[go: up one dir, main page]

WO2025221669A1 - Protéine de fusion comprenant il-12 et gm-csf - Google Patents

Protéine de fusion comprenant il-12 et gm-csf

Info

Publication number
WO2025221669A1
WO2025221669A1 PCT/US2025/024551 US2025024551W WO2025221669A1 WO 2025221669 A1 WO2025221669 A1 WO 2025221669A1 US 2025024551 W US2025024551 W US 2025024551W WO 2025221669 A1 WO2025221669 A1 WO 2025221669A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
mrna
csf
fusion protein
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2025/024551
Other languages
English (en)
Inventor
Lijun Wu
Vita Golubovskaya
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Forevertek Biotechnology Co Ltd
Promab Biotechnologies Inc
Original Assignee
Forevertek Biotechnology Co Ltd
Promab Biotechnologies Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Forevertek Biotechnology Co Ltd, Promab Biotechnologies Inc filed Critical Forevertek Biotechnology Co Ltd
Publication of WO2025221669A1 publication Critical patent/WO2025221669A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5434IL-12
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment

Definitions

  • the present invention relates to a fusion protein comprising IL-12 and GM-CSF, which are covalently attached to each other.
  • the present invention also relates to generating mRNA-LNPs (lipid nanoparticles) and delivering them into cells to generate cells expressing membrane-bound IL- 12 and GM-CSF fusion protein.
  • This membrane bound I1-12-GM-CSF protein can be used for inducing immune response and activating tumor microenvironment in vivo.
  • Interleukin- 12 is a heterodimer protein which was described in 1989 and consists of two subunits: p40 and p35 (1). L-12 regulates immune system response and is important for adaptive immunity (1). In response to pathogens, the heterodimeric protein is secreted by phagocytic cells (2). Binding of IL-12 to the IL-12 receptor (IL-12R) on T cells and natural killer (NK) cells leads to activator of transcription 4 (STAT4) and stimulates secretion of interferon gamma (IFN-y) (2).
  • IL-12R IL-12 receptor
  • NK natural killer cells
  • IFN-y Signaling downstream of IFN-y includes activation of T-box transcription factor TBX21 (Tbet) and induces pro-inflammatory functions of T helper 1 (Tul) cells, linking innate and adaptive immune responses(2).
  • II- 12 is also known to inhibit immunosuppressive tumor associated macrophages and MDSC (myeloid-derived suppressor cells. 11-12 showed anti-tumor activity in many pre- clinical studies (3). In spite of the anti-tumor effects of IL-12, systemic administration of IL- 12 has been too toxic.
  • the ideal targets of IL- 12 immunotherapy are immune cells within the tumor and nearby lymph nodes, including activated but exhausted T cells, NK cells, TAMs, and MDSCs.
  • Granulocyte-macrophage colony-stimulating factor is a well-known player of myelopoiesis and mobilization of hematopoietic stem cells (4).
  • GM-CSF can induce the differentiation of HPCs into different immune cells, including granulocytes, monocytemacrophages, T-cells, and natural killer (NK) cells in the bone marrow (4).
  • GM-CSF was identified in the 1960s as a myeloid growth factor, purified in the 1970s, molecularly-cloned in the 1980s, and clinically developed in the 1990s (5).
  • Sargramostim (Leukine®; Partner Therapeutics, Inc., Lexington, MA) is a glycosylated, recombinant human granulocytemacrophage colony-stimulating factor (rhu GM-CSF), FDA-approved for six different diseases based on its safe and efficacious hematopoietic growth factor function, differing from human GM-CSF by one amino acid at position 23, where leucine is substituted for arginine(5).
  • rhu GM-CSF granulocytemacrophage colony-stimulating factor
  • GM-CSF In addition to myelopoietic actions, GM-CSF possesses anti-apoptotic effects and is reported to induce proliferation, mobilization, and activation of hematopoietic stem cells, mesenchymal stromal cells, endothelial progenitor cells, pericytes, neural stem cells, and oligodendrocyte progenitor cells (5).
  • GM-CSF to IL- 12 membrane bound protein to express as a single protein with linker in between to induce immune cells.
  • FIG. 1 shows the scheme of DNA vector template (panel A) used for in vitro transcription to mRNA of IL-12-linker-GM-CSF-TM (panel B). The mRNA is then used to generate translated protein that is expressed on the cell surface. TM; transmembrane domain; 5’-UTR: 5’ untranslated region; 3’-UTR: 3 ’untranslated region; poly A tail for increased stability.
  • FIG. 2 shows a structure of membrane-bound IL- 12 linked to GM-CSF.
  • L linker
  • TM transmembrane domain
  • C a cytoplasmic tail.
  • IL-12 consists of IL-12 p40 domain- linker-IL12 p45 domain.
  • FIGs. 3A and 3B show protein expression 72 hours after delivery of IL-12-1-GM-CSF mRNA-LNP into different cell lines using mRNA-LNP.
  • 3A Expression of GM-CSF
  • 3B Expression of IL-12. FACS was done with mouse anti-GM-CSF or anti-IL-12 antibody, and then secondary APC-conjugated anti-mouse IgG antibody was used at 1:100 dilution.
  • FIG. 4 shows transfection of IL-12-GM-CSF LNPs to primary T cells resulted in increased expression of IFN-gamma.
  • T cells were used 3 days after activation with CD3/CD28 antibodies and 2 days post- transfection with mRNA-LNP. The cells were stained for intracellular IFN-g (Thl marker). The cells were treated with Brefeldin A for 2.5 hours before FACS to accumulate cytokines within the cells. CD3 staining is shown on X-axis, IFN-gamma expression is shown on Y-axis.
  • FIG. 5 shows that transfection of IL-12-GM-CSF to macrophages increased survival and morphology of macrophages. Morphology of cells is shown under X400 magnification.
  • FIG. 6 shows IL-12-GM-CSF mRNA-LNP transfection of monocytes with addition of IL-4 increases dendritic cell phenotype by expression of CD la dendritic marker and decreasing expression of monocyte marker CD 14.
  • FIG. 7 shows that IL-12-GM-CSF mRNA decreased OVCAR-5 xenograft tumor growth in vivo. *p ⁇ 0.05 (i) IL-12-GM-CSF mRNA-LNP, (ii) IL-12-GM-CSF+EpCAM-CD3 mRNA-LNP, versus GFP mRNA as a negative control.
  • FIGs. 8A and 8B show the efficacy of IL-12-GM-CSF in tumor in mice at a local treated side (8A), and at a distant non-treated site (8B).
  • PC3 tumor cells were injected subcutaneously into left and right side.
  • mRNA-LNP were injected intratumorally only to the left tumors. Tumor size was measured at left and right tumors with calipers. *p ⁇ 0.05 EpCAM-CD3+IL-12-GM-CSF vs GFP-LNP control, Student’s t-test.
  • the present invention is directed to a fusion protein comprising IL-12 and GM-CSF, which are covalently attached.
  • IL-12 is N-terminal to GM-CSF.
  • IL- 12 is C-terminal to GM-CSF.
  • IL- 12 and GM-CSF are attached through a linker.
  • the linker has 5-30 or 5-20 amino acids in length.
  • the fusion protein further comprises a transmembrane domain C- terminal to IL- 12 and GM-CSF. In another embodiment, the fusion protein also comprises a cytoplasmic tail at the C-terminal end of the fusion protein.
  • mRNA is transient and short-lived when delivered in vivo.
  • the present invention provides a method for producing IL-12-linker-GM-CSF protein attached to membrane by delivering lipid nanoparticle-encapsulated mRNA in cells. By encapsulating mRNA in lipid nanoparticles (LNPs), the stability of mRNA is improved.
  • LNPs lipid nanoparticles
  • membrane-bound IL-12 and GM-CSF provide better tolerated IL-12 with higher efficacy.
  • the transmembrane domain is selected from the group consisting of: a transmembrane domain of PGFRB receptor (platelet-derived growth factor receptor beta), a T cell receptor a or P chain, a CD3 zeta chain, CD28, CD3s., CD45, CD4, CDS, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS, CD 154, and GITR.
  • PGFRB receptor platelet-derived growth factor receptor beta
  • T cell receptor a or P chain a CD3 zeta chain
  • CD28, CD3s. CD45, CD4, CDS, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS, CD 154, and GITR.
  • the present invention is also directed to lipid nanoparticles have mRNA encapsulated, wherein the mRNA is transcribed from a DNA sequence comprising: (a) a promoter coding sequence, e.g., a T7 promoter coding sequence, (b) 5'-UTR (untranslated region) coding sequence, (c) a sequence encoding the present fusion protein as described above, (d) a 3'- UTR coding sequence, and (e) a poly A tail sequence.
  • a promoter coding sequence e.g., a T7 promoter coding sequence
  • 5'-UTR untranslated region
  • the present invention is further directed to a method for producing an IL-12-linker- GM-CSF protein expressed on the surface of cells.
  • the method comprises the steps of: (i) obtaining a DNA sequence comprising: (a) a promoter coding sequence, (b) 5'-UTR coding sequence, (c) a sequence to encode the fusion protein of the present invention, (d) a 3'-UTR coding sequence, and (e) a poly A tail sequence, (ii) transcribing the DNA sequence to mRNA with RNA polymerase in vitro, (iii) encapsulating the mRNA in lipid nanoparticles (LNPs), (iv) transfecting the mRNA-encapsulated LNPs into cells, and (v) translating the mRNA in the cells to produce cytokines of IL-12 and GM-CSF which are expressed on the surface of the cells.
  • a DNA sequence comprising: (a) a promoter coding sequence, (b) 5'-
  • the lipid nanoparticles comprise 8-[(2-hydroxyethyl)[6-oxo-6- (undecyloxy)hexyl] amino] -octanoic acid, 1-octylnonyl ester (SM-102), distearoylphosphatidylcholine (DSPC), Cholesterol, and l,2-dimyristoyl-rac-glycero-3- methoxypoly ethylene glycol-2000 (DMG-PEG2000).
  • SM-102 1-octylnonyl ester
  • DSPC distearoylphosphatidylcholine
  • Cholesterol Cholesterol
  • the lipid nanoparticles comprise 8-[(2-hydroxyethyl)[6-oxo-6- (undecyloxy)hexyl
  • the lipid nanoparticles comprise 2-hexyl-decanoic acid, 1, l'-[[(4- hydroxybutyl)imino]di-6,l -hexanediyl] ester (ALC-0315), DSPC, Cholesterol, and a-[2- (ditetradecylamino)-2-oxoethyl]-co-methoxy-poly(oxy-l,2-ethanediyl) (ALC-0159). [LNP- 315]
  • the lipid nanoparticles comprise 8-[(2-hydroxyethyl)[6-oxo-6- (undecyloxy)hexyl] amino] -octanoic acid, 1-octylnonyl ester (SM-102), distearoylphosphatidylcholine (DSPC), Cholesterol, and l,2-dimyristoyl-rac-glycero-3- methoxypolyethylene glycol-2000 (DMG-PEG2000).
  • LNP-102 is formulated at molar ratio of SM-102, DSPC, Cholesterol, and PEG2000 (50:10:38.5: 1.5 mol%).
  • mRNA-lipid nanoparticle preparation is described in Schoenmaker (International J. Pharmaceutics, 601: 120856, 2021), the article is incorporated herein by reference in its entirety, regarding the LNPs.
  • FIG. 1 shows linearized DNA template to be used for in vitro transcription with RNA polymerase and nucleotide triphosphate to generate mRNA.
  • the DNA template contains a promoter such as T7 promoter or SP6 promoter, then 5’UTR (untranslated region), the coding region of IL-12-linker-GM-CSF- transmembrane (TM) domain, then 3’UTR and poly A tail (for example, about 152 nucleotides) for RNA stability.
  • the generated RNA may contain 5’ cap ([m7G(5’)ppp(5')G], capl for increased stability.
  • the mRNA can be electroporated or transfected to cells.
  • mRNAs are preferably to be added to lipid nanoparticles, LNP-nanoparticles can be prepared according to the formula above.
  • the promoter may be T7 AG promoter.
  • Poly A tail sequence is from 20-170 nucleotides. Poly A tail sequence optionally comprises one or more linkers in between the poly A segments. If poly A tail is longer than 60 nucleotides, then it typically contains a linker which includes non-adenosine nucleotides. A linker is 5-30 or 5-25 nucleotides, e.g., 10 nucleotides or 20 nucleotides. In one example, poly A tails is 110 nucleotides in length, consisting of a stretch of 30 adenosine residues, followed by a 10- nucleotide linker sequence, and another 70 adenosine residues.
  • poly A tails is 90 nucleotides in length, consisting of a stretch of 40 adenosine residues, followed by a 30-nucleotide linker sequence, and another 30 adenosine residues.
  • Poly A tail can be up to 150-160 nucleotides in length, interrupted with the linker sequences.
  • UTRs of mRNAs may control their regulation, degradation and localization include stem-loop structures, upstream initiation codons and open reading frames, internal ribosome entry sites and various cis-acting elements that are bound by RNA-binding proteins. UTRs are important in the post- transcriptional regulation of RNA expression, including modulation of the transport of mRNAs out of the nucleus and of translation efficiency, subcellular localization, and stability.
  • 5’-UTR typically has 10-1000 nucleotides, or 20-500 nucleotides, or 30-200 nucleotides, or 30-100 nucleotides.
  • 5’-UTR is 50 nucleotides.
  • 3’-UTR typically has 10-3000 nucleotides, for example, 50-500 nucleotides, or 100-300 nucleotides.
  • Preferred 5’-UTRs and 3’-UTRs are UTRs of P-globin, or UTRs of Pfizer CO VID vaccine.
  • the 5 '-untranslated region is derived from human alphaglobin RNA with an optimized Kozak sequence.
  • the 3 ' untranslated region comprises two sequence elements derived from the amino-terminal enhancer of split (AES) mRNA and the mitochondrial encoded 12S ribosomal RNA to confer RNA stability and high total protein expression.
  • AES amino-terminal enhancer of split
  • Any suitable vector such as Vector pSP64 Poly(A) (Promega) or pGEM3Z-Vektor (Promega) can be used as a cloning vector for the DNA sequence described above.
  • the 3’-UTR of the P-globin molecule flanked by restriction enzyme site can be amplified from human bone marrow.
  • a single (pEM3Z-ip-globin-UTR-A[120]) or 2 serial fragments (pEM3Z-2p-globin-UTR-A[120]) can be inserted in front of the poly(A) tail.
  • the DNA sequence it preferably contains a transmembrane domain-coding sequence, which encodes the transmembrane domain that may be derived from a natural polypeptide, or may be artificially designed.
  • the transmembrane domain derived from a natural polypeptide can be obtained from any membrane-binding or transmembrane protein.
  • a transmembrane domain of a T cell receptor a or P chain, PGFRB receptor (platelet-derived growth factor receptor beta), a CD3 zeta chain, CD28, CD3e., CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, ICOS, CD 154, or a GITR can be used.
  • the artificially designed transmembrane domain is a polypeptide mainly comprising hydrophobic residues such as leucine and valine. It is preferable that a triplet of phenylalanine, tryptophan and valine is found at each end of the synthetic transmembrane domain.
  • a short oligopeptide linker or a polypeptide linker for example, a linker having a length of 2 to 10 amino acids can be arranged between the transmembrane domain and the intracellular domain.
  • DNA sequence it may contain a cytoplasmic tail-coding sequence, which encodes a short cytoplasmic tail.
  • IL- 12 cytokine sequence with a signaling peptide, a linker, GM-CSF, a transmembrane domain, and a small cytoplasmic tail to express IL- 12 and GM-CSF on the cell surface or tethered (membrane-bound).
  • a linker GM-CSF
  • transmembrane domain a transmembrane domain
  • small cytoplasmic tail a small cytoplasmic tail
  • IL-12 and GM-CSF that are membrane-bound.
  • the advantage of IL-12 and GM-CSF to be expressed on the surface of cells is that proteins are more stable, less toxic and can activate T cells and other immune cells.
  • 11-12 and GM-CSF can be expressed on tumor cells to activate immune cells or tumor microenvironment.
  • the present invention is directed to a method for producing a fusion protein on the surface of cells.
  • the method comprises the steps of: obtaining a DNA sequence comprising: (a) a promoter coding sequence, (b) 5'-UTR coding sequence, (c) a sequence encoding the fusion protein of IL-12 and GM-CSF, (d) a 3'-UTR coding sequence, and (e) a poly A tail sequence; transcribing the DNA sequence to mRNA with RNA polymerase in vitro, encapsulating the mRNA in lipid nanoparticles (LNPs), transfecting the mRNA-encapsulated LNPs into cells, and translating the mRNA in the cells to produce membrane-bound fusion protein on the surface of the cells.
  • the cells are cancer cells or blood cells, and the mRNA is transfected into the cancer cells or blood cells.
  • the present invention is directed to a method for treating cancer, comprising the step of administering a fusion protein comprising IL- 12 and GM-CSF to a subject suffering from cancer by intratumor injection, wherein the cancer is selected from the group consisting of breast, ovarian, colon cancer, prostate cancer, sarcoma, melanoma, hepatocellular carcinoma, thyroid, multiple myeloma and other cancers.
  • the method comprises the steps of obtaining an mRNA sequence comprising a coding sequence to encode a fusion protein comprising IL- 12 and GM-CSF; mixing the mRNA with lipid nanoparticles to form a mRNA-lipid nanoparticle complex; and injecting the mRNA- lipid nanoparticle complex into the cancer cells.
  • the fusion protein further comprises a transmembrane domain C-terminal to the two cytokines.
  • IL- 12 and GM-CSF are expressed on the surface of the cancer cells and are membrane bound.
  • the toxicity of IL- 12 and GM-CSF are reduced.
  • mRNA is embedded into LNPs and transfected inside tumors.
  • the nanoparticle-based drug delivery has many advantages, such as high bioavailability, solubility, stability, passage through the blood-brain barrier, and low toxicity and minimal side effects.
  • the mRNA or its template D A has a sequence as described in this application.
  • the present disclosure describes a safe local intratumoral delivery using mRNA-LNP technology to deliver IL- 12 and GM-CSF cytokine which are membrane tethered.
  • the present disclosure combines a cell therapy approach by using cytokine RNA-LNP cells to activate T cells and monocytes, macrophages, and other immune cells surrounding tumors to kill tumors.
  • the expressed cytokine is membrane tethered, which prevents it from circulating inside cells and causing toxicity.
  • This approach can be used similarly to stimulate expansion of NK cells or gamma-delta T cells.
  • the immune cells that express IL- 12 receptors will bind to IL-12-GM-CSF, and in intratumoral delivery, tumor cells will be lysed.
  • the lysed tumor releases neoantigens (natural vaccine), which can be recognized in the presence of immunomodulators by antigen-presenting cells or dendritic cells and promote the activity of memory T and B cells.
  • DNA was digested with appropriate restriction enzyme, which cuts DNA right 3’ after poly A tail, at 37°C overnight following manufacturer’s protocol.
  • the digested DNA was treated with 50-100 pg/mL Proteinase K and 0.5% SDS for 30 minutes at 50°C. Then phenol/chloroform extraction and ethanol precipitation of DNA was performed. The DNA was used for in vitro RNA transcription reaction.
  • the in vitro transcription reaction was done by the protocol below:
  • the DNA template for generating RNA had T7AG promoter in front of the coding sequence of protein.
  • the reaction was the following:
  • the reaction volume can be up to 50 pl with nuclease-free water. Add 2 pl of DNase I, mix well and incubate at 37°C for 15 minutes.
  • RNA Cleanup Kit T2050
  • RNA concentration can be determined by diluting an aliquot of the preparation (usually a 1:50 to 1: 100 dilution) in IxTE (10 mM Tris-HCl 1 mM EDTA, pH 8) buffer, and reading the absorbance in a spectrophotometer at 260 nm.
  • concentration (pg/mL) of RNA is therefore calculated as follows: A260 x dilution factor x 40 pg/mL.
  • LNP LNP
  • SM-102 SM-102
  • DSPC DSPC
  • Cholesterol a lipid that can be used.
  • PEG2000 50:10:38.5: 1 .5 mol%) mix.
  • MC3 a ionizing lipid that can be used.
  • Nanoparticles were prepared using the below stocks dissolved in Ethanol: SM-102 (Cayman) 8-[(2-hydroxyethyl)[6-oxo-6-(undecyloxy)hexyl]amino]-octanoic acid, 1- octylnonyl ester;
  • DMG-PEG-2000 (Cayman) l,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol-2000
  • Flex(M) and Flex(S) samples are prepared 2-fold diluted in PBS from their initial formulation volume.
  • RNA-LNP nanoparticles was confirmed using Dynamic Light Scattering (DLS) system.
  • the size of RNA-LNP nanoparticles was in the range of 90-105 nm for Flex S system and 75-100 nm for FlexM system.
  • 1-3 pg of encapsulated mRNA was added per 5xl0 5 -IxlO 6 293 cells. After transfection is completed, the cells were maintained at 37°C, 5.0% CO2. After 2 days their morphology and markers were checked by microscope and FACS.
  • Example 6 Sequence of DNA template to generate tethered (membranous) IL-12- linker-GM-CSF m-RNA
  • T7 AG promoter is underlined at 5’ ; 5’UTR is shown in small font, the IL-12-Flag tag-linker-GM-CSF coding sequence is shown in bold font; 3’UTR in small font, and poly A tail is shown in italics, capital letters.
  • the RNA is prepared by in vitro transcription using this template with 5’ capl for higher stability of RNA.
  • the sequence of DNA template to generate IL-12 mRNA is shown below (PMC2389).
  • T7 AG promoter-5 ’UTR- IL- 12 (P40-Flag tag-linker-P35)-linker-GM-CSF-PDGFR beta
  • IL- 12 (P40-Flag tag-linker-P35)-linker-GM-CSF-PDGFR beta TM:
  • PDGFR beta transmembrane domain-cytoplasmic tail
  • ETSCATQIITFESFKENLKDFLLVIPFDCWEPVQEAVGQDTQEVIVVPHSLPFKVVVI SAILALVVLTIISLIILIMLWQKKPRLEGSG SEQ ID NO: 7
  • VPGVGVPGVGA SEQ ID NO: 11
  • VPGVGVPGVGA SEQ ID NO: 11
  • Flag tag was used after IL-12 p40 for detection of IL-12.
  • Other tags such as transferrin tag can be added after IL- 12 protein sequence and before transmembrane domain to test expression of this protein.
  • Example 7 Expression of membranous IL-12 -linker-GM-CSF after transfecting IL- 12- linker-GM-CSF mRNA-LNP to 293 cells
  • IL-12-linker-GM-CSF mRNA generated from DNA template PMC2389 see Example 6) with CleanCap Reagent AG (NEB #E2080), and pseudo-UTP, incorporated into LNP, and delivered to 293, Daudi, Jurkat, Nalm-6 and Raji cell lines by transfection.
  • Transfected cells were shown GM-CSF positive and IL- 12-positive when tested with anti-GM-CSF antibody (FIG. 3A) and with anti-IL-12 antibody (FIG. 3B) at 72 posttransfections by FACS, which confirms expression of cytokines on the cell surface. There was variation in expression between cell lines from 15 to 95.7% of expression of GM-CSF (FIG. 3A) and from 22% to 96.8% of IL- 12 expression (FIG. 3B).
  • Transfection of IL-12-GM-CSF to T cells activated with CD3-CD28 beads increased Thl (effector) phenotype of T cells, which was confirmed by increased expression of IFN- gamma (FIG. 4).
  • IFN-gamma expression in transfected cells increased from 26% to 53% (Y- axis) (FIG. 4). This confirms the functional effect of IL- 12, which is known to stimulate Thl T cell subtype activation with increased IFN-gamma secretion.
  • Transfection of monocytes with IL-12-GM-CSF mRNA-LNP caused expression of IL-12 in 50% of LNP-treated cells, which was shown by staining with IL-12 Antibody, and expression of GM-CSF in 12% of cells, which was shown by staining with GM-CSF antibody.
  • IL-12-GM-CSF mRNA-LNP treated macrophages were bigger, with many vacuoles, more adherent, with higher MHCII expression, and lower CD 14 expression.
  • Treatment of monocytes with IL-12-GM-CSF mRNA-LNP and IL-4 promoted dendritic cell differentiation that was detected by FACS with CD la antibodies.
  • IL-12-GM-CSF mRNA-LNP (PMC2389) transfected monocytes lost monocyte CD 14 marker 6 days after transfection and increased dendritic cell marker CDla from 0.66% to 23% in monocytes (FIG. 6).
  • Monocyte CD14+ marker dropped from 67% to 1.4% (circle) in IL-12-GM-CSF mRNA-LNP+Il-4-treated cell during 6 days (FIG. 6).
  • IL-12-GM-CSF changed function of T cells, monocytes, macrophages and dendritic cells, which is important for anti-tumor therapy where functional Thl effector cells, monocytes and dendritic cells from tumor microenvironment play functional role.
  • Example 8 In vivo efficacy of IL-12-GM-CSF in mice locally
  • OVCAR-5 gastrointestinal (GI) cancer cells were injected subcutaneously into left and right flanks of NSG mice. Then IL-12-GM-CSF mRNA-LNPs were injected intratumorally (20 mcl, 1 mcg) on days 7, 14, and 21. Then T cells were injected intravenously on days 9 and 16. The results show that 1L-12-GM-CSF significantly inhibited tumor growth with T cells alone and in combination with EpCAM-CD3 mRNA-LNP. (FIG. 7).
  • EpCAM-CD3 mRNA-LNPs were prepared with the sequences and method described in WO2024/148107.
  • Example 9 In vivo efficacy of IL-12-GM-CSF in mice at local treated and untreated distant tumors
  • PC3 prostate cancer cells were injected at the left side (6xl0 6 cells) and right side (2x l0 6 cells) subcutaneously. Then EpCAM-CD3 and GM-CSF mRNA or EpCAM-CD3+IL-12-GM-CSF mRNA-LNPs were injected intratumorally at days 14, 21 and 28 at the left side only. T cells were injected intravenously on days 15, 22 and 29. GFP mRNA-LNP was used as negative control.
  • FIGs. 8A and 8B show that IL-12-GM-CSF mRNA in combination with EPCAM-CD3 mRNA eradicated tumors not only at a local side, but also at a distant untreated side. This is important for treating metastatic tumors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Toxicology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Cell Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne une protéine de fusion comprenant de l'IL-12 et du GM-CSF, qui sont liés de manière covalente. La protéine de fusion peut en outre comprendre un domaine transmembranaire C-terminal à IL-12 et GM-CSF. La présente invention concerne également l'administration par ARNm-LNP de la protéine de fusion. La présente invention concerne également un procédé de production d'IL-12 et de GM-CSF membranaires, et un domaine transmembranaire dans des cellules. Le procédé comprend les étapes de transcription d'ARNm à partir d'une séquence d'ADN codant pour la protéine de fusion avec de l'ARN polymérase, d'encapsulation de l'ARNm dans des nanoparticules lipidiques (LNP), de transfection des LNP encapsulés par ARNm dans des cellules, et de traduction de l'ARNm dans les cellules pour produire une IL-12 liée à une membrane fusionnée à une protéine de cytokine GM-CSF pour stimuler des cellules immunitaires et un microenvironnement tumoral.
PCT/US2025/024551 2024-04-18 2025-04-14 Protéine de fusion comprenant il-12 et gm-csf Pending WO2025221669A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202463636026P 2024-04-18 2024-04-18
US63/636,026 2024-04-18

Publications (1)

Publication Number Publication Date
WO2025221669A1 true WO2025221669A1 (fr) 2025-10-23

Family

ID=97404242

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2025/024551 Pending WO2025221669A1 (fr) 2024-04-18 2025-04-14 Protéine de fusion comprenant il-12 et gm-csf

Country Status (1)

Country Link
WO (1) WO2025221669A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7141651B2 (en) * 1999-08-09 2006-11-28 Emd Lexigen Research Center Corp. Multiple cytokine protein complexes
US20110182905A1 (en) * 2007-11-13 2011-07-28 Evec Inc. Monoclonal antibodies that bind to hgm-csf and medical compositions comprising same
US20190298769A1 (en) * 2018-03-08 2019-10-03 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating cancer and infectious diseases
US20220213161A1 (en) * 2015-09-09 2022-07-07 Beijing Bio-Targeting Therapeutics Technology Inc. Modified interleukin 12 and use thereof in preparing drugs for treating tumours
US20230116843A1 (en) * 2017-05-18 2023-04-13 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2025024674A2 (fr) * 2023-07-26 2025-01-30 Promab Biotechnologies, Inc. Méthode de traitement de tumeurs locales et distantes

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7141651B2 (en) * 1999-08-09 2006-11-28 Emd Lexigen Research Center Corp. Multiple cytokine protein complexes
US20110182905A1 (en) * 2007-11-13 2011-07-28 Evec Inc. Monoclonal antibodies that bind to hgm-csf and medical compositions comprising same
US20220213161A1 (en) * 2015-09-09 2022-07-07 Beijing Bio-Targeting Therapeutics Technology Inc. Modified interleukin 12 and use thereof in preparing drugs for treating tumours
US20230116843A1 (en) * 2017-05-18 2023-04-13 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
US20190298769A1 (en) * 2018-03-08 2019-10-03 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating cancer and infectious diseases
WO2025024674A2 (fr) * 2023-07-26 2025-01-30 Promab Biotechnologies, Inc. Méthode de traitement de tumeurs locales et distantes

Similar Documents

Publication Publication Date Title
Hu et al. Nucleofection with plasmid DNA for CRISPR/Cas9-mediated inactivation of programmed cell death protein 1 in CD133-specific CAR T cells
ES2774160T3 (es) Receptores de antígenos quiméricos biespecíficos y usos terapéuticos de los mismos
TW201842921A (zh) 治療性rna
CN112105648A (zh) 细胞介导的外泌体递送
JP2019532640A (ja) 免疫原性が低下したhlaクラスi欠損nk−92細胞
EP4135665A2 (fr) Protéines de liaison modulaire pour vésicules extracellulaires et leurs utilisations
CN106519037A (zh) 可活化的嵌合受体
KR102490457B1 (ko) 단백질 발현용 mRNA 구조체 및 이의 용도
CA3241014A1 (fr) Compositions therapeutiques d'arnm
JP2024521800A (ja) 操作されたキメラ融合タンパク質組成物およびそれらの使用の方法
WO2024023504A1 (fr) Vésicule extracellulaire chargée
US20250000975A1 (en) Preparation for chimeric antigen receptor immune cell constructed on basis of gas6 and use of chimeric antigen receptor immune cell
US20240277769A1 (en) Preparation and application of chimeric antigen receptor immune cell constructed on basis of lox1
EP4168562A2 (fr) Méthodes et compositions pour moduler des cellules et des membranes cellulaires
CA3193508A1 (fr) Procedes et compositions ameliores pour l'expression d'acides nucleiques dans des cellules
US11364265B1 (en) Recombinant erIL-15 NK cells
CN120390656A (zh) 环状rna组合物
WO2025221669A1 (fr) Protéine de fusion comprenant il-12 et gm-csf
US20220259284A1 (en) Ca2 compositions and methods for tunable regulation
Xu et al. PD-L1 antibody-modified plant-derived nanovesicles carrying a STING agonist for the combinational immunotherapy of melanoma
Burón et al. Natural killer cells in adoptive cell therapy: current landscape of genetic engineering strategies
WO2025024674A2 (fr) Méthode de traitement de tumeurs locales et distantes
CN119730863A (zh) 用car rna-lnp转染的nk细胞
US20210017246A1 (en) Il-13 receptor alpha 2 targeted, zetakine directed t cell immunotherapy
CN117230096A (zh) 一种用于构建mRNA CAR-T细胞的生物制剂、制备方法及应用