The exosomes of the WT HEK293SF cells did not bind to the BLI probe, but all the genetically modified constructs did. Both fragments of PTGFRN bound to the probe with greater affinity and remained stably linked (Figure 39, v). Anti-CD3 display exosomes have been tested for in vitro activity. T cell activation was measured by the positivity of the
[00340] [00340] The CD40 ligand (CD40L) is a ligand of the tumor necrosis superfamily (TNFSF) that binds to the co-stimulating CD40 receptor, which is highly expressed in B cells and other antigen presenting cells. Cellular activation mediated by the TNFSF ligand requires the formation of complex trimeric ligands that form on the cell surface and bind to cognate receptors. To investigate whether exosomes exhibiting different CD40L conformations on their surface were sufficient to activate B cells, more than 40 different CD40L expression constructs were designed and transfected individually into HEK293SF cells. CD40L was expressed as a fusion to the transmembrane domain of PDGFR, full size PTGFRN and a short fragment of single domain PTGFRN (Figure 41A, bottom). The CD40L-GFP PTGFRN fusions were expressed as a monomer (pCB-518 to pCB-526) or as a forced trimer (pCB-607 and pCB-527) (Figure 41A, lower). To promote the trimerization of monomeric CD40L, constructs were designed that expressed a fusion with TRAF2 multimerization domains (pCB-521 to pCB-523) or Collagen XV (pCB-524 to pCB-526). Among the CD40L monomeric constructs, pCB-518/521/524 contained full length N-terminal stem sequences from endogenous CD40L; pCB-519/522/525 contained an N-terminal stem sequence truncated from endogenous CD40L; and pCB-520/523/526 contained only the soluble portion of
[00341] [00341] The results shown in Figure 41, all used exosomes containing luminal GFP fused to the C-terminal of PTGFRN. In order to generate an unlabeled CD40L exosome, the same trimeric CD40L-PTGFRN construct as the main pCB-527 construct, but devoid of the C-terminal GFP, was stably expressed in HEK293SF (pCB- 766). The absolute concentration of CD40L on the surface of the genetically modified exosomes was quantified using ELISA (R&D Systems, Catalog No., DCDLA40), as shown in Table 10 below.
[00342] [00342] The purified exosomes of CD40L-PTGFRN were tested in B cell activation assays, as described above, in comparison with recombinant human CD40L of corresponding concentration (Biolegend, Catalog No. 591702). CD40L exosomes without label and containing GFP were comparable B cell activators when measured as a function of the number of particles or concentration of CD40L (Figure 42A), and both exosome preparations were more potent than the corresponding CD40L. concentration (Figure 42B). Native and unmodified exosomes of HEK293SF cells failed to activate B cells, demonstrating that the trimester constructs of genetically modified CD40L on the surface of the exosome were sufficient to potently activate B cells.
[00343] [00343] An alternative modality to agonize CD40 and activate B cells is to use an agonistic antibody cross-linked with a secondary antibody. To compare the potency of exosomes that express trimeric CD40L with an agonistic CD40L antibody, PBMC cultures were incubated with 2 µg / ml anti-CD40L antibody (Biology & Clone 5C3) with a secondary cross-linking antibody (Jackson Immuno Research, catalog number 115 -006-071). Maximum B cell activation is shown as the dotted line in Figures 43A and 43B. The pCB-527 exosomes (PT40-FRN trimeric CD40L-GFP) induced maximum B cell activation greater than the cross-linked agonistic antibody in two independent donor PBMC clusters (Figures 43A and 43B), demonstrating the superiority of trimeric CD40L exosomes in the activation of immune cells. EXAMPLE 15: SIMULTANEOUS DISPLAY OF MULTIPLE MOLECULES LAS OF IMMUNO-ONCOLOGY IN INDIVIDUAL EXOSOMES
[00344] [00344] The previous examples demonstrate that individual immune modulating proteins can be displayed on the surface of an exosome and induce functional changes in one or more types of immune cells. In certain applications, it may be necessary to use genetically modified exosomes in a combinatorial manner, that is, an exosome containing more than one molecule on the surface of the exosome, each capable of signaling a distinct immune cell pathway. HEK293SF cells were stably transfected with a plasmid that expresses fusion proteins both PTGFRN-IL-12 and PTGFRN-CDA40L. The exosomes were isolated and purified as described above. Exosomes from unmodified HEK293SF cells were used as negative controls.
[00345] [00345] In order to demonstrate the simultaneous loading of different binders, a down-tensile staining test was developed: REAGENTS:
[00346] [00346] Dynabeads (Exosomal Streptavidin Detection / Isolation Reagent for Termofisher, Catalog No. 10608D): 1x107 microspheres / mL, 50% slurry
[00347] [00347] “Insulation buffer: 0.5% BSA / PBS (1: 4 out of 2% BSA)
[00348] [00348] “Blocking buffer: 2% BSA / PBS (1gr / 5OmL, filter)
[00349] [00349] eLaundry 0.5 ml microspheres with 0.5 ml isolation buffer and resuspend in 0.5 ml! isolation buffer
[00350] [00350] e Add lug of biotinylated capture antibody (2.2 ul of stock to 0.5ug / ul)
[00351] [00351] e1howered, RT
[00352] [00352] and Wash 500u! L of insulation buffer
[00353] [00353] and Resuspend in 500ul blocking buffer, 10 min RT rotation
[00354] [00354] and Incubate in 500JUl of isolation buffer (1x10 Microspheres / mL, 50% slurry)
[00355] [00355] and Store ad4 ”C A. CAPTURE AND FLOW OF EXOSOMES
[00356] [00356] 1x10º microspheres per sample (10 µl microspheres, 20 µl of slurry)
[00357] [00357] 50,000 exosomes per microsphere; 5x10º exosomes per sample (1.2x10º exosomes / uL of stock)
[00358] [00358] South of each detection antibody fluorescently identified for flow
[00359] [00359] Mix 5x10º exosomes + 204! of Dyna-beads slurry + 0.7 ml of 0.1% BSA / PBS PROCEDURE:
[00360] [00360] 1. 120 ul of microspheres of slurry, remove excess, add 0.7 ml of blocking buffer, mix, rotate 10 min RT, remove supernatant
[00361] [00361] 2. Suspend the microspheres in 0.7 ml of isolation buffer + 25.2 ul of exosomes, turn ON at 4 ºC
[00362] [00362] 3. Next day: rapid rotation of exosomes and microspheres, 5s
[00363] [00363] 4. Place the tube on the magnet, remove supernatant
[00364] [00364] 5. Lock at 700yul, 10 min RT rotation
[00365] [00365] 6. Place the tube on the magnet, remove supernatant
[00366] [00366] 7. Resuspend in insulation buffer at 600ul!: 6 x 100ul! per tube
[00367] [00367] 8. Add 1ul of identified detection antibody, mix, incubate 30 min at 4ºC in the dark
[00368] [00368] 9 Spin2min500g, remove supernatant
[00369] [00369] 10. Wash 2x isolation buffer
[00370] [00370] 11. Resuspend in the isolation buffer at 200yl, perform the flow.
[00371] [00371] The native exosomes were isolated with microspheres decorated with anti-CD40L and fluorescent antibodies labeled against IL-12 and CD40L (Figure 44A) or CD81, an exosome marker present in native and genetically modified exosomes and CDA40L (Figure 44B ). The microspheres of CD40L did not pull down any of the native exosomes, since no fluorescent signal was detected for IL-12, CD40L or CD81. In contrast, the genetically modified double PTGFRN-CD40L / IL-12 exosomes were incubated with anti-CD40L microspheres and isolated as above. Staining for CD81 (Figure 45A), IL-12 or CD40L (Figure 45B) was detected with the genetically modified exosomes (more than 97% of the counted microspheres), indicating that CD40L-mediated isolation could also isolate IL-exosomes 12. Likewise, microspheres decorated with anti-IL-12 were incubated with exosomes genetically modified by IL-12 / CDA40L and stained for IL-12, CD40L and CD81. Over 98% of all microspheres were positive for CD40L and I | L-12 or for CD81 (Figures 46A and 46B), demonstrating that the exosomes contained | IL-12 and CD40L on their surface.
[00372] [00372] The concentration of IL-12 and CD40L was quantified by ELI-SA (Abcam Catalog Number ab119517) to test the genetically modified exosomes for in vitro potency. Equal concentrations of recombinant 11-12, recombinant 11-12 mixed with CD40L recombinant exosomes, PTGFRN-IL-12, doubly positive PTGFRN-CD40L / IL-12 exosomes or a mixture of PTGFRN-IL-12 and PTGFRN-CD40L exosomes were exosomes added to human PBMCs in increasing concentrations (rhlL-12 - BioLegend, Catalog No. 573004; rhnCD40L - Biolegend, Cat.
[00373] [00373] To further explore the possibility of combinatorial display exosomes from the surface, HEK293SF cells were stably transfected with three independent constructs that express PTGFRN-IL-12, PTGFRN-CD40L or PTGFRN-FLT3L fusion proteins. The exosomes were purified and isolated by the affinity microsphere methods, as described above, but were also interrogated for the presence of surface FLT3L using a conjugated anti-FLT3L-PE antibody. The exosomes isolated with anti-IL-12 microspheres were doubly positive for IL-12 and CD40L (Figure 51A), IL-12 and FLT3L (Figure 51B) and CD40L and FLT3L (Figure 51C). The exosomes isolated with anti-CD40L microspheres were doubly positive for IL-12 and CD40L (Figure 52A), I1L-12 and FLT3L (Figure 52B) and CD40L and FLT3L (Figure 52C), confirming that individual exosomes expressed each of the three binding immunomodulators. These results demonstrate that genetically modified immunomodulatory exosomes are a viable therapeutic modality and that they are comparable or more potent than soluble cytokines in activating immune cells.
[00374] [00374] Although the invention set out above has been described in detail by way of illustration and example for the sake of clarity of understanding, it is readily apparent to those skilled in the art in the light of the teachings of this disclosure that certain changes and modifications can be made without deviate from the spirit or the scope of the attached claims.
[00375] [00375] In this sense, the foregoing illustrates merely the principles of the invention. It will be realized that those skilled in the art will be able to plan various arrangements that, although not explicitly described or shown in this document, incorporate the principles of the invention and are included within its spirit and scope. In addition, all the examples and the conditional language cited here are primarily intended to help the reader understand the principles of the invention, without limitation to such examples and conditions specifically recited. In addition, all statements in this document that relate principles, aspects and modalities of the invention, as well as their specific examples, are intended to cover structural and functional equivalents thereof. In addition, it is intended that such equivalents include both the equivalents known today and the equivalents developed in the future, that is, any developed elements that perform the same function, regardless of the structure. The scope of the present invention, therefore, is not intended to be limited to the exemplary modalities shown and described in this document. Preferably, the scope and spirit of the present invention are incorporated by the appended claims.
SEQUENCE LISTING> SEQ ID NO: 1 MFHVSFRYIFGLPPLILVLLPVASSDCDIEGKDGKQYESVLMVSIDQLL DSMKEIGSNCLNNEFNFFKRHICDANKEGMFLFRAARKLRQFLKMNS TGDFDLHLLKVSEGTTILLNCTGQVKGRKPAALGEAQPTKSLEENKSL KEQKKLNDLCFLKRLLQEIKTCWNKILMGTKEHSAGGGGSDYKDDDD KGGGGSGPIFNASVHSDTPSVIRGDLIKLFCIITVEGAALDPDDMAFDV SWFAVHSFGLDKAPVLLSSLDRKGIVTTSRRDWKSDLSLERVSVLEF LLQVHGSEDQDFGNYYCSVTPWVKSPTGSWOQKEAEIHSKPVFITVK MDVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSHWCCKKEVQETRRER
RRLMSMEMD> SEQ ID NO: 2 MFHVSFRYIFGLPPLILVLLPVASSDCDIEGKDGKQYESVLMVSIDQLL DSMKEIGSNCLNNEFNFFKRHICDANKEGMFLFRAARKLRQFLKMNS TGDFDLHLLKVSEGTTILLNCTGQVKGRKPAALGEAQPTKSLEENKSL KEQKKLNDLCFLKRLLQEIKTCWNKILMGTKEHSAGGGGSGGGESG PIFNASVHSDTPSVIRGDLIKLFCIITVEGAALDPDDMAFDVSWFAVHS FGLDKAPVLLSSLDRKGIVTTSRRDWKSDLSLERVSVLEFLLQVHGS EDQDFGNYYCSVTPWVKSPTGSWQKEAEI / HSKPVFITVKMDVLNAF KYPLLIGVGLSTVIGLLSCLIGYCSSHWCCKKEVQETRRERRRLMSM
EMD> hIL-12-PTGFRN; 871 (SEQ ID NO: 3) MCHQQLVISW - FSLVFLASPL - VAIWELKKDV - YVVELDWYPD APGEMVVLTC DTPEEDGITW - TLDOSSEVLG SGKTLTIQVK EFGDAGQYTC HKGGEVLSHS LLLLHKKEDG .IWSTDILKDA KEPKNKTFLR - CEAKNYSGRF TCWWLTTIST DLTFSVKSSR “GSSDPQGVTC GAATLSAERV RGDNKEYEYS VECQEDSACP AAEESLPIEV MVDAVHKLKY ENYTSSFFIR DIIKPDPPKN LQLKPLKNSR QVEVSWEYPD TWSTPHSYFS LTFCVQVAGK SKREKKDRVF TDKTSATVIC RKNASISVRA QDRYYSSSWS EWASVPCSGG SGGGSGGGGS GGGGSGGGSG GRNLPVATPD PGMFPCLHHS QNLLRAVSNM LQKARQTLEF YPCTSEEIDH EDITKDKTST VEACLPLELT KNESCLNSRE TSFITNGSCL ASRKTSFMMA LCLSSIVYEDL - “KMYQVEFKTM NAKLLMDPKR —QIFLDQNMLA VIDELMQALN FNSETVPQKS SLEEPDFYKT KIKLCILLHA FRIRAVTIDR VMSYLNASSA GGGGSGGGGS RVVRVPTATL VRVVGTELVI PCNVSDYDGP SEQNFDWSFS SLGSSFVELA —STWEVGFPAQ LYQERLORGE ILLRRTANDA VELHIKNVQP SDQGHYKCST PSTDATVOGN YEDTVQVKVL ADSLHVGPSA RPPPSLSLRE GEPFELRCTA —ASASPLHTHL —ALLWEVHRGP —ARRSVLALTH EGRFHPGLGY EQRYHSGDVR LDTVGSDAYR LSVSRALSAD QGSYRCIVSE WIAEQGNWOQE IQEKAVEVAT VVIQPSVLRA AVPKNVSVAE GKELDLTCNI TTDRADDVRP EVTWSFSRMP - DSTLPGSRVL —ARLDRDSLVH —SSPHVALSHV DARSYHLLVR DVSKENSGYY YCHVSLWAPG HNRSWHKVAE AVSSPAGVGV TWLEPDYQVY LNASKVPGFA —DDPTELACRV VDTKSGEANV RFTVSWYYRM NRRSDNVVTS ELLAVMDGDW - “TLKYGERSKQ RAQDGDFIFS KEHTDTFNFR IQORTTEEDRG NYYCVWVSAWT - KORNNSWVKS KDVFSKPVNI FWALEDSVLV VKARQPKPFF AAGNTFEMTC - KVSSKNIKSP RYSVLIMAEK PVGDLSSPNE TKYIISLDQD SVVKLENWTD ASRVDGVVLE KVQEDEFRYR MYQTQVSDAG LYRCMVTAWS PVRGSLWREA - ATSLSNPIEI DFQTSGPIFN ASVHSDTPSV IRGDLIKLFC ITVEGAALD —PDDMAFDVSW —FAVHSFGLDK —APVLLSSLDR KGIVTTSRRD WKSDLSLERV —SVLEFLLOVH —GSEDQDFGNY YCSVTPWVKS PTGSWQKEAE IHSKPVFITV KMDVLNAFKY PLLIGVGLST VIGLLSCLIG
YCSSHWCCKK EVOQETRRERR RLMSMEMD *> mlL-12-PTGFRN; 872 (SEQ ID NO: 4) MCPQKLTISW - FAIVLLVSPL - MAMWELEKDV - YVWVEVDWTPD APGETVNLTC DTPEEDDITW. - TSDQORHGVIG - SGKTLTITVK EFLDAGQYTC HKGGETLSHS HLLLHKKENG IWSTEILKNF KNKTFLKCEA - PNYSGRFTCS WLVORNMDLK FNIKSSSSSP —DSRAVTCGMA SLSAEKVTLD QRDYEKYSVS CQEDVTCPTA EETLPIELAL EARQQNKYEN YSTSFFIRDI IKPDPPKNLQ MKPLKNSQVE VSWEYPDSWS TPHSYFSLKF FVRIORKKEK MKETEEGCNQ KGAFLVEKTS TEVQCKGGNV CVQOAQDRYYN SSCSKWACVP CRVRSGGSGG GSGGGGGSCGGÇ GSGGGSGGRV IPVSGPARCL SQOSRNLLKTT DDMVKTAREK LKHYSCTAED IDHEDITRDQ TSTLKTCLPL ELHKNESCLA TRETSSTTRG SCLPPQKTSL MMTLCLGSIY - EDLKMYQTEF - QAINAALQNH NHQQIILDKG MLVAIDELMQ SLNHNGETLR - QOKPPVGEADP - YRVKMKLCIL —LHAFSTRVVT INRVMGYLSS ASAGGGGSCGG —GGSRVVRVPT ATLVRVWVGTE LVIPCNVSDY DGPSEQNFDW SFSSLGSSFV —ELASTWEVGF PAQLYQERLAQ —RGEILLRRTA NDAVELHIKN VQAQPSDAGHYK CSTPSTDATIV QGNYEDTVOQV KVLADSLHVG PSARPPPSLS LREGEPFELR - CTAASASPLH —THLALLWEVH —RGPARRSVLA LTHEGRFHPG LGYEQRYHSG IS DVRLDTVGSD AYRLSVSRAL SADQGSYRCI VSEWIAEQGN WQEIQEKAVE - VATVVIQPSV - LRAAVPKNVS - VAEGKELDLT CNITTDRADD VRPEVTWSFS ——RMPDSTLPGS RVLARLDRDS LVHSSPHVAL SHVDARSYHL LVRDVSKENS - GYYYCHVSLW —APGHNRSWHK VAEAVSSPAG VGVTWLEPDY QVYLNASKVP - GFADDPTELA —CRVVDTKSGE ANVRFTVSWY YRMNRRSDNV VTSELLAVMD - “GDWTLKYGER SKQRAQDGDF IFSKEHTDTF NFRIQRTTEE DRGNYYCVWS AWTKORNNSW VKSKDVFSKP VNIFWALEDS VLVVKARQPK PFFAAGNTFE - MTCKVSSKNI —KSPRYSVLIM —AEKPVGDLSS PNETKYIISL DQDSVVKLEN WTDASRVDGV VLEKVQEDEF RYRMYQTOVS DAGLYRCMVT AWSPVRGSLW - REAATSLSNP IEIDFATSGP —IFNASVHSDT PSVIRGDLIK LFCIITVEGA - ALDPDDMAFD - VSWFAVHSFG LDKAPVLLSS LDRKGIVTTS RRDWKSDLSL ERVSVLEFLL QVHGSEDQDF GNYYCSVTPW VKSPTGSWQK EAEIHSKPVF ITVKMDVLNA FKYPLLIGVG LSTVIGLLSC
LIGYCSSHWC CKKEVQETRR ERRRLMSMEM D *> hIL-12-PTGFRN short; 873 (SEQ ID NO: 5) MCHQQLVISW. - FSLVFLASPL - VAIWELKKDV - YVVELDWYPD APGEMVVLTC DTPEEDGITW - TLDOSSEVLG IS SGKTLTIQVK EFGDAGQYTC HKGGEVLSHS LLLLHKKEDG INVSTDILKDQ - KEPKNKTFLR - CEAKNYSGRF TCWWLTTIST DLTFSVKSSR - “GSSDPQGVTC GAATLSAERV RGDNKEYEYS VECQEDSACP AAEESLPIEV MVDAVHKLKY ENYTSSFFIR DIIKPDPPKN LQLKPLKNSR QVEVSWEYPD TWSTPHSYFS LTFCVQVAGK SKREKKDRVF TDKTSATVIC RKNASISVRA QDRYYSSSWS EWASVPCSGG SGGGSGGGGS GGGGSGGGSGÇ GRNLPVATPD PGMFPCLHHS QNLLRAVSNM LQKARQTLEF YPCTSEEIDH EDITKDKTST VEACLPLELT KNESCLNSRE TSFITNGSCL ASRKTSFMMA LCLSSIVYEDL - “KMYQVEFKTM NAKLLMDPKR —QIFLDAQNMLA VIDELMQALN FNSETVPQKS SLEEPDFYKT KIKLCILLHA FRIRAVTIDR VMSYLNASSA GGGGSGCGGGGS GPIFNASVHS DTPSVIRGDL IKLFCIITVE GAALDPDDMA FDVSWFAVHS - FGLDKAPVLL —SSLDRKGIVT TSRRDWKSDL SLERVSVLEF LLAVHGSEDQ “DFGNYYCSVT PWVKSPTGSW QKEAEIHSKP VFITVKMDVL NAFKYPLLIG VGLSTVIGLL SCLIGYCSSH WECCKKEVQET
RRERRRLMSM EMD *> mlL-12-PTGFRN short; 874 (SEQ ID NO: 6) MCPQKLTISW - FAIVLLVSPL - MAMWELEKDV - YVWVEVDWTPD APGETVNLTC DTPEEDDITW. - TSDQORHGVIG - SGKTLTITVK EFLDAGQYTC HKGGETLSHS HLLLHKKENG IWSTEILKNF KNKTFLKCEA - PNYSGRFTCS WLVORNMDLK FNIKSSSSSP —DSRAVTCGMA SLSAEKVTLD QRDYEKYSVS CQEDVTCPTA EETLPIELAL EARQQNKYEN YSTSFFIRDI IKPDPPKNLQ MKPLKNSQVE VSWEYPDSWS TPHSYFSLKF FVRIORKKEK MKETEEGCNQ KGAFLVEKTS TEVQCKGGNV CVOAQDRYYN SSCSKWACVP CRVRSGGSGG GSGGGGGSGCGGGÇ GSGGGSGGRV IPVSGPARCL SOQOSRNLLKTT DDMVKTAREK LKHYSCTAED IDHEDITRDQ TSTLKTCLPL ELHKNESCLA TRETSSTTRG SCLPPQKTSL MMTLCLGSIY - “EDLKMYQTEF - QAINAALQNH NHQQIILDKG MLVAIDELMQ SLNHNGETLR - QOKPPVGEADP - YRVKMKLCIL LHAFSTRVVT INRVMGYLSS ASAGGGGSGG GGSGPIFNAS VHSDTPSVIR GDLIKLFCII TVEGAALDPD DMAFDVSWFA VHSFGLDKAP VLLSSLDRKG IVTTSRRDWK SDLSLERVSV LEFLLQOVHGS —— “EDQDFGNYYC SVTPWVKSPT GSWQKEAEIH SKPVFITVKM DVLNAFKYPL LIGVGLSTVI GLLSCLIGYC SSHWCCKKEV
QETRRERRRL MSMEMD * SEQ ID NO: monomer 7 PTGFRN IFN gamma MGRLASRPLLLALLSLALCRGQDPYVKEAENLKKYFNAGHSDVADN GTLFLGILKNWKEESDRKIMQSQIVSFYFKLFKNFKDDOQSIQKSVETIK EDMNVKFFNSNKKKRDDFEKLTNYSVTDLNVORKAIHELIQVMAELS PAAKTGSAGGGGSGGGGSRVVRVPTATLVRVVGTELVIPCNVSDYD GPSEQNFDWSFSSLGSSFVELASTWEVGFPAQLYQERLQRGEILLR RTANDAVELHIKNVQPSDQGHYKCSTPSTDATVQGNYEDTVQVKVL ADSLHVGPSARPPPSLSLREGEPFELRCTAASASPLHTHLALLWEVH RGPARRSVLALTHEGRFHPGLGYEQRYHSGDVRLDTVGSDAYRLSV SRALSADQGSYRCIVSEWIAEQGNWQEIQEKAVEVATVVIQPSVLRA AVPKNVSVAEGKELDLTCNITTDRADDVRPEVTWSFSRMPDSTLPGS RVLARLDRDSLVHSSPHVALSHVDARSYHLLVRDVSKENSGYYYCH VSLWAPGHNRSWHKVAEAVSSPAGVGVTWLEPDYQVYLNASKVPG FADDPTELACRVVDTKSGEANVRFTVSWYYRMNRRSDNVVTSELLA VMDGDWTLKYGERSKQRAQDGDFIFSKEHTDTFNFRIQORTTEEDRG NYYCVVSAWTKQRNNSWVKSKDVFSKPVNIFWALEDSVLVVKARQP KPFFAAGNTFEMTCKVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYI SLDQDSVVKLENWTDASRVDGVVLEKVQEDEFRYRMYQTQVSDAG LYRCMVTAWSPVRGSLWREAATSLSNPIEIDFQTSGPIFNASVHSDT PSVIRGDLIKLFCIITVEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLS SLDRKGIVTTSRRDWKSDLSLERVSVLEFLLQOVHGSEDQDFGNYYCS VTPWVKSPTGSWQKEAEIHSKPVFITVKMDVLNAFKYPLLIGVGLSTV
IGLLSCLIGYCSSHWCCKKEVQETRRERRRLMSMEMD SEQ ID NO: 8 dimer PTGFRN IFN range MGRLASRPLLLALLSLALCRGQDPYVKEAENLKKYFNAGHSDVADN GTLFLGILKNWKEESDRKIMQSQIVSFYFKLFKNFKDDOQSIQKSVETIK EDMNVKFFNSNKKKRDDFEKLTNYSVTDLNVQRKAIHELIQVMAELS PAAKTGGSGGSGGSGGSGCGQDPYVKEAENLKKYFNAGHSDVADNGT LFLGILKNWKEESDRKIMQSQIVSFYFKLFKNFKDDQSIQKSVETIKED MNVKFFNSNKKKRDDFEKLTNYSVTDLNVOQRKAIHELIQVMAELSPA AKTGSAGGGGSGGGGSRVVRVPTATLVRVVGTELVIPCNVSDYDGP SEQNFDWSFSSLGSSFVELASTWEVGFPAQLYQERLQRGEILLRRTA NDAVELHIKNVQPSDQGHYKCSTPSTDATVQGNYEDTVQVKVLADS LHVGPSARPPPSLSLREGEPFELRCTAASASPLHTHLALLWEVHRGP ARRSVLALTHEGRFHPGLGYEQRYHSGDVRLDTVGSDAYRLSVSRA LSADQGSYRCIVSEWIAEQGNWQEIQEKAVEVATVVIQPSVLRAAVP KNVSVAEGKELDLTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVL ARLDRDSLVHSSPHVALSHVDARSYHLLVRDVSKENSGYYYCHVSL WAPGHNRSWHKVAEAVSSPAGVGVTWLEPDYQVYLNASKVPGFAD DPTELACRVVDTKSGEANVRFTVSWYYRMNRRSDNVVTSELLAVMD GDWTLKYGERSKQRAQDGDFIFSKEHTDTFNFRIQORTTEEDRGNYY CVVSAWTKQRNNSWVKSKDVFSKPVNIFWALEDSVLVVKARQPKPF FAAGNTFEMTCKVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYIISLD QDSVVKLENWTDASRVDGVVLEKVQEDEFRYRMYQTQVSDAGLYR CMVTAWSPVRGSLWREAATSLSNPIEIDFOTSGPIFNASVHSDTPSVI RGDLIKLFCIITVEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLD RKGIVTTSRRDWKSDLSLERVSVLEFLLQOVHGSEDQDFGNYYCSVTP WVKSPTGSWQKEAEIHSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGL
LSCLIGYCSSHWCCKKEVQETRRERRRLMSMEMD SEQ ID NO: 9 PTGFRN IFN mouse range monomer MGRLASRPLLLALLSLALCRGRHGTVIESLESLNNYFNSSGIDVEEKS LFLDIWRNWQKDGDMKILQSQIISFYLRLFEVLKDNQAISNNISVIESHL ITTFFSNSKAKKDAFMSIAKFEVNNPQVQRQAFNELIRVVHQLLPESS LRSAGGGGSGGGGSRVVRVPTATLVRVVGTELVIPCNVSDYDGPSE QNFDWSFSSLGSSFVELASTWEVGFPAQLYQERLQORGEILLRRTAN DAVELHIKNVQPSDQGHYKCSTPSTDATVAQGNYEDTVQVKVLADSL HVGPSARPPPSLSLREGEPFELRCTAASASPLHTHLALLWEVHRGPA RRSVLALTHEGRFHPGLGYEQRYHSGDVRLDTVGSDAYRLSVSRAL SADQGSYRCIVSEWIAE QGNWQEIQEKAVEVATVVIQPSVLRAAVPK NVSVAEGKELDLTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVLA RLDRDSLVHSSPHVALSHVDARSYHLLVRDVSKENSGYYYCHVSLW APGHNRSWHKVAEAVSSPAGVGVTWLEPDYQVYLNASKVPGFADD PTELACRVVDTKSGEANVRFTVSWYYRMNRRSDNVVTSELLAVMDG DWTLKYGERSKQRAQDGDFIFSKEHTDTFNFRIQRTTEEDRGNYYC VVSAWTKQRNNSWVKSKDVFSKPVNIFWALEDSVLVVKARQPKPFF AAGNTFEMTCKVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYIISLD QDSVVKLENWTDASRVDGVVLEKVQEDEFRYRMYQTQVSDAGLYR CMVTAWSPVRGSLWREAATSLSNPIEIDFQTSGPIFNASVHSDTPSVI RGDLIKLFCIITVEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLD RKGIVTTSRRDWKSDLSLERVSVLEFLLQVHGSEDQDFGNYYCSVTP WVKSPTGSWQKEAEIHSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGL
LSCLIGYCSSHWCCKKEVQETRRERRRLMSMEMD SEQ ID NO: 10 PTGFRN IFN gamma mouse dimer MGRLASRPLLLALLSLALCRGRHGTVIESLESLNNYFNSSGIDVEEKS LFLDIWRNWQKDGDMKILQASQIISFYLRLFEVLKDNQAISNNISVIESHL ITTFFSNSKAKKDAFMSIAKFEVNNPQVQRQAFNELIRVVHQLLPESS LRGSGGSGGSGGSGHGTVIESLESLNNYFNSSGIDVEEKSLFLDIWR NWOQKDGDMKILQSQIISFYLRLFEVLKDNQAISNNISVIESHLITTFFSN SKAKKDAFMSIAKFEVNNPQVQRQAFNELIRVVHQLLPESSLRSAGG GGSGGGGSRVVRVPTATLVRVVGTELVIPCNVSDYDGPSEQNFDWS FSSLGSSFVELASTWEVGFPAQLYQERLQRGEILLRRTANDAVELHIK NVAQPSDAQGHYKCSTPSTDATVAGNYEDTVQVKVLADSLHVGPSARP PPSLSLREGEPFELRCTAASASPLHTHLALLWEVHRGPARRSVLALT HEGRFHPGLGYEQRYHSGDVRLDTVGSDAYRLSVSRALSADQGSY RCIVSEWIAEQGNWQEIQEKAVEVATVVIQPSVLRAAVPKNVSVAEG KELDLTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVLARLDRDSL VHSSPHVALSHVDARSYHLLVRDVSKENSGYYYCHVSLWAPGHNRS WHKVAEAVSSPAGVGVTWLEPDYQVYLNASKVPGFADDPTELACRV VDTKSGEANVRFTVSWYYRMNRRSDNVVTSELLAVMDGDWTLKYG ERSKQRAQDGDFIFSKEHTDTFNFRIQRTTEEDRGNYYCVVSAWTK QRNNSWVKSKDVFSKPVNIFWALEDSVLVVKARQPKPFFAAGNTFE MTCKVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYIISLDQDSVVKLE NWTDASRVDGVVLEKVQEDEFRYRMYQTQVSDAGLYRCMVTAWSP VRGSLWREAATSLSNPIEIDFQTSGPIFNASVHSDTPSVIRGDLIKLFCI ITVEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLDRKGIVTTSR RDWKSDLSLERVSVLEFLLQVHGSEDQDFGNYYCSVTPWVKSPTGS WQKEAEIHSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGLLSCLIGYCS
SHWCCKKEVQETRRERRRLMSMEMD SEQ ID NO: 11 I1L-15 441 MAPRRARGCRTLGLPALLLLLLLRPPATRGITCPPPMSVEHADIWVKS YSLYSRERYICNSGFKRKAGTSSLTECVLNKATNVAHWTTPSLKCIRD PALVHQRPAPPSTVTTAGVTPQPESLSPSGKEPAASSPSSNNTAATT AAIVPGSQLMPSKSPSTGTTEISSHESSHGTPSQTTAKNWELTASAS HQPPGVYPQGHSDTTGGSGEGGSGGEGSCGSGESGESGSGGSNWVN VISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQVISLES GDASIHDTVENLIILANNSLSSNGNVTESGCKECEELEEKNIKEFLOSF VHIVQMFINTSSADYKDDDDKFEGGGGSGGGGSAVGQDTQEVIVVYP
HSLPFKVVVISAILALVVLTIISLIILIMAINWQKKPRSGLLTGRT SEQ ID NO: 12 I1L-15 442 MAPRRARGCRTLGLPALLLLLLLRPPATRGHHHHHHITCPPPMSVEH ADIWVKSYSLYSRERYICNSGFKRKAGTSSLTECVLNKATNVAHWTT PSLKCIRDPALVHQORPAPPSTVTTAGVTPQPESLSPSGKEPAASSPS SNNTAATTAAIVPGSQLMPSKSPSTGTTEISSHESSHGTPSQTTAKN WELTASASHQPPGVYPQGHSDTTGGSGGGSGGGGSTLDPRSFLLR NPNDKYEPFWEDEEKNESGGGGSGGGSGGSNWVNVISDLKKIEDLI QSMHIDATLYTESDVHPSCKVTAMKCFLLELQVISLESGDASIHDTVE NLIILANNSLSSNGNVTESGCKECEELEEKNIKEFLQSFVHIVQMFINT SSADYKDDDDKFEGGGGSGGGGSAVGQDTQEVIVVPHSLPFKVVVI
SAILALVVLTIISLIILIMANWOQKKPRSGLLTGRT SEQ ID NO: 13 | L-15 443 METDTLLLWVLLLWVPGSTGNWVNVISDLKKIEDLIQSMHIDATLYTE SDVHPSCKVTAMKCFLLELQVISLESGDASIHDTVENLIILANNSLSSN GNVTESGCKECEELEEKNIKEFLOSFVHIVQMFINTSGGSGGGSCGG GSGGGGSGGGSGGSITCPPPMSVEHADIWVKSYSLYSRERYICNSG FKRKAGTSSLTECVLNKATNVAHWTTPSLKCIRDPALVHQRPAPPST VTTAGVTPQPESLSPSGKEPAASSPSSNNTAATTAAIVPGSQLMPSK SPSTGTTEISSHESSHGTPSQTTAKNWELTASASHQPPGVYPQGHS DTTSADYKDDDDKFEGGGGSGGGGSAVGQDTQEVIVVPHSLPFKVV VISAILALVVLTIISLIILIMLNWQKKPRSGLLTGRT
SEQ ID NO: 14 IL-15 444 METDTLLLWVLLLWVPGSTGNWVNVISDLKKIEDLIQSMHIDATLYTE SDVHPSCKVTAMKCFLLELQVISLESGDASIHDTVENLIILANNSLSSN GNVTESGCKECEELEEKNIKEFLQOSFVHIVQMFINTSDYKDDDDKGG SGGGSGGGGSTLDPRSFLLRNPNDKYEPFWEDEEKNESGGGGSG GGSGGSITCPPPMSVEHADIWVKSYSLYSRERYICNSGFKRKAGTSS LTECVLNKATNVAHWTTPSLKCIRDPALVHQRPAPPSTVTTAGVTPQ PESLSPSGKEPAASSPSSNNTAATTAAIVPGSQLMPSKSPSTGTTEIS SHESSHGTPSQTTAKNWELTASASHQPPGVYPQGHSDTTSAFEGG GGSGGGGSAVGQDTQAEVIVVPHSLPFKVVVISAILALVVLTIISLIILIML
WQKKPRSGLLTGRTHHHHHH SEQ ID NO: 15 IL-15 1009 METDTLLLWVLLLWVPGSTGNWVNVISDLKKIEDLIQSMHIDATLYTE SDVHPSCKVTAMKCFLLELQVISLESGDASIHDTVENLIILANNSLSSN GNVTESGCKECEELEEKNIKEFLQOSFVHIVQMFINTSGGSSGSGSGS TGTSSSGTGTSAGTTGTSASTSGSGSGGGGGSGGGGSAGGTATAG ASSGSITCPPPMSVEHADIWVKSYSLYSRERYICNSGFKRKAGTSSLT ECVLNKATNVAHWTTPSLKCIRDPALVHQRPAPPSTVTTAGVTPQPE SLSPSGKEPAASSPSSNNTAATTAAIVPGSQLMPSKSPSTGTTEISSH ESSHGTPSQTTAKNWELTASASHQPPGVYPQGHSDTTSAGGGGSG GGGSRVVRVPTATLVRVVGTELVIPCNVSDYDGPSEQNFDWSFSSL GSSFVELASTWEVGFPAQLYQERLQRGEILLRRTANDAVELHIKNVQ PSDQGHYKCSTPSTDATVAGNYEDTVQVKVLADSLHVGPSARPPPS LSLREGEPFELRCTAASASPLHTHLALLWEVHRGPARRSVLALTHEG RFHPGLGYEQRYHSGDVRLDTVGSDAYRLSVSRALSADQGSYRCIV SEWIAEQGNWQEIQEKAVEVATVVIQPSVLRAAVPKNVSVAEGKELD LTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVLARLDRDSLVHSS PHVALSHVDARSYHLLVRDVSKENSGYYYCHVSLWAPGHNRSWHK VAEAVSSPAGVGVTWLEPDYQVYLNASKVPGFADDPTELACRVVDT KSGEANVRFTVSWYYRMNRRSDNVVTSELLAVMDGDWTLKYGERS KQRAQDGDFIFSKEHTDTFNFRIQRTTEEDRGNYYCVVSAWTKQRN NSWVKSKDVFSKPVNIFWALEDSVLVVKARQPKPFFAAGNTFEMTC KVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYIISLDQDSVVKLENW TDASRVDGVVLEKVQEDEFRYRMYQTQVSDAGLYRCMVTAWSPVR GSLWREAATSLSNPIEIDFQTSGPIFNASVHSDTPSVIRGDLIKLFCIIT VEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLDRKGIVTTSRRD WKSDLSLERVSVLEFLLQOVHGSEDQDFGNYYCSVTPWVKSPTGSW QKEAEI! HSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSH
WCCKKEVQETRRERRRLMSMEMD SEQ ID NO: 16 IL-15 1010 METDTLLLWVLLLWVPGSTGNWVNVISDLKKIEDLIQSMHIDATLYTE SDVHPSCKVTAMKCFLLELQVISLESGDASIHDTVENLIILANDSLSSN GNVTESGCKECEELEEKNIKEFLQOSFVHIVQMFINTSGGSSGSGSGS TGTSSSGTGTSAGTTGTSASTSGSGSGGGGGSCGSGGGSAGGTATAG ASSGSITCPPPMSVEHADIWVKSYSLYSRERYICNSGFKRKAGTSSLT ECVLNKATNVAHWTTPSLKCIRDPALVHORPAPPSTVTTAGVTPQPE SLSPSGKEPAASSPSSNNTAATTAAIVPGSQLMPSKSPSTGTTEISSH ESSHGTPSQTTAKNWELTASASHQPPGVYPQGHSDTTSAGGGGSG GGGSRVVRVPTATLVRVVGTELVIPCNVSDYDGPSEQNFDWSFSSL GSSFVELASTWEVGFPAQLYQERLQRGEILLRRTANDAVELHIKNVQ PSDQGHYKCSTPSTDATVAGNYEDTVQVKVLADSLHVGPSARPPPS LSLREGEPFELRCTAASASPLHTHLALLWEVHRGPARRSVLALTHEG RFHPGLGYEQRYHSGDVRLDTVGSDAYRLSVSRALSADQGSYRCIV SEWIAEQGNWQEIQEKAVEVATVVIQPSVLRAAVPKNVSVAEGKELD LTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVLARLDRDSLVHSS PHVALSHVDARSYHLLVRDVSKENSGYYYCHVSLWAPGHNRSWHK VAEAVSSPAGVGVTWLEPDYQVYLNASKVPGFADDPTELACRVVDT KSGEANVRFTVSWYYRMNRRSDNVVTSELLAVMDGDWTLKYGERS KQRAQDGDFIFSKEHTDTFNFRIQRTTEEDRGNYYCVVSAWTKQRN NSWVKSKDVFSKPVNIFWALEDSVLVVKARQPKPFFAAGNTFEMTC KVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYIISLDQDSVVKLENW TDASRVDGVVLEKVQEDEFRYRMYQTQVSDAGLYRCMVTAWSPVR GSLWREAATSLSNPIEIDFQTSGPIFNASVHSDTPSVIRGDLIKLFCIIT VEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLDRKGIVTTSRRD WKSDLSLERVSVLEFLLQVHGSEDQDFGNYYCSVTPWVKSPTGSW QKEAEIHSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSH
WCCKKEVQETRRERRRLMSMEMD SEQ ID NO: 17 pDisplay-anti-CD3 MKIICLALVALLLTAQPAMAEIVLTQOSPATLSLSPGERATLSCRASQSV SSYLAWYQQKPGQAPRLLIYDASNRATGIPARFSGSGSGTDFTLTISS LEPEDFAVYYCQQAQRSNWPPLTFGGGTKVEIKRTVAAPSVFIFPPSDE QLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSK DSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECGG SSGSGSGSTGTSSSGTGTSAGTTGTSASTSGSGSGGGGGSCSCGÇE SAGGTATAGASSGSQVQLVESGGGVVQPGRSLRLSCAASGFKFSG YGMHWVRQAPGKGLEWVAVIWYDGSKKYYVDSVKGRFTISRDNSK NTLYLQMNSLRAEDTAVYYCARQMGYWHFDLWGRGTLVTVSSAST KGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLOSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDK KVEPKSCDKTHTGGSGGGSGGGGSGGGGSGGGSGGSAVGADTA
EVIVVPHSLPFKVVVISAILALVVLTIISLIILIMIWOKKPRDYKDDDDK SEQ ID NO: 18 PTGFRN-anti-CD3 MKIICLALVALLLTAQPAMAEIVLTOSPATLSLSPGERATLSCRASQSV SSYLAWYQQKPGQAPRLLIYDASNRATGIPARFSGSGSGTDFTLTISS LEPEDFAVYYCQQRSNWPPLTFGGGTKVEIKRTVAAPSVFIFPPSDE QLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSK DSTYSLSSTLTLSKADYEKHKVYACEVTHQOGLSSPVTKSFNRGECGG SSGSGSGSTGTSSSGTGTSAGTTGTSASTSGSGSGGGGGCGSCGGGÇ SAGGTATAGASSGSQVQLVESGGGVVQPGRSLRLSCAASGFKFSG YGMHWVRQAPGKGLEWVAVIWYDGSKKYYVDSVKGRFTISRDNSK NTLYLQMNSLRAEDTAVYYCARQMGYWHFDLWGRGTLVTVSSAST KGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLOSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDK KVEPKSCDKTHTGGSGGGSGGGESGSGGESGGGESGGSRVVRVPTA TLVRVVGTELVIPCNVSDYDGPSEQNFDWSFSSLGSSFVELASTWEV GFPAQLYQERLQRGEILLRRTANDAVELHIKNVQPSDQGHYKCSTPS TDATVQGNYEDTVQVKVLADSLHVGPSARPPPSLSLREGEPFELRCT AASASPLHTHLALLWEVHRGPARRSVLALTHEGRFHPGLGYEQRYH SGDVRLDTVGSDAYRLSVSRALSADQGSYRCIVSEWIAEQGNWOQEI QEKAVEVATVVIQPSVLRAAVPKNVSVAEGKELDLTCNITTDRADDVR PEVTWSFSRMPDSTLPGSRVLARLDRDSLVHSSPHVALSHVDARSY HLLVRDVSKENSGYYYCHVSLWAPGHNRSWHKVAEAVSSPAGVGV TWLEPDYQVYLNASKVPGFADDPTELACRVVDTKSGEANVRFTVSW YYRMNRRSDNVVTSELLAVMDGDWTLKYGERSKQRAQDGDFIFSKE HTDTFNFRIQORTTEEDRGNYYCVVSAWTKQRNNSWVKSKDVFSKPV NIFWALEDSVLVVKARQPKPFFAAGNTFEMTCKVSSKNIKSPRYSVLI MAEKPVGDLSSPNETKYIISLDQDSVVKLENWTDASRVDGVVLEKVQ EDEFRYRMYQTQVSDAGLYRCMVTAWSPVRGSLWREAATSLSNPIE IDFQTSGPIFNASVHSDTPSVIRGDLIKLFCIITVEGAALDPDDMAFDVS WFAVHSFGLDKAPVLLSSLDRKGIVTTSRRDWKSDLSLERVSVLEFL LQVHGSEDQDFGNYYCSVTPWVKSPTGSWQKEAEIHSKPVFITVKM DVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSHWCCKKEVQETRRERR RLMSMEMDTGGSGGSVSKGEELFTGVVPILVELDGDVNGHKFSVSG EGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTYGVQCFSRYPDHM KQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIEL KGIDFKEDGNILGHKLEYNYNSHNVYIMADKQKNGIKVNFKIRHNIED GSVQLADHYQQNTPIGDGPVLLPDNHYLSTAQSKLSKDPNEKRDHMV
LLEFVTAAGITLGMDELYKDYKDDDDK SEQ ID NO: 19 PTGFRN CD40L mouse dimer METDTLLLWVLLLWVPGSTGMQRGDEDPQIAAHVVSEANSNAASVL QWAKKGYYTMKSNLVMLENGKQLTVKREGLYYVYTQVTFCSNREPS SQRPFIVGLWLKPSSGSERILLKAANTHSSSQLCEQQSVHLGGVFEL QAGASVFVNVTEASQVIHRVGFSSFGLLKLGSGGSGGSGGSGMAR GDEDPQIAAHVVSEANSNAASVLQWAKKGYYTMKSNLVMLENGKQL TVKREGLYYVYTQVTFCSNREPSSQRPFIVGLWLKPSSGSERILLKAA NTHSSSQLCEQQASVHLGGVFELQAGASVFVNVTEASQVIHRVGFSS FGLLKLGSGGSGGSGGSGMQRGDEDPQIAAHVVSEANSNAASVLQ WAKKGYYTMKSNLVMLENGKQLTVKREGLYYVYTQVTFCSNREPSS QRPFIVGLWLKPSSGSERILLKAANTHSSSQLCEQQSVHLGGVFELQ AGASVFVNVTEASQVIHRVGFSSFGLLKLSAGGGGSGGGGSRVVRV PTATLVRVVGTELVIPCNVSDYDGPSEQNFDWSFSSLGSSFVELAST WEVGFPAQLYQERLQRGEILLRRTANDAVELHIKNVQPSDQGHYKC STPSTDATVAQGNYEDTVQVKVLADSLHVGPSARPPPSLSLREGEPFE LRCTAASASPLHTHLALLWEVHRGPARRSVLALTHEGRFHPGLGYE QRYHSGDVRLDTVGSDAYRLSVSRALSADQGSYRCIVSEWIAEQGN WQEIQEKAVEVATVVIQPSVLRAAVPKNVSVAEGKELDLTCNITTDRA DDVRPEVTWSFSRMPDSTLPGSRVLARLDRDSLVHSSPHVALSHVD ARSYHLLVRDVSKENSGYYYCHVSLWAPGHNRSWHKVAEAVSSPA GVGVTWLEPDYQVYLNASKVPGFADDPTELACRVVDTKSGEANVRF TVSWYYRMNRRSDNVVTSELLAVMDGDWTLKYGERSKQRAQDGDF IFSKEHTDTFNFRIQRTTEEDRGNYYCVVSAWTKQRNNSWVKSKDV FSKPVNIFWALEDSVLVVKARQPKPFFAAGNTFEMTCKVSSKNIKSP RYSVLIMAEKPVGDLSSPNETKYIISLDQDSVVKLENWTDASRVDGVV LEKVQEDEFRYRMYQTQVSDAGLYRCMVTAWSPVRGSLWREAATS LSNPIEIDFQTSGPIFNASVHSDTPSVIRGDLIKLFCIITVEGAALDPDD MAFDVSWFAVHSFGLDKAPVLLSSLDRKGIVTTSRRDWKSDLSLERV SVLEFLLQOVHGSEDQDFGNYYCSVTPWVKSPTGSWQKEAEIHSKPV FITVKMDVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSHWCCKKEVQET
RRERRRLMSMEMD SEQ ID NO: 20 PTGFRN CD40L human trimer METDTLLLWVLLLWVPGSTGMQKGDQNPQIAAHVISEASSKTTSVLQ WAEKGYYTMSNNLVTLENGKQLTVKRQGLYYIYAQVTFCSNREASS QAPFIASLCLKSPGRFERILLRAANTHSSAKPCGQQSIHLGGVFELQP GASVFVNVTDPSQVSHGTGFTSFGLLKLGSGGSGGSGCECSGMQAKGD QNPQIAAHVISEASSKTTSVLQWAEKGYYTMSNNLVTLENGKQLTVK RQGLYYIYAQVTFCESNREASSQAPFIASLCLKSPGRFERILLRAANTH SSAKPCGQQSIHLGGVFELQPGASVFVNVTDPSQVSHGTGFTSFGLL KLGSGGSGESGGS GMQKGDQNPQIAAHVISEASSKTTSVLQWAEK GYYTMSNNLVTLENGKQLTVKRQGLYYIYAQVTFCSNREASSQAPFI ASLCLKSPGRFERILLRAANTHSSAKPCGQQSIHLGGVFELQPGASV FVNVTDPSQVSHGTGFTSFGLLKLSAGGGGSGGGGSRVVRVPTATL VRVVGTELVIPCNVSDYDGPSEQNFDWSFSSLGSSFVELASTWEVG FPAQLYQERLQRGEILLRRTANDAVELHIKNVQPSDQGHYKCSTPST DATVAQGNYEDTVQVKVLADSLHVGPSARPPPSLSLREGEPFELRCT AASASPLHTHLALLWEVHRGPARRSVLALTHEGRFHPGLGYEQRYH SGDVRLDTVGSDAYRLSVSRALSADQGSYRCIVSEWIAEQGNWQEI QEKAVEVATVVIQPSVLRAAVPKNVSVAEGKELDLTCNITTDRADDVR PEVTWSFSRMPDSTLPGSRVLARLDRDSLVHSSPHVALSHVDARSY HLLVRDVSKENSGYYYCHVSLWAPGHNRSWHKVAEAVSSPAGVGV TWLEPDYQVYLNASKVPGFADDPTELACRVVDTKSGEANVRFTVSW YYRMNRRSDNVVTSELLAVMDGDWTLKYGERSKQRAQDGDFIFSKE HTDTFNFRIQORTTEEDRGNYYCVVSAWTKQRNNSWVKSKDVFSKPV NIFWALEDSVLVVKARQPKPFFAAGNTFEMTCKVSSKNIKSPRYSVLI MAEKPVGDLSSPNETKYIISLDQDSVVKLENWTDASRVDGVVLEKVQ EDEFRYRMYQTQVSDAGLYRCMVTAWSPVRGSLWREAATSLSNPIE IDFQTSGPIFNASVHSDTPSVIRGDLIKLFCIITVEGAALDPDDMAFDVS WFAVHSFGLDKAPVLLSSLDRKGIVTTSRRDWKSDLSLERVSVLEFL LQVHGSEDQDFGNYYCSVTPWVKSPTGSWQKEAEIHSKPVFITVKM DVLNAFKYPLLIGVGLSTVIGLLSCLIGYCSSHWCCKKEVQETRRERR RLMSMEMD
SEQ ID NO: 21 PTGFRN short-anti-CD3 MKIICLALVALLLTAQPAMAEIVLTQOSPATLSLSPGERATLSCRASQSV SSYLAWYQQKPGQAPRLLIYDASNRATGIPARFSGSGSGTDFTLTISS LEPEDFAVYYCQQRSNWPPLTFGGGTKVEIKRTVAAPSVFIFPPSDE QLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSK DSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECGG SSGSGSGSTGTSSSGTGTSAGTTGTSASTSGSGSCGGGGSCGGGÇ SAGGTATAGASSGSQVQLVESGGGVVQPGRSLRLSCAASGFKFSG YGMHWVRQAPGKGLEWVAVIWYDGSKKYYVDSVKGRFTISRDNSK NTLYLQMNSLRAEDTAVYYCARQMGYWHFDLWGRGTLVTVSSAST KGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG VHTFPAVLOSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDK KVEPKSCDKTHTGGSGGGSGEGGGSGEGESGGGSGGSGPIFNASV HSDTPSVIRGDLIKLFCIITVEGAALDPDDMAFDVSWFAVHSFGLDKA PVLLSSLDRKGIVTTSRRDWKSDLSLERVSVLEFLLQOVHGSEDQDFG NYYCSVTPWVKSPTGSWQKEAEIHSKPVFITVKMDVLNAFKYPLLIGV GLSTVIGLLSCLIGYCSSHWCCKKEVQETRRERRRLMSMEMDTGGS GGSVSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTL KFICTTGKLPVPWPTLVTTLTYGVQCFSRYPDHMKQHDFFKSAMPEG YVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGH KLEYNYNSHNVYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNT PIGDGPVLLPDNHYLSTAOSKLSKDPNEKRDHMVLLEFVTAAGITLGM
DELYKDYKDDDDK SEQ ID NO: 22 FLT3L-PTGFRN MTVLAPAWSPTTYLLLLLLLSSGLSGTQDCSFQHSPISSDFAVKIREL SDYLLQDYPVTVASNLQDEELCGGLWRLVLAQRWMERLKTVAGSK MQGLLERVNTEIHFVTKCAFQPPPSCLRFVQTNISRLLQETSEQLVAL KPWITRQNFSRCLELQCQPDSSTLPPPWSPRPLEATAPTAPQPPSA GGGGSCGGGGSRVVRVPTATLVRVVGTELVIPCNVSDYDGPSEQNFD WSFSSLGSSFVELASTWEVGFPAQLYQERLQRGEI / LLRRTANDAVEL HIKNVQPSDQGHYKCSTPSTDATVQGNYEDTVQVKVLADSLHVGPS ARPPPSLSLREGEPFELRCTAASASPLHTHLALLWEVHRGPARRSVL ALTHEGRFHPGLGYEQRYHSGDVRLDTVGSDAYRLSVSRALSADOQG SYRCIVSEWIAE QGNWQEIQEKAVEVATVVIQPSVLRAAVPKNVSVA EGKELDLTCNITTDRADDVRPEVTWSFSRMPDSTLPGSRVLARLDRD SLVHSSPHVALSHVDARSYHLLVRDVSKENSGYYYCHVSLWAPGHN RSWHKVAEAVSSPAGVGVTWLEPDYQVYLNASKVPGFADDPTELAC RVVDTKSGEANVRFTVSWYYRMNRRSDNVVTSELLAVMDGDWTLK YGERSKQRAQDGDFIFSKEHTDTFNFRIQRTTEEDRGNYYCVVSAW TKQRNNSWVKSKDVFSKPVNIFWALEDSVLVVKARQPKPFFAAGNT FEMTCKVSSKNIKSPRYSVLIMAEKPVGDLSSPNETKYIISLDQDSVVK LENWTDASRVDGVVLEKVQEDEFRYRMYQTQVSDAGLYRCMVTAW SPVRGSLWREAATSLSNPIEIDFQOTSGPIFNASVHSDTPSVIRGDLIKL FCIITVEGAALDPDDMAFDVSWFAVHSFGLDKAPVLLSSLDRKGIVTT SRRDWKSDLSLERVSVLEFLLQOVHGSEDQDFGNYYCSVTPWVKSPT GSWQKEAEIHSKPVFITVKMDVLNAFKYPLLIGVGLSTVIGLLSCLIGY CSSHWCCKKEVQETRRERRRLMSMEMD TABLES
C nsPeoasaP [aPovIo RAE
PTPRo - [acres - [eme - [MP UBE2V1 emo Neo oem
Cao fes cloth
PEBRT [BA [were —sRaN
[ram - | maRvELDE | stecALNACS |
Not EE are - [Promo er rem reta
CPA women BT
AGE [PTANS | roca Rover AA [fan vamee PTARO | fe eu - rocraaso TMB
EE - MARAGPT fom = [tail - es war TnFRSAZA
Fibronectin repeat proteins | Ankyrin Liases Antibodies Polypeptide Receptors Complementary Bodies to GP! Aptamers Cyclic peptides | HEAT repeat Nucleic Acids proteins cos ARM repeat DARPINS Hydrolases Polypeptides proteins carbohydrates | DNAses Kinase Single-chain variable fragments (scFv) Receptors from | Enzymes Lipoproteins Proteins with cellulite re- surface surface tricopeptide C1 inhibitor | Alloy protein | CR3 Factor | C4 CD59 receptor CR4 C3 beta chain restriction factor homologous C3aR CR1 Acceleration factor | Memory deficiency heart protein (DAF) | brana (MCP) | Triacylglycerol enzymes | li- | bile acid-CoA feruloyl esterase | phosphatidate phospase hydrolase fatase (S) -methylmalonyl- | bis (2-ethylhexyl) | formalin-CoA hi- | phosphatidylglycerol- CoA Hydrolase | phthalate esterase | phosphatase drolase [carrier protein | fructose phosphatase- phosphatidylinositol adores bisphosphoglycer- | bisphosphatase deacylase acillfosfodiestera | I'm se
[hosphorylase] fos- | carboxy ester | fumarylacetoa- | phosphodiesterase fatase lico hydrolases - ketase | 1,4-lactonase carboxymethyl- - fusarinin-C or- | phosphoglycerate nobutenolidase | nitinesterase phosphatase 11-cis-retinyl-cellulose-galactolipase phosphoglycolate palmitate Hydro- | polysulfatase phosphatase lase 1-alkyl-2-cephalosporin-C | gluconolacto- phosphoinositide acetylglycerophos- | deacetylase nase phospholipase C focololine 2'-hydroxybiphenyl- esterase | cerebroside-glucose-1- Phospholipase A1 2-sulfatase sulfatase phosphatase desulfinase 2- Lactonase | benzi- cetraxate | glucose-6-phospholipase A2 pyrone-4,6-lesterase phosphatase dicarboxylate Nucleotidase de | hydrogenase from | thioles- glutathione | phospholipase C 3 ', 5'-bisphosphate chlorogenate terase Hydrolase 3- | chlorophyllase glycerol-1-phospholipase D hydroxyphosphatase isobutyliri-CoA 3'-nucleotidase | Cholinesterase - glycerol-2-phosphatase phosphonacetaldehyde 3-oxoadipate choline sulfatase hydrolase | glycerophosfocolo- | enol-lactonase lina phosphodieste- hydrolase | phosphonoacetate rase 3 co-phytase hydrolase | Glycosidases, loil-CoA hydrolase i.e. enzymes that | phosphonopyruvate hydrolyzes O and S-glycosyl compounds
Thioesterase from | chondro-4-glycosulfatase phosphoprotein 4-hydroxybenzoyl- | sulfatase phosphatase CoA Esterase 4- | chondro-6- Glycosylases Methyloxaloacet hydrolases | phosphoric diester sulfatase to 4-phytase citrate lyase | histidinol- Sacetylase phosphatase hydrolases phosphoric monoester 4- co- esterase | sensitive lipase | Pyridoxolactacine hydrolases to phosphorine triester hormones cas B'-nucleotidase | cutinase Hydrolyzing phosphoserine phosphorus compounds | N-glycosyl deacetylase fatase from | sulfo- cyclamate | Hydrolyzing depolymerase 6-acetylglucose | hydrolase compounds from | poly (3-N-glycosyl hydroxybutyrate) 6- Endopeptidases, hydroxyacylglut- | depolymerase Phosphoglycololac- | of cysteine thione hydrolase | poly (3-hydroxyoctanoate to) esterase a- Carboxypeptide hydrolase of po- amino acid doses of the type of hylineuridine dimer- cysteine droxybutyrate aldehyde hydrolases of a- | D- hydroxymethylglu- | amino-acyl-arabinonolacto- methylesterase | hydro-CoA taryl | protein-peptide nasase lase glutamate acetoacetyl-CoA | ring A lacto-iduronate-2- N-acylhydrolase nase deoxy- | homoserine sulphatase limonate lactonase which extinguishes the acetoxybutynylbi- quorum | dGTPase phosphatase of re-thiophene esterase deacetininositol-phosphate | tinyl-palmitate lase esterase aceti- | Serine dehydro-lajmaline hydrocoumarin - juvenile hormone diesterase tase or serine hydrolase hydroxymethyl transferase acetylalkylglice- | Quinureninase dipeptidases | Endopeptide-acetylsesine serines hydrolase Acetylcholinester- | Hy dipeptide | L- serine rase drolases arabinonolacto- | ethanolamine-nase phosphate phosphodiesterase acetyl-CoA hydro- | Dipeptidyl-limonine-D-ring | Carboxypeptylase peptidases and lactonase doses of the tripeptidyl serine peptidases type Acetylesterase Hydrolases di- | lipo- S- phosphoric- lipase protein formylglutathione monoester hydrolase disulfogluco- hydrolase | L-ramnono-1,4- | O- acetylpyruvate samine-6-lactonase acetylesterase sialate acetylsalicylate dodecanoyl-lysophospholipase | si-deacetylase esterase [carcinapine protein adores acyl] estyl acetylxylan hydrolase | Endodesoxirri- | mannitol-1 endodesoxirrase bonucleases phosphatase bonucleases producing specific 3 "- site: cleavage phosphomonoesthesia is not specific in terms of sequence
Acid phosphatase | Endodesoxirri- | Metalocarboxi- | Endodesoxyribonucleases peptidases bonucleases producing specific 5-site, specific phosphomonoesthesia for altered res bases.
Acting on Endopeptidases, Metaloendopep- | Endodesoxyrri-acid anhydrides | of mechanism | tidases. bonucleases to catalyze the | catalytic nonspecific movement known site: the cleavage-transmembrane gem is specific for substances as to the sequence Acting on Endoribonu- methylphosphothiogli- | sphingomyelin acid anhydrides | product cleases | phosphate kerate | phosphodiesterase to facilitate 3'- cell movement logs | phosphomonoesteral and subcellular | res Acting on Endoribonu- metilumbeliferi- | S- GTP to facilitate | product cleases | lacetate desace- | succinylglutathione movement | 5'-tylase logs in cell hydrolase and sub- | phosphomono-stellar res Performance in the li- | Endoribonu- monoterpene e- | steroid- phosphorus | active cleases | lactone hydro-lactonase nitrogen with ri- acids | lase bo- or deoxyron-ribonucleic and produce 3'-phosphomonoesters
Performance in | i- Endoribonu- N- esterase sulfur stagnations- | acetylgalactol-nitrogen cleases are active with | samina-4-
ribo- or | deoxyribonium sulfatase
cleic and pro-
lead 5'-
phosphomonoester
res actinomycin lac- | Enzymes that N- sterile sulfatase tonase act on acetylgalacto-
anhydrousamine-6- acids
of Enzymes Atu-Deacetylase Sulfatase Hydrolase | succinyl-CoA acylcarnitine and carbon-acetylgalacto- N- hydrolase bonds
carbon acin- saminoglycan hydrolase | Enzymes that N- Sucrose- CoA act on alloys | acetylglucosa- phosphate-
carbon- | mine-6- nitrogen tase, ex- | keto sulfatase in the
acylgli- lipase peptide tions | Enzymes that N-phosphatase of cerol act on alloys - sulfoglucosami- | carbon- sugar tions | at sulfohydro-
hydroxyl acyloxyacryl phosphorus | Enzymes that oleoil- [rotein | Lase hydrolases act on alloys | carrier of | sulfuric ester carbon-acyl tions] sulfur hydrolase Enzymes atuan- | Peptidases tannyl acylpyruvate in bonds | ether omega
ADAMTS13 Enzymes that orselinate- Hydrolases act on alloys- | hydro depside | thioester halolase genotypes Adenosine de- Enzymes that oxaloacetase Saminase hydrolases act on adenylyl- [gluta- | Enzymes that palmitoil [rotein | Endopeptide- bush - ammonia act in the alloys | na] threonine ligase hydrolase] hydrolase | phosphorus-nitrogen Ca- hydrolase | Palmitoyl-CoA thymidine enzymes medium-acyl- | acting in li- | CoA-dependent lase hydrolase | sulfur-additions of ADP nitrogen Ca- hydrolase | Enzymes that pectinesterase | trehalose- short-acyl- | act on CoA-dependent phosphatase | sulfur-additions of ADP sulfur-ADP-ether hydrolases. | Peptidyl peptide-triacetate-phosphoglycerate deo hydrolases | lactonase phosphatase Alkali phosphatase | Exodesoxirribo- | Peptidyl- Hydrolases tri-nucleases pro- | hy- amino acid | phosphoric-producing 5 | to- monoester phosphomonoester hydrolase drolases | Exonucleases Peptidyl- Trithionate hydro- | of those who are active | hy- amino acid | retinyl palmitate lase | with acids | drolases or acci-
ribo- or desolaminino acid
xyribonucleic | drolases and produce 3 '; -
phosphomonoester
aminoacyl-RNA | Peptidyl tropinesterase hydrolase exonucleases that are active | dipeptidases with rich acids
bo- or deoxyrize-
ribonucleic and produce 5 '; -
phosphomonoester
res Aminopeptida- | Exorribonuclea- | phenylacetyl-CoA | Ubiquitin unions producing companies | 3 'lesterase hydrolase; -
phosphomonoester
res arylesterase Exorribonuclea-, Phenylalanine UDP-
producing countries | ammonia lyase sulfoquinovose 5 '; - phosphomonoester synthase
res. arylsulfatase Factor IX Phenylalanine Hi- | uricase droxylase (PAH) Endopeptidases | ester-ethyl-acyl- | hydro floretine | wax aspartic fatty acid synthase lase hydrolase b-diketone hydrolase phorbol-diester xylono-1,4-lactonase hydrolase
Virus Cells Cell debris
Lipopolissaca- | Invasion protein | Intermedylisin | Protein effetides are cellular secreted sptP Toxic zone | Protein enterotoxin in- | Seeligeriolisina na occludens | cholera vasospasm sipA cysteine protease protein | Component of | Serine protease polymerization | toxin ota lotin la actin RickA Toxin protein distender | Ivanolisina Toxina Shiga polymerization | actin citoletal RickA Adenosine Cytolysin LepB Sphingomyeli- monophosphate-nase protein transferase vopS adenylate- Necrotizing factor Lethal factor Staphylocinase cytotoxic cyclase Adenylate cy- | Cytotoxin Leukotoxin Streptokylase ExoY nase Component | Dermone- toxin | Listeriolysin Streptolysin enzyme from | cryptic ADP- ribosyltransferase
Aerolysin Deubiquitinase Collagenase Microbial streptopain Alpha toxin Diphtheria toxin | Self-transport | External membrane protein suilisin IlcsA Enterohemolysin Alveolysin | Leukocidin F | Superantigen of Panton- Valentine Alveolysin Enterotoxin Perfringolysin | Secretory effector of T3SS EspF Anthrolysin O | Co-toxin inhibitor | Tetanus toxin cell resonance | epidermal queluche Exoenzyme protein phospholipase Tir activation of the Arp2 / 3 rickA complex Binary toxin | Exotoxin ADC TolC Activator - plasminogen ribosyltransferase CDT Neurotoxin | change factor | Pneumolysin | Toxin of syn- botulinum nucleotide G toxic shock syndrome Toxin C2, Exchange factor | Protein antigen | Carboxypepti- component Il | nucleotides of guanine zinc oxide sopE CagA Enterotoxin is- | protein kinase | Carboxypeptible in heat zinc adenylate | Autotransporter | Pyrolysin Peptidase declase sensitive | of endo- dependent serine from Zn to calmodulin | IgA-specific peptidase
Inhibitory factor | inosi- phosphatase | Cell cycle RTX toxin | tol-phosphate lar Tumo- cells | densi- lipid | Triglycerides | Circulating fatty acids | very low test (VLDL) Metastases Al-- chylomicron lipoprotein Cholesterol at density Euca- cells | low density apolipoprotein lipoprotein leukemia lin- | Colorectal cancer | Macroglobuline- | Pleu- foblastic mias, Waldenstrôm, ropulmonary blastoma in acute (ALL) childhood Leukemia Craniopharyngioma | breast cancer | Pregnancy and acute myeloid | in childhood Male breast cancer (AML) cancer Carcinoma cutaneous lymphoma | Histiocytoma- Adrenocortical lymphoma | malignant t-cell analysis | Ner- bone and osteosar- system | Central coma (SNC) Sarcoma de | Duc- carcinoma | Kaposi's cancer-related melanoma | tal / n Situ (DCIS) prostate linked to AIDS Lymphoma related | Embry Tumors | Carcinoma of the brain | Cancer caused by cancer in childhood | Merkel ros squids
AIDS trial
Ependymoma cancer, In- | Neck cancer | squamous appendix carcinoma me- | tastatic renal cell with hidden primary Astrocytomas | Epithelial cancer | Trauma carcinoma | Renal pelvis and midline childhood | ureter, cancer involving the ge- | teratoi- tumor transitional NUT cells | esophageal cancer | Retinoblastoma molar pregnancy of atypical / rhabdoid go in childhood carcinoma of | Estesioneu- Cancer of the mouth and | Rhabdomyos- basal cell roblastoma, In- oropharynx sarcoma childhood Cancer of the du- | Sarcoma of Cancer Syndromes of Biliary Ewing Neoplasm Endó- | Multiple salivary gland in | var Childhood cancer | Extrago- tumor | Multiple myeloma Plasma cell xarcomal sarcoma / germ cell neoplasia plasma cells Cancer in Extra-fungal mycosis cancer | Bile duct secondary hepatic bone cancers Inte- cancer | Eye cancer | Myelo- Syndromes | Dysplastic tinal syndrome Sézary Glioma da Vesí cancer | Neoplasms (Skin cancer) Stem Elodisplastic gallbladder in childhood / myeloproliferation Tumors ce- | gastric cancer | Myelo- disorders | Proliferative renal skin cancer (non-melano- chronic Ch)
Carcinoid Tumor Cancer | Cavi- cancer | Gastrointesti- breast cancer | nasal and sinus disease | Paranasal Lung Cancer Small squid Tumors Cell tumor | nasopharyngeal cancer | Cancer of In- Bronchial na) germinative geo testino Delga- Leukemia childhood | Tropho-neuroblastoma Burkitt's sarcoma blast gestational soft tissue (GTT) Glioma cancer Non-Ho- lymphoma | Primary des-dakin cell carcinoma Esc- known mosquitoes Cancer es- | Brain leukemia | lung cancer, Cancer has spread to non-skin cell hairy squids | of the people- BONES that itch with a hidden primary, metastatic Cancer is es | head cancer | Oesophageal cancer | Cancer spread to the | stomach and neck (gas-brain) Cancer is es- | Cora- Cancer | oral cancer Cancer spread to the | Childhood, liver, liver Cancer is es- | Hepato- cancer | Cavi- cancer | Cutaneous lymphoma for cell (Liver) | oral neoplasm of T cells - see Mycosis) fungus and Sézary Carci- syndrome | Histiocytosis, sky | Oropa- cancer | Lange-ringe rhans tula squid cancer
Carcinoma of | Hod- lymphoma | Osteosarcoma | Primary Des- Cancer | gkin (bone cancer) throat known Tumors car- | hypopharyngeal cancer | Osteosarcoma and | Thymoma and deacons (heart | fibro- histiocytoma | Carcinoma tion) in malignant childhood Tímico cia Tumor teratoi- | Intra- melanoma | ovarian cancer | thyroid cancer / ocular rhabdoid atypical oide of the central nervous system in childhood Tumors Em- | Cell tumors, Pancreatic Cancer | Cancer of islet lasers, typical of trans- Ner- System | neuronal tumors of the central pelvis, | endocrine endocrine and urea- Childhood cremas having ner- System | kidney cancer Neuro- tumors | Pan endocrine central primitive cancer | unknown rivers (tumor cells | islets) cervical cancer | Histiocytosis of | Papillomatosis in | Lan- ureter and pelvis lime cells | Renal childhood, cancer germs of transitional cells Chordoma, In- | laryngeal cancer, Paraganglioma Choriocarcino- urethral cancer | Leukemia Cancer of Parati- | Endometrial uterine mareoid cancer lin- leukemia | Lip cancer | Penile cancer | Chronic uteri-focitic sarcoma | and oral cavity in ca (CLL)
Leukemia Liver cancer | Pharynx cancer | Chronic myeloid vaginal cancer (CML) Mi- disorders | Lobu- carcinoma |! Pheochromocytoma | Vulvar cancer eloproliferati- | chronic in situ home (LCIS) colon cancer | Potential tumor | Pituitary tumor] Malignant macroglobulitis low Wal-denstróm lymphoma Pulmonary cancer- Neoplasm of the brain | Plasmid squid hand tumor- Wilms; cas / multiple myeloma
权利要求:
Claims (83)
[1]
1. Composition, characterized by the fact that it comprises: an extracellular vesicle comprising a cell membrane that connects a closed volume, said cell membrane having an internal surface and an external surface; and a first immunomodulatory component associated with said cell membrane or closed within said closed volume.
[2]
2. Composition according to claim 1, characterized by the fact that said first immunomodulatory component is an inhibitor for a negative checkpoint regulator or an inhibitor for a binding partner of a negative checkpoint regulator verification.
[3]
3. Composition, according to claim 2, characterized by the fact that the negative checkpoint regulator is selected from the group consisting of: protein 4 associated with cytotoxic T lymphocytes (CTLA-4), protein 1 programmed cell death (PD-1), lymphocyte-activated gene 3 (LAG-3), protein 3 containing T cell immunoglobulin mucin (TIM-3), B and T lymphocyte attenuator (BTLA), T cell immunoreceptor with lg and ITIM domains (TI-GIT), Ig suppressor of T cell activation domain V (VISTA), A2a adenosine receptor (A2aR), killer cell immunoglobulin-like receptor (KIR), indoleamine 2, 3-dioxigenase (IDO), CD20, CD39 and CD73.
[4]
4. Composition according to claim 1, characterized by the fact that said first immunomodulatory component is an activator for a positive co-stimulating molecule or an activator for a binding partner of a positive co-stimulating molecule.
[5]
5. Composition according to claim 4, characterized by the fact that said positive co-stimulating molecule is a member of the TNF receptor superfamily.
[6]
6. Composition, according to claim 5, characterized by the fact that said member of the TNF receptor superfamily is selected from the group consisting of: CD120a, CD120b, CD18, OX40, CD40, Fas receptor, M68, CD27, CD30, 4-1BB, TRAILR1I, TRAILR2, TRAILR3, TRAILR4, RANK, OCIF, TWEAK receiver, TACI, BAFF receiver, ATAR, CD271, CD269, AITR, TROY, CD358, TRAMP and XEDAR.
[7]
7. Composition according to claim 4, characterized by the fact that said activator for a positive co-stimulating molecule is a member of the TNF superfamily.
[8]
8. Composition according to claim 7, characterized by the fact that said member of the TNF superfamily is selected from the group consisting of: TNFa, TNF-C, OX40L, CD40L, FasL, LIGHT, TLIA, CD27L , Siva, CD153, 4-1BB ligand, TRAIL, RANKL, TWEAK, APRIL, BAFF, CAMLG, NGF, BDNF, NT-3, NT, GITR ligand and EDA-2.
[9]
9. Composition according to claim 8, characterized by the fact that said member of the TNF superfamily is CDA40L.
[10]
10. Composition according to claim 8, characterized by the fact that said member of the TNF superfamily is CD27L.
[11]
11. Composition according to claim 8, characterized by the fact that said member of the superfamily TNF is OXA40L.
[12]
12. Composition according to claim 4, characterized by the fact that said positive co-stimulating molecule is a co-stimulating molecule of the CD28 superfamily.
[13]
13. Composition according to claim 12, characterized by the fact that the said co-stimulating molecule of the CD28 superfamily is ICOS or CD28.
[14]
14. Composition, according to claim 4, characterized by the fact that said activator for a positive co-stimulating molecule is ICOSL, CD80 or CD86.
[15]
15. Composition, according to claim 14, characterized by the fact that said activator for a positive co-stimulating molecule is CD80.
[16]
16. Composition according to claim 1, characterized by the fact that said first immunomodulatory component is a cytokine or a cytokine binding partner.
[17]
17. Composition, according to claim 16, characterized by the fact that said cytokine is selected from the group consisting of: 1L-2, IL-7, I1L-10, 11-12 and I1L-15 .
[18]
18. Composition according to claim 16, characterized by the fact that said cytokine is IL-7.
[19]
19. Composition, according to claim 16, characterized by the fact that said cytokine is 11-12.
[20]
20. Composition according to claim 16, characterized by the fact that the referred cytokine is IL-15.
[21]
21. Composition according to claim 1, characterized by the fact that the first immunomodulatory component is a T cell receptor (TCR), a T cell coreceptor, a major histocompatibility complex (MHC), a human leukocyte antigen (HLA) or a derivative thereof.
[22]
22. Composition according to claim 1, characterized by the fact that said first immunomodulatory component is an activator of a T cell receptor or coreceptor.
[23]
23. The composition according to claim 22, characterized in that said activator of a T cell receptor or coreceptor is a CD3 activator, optionally a CD3 agonist antibody.
[24]
24. Composition according to claim 1, characterized by the fact that said first immunomodulatory component is a tumor antigen.
[25]
25. Composition according to claim 24, characterized by the fact that said tumor antigen is selected from the group consisting of: alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), tumor epithelial antigen ( ETA), mucin 1 (MUC1), Tn-MUC1, mucin 16 (MUC16), tyrosinase, melanoma-associated antigen (MAGE), tumor protein p53 (p53), CD4, CD8, CD45, CD80, CDB86, ligand programmed death 1 (PD-L1), programmed death ligand 2 (PD-L2), NY-ESO-1, PSMA, TAG-72, HER2, GD2, cMET, EGFR, Mesothelin, VEGFR, alpha-receptor folate, CE7R, IL-3, cancerous testis antigen, MART-1 gp100 and TNF-related apoptosis-inducing ligand.
[26]
26. Composition according to claim 24 or 25, characterized in that said tumor antigen is derived from a reference genome sequence.
[27]
27. Composition according to claim 24 or 25, characterized by the fact that said tumor antigen is an individual's genome sequence.
[28]
28. Composition according to claims 1 to 27, characterized in that said first immunomodulating component is an agonist or an antagonist.
[29]
29. Composition according to any one of claims 1 to 27, characterized by the fact that said first immunomodulatory component is an antibody or a fragment of |
antigen.
[30]
30. Composition, according to any of the claims | to 27, characterized by the fact that said first immunomodulatory component is a polynucleotide.
[31]
31. Composition according to claim 30, characterized by the fact that said polynucleotide is selected from the group consisting of: an mMRNA, a miRNA, a siRNA, an antisense RNA, a ShRNA, an IncRNA and a dsDNA .
[32]
32. Composition according to claims 1 to 27, characterized in that said first immunomodulating component is a protein, a peptide, a glycolipid or a glycoprotein.
[33]
33. Composition according to any one of claims 1 to 32, characterized by the fact that said first immunomodulatory component is expressed as a fusion protein displayed on the external surface of said extracellular vesicle.
[34]
34. Composition according to claim 33, characterized in that said fusion protein comprises PTGFRN or a fragment or variant thereof.
[35]
35. Composition according to claim 34, characterized by the fact that the sequence of said fusion protein is SEQ ID NO: 3.
[36]
36. Composition, according to any one of claims 1 to 35, characterized by the fact that the said extracellular vesicle is an exosome.
[37]
37. Composition, according to any one of claims 1 to 32, characterized by the fact that the said extracellular vesicle is a nanovesicle.
[38]
38. Composition according to any one of claims 1 to 37, characterized by the fact that it also comprises a pharmaceutically acceptable carrier.
[39]
39. Composition according to any one of claims 1 to 38, characterized by the fact that the extracellular vesicle additionally comprises a second immunomodulatory component.
[40]
40. Composition according to claim 39, characterized in that said second immunomodulating component is an inhibitor for a negative checkpoint regulator or an inhibitor for a binding partner of a negative checkpoint regulator verification.
[41]
41. Composition according to claim 40, characterized by the fact that the negative checkpoint regulator is selected from the group consisting of: protein 4 associated with cytotoxic T lymphocytes (CTLA-4), protein 1 programmed cell death (PD-1), lymphocyte-activated gene 3 (LAG-3), protein 3 containing T cell immunoglobulin mucin (TIM-3), B and T lymphocyte attenuator (BTLA), T cell immunoreceptor with Ig and ITIM domains (TI-GIT), Ig suppressor of T cell activation domain V (VISTA), A2a adenosine receptor (A2aR), killer cell immunoglobulin-like receptor (KIR), indoleamine 2, 3-dioxigenase (IDO), CD20, CD39 and CD73.
[42]
42. Composition according to claim 39, characterized in that said second immunomodulatory component is an activator for a positive co-stimulatory molecule or an activator for a binding partner of a positive co-stimulatory molecule.
[43]
43. Composition according to claim 42, characterized in that said positive co-stimulating molecule is a member of the TNF receptor superfamily.
[44]
44. Composition according to claim 43, characterized by
characterized by the fact that said member of the TNF receptor superfamily is selected from the group consisting of: CD120a, CD120b, CD18, OX40, CD40, Fas receptor, M68, CD27, CD30, 4-1BB, TRAILR1I, TRAILR2 , TRAILR3, TRAILR4, RANK, OCIF, TWEAK receiver, TACI, BAFF receiver, ATAR, CD271, CD269, AITR, TROY, CD358, TRAMP and XEDAR.
[45]
45. Composition according to claim 42, characterized by the fact that said activator for a positive co-stimulating molecule is a member of the TNF superfamily.
[46]
46. Composition according to claim 45, characterized by the fact that said member of the TNF superfamily is selected from the group consisting of: TNFa, TNF-C, OX40L, CD40L, FasL, LIGHT, TLIA, CD27L , Siva, CD153, 4-1BB ligand, TRAIL, RANKL, TWEAK, APRIL, BAFF, CAMLG, NGF, BDNF, NT-3, NT, GITR ligand and EDA-2.
[47]
47. Composition according to claim 46, characterized by the fact that said member of the superfamily TNF is CDA40L.
[48]
48. Composition according to claim 46, characterized by the fact that said member of the TNF superfamily is CD27L.
[49]
49. Composition according to claim 46, characterized by the fact that said member of the superfamily TNF is OXA40L.
[50]
50. Composition according to claim 42, characterized by the fact that said positive co-stimulating molecule is a co-stimulating molecule of the CD28 superfamily.
[51]
51. Composition according to claim 50, characterized by the fact that said co-stimulating molecule of the CD28 superfamily is ICOS or CD28.
[52]
52. Composition, according to claim 42, characterized by the fact that said activator for a positive co-stimulating molecule is ICOSL, CD80 or CD86.
[53]
53. Composition according to claim 52, characterized by the fact that said activator for a positive co-stimulating molecule is CD80.
[54]
54. Composition according to claim 39, characterized in that said second immunomodulatory component is a cytokine or a cytokine binding partner.
[55]
55. Composition according to claim 54, characterized by the fact that said cytokine is selected from the group consisting of: 1L-2, IL-7, I1L-10, 11-12 and I1L-15 .
[56]
56. Composition according to claim 54, characterized by the fact that said cytokine is IL-7.
[57]
57. Composition according to claim 54, characterized by the fact that said cytokine is 11-12.
[58]
58. Composition according to claim 54, characterized by the fact that the referred cytokine is IL-15.
[59]
59. Composition according to claim 39, characterized by the fact that the second immunomodulatory component is a T cell receptor (TCR), a T cell coreceptor, a major histocompatibility complex (MHC), a leuk antigen - human cytoplasm (HLA) or a derivative thereof.
[60]
60. Composition according to claim 39, characterized in that said second immunomodulatory component is an activator of a T cell receptor or coreceptor.
[61]
61. Composition according to claim 60, characterized by the fact that said activator of a T cell receptor or coreceptor is a CD3 activator, optionally a CD3 agonist antibody.
[62]
62. Composition according to claim 39, characterized by the fact that said second immunomodulatory component is a tumor antigen.
[63]
63. Composition according to claim 62, characterized by the fact that said tumor antigen is selected from the group consisting of: alpha-fetoprotein (AFP), carcinoembryonic antigen (CEA), tumor epithelial antigen ( ETA), mucin 1 (MUC1), Tn-MUC1, mucin 16 (MUC16), tyrosinase, melanoma-associated antigen (MAGE), tumor protein p53 (p53), CD4, CD8, CD45, CD80, CDB86, ligand programmed death 1 (PD-L1), programmed death ligand 2 (PD-L2), NY-ESO-1, PSMA, TAG-72, HER2, GD2, cMET, EGFR, Mesothelin, VEGFR, alpha-receptor folate, CE7R, IL-3, cancerous testis antigen, MART-1 gp100 and TNF-related apoptosis-inducing ligand.
[64]
64. Composition according to claim 62 or 63, characterized in that said tumor antigen is derived from a reference genome sequence.
[65]
65. Composition according to claim 62 or 63, characterized by the fact that said tumor antigen is a genome sequence of an individual.
[66]
66. Composition according to any one of claims 39 to 65, characterized by the fact that said second immunomodulating component is an agonist or an antagonist.
[67]
67. Composition according to any one of claims 39 to 65, characterized by the fact that said second immunomodulatory component is an antibody or antigen-binding fragment.
[68]
68. Composition according to any one of claims 39 to 65, characterized by the fact that said second immunomodulatory component is a polynucleotide.
[69]
69. Composition according to claim 68, characterized by the fact that said polynucleotide is selected from the group consisting of: an mMRNA, a miRNA, a siRNA, an antisense RNA, a ShRNA, an IncRNA and a dsDNA .
[70]
70 Composition according to any one of claims 39 to 65, characterized in that said second immunomodulating component is a protein, a peptide, a glyco-lipid or a glycoprotein.
[71]
71. Composition according to any one of claims 39 to 70, characterized by the fact that said second immunomodulatory component is expressed as a fusion protein displayed on the external surface of said extracellular vesicle.
[72]
72. Composition according to claim 71, characterized in that said fusion protein comprises PTGFRN or a fragment or variant thereof.
[73]
73. Composition according to claim 72, characterized in that the sequence of said fusion protein is SEQ ID NO: 3.
[74]
74. Composition according to any one of claims 39 to 73, characterized by the fact that said second immunomodulatory component is different from said first immunomodulatory component.
[75]
75. Composition according to any one of claims 39 to 74, characterized by the fact that the extracellular vesicle additionally comprises a second immunomodulating component.
[76]
76. The composition of claim 75, characterized by the fact that said third immunomodulatory component is different from said first and second immunomodulatory components.
[77]
77. Method for producing said composition, as defined in any one of claims 1 to 38, characterized in that it comprises: modifying a producing cell with said first, second and / or third immunomodulatory components; obtaining said extracellular vesicle from said producing cell; and optionally isolating said obtained extracellular vesicles.
[78]
78. Method for producing said composition, as defined in any of claims 1 to 38, characterized by the fact that it comprises: obtaining said extracellular vesicle from said producing cell; isolating said extracellular vesicles obtained; and modifying said isolated extracellular vesicle with said first, second and / or third immunomodulatory components.
[79]
79. The method of claim 77 or 78, characterized by the fact that it further comprises the formulation of said isolated extracellular vesicles in a pharmaceutical composition.
[80]
80. Method for treating cancer in an individual, characterized by the fact that it comprises: administering to said subject a therapeutically effective amount of said composition, as defined in any one of claims 1 to 38, wherein said composition is capable of to increasingly regulate an immune response in said individual, thereby improving targeting of the tumor of the immune system of said individual.
[81]
81. Method for treating graft versus host disease (GvHD) in an individual, characterized by the fact that it comprises: administering to said individual a therapeutically effective amount of said composition, as defined in any of the claims 1 to 38, in which said composition is able to downwardly regulate an immune response in said individual, thus alleviating the symptoms of GvHD.
[82]
82. Method for treating an autoimmune disease in an individual, characterized in that it comprises: administering to said individual a therapeutically effective amount of said composition, as defined in any one of claims 1 to 38, wherein said composition is able to downwardly regulate an immune response in that individual, thus suppressing that individual's immune activity.
[83]
83. Method for treating or preventing cancer in an individual, characterized by the fact that it comprises: administering to said individual a therapeutically effective amount of said composition, as defined in any one of claims 24 to 27, wherein said composition it is capable of potentiating an immune response to the referred tumor antigen, thus improving the individual's immune response to cancer.
类似技术:
公开号 | 公开日 | 专利标题
BR112020013131A2|2020-12-08|EXOSOMES FOR IMMUNO-ONCOLOGY AND ANTI-INFLAMMATORY THERAPY
JP2021518386A|2021-08-02|Extracellular vesicles containing STING agonists
KR20200120624A|2020-10-21|Methods and compositions for polarization of macrophages
TW202024120A|2020-07-01|Extracellular vesicles targeting dendritic cells and uses thereof
EP3774864A1|2021-02-17|Chimeric engulfment receptors and uses thereof for neurodegenerative diseases
Jang et al.2021|ExoSTING, an extracellular vesicle loaded with STING agonists, promotes tumor immune surveillance
CN110709416A|2020-01-17|TGF-beta decoy receptors
US20210299172A1|2021-09-30|Engineered phagocytic receptor compositions and methods of use thereof
EP3288564A1|2018-03-07|Methods for enhancing an immune response with a ctla-4 antagonist
JP2019516768A|2019-06-20|Immune checkpoint inhibitors and cytotoxic T cells for the treatment of cancer
WO2021184017A1|2021-09-16|Extracellular vesicles for treating neurological disorders
KR20210044243A|2021-04-22|OX40 binding polypeptides and uses thereof
Kusumoto et al.2018|Epithelial membrane protein 3 | downregulates induction and function of cytotoxic T lymphocytes by macrophages via TNF-α production
WO2011129379A1|2011-10-20|Novel anti-hsp90 monoclonal antibody
WO2020163370A1|2020-08-13|Membrane protein scaffolds for exosome engineering
CA3090659A1|2019-08-15|Modulation of p62 and sting activity
JP2022519275A|2022-03-22|Membrane protein scaffolding for exosome manipulation
US11090336B2|2021-08-17|Tn-MUC1 chimeric antigen receptor | T cell therapy
JP7034934B2|2022-03-14|Chimeric antigen and T cell receptor, and how to use
KR20200036874A|2020-04-07|Methods and compositions for the treatment of cancer
CN114072157A|2022-02-18|Engineered chimeric fusion protein compositions and methods of use thereof
CN113874389A|2021-12-31|Interleukin-2 receptor | and interleukin-2 | variants for specific activation of immune effector cells
WO2021248133A1|2021-12-09|Anti-transferrin extracellular vesicles
Anyaegbu2016|The Effect of Tumour Antigen Properties on the Efficiency of Cross-presentation
Kar et al.2011|Novel CCL21-Vault Nanocapsule Intratumoral Delivery Inhibits Lung Cancer
同族专利:
公开号 | 公开日
EP3731849A2|2020-11-04|
PH12020550805A1|2021-05-31|
CA3085471A1|2019-07-04|
CN111655271A|2020-09-11|
IL275600D0|2020-08-31|
EP3731849A4|2021-12-01|
EA202091134A1|2020-11-20|
US10723782B2|2020-07-28|
US20200407419A1|2020-12-31|
KR20200108275A|2020-09-17|
CO2020009091A2|2020-12-21|
AU2018394238A1|2020-06-18|
WO2019133934A3|2019-08-29|
JP2021508691A|2021-03-11|
CL2020001691A1|2020-11-13|
SG11202005049QA|2020-07-29|
WO2019133934A2|2019-07-04|
US20190202892A1|2019-07-04|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

CN1254234C|2000-06-09|2006-05-03|莱古伦公司|Plasmid DNA and nucleas-containing location signal/fusogene conjogates drug encapsulating into targeted liposomes complex|
JP4662708B2|2001-08-17|2011-03-30|エクソセラ・エルエルシー|Methods and compositions for targeting proteins to exosomes|
US7803906B2|2002-03-22|2010-09-28|Gene Signal International Sa|Composition comprising an angiogenesis related protein|
US9085778B2|2006-05-03|2015-07-21|VL27, Inc.|Exosome transfer of nucleic acids to cells|
AU2011316477A1|2010-10-11|2013-05-02|Medsaic Pty Ltd|Assay for disease detection|
WO2013043569A1|2011-09-20|2013-03-28|Vical Incorporated|Synergistic anti-tumor efficacy using alloantigen combination immunotherapy|
US9777042B2|2011-12-15|2017-10-03|Morehouse School Of Medicine|Method of purifying HIV/SIV Nef from exosomal fusion proteins|
EP2687219A1|2012-07-18|2014-01-22|Universität Duisburg-Essen|Use of preparations comprising exosomes derived from mesenchymal stem cells in the prevention and therapy of inflammatory conditions|
US20150301058A1|2012-11-26|2015-10-22|Caris Science, Inc.|Biomarker compositions and methods|
CA2894668A1|2012-12-12|2014-06-19|The Broad Institute, Inc.|Crispr-cas component systems, methods and compositions for sequence manipulation|
KR20150043937A|2013-10-15|2015-04-23|삼성전자주식회사|A compositon for diagnosing a liver cancer in a subject, method for diagnosing a liver cancer in a subject and method for obtaining information for diagnosing a liver cancer in a subject|
US10772911B2|2013-12-20|2020-09-15|Advanced ReGen Medical Technologies, LLC|Cell free compositions for cellular restoration and methods of making and using same|
EP3083939A4|2013-12-20|2017-05-17|Advanced Regen Medical Technologies, LLC|Compositions for cellular restoration and methods of making and using same|
JP2017536096A|2014-10-09|2017-12-07|アントフロゲネシス コーポレーション|Placenta-derived adherent cell exosomes and their use|
CN113151167A|2014-10-27|2021-07-23|弗罗里达中央大学研究基金会|Methods and compositions for natural killer cells|
WO2016077639A2|2014-11-12|2016-05-19|VL27, Inc.|Nanovesicular therapies|
CA3017571A1|2015-03-16|2016-09-22|Duncan Ross|Method of treatment comprising membrane-enclosed vesicle|
US10427129B2|2015-04-17|2019-10-01|LLT International Ltd.|Systems and methods for facilitating reactions in gases using shockwaves produced in a supersonic gaseous vortex|
WO2016168680A1|2015-04-17|2016-10-20|Exocyte Therapeutics Pte Ltd.|Method for developing exosome-based vaccines|
JP2019513019A|2016-03-15|2019-05-23|コディアック バイオサイエンシズ インコーポレイテッド|Therapeutic membrane vesicles|
JP2020531018A|2017-08-25|2020-11-05|コディアック バイオサイエンシズ インコーポレイテッド|Preparation of therapeutic exosomes using membrane proteins|
BR112020013131A2|2017-12-28|2020-12-08|Codiak Biosciences, Inc.|EXOSOMES FOR IMMUNO-ONCOLOGY AND ANTI-INFLAMMATORY THERAPY|GB2597623A|2017-08-07|2022-02-02|Univ California|Platform for generating safe cell therapeutics|
BR112020013131A2|2017-12-28|2020-12-08|Codiak Biosciences, Inc.|EXOSOMES FOR IMMUNO-ONCOLOGY AND ANTI-INFLAMMATORY THERAPY|
AU2020289326A1|2019-06-04|2022-01-27|Thomas Jefferson University|Oligodendrocyte-derived extracellular vesicles for therapy of multiple sclerosis|
WO2021003425A1|2019-07-03|2021-01-07|Codiak Biosciences, Inc.|Methods of inducing hematopoiesis|
WO2021062060A1|2019-09-25|2021-04-01|Codiak Biosciences, Inc.|Sting agonist comprising exosomes combined with il-12 displaying exosomes for treating a tumour|
WO2021062317A1|2019-09-25|2021-04-01|Codiak Biosciences, Inc.|Extracellular vesicle compositions|
WO2021076808A1|2019-10-15|2021-04-22|Cornell University|Methods for detecting and inhibiting brain metastasis|
WO2021146616A1|2020-01-17|2021-07-22|Codiak Biosciences, Inc.|Cholesterol assays for quantifying extracellular vesicles|
EP3858332A1|2020-01-31|2021-08-04|Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V.|Bottom-up assembly of synthetic extracellular vesicles|
WO2021231425A1|2020-05-11|2021-11-18|Strm.Bio Incorporated|Compositions and methods related to megakaryocyte-derived extracellular vesicles|
KR102341138B1|2020-05-31|2021-12-21|주식회사 엑소코바이오|Exosomes comprising exosomal membrane protein variant and manufacturing method thereof|
WO2021262879A1|2020-06-24|2021-12-30|Chameleon Biosciences, Inc.|Extracellular vesicles with immune modulators|
WO2022040223A1|2020-08-17|2022-02-24|Codiak Biosciences, Inc.|Methods of treating cancer|
CN113040877A|2021-03-11|2021-06-29|上海市东方医院(同济大学附属东方医院)|Method for treating abdominal diseases by infusing exosomes based on fascia clearance concept|
法律状态:
2021-12-07| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
申请号 | 申请日 | 专利标题
US201762611140P| true| 2017-12-28|2017-12-28|
US62/611,140|2017-12-28|
US201862723267P| true| 2018-08-27|2018-08-27|
US62/723,267|2018-08-27|
PCT/US2018/068062|WO2019133934A2|2017-12-28|2018-12-28|Exosomes for immuno-oncology and anti-inflammatory therapy|
[返回顶部]