WO2018030806A1 - Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same - Google Patents

Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same Download PDF

Info

Publication number
WO2018030806A1
WO2018030806A1 PCT/KR2017/008676 KR2017008676W WO2018030806A1 WO 2018030806 A1 WO2018030806 A1 WO 2018030806A1 KR 2017008676 W KR2017008676 W KR 2017008676W WO 2018030806 A1 WO2018030806 A1 WO 2018030806A1
Authority
WO
WIPO (PCT)
Prior art keywords
region
cells
protein
domain
mil
Prior art date
Application number
PCT/KR2017/008676
Other languages
French (fr)
Korean (ko)
Inventor
김용성
정근옥
하지희
Original Assignee
아주대학교산학협력단
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
Priority to BR112019002394-1A priority Critical patent/BR112019002394B1/en
Priority to EP17839824.4A priority patent/EP3511340A4/en
Application filed by 아주대학교산학협력단 filed Critical 아주대학교산학협력단
Priority to US16/323,839 priority patent/US10696722B2/en
Priority to JP2019506697A priority patent/JP6993403B2/en
Priority to MX2019001651A priority patent/MX2019001651A/en
Priority to AU2017310163A priority patent/AU2017310163B2/en
Priority claimed from KR1020170101594A external-priority patent/KR102050463B1/en
Priority to SG11201901071TA priority patent/SG11201901071TA/en
Priority to CN201780062851.0A priority patent/CN110267977A/en
Priority to CA3033475A priority patent/CA3033475A1/en
Publication of WO2018030806A1 publication Critical patent/WO2018030806A1/en
Priority to ZA2019/00772A priority patent/ZA201900772B/en
Priority to US16/886,177 priority patent/US11078249B2/en
Priority to US16/886,184 priority patent/US11692019B2/en
Priority to AU2021273642A priority patent/AU2021273642A1/en
Priority to US18/323,124 priority patent/US20230416325A1/en

Links

Images

Classifications

    • 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]
    • 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
    • A61K38/22Hormones
    • A61K38/24Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g. HCG; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]
    • 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]
    • 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
    • C07K14/575Hormones
    • C07K14/59Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g.hCG [human chorionic gonadotropin]; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]

Definitions

  • the present invention includes a first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
  • Heterodimeric Fc-fused protein having a subunit of a bioactive protein bound to at least one of the N-terminus or C-terminus of the first Fc region and / or the second Fc region.
  • the first Fc region and the second Fc region are heterodimeric Fc-fused proteins, characterized in that the CH3 domain is mutated to promote the formation of heterodimers and
  • It relates to a pharmaceutical composition comprising the heterodimer-fusion protein.
  • Heterodimeric Fc-fused protein according to the present invention is a form and structure in which two or more subunits form a protein complex so that a protein constituting one bioactive protein exists in nature. It can be fused to the antibody heavy chain constant region (Fc), there is an advantage that can maintain the activity as it exists in nature.
  • Fc antibody heavy chain constant region
  • the half-life of the bioactive protein contained in the heterodimer-fusion protein is significantly increased, and various biological activities in the body are maintained for a long time. There is an advantage that can be sustained.
  • heterodimer-fusion protein in which the subunit of the bioactive protein is fused to the N-terminus or C-terminus of the antibody heavy chain constant region heterodimer Fc according to the present invention is expressed after purification, compared with the wild type Fc-based fusion protein. This is easy.
  • Immunoglobulin G IgG
  • IgM immunoglobulin M
  • IgD immunoglobulin D
  • IgE immunoglobulin A
  • homodimerization between two identical heavy chains is performed by the final domain of the constant region of the antibody (CH3 domain for IgG, IgD, IgA, CH4 domain for IgM, CH2 and CH4 domain for IgE). It is induced by non-covalent interactions between and disulfide bonds between hinge regions.
  • Antibody-derived heterodimeric heavy chain constant region (Hterodimeric Fc) technology allows specific non-covalent linkages between the last domain of the constant region (CH3 domain), which contributes significantly to the homologous duplication of earlier spontaneous antibodies (IgG, IgM, IgA, IgD, IgE). Heterogeneous redundancy is preferred, and it is a technique to create heterologous heavy chain invariant regions by engineering them to have a bond that is not preferred or rejected. More specifically, genetic modifications induce mutations in the CH3 domains of two different antibody heavy chains, forming two heterozygotes with very similar structures to naturally occurring antibodies and minimal variation in sequence. (US Patent No. 7,695,936; Korean Patent No. 1,522,954).
  • the heterodimeric heavy chain constant region technology is a basic technology for making double antibodies.
  • CH3 domain mutants known to induce heterodimers are known to be mostly asymmetric by structure-based rational design of the antibody on the surface of CH3 domain interaction. It was prepared by introducing a red pair of mutations (Spreter Von Kreudenstein et al., 2014).
  • the A107 mutant used in the present invention is a high yield heterodimeric heavy chain constant region selected from a human antibody heteroduplex heavy chain constant region library prepared using a yeast cell surface expression system, and a hydrophobic core of the CH3 domain interaction surface. Induces mutations in the conserved and charged amino acids to form complementary hydrophobic bonds (K409W CH3A -D399V / F405T CH3B ) and to form hydrogen bonds (K370E CH3A -E357N CH3B ) to form heterodimers (Choi et al. 2016; Korean Patent Application No. 2015-0142181).
  • the native protein complex structure existing in nature is formed as it is, and the activity of the original protein is not only properly expressed. There is no such form of development that can be sustained for a long time.
  • heterodimer variants comprising Fc regions from IgG1 as well as other previously reported isotype antibodies such as IgG2, IgG3, and IgG4, Using this, two or more different subunits are formed, and two or more subunits form one protein complex to bind one or more subunits of a protein showing physiological activity to the ends of the Fc region.
  • the present invention was completed by developing a novel therapeutic fusion protein in the form of a heterodimeric Fc-fused protein.
  • a protein consisting of two or more different subunits in the present invention, wherein the two or more subunits form a protein complex and exhibit physiological activity is preferably Interleukin-12, IL-12) can be used.
  • Interleukin-12 increases the activity of immune cells, such as cytotoxic T Lymphocytes (CTL) and natural killer cells (NK), among other immune cells, to directly kill tumors or interferon Inhibits tumorigenesis by activating the immune response in the tumor microenvironment where the immune response is suppressed through the secretion of pro-inflammatory cytokine such as gamma (IFN- ⁇ )
  • CTL cytotoxic T Lymphocytes
  • NK natural killer cells
  • an optional tag for further purification only at the C terminus of one Fc region as shown in FIGS.
  • the method of constructing a fusion protein has been used by fusion or by purifying the Fc region and protein separately in high purity.
  • this form is not only costly in producing large amounts of protein, but also requires research to further optimize the purification process.
  • the problem to be solved in the present invention is a heterozygote-fusion heterozygous form of a new type of antibody heavy chain constant region in which two or more subunits form a protein complex so that the activity of the protein showing physiological activity can be sufficiently maintained for a long time. It is to provide a protein (heterodimeric Fc-fused protein).
  • the antibody heavy chain constant region heterodimeric Fc-fused protein according to the present invention has two or more subunits that form a protein complex to form a protein complex that exhibits physiological activity as it exists in nature. By simulating as much as possible, it is a form which can maintain the activity as it exists in nature.
  • the half-life of the fusion protein is significantly increased by fusion of the antibody heavy chain constant region, so that various physiological activities in the body can be maintained for a long time.
  • the present invention also provides a pharmaceutical composition comprising the antibody heavy chain constant region heterodimeric Fc-fused protein, and a composition and treatment method for the treatment of diseases, in particular cancer, using the same. do.
  • the present invention includes a first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
  • heterodimeric Fc-fused protein in which a bioactive protein is bound to at least one of the N-terminus or C-terminus of the first Fc region and / or the second Fc region,
  • the physiologically active protein is composed of two or more different subunits (subunit), two or more different subunits (subunit) is characterized in that to form a protein complex (protein complex) to show the physiological activity,
  • the first Fc region and the second Fc region provide a heterodimeric Fc-fused protein, characterized in that the CH3 domain is mutated to promote formation of a heterodimeric Fc.
  • the present invention also provides a pharmaceutical composition comprising the heterologous-fusion protein and a composition and a method for the treatment of diseases, in particular cancer, using the same.
  • 1 (A) to 1 (C) is a diagram showing a strategy for obtaining a fusion protein in the form of monomers and heterodimers using existing wild-type human antibody heavy chain constant region.
  • FIG. 1 (D) shows an antibody-cytokine (Immunocytokine) construct by fusing monomeric cytokines to an IgG-type antibody containing a KnoH-in-hole heterodimeric heavy chain constant region (KiH heterodimeric Fc) variant in the existing literature. It is a figure which shows an example.
  • Figure 2 (A) and 2 (B) is a diagram showing the monomer and heterodimer fusion protein forms that can be constructed using heterodimeric heavy chain constant region.
  • Figure 2 (C) is a diagram showing a heterodimeric fusion protein form in a human antibody of the IgG form containing a heterodimeric heavy chain constant region.
  • Figure 3 is a view showing the results of selecting and comparing the sequence of the sequence of each human antibody immunoglobulin G isotype CH3 domain for the production of CH3 domain variants for heterodimer formation by human antibody isotype.
  • FIG. 4 is a structural modeling of heterologous heavy double chain constant region variants of the heterologous type having the mutation-induced sequence at the position selected in FIG. 3, and the resulting modeling structure is compared with the wild type IgG1 based A107 mutant and It is a figure which shows the result of an analysis.
  • FIG. 5 is a schematic diagram of a vector for expressing heterologous heavy chain constant regions of homologous type constructed by sequence and structural analysis in animal cells. Heterozygous heavy chain constant region variants of each homotype, including the mutated hinge region, were cloned into the vector using the restriction enzyme NotI / HindIII.
  • FIG. 6 is a schematic diagram schematically illustrating a scFv-Fc CH3A / Fc CH3B co-expression system for evaluating the degree of heterodimer formation of heteroduplex heavy chain constant region variants due to the duplex size difference of the expressed protein. to be.
  • FIG. 7 shows a single-chain antibody fragment (scFv) fused with a single chain antibody fragment (scFv) constructed to evaluate the formation yield of heteroduplex of the antibody heavy chain constant region by the CH3 mutation pair as shown in FIG. 6. .1 Schematic diagram for cloning into vectors.
  • Figure 8 is a transient expression of co-transformation in HEK293F cells for evaluating the formation ability of the heteroduplex described in Figure 6 animal cell expression vector introduced with a CH3 mutation pair constructed in accordance with the expression system described in Figures 5 and 7 And, after purification, 5 ⁇ g of protein was isolated on SDS-PAGE under non-reducing conditions for evaluation of heterodimer antibody formation ability, and analyzed by size and combination through Coomasie Blue staining. At this time, wild type Fc using wild type CH3 was used as a negative control.
  • FIG. 9 is a diagram showing the result of Western blot using the anti-human IgG-AP conjugated antibody with AP enzyme after protein separation by SDS-PAGE in the same manner as in FIG. 8. .
  • Figure 10 (A) is a schematic diagram showing the form of the endogenous interleukin 12 cytokine unfused to the heavy chain constant region, which is a control of the present invention.
  • FIG. 10 (B) is a schematic diagram showing the form of a bi-IL-12-Fc fusion protein in which an interleukin 12 cytokine linked with an amino acid linker is fused to a wild-type IgG4 Fc as a comparative example of the present invention.
  • FIG. 10 (C) is a schematic diagram showing the form of a mono-IL-12-Fc fusion protein in which the interleukin 12 cytokine is fused with a ⁇ 4-A107 variant made of IgG4 in a heterologous heavy double chain constant region variant of the present invention. to be.
  • FIG. 10C are schematic diagrams of vectors for expressing and purifying the fusion protein of the embodiment of the present invention (FIG. 10C) in animal cells.
  • Figure 12 is a schematic diagram of a vector for the expression and purification of the fusion protein of the comparative example (Fig. 10 (B) in the animal cell of the present invention.
  • FIG. 13 transiently expresses and purifies the animal cell expression vectors of FIGS. 11 (A) and (B) constructed with human and mouse interleukin genes through cotransformation into HEK293F cells, and then amplifies 5 ⁇ g of protein. It is a diagram showing the results of the analysis on the size and combination form by separating on the SDS-PAGE of the condition, Coomasie Blue staining.
  • FIG. 14 shows the results of analyzing the fusion proteins of FIG. 13 using size exclusion chromatography.
  • FIG. 15 shows peripheral blood immune cells (PHA-activated PBMCs) that induced receptors for IL-12 by treating normal peripheral blood immune cells (normal PBMCs) having no receptor for IL-12 and phytohaemagglutinin (PHA). It is a figure which shows the result of having confirmed the binding ability of the mono-hIL-12-Fc and the wild type bi-hIL-12-Fc which were constructed about with the flow cytometer (FACS).
  • PHA-activated PBMCs peripheral blood immune cells
  • FACS flow cytometer
  • FIG. 16 shows Fc (A107), recombinant human IL-12 (rhIL-12), and bi-hIL-12 in peripheral blood immune cells (PHA-activated PBMCs) inducing receptors for IL-12 by treating PHA, a mitogen.
  • Figure showing the results of measuring the cell proliferation according to the concentration-specific treatment of -Fc and mono-hIL-12-Fc through the WST-1 cell proliferation assay.
  • FIG. 17 is a view showing the results of measuring the concentration of IFN- ⁇ in the culture supernatant cultured in Figure 16 by ELISA
  • FIG. 18 shows normal peripheral blood binding ability of mono-mIL-12-Fc and bi-mIL-12-Fc constructed using the property that mouse IL-12 binds to human IL-12 receptor as well as mouse IL-12 receptor.
  • PHA peripheral blood immune cells
  • FIG. 19 shows Fc (A107), recombinant mouse IL-12 (rmIL-12), bi-mIL-12-Fc in peripheral blood immune cells (PHA-activated PBMCs) inducing receptors for IL-12 by PHA treatment.
  • cell proliferation according to the concentration-specific treatment of mono-mIL-12-Fc is a diagram showing the results measured by the WST-1 cell proliferation assay.
  • Figure 20 (A) shows Fc (A107), rmIL-12, bi-mIL-12-Fc and mono-mIL-12-Fc when the tumor size is 100 mm3 in Balb / c mice transplanted with CT26 HER2 / Neu cancer cells It is a diagram showing the results of confirming the change in tumor volume measured during intraperitoneal administration and the size of tumor after lethality of mice at the end of administration.
  • FIG. 20 (B) is a graph showing the weight change of the mouse periodically measured during the experiment of FIG. 20 (A).
  • FIG. 21 (A) shows bi-mIL-12-Fc and mono-mIL-12-Fc intraperitoneally administered twice a week at the tumor size of 300 mm 3 in CT26 HER2 / Neu transplanted Balb / c mice. It is a figure which measured the change of the tumor volume.
  • Figure 21 (B) is a graph showing the change in the volume of the tumor of the individual mice periodically measured in the experimental procedure of Figure 21 (A).
  • Figure 21 (C) is a diagram showing the result of confirming the tumor size by killing the mouse 3 days after the last administration of Figure 21 (A).
  • Figure 21 (D) is a graph showing the weight change of the mouse periodically measured in the experimental procedure of Figure 21 (A).
  • FIG. 21 (E) is a graph of alanine aminotransferase (ALT) which is an indicator of liver toxicity by collecting blood from the facial vein of the mouse on day 1 after the last administration of FIG. 21 (A).
  • ALT alanine aminotransferase
  • Figure 22 (A) is a graph measuring the increase in the number of CD4 + T cells, CD8 + T cells and NK cells in the spleen after 3 days of the last administration of Figure 21 (A).
  • Figure 22 (B) is a view showing the total number of immune cells, CD4 + T cells, CD8 + T cells infiltrated into the tumor after 3 days of the third administration of Figure 21 (A).
  • Figure 23 (A) is the result of measuring the IFN-g concentration in the serum isolated by collecting blood from the facial vein of the mouse 24 hours after the last administration of Figure 21 (A) by ELISA.
  • FIG. 23 (B) shows bi-mIL-12-Fc and mono-mIL-12 at a molar concentration of 1 ⁇ g rmIL-12 when the tumor size was 300 mm 3 in Balb / c mice transplanted with CT26 HER2 / Neu cancer cells.
  • Figure 23 (C) is a graph measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of cytotoxic T cells isolated from the spleen by killing the mouse 3 days after the last administration of Figure 21 (A).
  • Figure 23 (D) shows the cytotoxic effect of cytotoxic T cells isolated from the spleen by killing mice 3 days after the third administration of Figure 21 (A) does not express CT26 HER2 / Neu cancer cells expressing tumor antigens It is a figure analyzed by the flow cytometer using 4T1 cells.
  • Figure 23 (E) is a graph measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of natural killer cells isolated from the spleen by killing the mouse 3 days after the third administration of Figure 21 (A).
  • Figure 24 (A) is a graph measuring the number of CD8 + effect T cells isolated from the spleen by lethal mice three days after the last administration of Figure 21 (A).
  • Figure 24 (B) is a graph measuring the number of CD8 + effect memory T cells isolated from the spleen by lethal mice three days after the last administration of Figure 21 (A).
  • Figure 24 (C) is a graph measuring the number of CD8 + central memory T cells isolated from the spleen by killing the mouse three days after the last administration of Figure 21 (A).
  • FIG. 24 (D) shows CT26 HER2 / Neu in Balb / c mice of the same age as those surviving 120 days after administration of 1 ⁇ g mono-IL-12-Fc in FIG. 21 (A). Cancer cells are transplanted to measure the change in the tumor volume of the mouse.
  • FIG. 24 (E) shows memory precursor effect cells (KLRG1 - IL-7R + ) and short-lived effect cells (KLRG1) among CD8 + T cells present in the spleen after 3 days of the third administration of FIG. 21 (A).
  • + IL-7R - is a diagram of analyzing the percentage of by flow cytometry (flow cytometry).
  • FIG. 25 (A) shows the ratio of CD8 + T cells with high expression of T-bet, a transcription factor that inhibits the differentiation of memory cells by killing mice after 3 days of the third administration of FIG. 21 (A). It is a graph analyzed by measuring with a flow cytometer.
  • FIG. 25 (B) shows CD8 + T with high expression of Eomes and low expression of T-bet that promote the differentiation of memory cells by killing mice after 3 days of the third administration of FIG. 21 (A). It is a graph analyzed by measuring the percentage of cells by flow cytometry.
  • Figure 25 (C) shows CT26 HER2 / Neu Balb / c mice transplanted with cancer cells were intraperitoneally administered with bi-mIL-12-Fc and mono-mIL-12-Fc at a molar concentration of 1 ⁇ g rmIL-12 when the tumor size was 300 mm 3. After time, the expression level of phosphorylated STAT4 in CD8 + T cells isolated from inguinal lymph nodes was measured by flow cytometry.
  • Figure 25 (D) is a graph measuring the ratio of CD8 + T cells expressing T-bet inhibiting the differentiation of memory cells in the inguinal lymph nodes 72 hours after one intraperitoneal administration of Figure 25 (C) by flow cytometry.
  • FIG. 25 (F) shows that CD8 + T cells isolated from the spleen and inguinal lymph nodes of normal Balb / c mice were stimulated with mono-mIL-12-Fc and bi-mIL-12-Fc cross-linked with an antibody that recognizes Fc.
  • the ratio of CD8 + T cells expressing T-bet is a graph measured by flow cytometry.
  • FIG. 26 is a schematic diagram showing the differentiation-induced mechanism of memory precursor effector and memory cells induced by mono-mIL-12-Fc and the differentiation-induced mechanism of short-lived effect cells induced by bi-mIL-12-Fc.
  • the present invention includes a first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
  • heterodimeric Fc-fused protein in which a bioactive protein is bound to at least one of the N-terminus or C-terminus of the first Fc region and / or the second Fc region,
  • the bioactive protein is composed of two or more different subunits (subunit), the two or more different subunits (subunit) is characterized in that the protein complex (protein complex) to form a physiological activity,
  • the first Fc region and the second Fc region are for a heterodimeric Fc-fused protein, characterized in that the CH3 domain is mutated to promote formation of a heterodimer.
  • Fc region or “heavy chain constant region” means a region including a CH2 domain, a CH3 domain, and a hinge domain derived from an antibody. However, in the case of IgE, it means a region including a CH2 domain, a CH3 domain, a CH4 domain, and a hinge domain.
  • the expression “the first Fc region and the second Fc region are mutated to promote the formation of heterodimers” indicates that antibodies present in nature are homodimers in which two Fc regions have the same sequence. (homodimer) form, which causes mutations in some sequences of these Fc regions, thereby promoting the formation of heterodimers through specific non-covalent linkages between the first and second Fc regions and homodimers. It means that the formation of is reduced or preferably mutated so that it hardly occurs.
  • the mutations to facilitate the formation of a heterodimer of the first Fc region and the second Fc region according to the present invention are each of the CH3 domains included in the antibody-derived first and second Fc regions. May include mutations that facilitate the formation of this heterodimer.
  • heterodimeric Fc or Fc heterodimer includes a first Fc region and a second Fc region, and the first Fc region and the second Fc region are heterodimers. It means a heterodimer, characterized in that the CH3 domain is modified to promote the formation of).
  • the first Fc region and the second Fc region in the present invention may be derived from an Fc region selected from the group consisting of human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, and IgE, respectively, preferably Wherein the first Fc region and the second Fc region are derived from IgG1, IgG2, IgG3, or IgG4, respectively,
  • first Fc region and the second Fc region may be characterized in that it is derived from an isotype antibody.
  • the mutation of the CH3 domain may be characterized in that it comprises one or more mutations selected from the following group. All mutation positions in the present invention are according to the EU index.
  • substitution of the amino acid residue at the K370 position of the CH3 domain of the first Fc region is K370E, K370R, K370M, K370D or K370H,
  • substitution of the amino acid residue at the E357 position of the CH3 domain of the second Fc region may be E357N, E357D, E357A, E357I, E357G or E357M, and the substitution of the amino acid residue at the S364 position may be S364T or S364W. .
  • substitution of the amino acid residue at the K409 position in the CH3 domain of the first Fc region is K409W
  • the substitution of the amino acid residue at the F405 position of the CH3 domain of the second Fc region is F405T
  • amino acid residue at the D399 position is The substitution may be characterized as being D399V.
  • amino acid residue change such as K370E means that the K at position 370 is changed to E, and all amino acid residue changes in the present invention are used as the same meaning.
  • the mutation of the CH3 domain of the first Fc region or the second Fc region may include one or more mutations selected from the following group. (However, the mutation position is according to the EU index)
  • the CH3 domains of the first Fc region and the second Fc region as described above may further include the following bonds.
  • the mutation of the CH3 domain may be characterized in that it comprises one or more mutations selected from the following group.
  • the substitution of the amino acid residue at the K360 position of the CH3 domain of the first Fc region is K360E
  • the substitution of the amino acid residue at the E347 position of the CH3 domain of the second Fc region may be E347R.
  • Substitution of the amino acid residue at the K409 position of the CH3 domain of the first Fc region is K409W
  • Substitution of the amino acid residue at the F405 position of the CH3 domain of the second Fc region is F405T
  • Substitution of the amino acid at the D399 position is It may be characterized by the D399V.
  • the mutation of the CH3 domain of the first Fc region or the second Fc region may include one or more mutations selected from the following group. (However, the mutation position is according to the EU index)
  • the CH3 domains of the first Fc region and the second Fc region as described above may further include the following bonds.
  • the CH3 domains included in the antibody-derived first and second Fc regions of the present invention are each
  • It may be characterized by having a sequence selected from the group consisting of the amino acid sequence represented by the sequence number of.
  • the antibody-derived first Fc region and the second Fc region preferably have a sequence of the CH3 domain described in Table 1 derived from IgG4.
  • the subunit of the bioactive protein may be bound only to one of the N-terminus or the C-terminus of the first Fc region or the second Fc region,
  • One or more different subunits of one (one) bioactive protein may be respectively bound to the N-terminus or C-terminus of the first Fc region and the second Fc region (FIG. 2 ( B) and FIG. 2 (C)).
  • the "subunit of the bioactive protein is bonded only to one terminal of either the N-terminus or the C-terminus of the first Fc region or the second Fc region", the first Fc region or the second Fc One of the subunit (s) of the bioactive protein is bound to either end of the N-terminal or C-terminal end of the region, and the remaining subunit (s) of the bioactive protein are linker-mediated.
  • the linker is preferably an amino acid linker, but is not limited thereto.
  • the one or more different subunits of one (one) bioactive protein is respectively bonded to each of the N-terminal or C-terminal of the first Fc region and the second Fc region.
  • One or more different subunit (s) of the bioactive protein are bound to both the first Fc region and the second Fc region N-terminus, respectively, or the bioactive protein is bound to both the C-terminus of the first Fc region and the second Fc region.
  • one or more different subunits of the bioactive protein are respectively bound to both the N-terminus and the C-terminus of the first Fc region and the second Fc region.
  • the binding of the terminal of the first Fc region and / or the second Fc region and the subunit of the bioactive protein is genetic fusion. It can be characterized by fused by
  • first Fc region and the second Fc region and a subunit of the bioactive protein may be combined in a linker-mediated form.
  • the linker is preferably an amino acid linker, but is not limited thereto.
  • heterodimeric-fusion protein heterodimeric Fc-fused protein
  • the bioactive protein is composed of two or more different subunits, and the two or more different subunits are characterized by forming a (one) protein complex to represent the biological activity.
  • the bioactive protein is selected from the group consisting of interleukin 12 (IL-12), interleukin 23 (IL-23), interleukin 27 (IL-27), interleukin 35 (IL-35), and follicle stimulating hormone (FSH).
  • IL-12 interleukin 12
  • IL-23 interleukin 23
  • IL-27 interleukin 27
  • IL-35 interleukin 35
  • FSH follicle stimulating hormone
  • the most preferred bioactive protein according to the present invention is interleukin 12 (IL-12).
  • IL-12 Interleukin-12
  • IL-12 consists of two subunits of p35 (IL-12A) and p40 (IL-12B), and its bioactive form is p70, a heterodimer of p35 and p40. . In nature, IL-12 must be present in the form of p70, a heterodimer of p35 and p40 in order to have activity.
  • the antibody heavy chain constant region heterologous double-fusion protein according to the present invention was implemented.
  • At least one subunit of a bioactive protein comprising a first Fc region and a second Fc region according to the present invention, at one or more of the ends of the first and second Fc regions Heterodimeric Fc-fused protein in the antibody heavy chain constant region to which (subunit) is bound,
  • At least one subunit (s) constituting one bioactive protein is bound only at either end of the N-terminus or C-terminus of the first Fc region and the second Fc region, and the rest
  • the subunit (s) may be linked by a linker
  • one or more different subunit (s) of one bioactive protein may be bound to each of the N-terminus and / or C-terminus of the first Fc region and the second Fc region, respectively. have.
  • the p35 or p40 subunit is attached to only one terminal of either the N-terminus or the C-terminus of the first Fc region or the second Fc region, and the remaining subunits are the first Fc region or the second Fc region.
  • the linker may be linked to a subunit of p35 or p40 that is bound to either the N-terminus or C-terminus of the Fc region to form a heterodimer-fusion protein (FIGS. 2B and 2C). ) Reference).
  • any one subunit selected from p35 and p40 is coupled to the N-terminus or C-terminus of the first Fc region, and the other subunit is N-terminus or the C-terminus of the second Fc region.
  • Units can form heterodimer-fusion proteins having a combined form (see FIGS. 2 (B) and 2C).
  • the physiologically active protein is interleukin 12 (IL-12),
  • the p35 or p40 subunit of interleukin 12 is bound to only one of the N-terminus or C-terminus of the first Fc region or the second Fc region, and the remaining subunits are linker-mediated. Form a subunit bound to either the N-terminus or C-terminus of the 1 Fc region or the second Fc region, or
  • P35 and p40 subunits of interleukin 12 are respectively bound to the N-terminus or C-terminus of the first Fc region and the second Fc region.
  • heterodimeric-fusion protein heterodimeric Fc-fused protein
  • the hinge region included in the N-terminus in the first Fc region and the second Fc region may be characterized by mutation of a cysteine residue included in the hinge region.
  • the mutation of the cysteine residue in the hinge region is a cysteine residue of the upper hinge region except for the cysteine residue in the core hinge region for heterodimer formation.
  • the first Fc region and the second Fc region are included in a whole antibody (whole antibody) form consisting of human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD and IgE. can do.
  • the "whole antibody form” means the CH2 domain, CH3 domain and hinge region (including CH4 domain in IgE) in the Fc region in the case of IgG, IgA and IgD, in addition to the CH1 domain, VH.
  • intact form is meant an antibody comprising a domain, a CL domain and a VL domain.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the antibody heavy chain constant region heterodimer-fusion protein according to the present invention.
  • the use of the pharmaceutical composition according to the invention is characterized in that it depends on the use of the bioactive protein contained in the antibody heavy chain constant heterodimer-fusion protein.
  • the bioactive protein contained in the antibody heavy chain constant region heterodimer-fusion protein according to the present invention is IL-12 or one or more subunits thereof, and thus the present invention includes IL-12 as a bioactive protein. It provides a pharmaceutical composition for the treatment of cancer comprising an antibody heavy chain constant region heterodimer-fusion protein.
  • Cancers treatable with a pharmaceutical composition for treating cancer comprising an antibody heavy chain constant region heterodimer-fusion protein comprising IL-12 or one or more subunits thereof as the bioactive protein include colorectal cancer, melanoma, breast cancer, Pancreatic cancer, kidney cancer, prostate cancer, ovarian cancer, small intestine cancer, esophageal cancer, cervical cancer, lung cancer, lymphoma and hematologic cancer may be selected from the group consisting of, but not limited to.
  • compositions according to the invention may additionally include a pharmaceutically acceptable carrier.
  • a “pharmaceutically acceptable carrier” is a substance that can be added to the active ingredient to help formulate or stabilize a formulation and does not cause significant deleterious toxic effects on the patient.
  • the carrier refers to a carrier or diluent that does not irritate the patient and does not inhibit the biological activity and properties of the heterodimer-fusion protein according to the present invention.
  • Acceptable pharmaceutical carriers in compositions formulated as liquid solutions are sterile and physiologically compatible, including saline, sterile water, Ringer's solution, buffered saline, albumin injectable solutions, dextrose solution, maltodextrin solution, glycerol, ethanol and One or more of these components may be mixed and used, and other conventional additives such as antioxidants, buffers and bacteriostatic agents may be added as necessary.
  • diluents may be additionally added to formulate injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like.
  • Other carriers are described, for example, in Remington's Pharmaceutical Sciences (E. W. Martin).
  • compositions include sterile aqueous solutions or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersions for immediate administration.
  • the use of such media and agents for pharmaceutically active substances is known in the art.
  • the composition is preferably formulated for parenteral injection.
  • the compositions may be formulated as solutions, microemulsions, liposomes, or other ordered structures suitable for high drug concentrations.
  • the carrier can be, for example, a solvent or dispersion medium containing water, ethanol, polyols (eg glycerol, propylene glycol and liquid polyethylene glycols, etc.) and suitable mixtures thereof.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol or sodium chloride in the composition.
  • Sterile injectable solutions can be prepared by incorporating the required amount of the heterodimer-fusion protein in an appropriate solvent with one or a combination of ingredients described above as required, followed by sterile microfiltration.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those described above.
  • sterile powders for the preparation of sterile injectable solutions some methods of preparation include vacuum drying and freeze-drying (freeze-drying), which produce a powder of the active ingredient and any further desired ingredients from its presterilized-filtered solution. )to be.
  • compositions according to the invention may be administered orally or parenterally at dosages and frequencies that may vary depending on the severity of the suffering patient.
  • the composition may be administered to the patient as a bolus or by continuous infusion as needed.
  • the pharmaceutical compositions according to the invention may be administered rectally, intravenously, subcutaneously, intrauterinely or intratracerebrovascularly, but are not limited thereto.
  • the pharmaceutical composition for treating cancer comprising the antibody heavy chain constant region heterodimer-fusion protein comprising IL-12 may be used for combination therapy with other anticancer agents, and the other anticancer agents may be cytotoxic T.
  • Cells and / or Natural Killer (NK) cells are preferred, but are not limited to any of the other anticancer agents that can be used in the art and can be used for combination therapy.
  • a pharmaceutical composition for treating cancer comprising an antibody heavy chain constant region heterodimer-fusion protein comprising interleukin 12 is used for combination treatment with cytotoxic T cells and / or Natural Killer (NK) cells. Occation,
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CTL cytotoxic T lymphocytes
  • CTLs cytotoxic T lymphocytes
  • the present invention provides a method of treating or preventing a disease comprising administering to a patient in need thereof a pharmaceutical composition comprising an antibody heavy chain constant region heterodimer-fusion protein according to the present invention.
  • the treatable or preventable disease depends on the use of the bioactive protein contained in the antibody heavy chain constant heterodimer-fusion protein,
  • the present invention provides a method of treating or preventing a patient suffering from cancer selected from the group consisting of breast cancer, pancreatic cancer, kidney cancer, prostate cancer, ovarian cancer, small intestine cancer, esophageal cancer, cervical cancer, lung cancer, lymphoma and hematologic cancer.
  • Example 1 Design of antibody heavy chain constant region CH3 domain variants for heterodimer formation by human antibody isotype (sequence analysis)
  • Figure 3 is a comparison of the sequence of each human antibody immunoglobulin G (IgG) isotype CH3 domains listed.
  • IgG human antibody immunoglobulin G
  • Each amino acid sequence was identified in the International ImMunoGeneTics information system (IMGT; URL: http://www.imgt.org/).
  • IMGT International ImMunoGeneTics information system
  • the sequence of G3m (s, t) which has been reported to maintain serum half-life similar to other IgG isotypes among various Gm allotypes, was used (Stapleton NM et al., 2011). ).
  • the 409th amino acid sequence unlike IgG1, IgG2, and IgG3, had a sequence conserved in all homotypes at the positions of the other mutations except that it had a different sequence from IgG4 to arginine. It was.
  • the position having the same amino acid sequence number was selected as a position for transplanting the A107 mutant pair into an isoform other than IgG1. All amino acid position notations in the present invention follow the EU index (numbering).
  • Example 2 Design of antibody heavy chain constant region CH3 domain variants for heterodimer formation by human antibody isotype (structural modeling)
  • each mutant shown in FIG. 3 shows whether the mutation can be stably introduced to form a heterodimer.
  • Structural modeling uses an online modeling server (URL: https://swissmodel.expasy.org/; Biasini M et al., 2014) as a template of the structure of an already known Immunoglobulin Fc heterodimer variant (PDB ID: 4X98). Predicted through.
  • the heterologous mutant A107 heavy chain constant region of each isotype designed through the sequence analysis of Example 1 and the structural analysis of Example 2 is a site-directed mutation performed by a person skilled in the art using synthetic oligonucleotides (Macrogen, Korea) Site-directed mutagenesis) was performed using the NotI / HindIII restriction enzyme in-frame to sequence the signal sequence-hinge-CH2-CH3 to the animal cell expression vector pcDNA3.1 (+) (Invitrogen, USA). Cloning was carried out (see FIG. 5).
  • the hinge region used here is a cysteine residue of the upper hinge region except for the cysteine residue in the core hinge region for duplex formation. Substituted with serine residues to prevent formation.
  • serine residues to prevent formation.
  • only 15 amino acids in the C-terminal region of the central hinge region among the 47 amino acid sequences of the G3m (s, t) allotypes maintain high antibody-specific effect functions (ADCC, CDC) of IgG3. This was confirmed through the literature (Dall'Acqua WF et al., 2006), whereby only 15 amino acids of the C terminus of the sequence shown in FIG. 5 were used.
  • Table 2 shows amino acid sequence information of the CH3 region in the wild type and A107 heavy chain constant heterodimeric variant pairs of the present invention.
  • Purified antibodies in the scFv-Fc CH3A / Fc CH3B co-expression system were scFv-Fc CH3A homodimer (103 kDa), scFv-Fc CH3A / Fc CH3B heterodimer (78 kDa), Fc CH3B homodimer (53 kDa) Since the molecular weights of) are different, it is possible to compare the degree of heterodimer formation on SDS-PAGE.
  • Fc CH3B vector a vector constructed in Example 3 was used, and additionally, scFv was introduced only to the N terminus of Fc CH3A , that is, of pcDNA3.1 (+)-scFv-hinge-CH2-CH3A (scFv-Fc CH3A ). Vectors expressed in the format were cloned.
  • Figure 7 shows a schematic diagram of the animal cell expression vector pcDNA3.1 (+)-scFv-hinge-CH2-CH3A (scFv-Fc CH3A ) vector used in the scFv-Fc CH3A / Fc CH3B co-expression system.
  • the scFv antibody used is an antibody connecting the VH and VL regions of hAY4a, which is an enhanced version of the humanized antibody hAY4 that specifically binds DR4 (Lee, Park et al. 2010). Cloning was performed using NotI restriction enzyme and BsiWI restriction enzyme located immediately before the hinge region. Wild type Fc was constructed in the same format (scFv-Fc / Fc) as a control for the variants.
  • HEK293-F cells Upon 200 mL transfection in a shake flask, HEK293-F cells were seeded in 100 ml of medium at a density of 2.0 ⁇ 10 6 cells / ml and incubated at 150 rpm, 8% CO 2 .
  • the resulting heavy and light chain plasmids were diluted in 10 ml FreeStyle 293 expression medium (Invitrogen) with 125 ⁇ g of heavy chain and 125 ⁇ g of light chain in total, 250 ⁇ g (2.5 ⁇ g / ml), with 10 ml of dilute 750 ⁇ g of PEI. The mixture was mixed with the medium (7.5 ⁇ g / ml) and reacted for 10 minutes at room temperature.
  • the reacted mixed medium was put in the cells seeded with 100 ml in advance, incubated at 150 rpm and 8% CO 2 for 4 hours, and then the remaining 100 ml of FreeStyle 293 expression medium was added.
  • the proteins produced by the cells ie antibodies containing heavy chain constant region variants, are secreted out of the cells by the cells and accumulated in the medium. Therefore, the protein was purified using a protein A Sepharose column (GE healthcare) from the cell culture supernatant collected by centrifugation at 2500 rpm for 20 minutes after cell culture.
  • the purification method referred to the standard protocol provided by the Protein A column company, and the purified protein was measured by absorbance at a wavelength of 562 nm using a solution in a BCA protein assay kit (Thermo), and the amount was determined according to the drawn standard curve. was quantified.
  • Example 5 5 ⁇ g of the antibody including the heterologous heterologous heterologous heterologous heterologous variant A107 heavy chain in Example 5 was analyzed on SDS-PAGE under 12% non-reducing conditions (FIG. 8).
  • the homodimer of the CH3A variant was 103 kD
  • the homodimer of the CH3B variant was 53 kD
  • the monomer of the CH3B variant was 25 kD
  • the heterodimer of the CH3A and CH3B variants was 78 kD.
  • Western blots were performed together to determine the extent of more sophisticated homodimers.
  • Western blot was performed by separating anti-human IgG-AP conjugated antibody (Sigma) using a method performed by those skilled in the art after separating 0.1 ⁇ g of protein, which is less than SDS-PAGE analysis, in 12% non-reducing conditions ( 9).
  • the IgG1 heterodimer into which the wild-type CH3 domain was introduced as a control group showed all homodimers of CH3A / CH3B and CH3A: CH3B heterodimers on SDS-PAGE, whereas IgG2, IgG3, Human antibody isotype A107 heavy chain constant region heterodimer variants incorporating A107 heterodimerization mutations into IgG4 were found to form heterodimers with similar or higher yields than previously reported IgG1-based A107 mutants. .
  • the Fc monomer (Half Fc) containing CH3A or CH3B was also observed, which is one of the characteristics of naturally occurring IgG4, and the hinge region (particularly, before the Fab-arm exchange in the blood) occurs. , 228 th serine in the central hinge region). This is due to the characteristic of forming half Fc (Liu H et al., 2012).
  • Example 7 Construction of human / mouse IL-12 fusion protein
  • the IgG4-based variant ( ⁇ 4-A107) was used.
  • Persistent Interleukin 12 fusion protein was constructed.
  • Interleukin 12 in nature consists of two different units of p35 monomer (p35; IL-12A) and p40 monomer (p40; IL-12B), which are active by interacting with each other to form heterodimers.
  • p35; IL-12A p35 monomer
  • p40 monomer p40 monomer
  • Has Formation of such heterodimers is achieved by binding more stably with one disulfide bond present between two units. Therefore, we tried to maintain the heterodimer form of cytokines in nature by connecting each monomer to different heterodimeric Fc variants (CH3A or CH3B).
  • the heavy chain constant region heterodimeric variant for the construction of the fusion protein was used ⁇ 4-A107 to introduce a heterodimer by introducing an A107 mutation based on IgG4.
  • intrinsic functions such as ADCC / CDC of IgG1 in the construction of immunocytokine, which is a fusion form of antibody and cytokine, promote the clearance in vivo.
  • the fusion protein was constructed using an IgG4 isotype that shows little function of ADCC / CDC compared to IgG1 (Gillies SD et al., 1999).
  • (C) is a fusion protein to which the CH3 mutant pair produced in the present invention is introduced.
  • Human interleukin 12 (hIL-12, Uniprot entry name P29460, P29459; SEQ ID NO: 17-18) and mouse interleukin 12 (mIL-12, Uniprot entry name P43432, P43431; SEQ ID NO: 19-20) are both mature except signal sequence Only the DNA sequence encoding the form was amplified and cloned using an NotI / BsiWI restriction enzyme in an in-frame as shown in FIGS. 11 (A) and (B) in an animal cell expression vector containing a ⁇ 4-A107 variant. They were named mono-hIL-12-Fc and mono-mIL-12-Fc, respectively.
  • the human / mouse p35 monomer was assigned 15 flexible peptide linkers (G 4 S) 3 Linker between the p35 monomer and the hinge region to allow sufficient interaction with the p40 monomer.
  • G 4 S flexible peptide linkers
  • bi-hIL-12-Fc and bi-mIL-12- fused with human interleukin 12 (hIL-12) and mouse interleukin 12 (mIL-12) were fused with wild-type IgG4 Fc (wt IgG4).
  • Fc was constructed.
  • IL-12 which is active only by heteroduplexing
  • two units are connected to the 15 peptide linkers, and then the animal cell expression vector containing the ⁇ 4-A107 variant is shown in FIG. 12. It was cloned in-frame using NotI / BsiWI restriction enzyme.
  • the comparative example is a fusion protein of the type used in previous studies to make IL-12 fusion proteins (Lisan S. Peng et al., 1999).
  • Table 3 shows the amino acid sequences for the mature forms of the monomers of human and mouse interleukin 12 used to construct the fusion protein.
  • Example 8 IL-12 fusion protein expression / purification
  • the mono-IL-12-Fc fusion protein of FIG. 10C shows 1: 1 ratio of expression vectors of human / mouse IL-12.p40- ⁇ 4-A107A and human / mouse IL-12.p35- ⁇ 4-A107B. It was expressed / purified in the same manner as in Example 5.
  • the bi-IL-12-Fc fusion protein of FIG. 10 (B) was expressed / purified via a single transfection of the human / mouse scIL-12-IgG4 Fc (wt) expression vector. All fusion proteins were expressed / purified at a similar level of 12-13 mg per 100 ml HEK293F cell culture.
  • SEC 14 is a result of size-exclusion chromatography (SEC) of the fusion proteins. Some polymers were observed in the Mono-hIL-12-Fc fusion protein.
  • Example 8 The binding ability of the mono-hIL-12-Fc expressed and purified in Example 8 to the IL-12 receptor was compared with bi-hIL-12-Fc.
  • FIG. 15 shows the result of confirming that the mono-hIL-12-Fc constructed as compared with bi-hIL-12-Fc showed binding ability to the IL-12 receptor by FACS Calibur (BD Biosciences).
  • PBMC immune cells
  • PHA Sigma-Aldrich
  • T and NK cells were treated for 72 h to stimulate T and NK cells.
  • PHA treatment has been reported to express IL-12 receptors on T cells and NK cells as the immune cells divide.
  • 1 ⁇ 10 6 cells / ml of PBMC was added to RPMI1640 medium containing 10% FBS, and PHA was added at a concentration of 10 ⁇ g / ml as a mitogen, followed by incubation for 72 hours at 37 degrees and 5% CO 2 incubator.
  • Normal PBMC and PHA activated PBMC were washed with cold PBS (pH 7.4) and 5 ⁇ 10 5 cells were prepared for each sample.
  • Fc (A107), bi-hIL-12-Fc and mono-hIL-12-Fc were added at a concentration of 1 ⁇ M, reacted at 4 ° C. for 30 minutes, and washed with cold PBS (pH 7.4).
  • FITC fluorescence-linked secondary antibody Sigma-Aldrich
  • FACS Calibur BD Bioscience
  • bi-hIL-12-Fc and mono-hIL-12-Fc did not bind to normal PBMCs that did not express IL-12 receptors, but only to PBMCs that were activated by PHA and expressed IL-12 receptors. Therefore, the binding capacity of mono-hIL-12-Fc to IL-12 receptor was confirmed to be the same as bi-hIL-12-Fc.
  • IL-12 recombinant human IL-12
  • FIG. 16 shows WST-1 cell proliferation test results confirming cell proliferation ability by Fc (A107), rhIL-12, bi-hIL-12-Fc and mono-hIL-12-Fc in PHA-activated PBMCs.
  • PBMC PBMC (2 ⁇ 10 4, 50 ⁇ l) activated with PHA was added to 96-well plate (SPL, Korea) and serially diluted with RPMI1640 medium containing 10% FBS as in Example 9.
  • Example 11 Evaluation of the ability to induce IFN- ⁇ secretion of mono-hIL-12-Fc fusion protein against PBMC
  • FIG. 17 shows ELISA results of measuring the amount of IFN- ⁇ secretion by Fc (A107), rhIL-12, bi-hIL-12-Fc and mono-hIL-12-Fc in PHA activated PBMC.
  • Example 10 in order to measure IFN- ⁇ concentration in cell culture cultured for 72 hours in Example 10, a human IFN- ⁇ capture antibody (Thermo Fisher Scientific) was placed in a 96-well plate (Thermo Fisher Scientific, Korea) for ELISA. After coating for 12 hours, washed with PBST (PBS with 0.1% Tween-20), 1% BSA (PBS with 1% bovine serum albumin) was added and blocked for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), the culture cultured in Example 2 was diluted 5 times with 1% BSA, added 100 ⁇ l, and then reacted at room temperature for 2 hours.
  • PBST PBS with 0.1% Tween-20
  • BSA PBS with 1% bovine serum albumin
  • mono-hIL-12-Fc showed that IFN- ⁇ secretion ability to PBMC is similar or higher than rhIL-12.
  • Example 8 The binding ability of the mono-mIL-12-Fc expressed and purified in Example 8 to the IL-12 receptor was compared with bi-mIL-12-Fc.
  • FIG. 18 is a flow cytometry result confirming that mono-mIL-12-Fc constructed as compared with bi-mIL-12-Fc shows binding ability to IL-12 receptor.
  • mouse IL-12 was reported to bind not only mouse IL-12 receptor but also human IL-12 receptor, and analyzed in the same manner as in Example 9.
  • bi-mIL-12-Fc and mono-mIL-12-Fc did not bind to normal PBMCs that did not express IL-12 receptors, but only to PBMCs that were activated by PHA and expressed IL-12 receptors. Therefore, the binding capacity of mono-mIL-12-Fc to IL-12 receptor was confirmed to be the same as bi-mIL-12-Fc.
  • WST-1 confirmed the cell proliferation ability by Fc (A107), recombinant mouse IL-12 (rmIL-12), bi-mIL-12-Fc and mono-mIL-12-Fc in PHA activated PBMC Cell proliferation assay results.
  • Example 13 cell proliferation in PBMCs activated with PHA of mono-mIL-12-Fc was confirmed. It was confirmed whether the effect of mono-mIL-12-Fc is the same in vivo.
  • 20 (A) and 20 (B) show the results of measuring tumor growth inhibitory activity of mono-mIL-12-Fc in 100 mm 3 tumors.
  • mice hair of 4 week old female Balb / c mice (NARA Biotech, Korea) was removed with a razor and CT26 HER2 / Neu colorectal cancer cells (1 ⁇ 10 6 cells / mouse) were diluted in 150 ⁇ L PBS and transplanted into the mouse subcutaneous. It was.
  • Figure 21 (A), 21 (B) and 21 (C) is the result of measuring the tumor growth inhibitory activity in mice in vivo of mono-mIL-12-Fc administered at different concentrations in mice with 300 mm 3 tumor size to be.
  • mice hair of 4 week old female Balb / c mice (NARA Biotech, Korea) was removed with a razor and CT26 HER2 / Neu colorectal cancer cells (1 ⁇ 10 6 cells / mouse) were diluted in 150 ⁇ L PBS and transplanted into the mouse subcutaneous. It was.
  • bi-IL-12-Fc was mono-mIL-12-Fc at a molar concentration of 1 ⁇ g IL-12 or less even for large tumors. Compared with that, the effect of inhibiting tumor growth was significantly higher.
  • the same molar concentration of mono-mIL-12-Fc was effective in removing tumors in 40% of mice.
  • 73% of the tumors were cleared by only 5 doses of mono-mIL-12-Fc at a concentration of 0.5 ⁇ g IL-12 where bi-mIL-12-Fc did not remove tumors.
  • Figure 21 (D) is the result of measuring the weight change in vivo toxicity of mono-mIL-12-Fc administered by concentration.
  • the weight of the mice administered as shown in Figure 21 (A) was measured twice a week to observe the weight loss.
  • the weight of the tumor was increased as the tumor size increased, but it was confirmed that there was no weight loss in the mice administered with all concentrations of bi-mIL-12-Fc and mono-mIL-12-Fc. Therefore, mono-mIL-12-Fc was not considered to be in vivo toxic to induce weight loss.
  • Figure 21 (D) is the result of measuring alanine aminotransferase (ALT) which is an indicator of liver toxicity.
  • blood was collected from the facial vein at 24 hours of the last administration in the mouse of FIG. 21 (A). Blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer.
  • blood was collected from the facial vein of the mouse 24 hours after the last IL-12-Fc fusion protein administration. The blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer.
  • ALT measurement substrate solution (alanine and ⁇ -ketoglutarate mixed solution) was taken in a 15 ml test tube and incubated in a 37 ° C constant temperature water bath for 5 minutes.
  • the mice transplanted with the tumor were treated with bi-mIL-12-Fc and mono-mIL-12-Fc, and blood samples were collected by diluting the serum 10-fold and adding 200 ⁇ l and shaking. Incubate for 30 minutes in a water bath. 1 ml of color solution (2,4-dinitrophenyl-1-hydrazone) was added to the test tube taken out of the constant temperature water bath, followed by mixing at room temperature for 20 minutes.
  • Example 16 Evaluation of immune cell proliferation induction capacity of mono-mIL-12-Fc in vivo
  • Figure 22 (A) is the result of confirming the increase in the number of CD4 + T cells, CD8 + T cells and NK cells in the spleen after 3 days of the last administration of Figure 21 (A).
  • the mouse spleen was extracted at 34 days after tumor transplantation, pulverized with a wire mesh in Petri dishes and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to erythrocytes, and red blood cells were lysed and washed with PBS to prepare splenocyte turbidity, and the number of cells was counted with a hemocytometer. Spleen lymphocytes were stained at 4 ° C. for 30 minutes with antibodies recognizing CD45, CD3, CD4, CD8 and CD49b bound to APC, FITC, PE or PE-cy5 and washed with cold PBS (pH 7.4).
  • CD45 + CD3 + CD4 + cell populations CD45 + CD3 + CD8 + cell populations
  • CD45 + CD3 - CD49b + cell populations CD4 + T cells, CD8 + T cells, and NK cells.
  • the ratio of total splenocytes was calculated and the number of CD4 + T cells, CD8 + T cells and NK cells increased after mono-mIL-12-Fc administration by multiplying the number of cells counted with hemocytometer. .
  • mono-mIL-12-Fc infiltrated tumors based on the report that the increase in the number of CD4 + T cells and CD8 + T cells, which are adaptive immune cells infiltrated into tumors, is important (Schreiber et al., 2011). It was analyzed whether the number of adaptive immune cells increased. When 6 mono-mIL-12-Fc were administered to a large number of mice without tumors, the number of immune cells infiltrated into the tumors of mice after 3 administrations was analyzed.
  • Figure 22 (B) is the result showing the total number of immune cells, CD4 + T cells and CD8 + T cells infiltrated into the tumor by 3 days after the third administration of the 21 (A).
  • the tumor of the mouse was extracted and weighed on the 24th day of tumor transplantation, and then pulverized using a wire mesh and collagenase (100 ⁇ g / ml) in Petri dish, followed by 2% FBS. 10 ml of the medium containing the was added and centrifuged at 50 g for 5 minutes to remove the parenchyma. Thereafter, 1 ml of red blood cell lysis buffer was added to erythrocytes, and red blood cells were lysed, washed with PBS to prepare a cell suspension, and the number of cells was counted with a hemocytometer. Cells isolated from tumors were stained at 4 ° C.
  • CD45 + T cells and tumor infiltrating CD8 + T cells were defined.
  • bi-mIL-12-Fc and mono-mIL-12-Fc showed the number of total immune cells, CD4 + T cells, and CD8 + T cells infiltrating tumors in tumor-transplanted mice. It can be confirmed that increased.
  • Mono-mIL-12-Fc increased the number of total immune cells, CD4 + T cells, and CD8 + T cells infiltrated tumors more significantly than bi-mIL-12-Fc. Therefore, mono-mIL-12-Fc significantly increased the number of CD4 + T cells and CD8 + T cells infiltrating tumors than bi-mIL-12-Fc.
  • Example 17 Evaluation of cytokine secretion and cytotoxicity function of mono-mIL-12-Fc in vivo immune cells
  • IL-12 is known to inhibit the growth of cancer cells by increasing IFN-g secretion of T cells and NK cells (Trinchieri, 2003). IL-12 also has an anticancer effect by enhancing direct cytotoxic effects on cancer cells of cytotoxic T cells and natural killer cells. Therefore, we analyzed whether the anti-cancer effect of Mono-IL-12-Fc was due to the increase of serum IFN-g concentration and the direct cytotoxic effect of cytotoxic T cells and natural killer cells on cancer cells.
  • Figure 23 (A) is the result of measuring the IFN-g concentration in the serum isolated by collecting blood from the facial vein of the mouse 24 hours after the last administration of Figure 21 (A) by ELISA.
  • FIG. 20 blood was collected from the facial vein of the mouse 24 hours after the last mIL-12-Fc fusion protein administration in FIG. 20 (A).
  • the blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer.
  • a mouse IFN- ⁇ capture antibody was coated in a 96-well plate for ELISA (Thermo Fisher Scientific) for 12 hours, followed by washing with PBST (PBS with 0.1% Tween-20).
  • 1% BSA PBS with 1% bovine serum albumin
  • bi-mIL-12-Fc was not capable of inducing IFN- ⁇ secretion by NK cells and T cells, and thus, bi-mIL-12-Fc was administered in FIG. 23 (A).
  • mono-mIL-12-Fc and bi-mIL-12-Fc were administered only once, and then blood IFN- ⁇ levels were measured over time.
  • FIG. 23 (B) shows IFN- ⁇ in serum by time after intraperitoneal administration of bi-mIL-12-Fc and mono-mIL-12-Fc to Balb / c mice transplanted with CT26 HER2 / Ne colorectal cancer cells. Concentration was measured by ELISA.
  • a mouse IFN- ⁇ capture antibody was coated in a 96-well plate for ELISA (Thermo Fisher Scientific) for 12 hours, followed by washing with PBST (PBS with 0.1% Tween-20).
  • PBST PBS with 0.1% Tween-20
  • 1% BSA PBS with 1% bovine serum albumin
  • the serum was diluted 10-fold with 1% BSA and reacted at room temperature for 2 hours.
  • biotin-coupled mouse IFN- ⁇ detection antibody was bound for 1 hour at room temperature.
  • the bi-mIL-12-Fc-administered group showed a similar concentration of IFN- ⁇ in the blood to the mono-mIL-12-Fc-treated group until day 5 in the tumor-grafted mice. There was no inherent deficiency in the ability of -Fc to induce the secretion of IFN- ⁇ in effector cells.
  • Figure 23 (C) is a graph measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of cytotoxic T cells isolated from the spleen by killing the mouse 3 days after the last administration of Figure 21 (A).
  • mice 72 hours after the last cytokine administration in FIG. 21 (A), mice were killed and spleens were removed and spleens and PBS were put into a 60 mm dish containing 70 micron mesh and ground.
  • the cells obtained by centrifugation were lysed with red blood cell hemolysis buffer, and then washed with PBS and washed with PBS, an APC conjugated antibody that recognizes CD3 (Thermo Fisher Scientific), and a PE conjugated antibody that recognizes CD8 (Thermo Fisher Scientific).
  • the reaction was carried out for 30 minutes at 4 °C. After washing with PBS, cytotoxic T cells (CD3 + CD8 + ) were isolated by FACS Aria III (BD biosciences, Korea).
  • CT26 HER2 / Neu cancer cells were stained with calcein AM (Thermo Fisher Scientific Inc., 10 ⁇ M). After CT26 HER2 / Neu cancer cells (2 ⁇ 10 6 ) were suspended in 2 ml of DPBS, 2 ⁇ l of calcein AM (10 mM) was added and mixed, followed by reaction for 45 minutes at 5% CO 2 and 37 ° C.
  • a RPMI1640 with 10% FBS was added 10 ml into three times, washed, and then put into a 2 ⁇ 10 4 cells per well in a 96-well plate cytotoxic T cells (1 ⁇ 10 5/100 ⁇ l / well) into each 5 % CO 2 , incubated for 4 hours at 37 °C incubator.
  • Live CT26 HER2 / Neu cancer cells that fluoresce green and dead CT26 HER2 / Neu cancer cells that do not fluoresce green were analyzed by flow cytometry to express the cytotoxic effect of cytotoxic T cells in percentage.
  • Cytotoxic T cells isolated from tumor-grafted mice treated with Mono-mIL-12-Fc were cytotoxic T cells isolated from tumor-grafted mice administered bi-mIL-12-Fc or cytotoxic T cells isolated from the control group.
  • the cytotoxic effect on CT26 HER2 / Neu cancer cells, which are more target cells, was higher, and the tumor suppression effect of mono-mIL-12-Fc was a direct cytotoxic effect of cancer cells by some cytotoxic T cells.
  • Figure 23 (D) is the third dose of Figure 21 (A) to determine whether the cytotoxic effect of cytotoxic T cells enhanced by administration of mono-IL-12-Fc in tumor-grafted mice is cancer antigen specific. Three days later, the cytotoxic effect of cytotoxic T cells isolated from the spleen by lethal mice was measured using CT26 HER2 / Neu cancer cells expressing tumor antigen and 4T1 cells not expressing tumor antigen.
  • mice 72 hours after the three-time administration of mono-IL-12-Fc in FIG. 20 (A), mice were killed and spleens were removed, and spleen and PBS were placed in a 60 mm dish containing 70 micron mesh. Pulverized. Cytotoxic T target cells in CT26 HER2 / Neu tumor cells and cytotoxic T Target cells 4T1 cytotoxic effect in the same way as FIG. 21 (C) to measure CT26 HER2 / Neu cancer with 4T1 tumor cells to a non-cellular in the cell Were stained with calcein AM (Thermo Fisher Scientific Inc., 10 ⁇ ).
  • calcein AM Thermo Fisher Scientific Inc., 10 ⁇
  • the RPMI1640 with 10% FBS was added 10 ml into three times, washed, and then put into a 2 ⁇ 10 4 cells per well in a 96-well plate cytotoxic T cells (1 ⁇ 10 5/100 ⁇ l / well) into each 5 % CO 2 , incubated for 4 hours at 37 °C incubator.
  • Green Fluorescent Live CT26 HER2 / Neu Dead CT26 HER2 / Neu with no green fluorescence with cancer cells or 4T1 cancer cells Cancer cells or 4T1 cancer cells were analyzed by flow cytometry to express the cytotoxic effects of cytotoxic T cells in percentage. As a result, it was confirmed that the cytotoxic effect of the cytotoxic T cells enhanced by the administration of mono-mIL-12-Fc was target cell specific.
  • Figure 23 (E) is a result of measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of natural killer cells isolated from the spleen by killing the mouse 3 days after the third administration of Figure 21 (A).
  • mice were killed and spleens were removed, and spleens and PBS were put in a 60 mm dish containing 70 micron mesh and ground.
  • the cells obtained by centrifugation were lysed with red blood cell hemolysis buffer, washed with PBS, and washed with PBS, and APC-bound antibody (Thermo Fisher Scientific) that recognizes CD3 and PE-bound antibody (Thermo Fisher Scientific) that recognizes CD49b.
  • the reaction was carried out for 30 minutes at 4 °C. After washing with PBS, natural killer cells (CD3 - CD49b + ) were separated by FACS Aria III (BD biosciences, Korea).
  • CT26 HER2 / Neu Cancer cells were stained with calcein AM (Thermo Fisher Scientific Inc., 10 ⁇ M) to measure the cytotoxicity of the target cells in CT26 HER2 / Neu cancer cells of NK cells.
  • CT26 HER2 / Neu cancer cells (2 ⁇ 10 6 ) were suspended in 2 ml of DPBS, 2 ⁇ l of calcein AM (10 mM) were mixed and allowed to react for 45 minutes at 5% CO 2 , 37 ° C.
  • a RPMI1640 with 10% FBS was added 10 ml into three by three times, and then each well in a 96-well plate into a 2 ⁇ 10 4 cells into natural killer cells (1 ⁇ 10 5/100 ⁇ l / well) , respectively 5% Incubated for 4 hours in a CO 2 , 37 °C incubator.
  • Live CT26 HER2 / Neu cancer cells that fluoresce green and dead CT26 HER2 / Neu cancer cells that do not fluoresce green were analyzed by flow cytometry to express the cytotoxic effect of natural killer cells in percentage.
  • Natural killer cells isolated from tumor-grafted mice treated with Mono-mIL-12-Fc were more targeted than natural killer cells isolated from bi-mIL-12-Fc-treated mice or with cytotoxic T cells isolated from the control group.
  • the cytotoxic effect on CT26 HER2 / Neu cancer cells, which are the cells, was high, and the tumor suppression effect of mono-mIL-12-Fc was a direct cytotoxic effect of cancer cells by some natural killer cells.
  • the generation of adaptive immunity in tumor-grafted mice is assessed by effector memory CD8 + T cells and memory CD8 + T cell formation.
  • Tumor elimination effect by Mono-mIL-12-Fc was determined by the effect memory CD8 + T cells and memory CD8 + T cell formation.
  • 24 (A), 24 (B) and 24 (C) show effect CD8 + T cells, effect memory CD8 + T cells and memory CD8 generated when mono-mIL-12-Fc was administered to mice with tumors, respectively. + The result of measuring the number of T cells.
  • the mouse spleen was extracted at 34 days after tumor transplantation, pulverized using a wire mesh in Petri dishes, washed with 10 ml of medium containing 2% FBS, and then erythrocyte hemolysis buffer 1 ml of red blood cell lysis buffer was added to erythrocytes, and then washed with PBS to prepare a splenocyte suspension, and the number of cells was counted with a hemocytometer.
  • Splenocytes were stained at 4 ° C. for 30 minutes with antibodies recognizing CD3, CD8, CD62L and IL-7 receptor (IL-7R) bound to PE-cy5, PE, FITC or APC, and cold stained with PBS (pH 7.4).
  • CD3 + CD8 + CD62L low IL-7R low Cell groups CD3 + CD8 + CD62L low IL-7R hi cell populations and CD3 + CD8 + CD62L hi IL-7R hi cell populations were defined as effect CD8 + T cells, effect memory CD8 + T cells and memory CD8 + T cells, respectively.
  • the number of effect CD8 + T cells, effect memory CD8 + T cells, and memory CD8 + T cells after mono-mIL-12-Fc administration was analyzed by multiplying the number of cells counted by hemocytometer. .
  • mono-mIL-12-Fc increased the number of effect memory CD8 + T cells and memory CD8 + T cells in a tumor-grafted mouse compared with the control group.
  • bi-mIL-12-Fc increased the number of effect memory CD8 + T cells and memory CD8 + T cells only in the group administered with a molar concentration such as 0.5 ⁇ g IL-12, and the molar like 1 ⁇ g IL-12.
  • the concentration group did not increase the number of effect memory CD8 + T cells and memory CD8 + T cells. Therefore, mono-mIL-12-Fc significantly increased the number of effect memory CD8 + T cells and memory CD8 + T cells than bi-mIL-12-Fc.
  • Figure 24 (D) is in the mice survived 120 days after the administration in 21 (A) to FIG 1 ⁇ g mono-IL-12-Fc CT26 HER2 / Neu Cancer cells are transplanted to measure the change in the tumor volume of the mouse.
  • FIG. 21 (A) the last 1 ⁇ g mono-IL-12- was applied to female Balb / c mice (NARA Biotech, Korea) that matched the age of the surviving mice after administration of 1 ⁇ g mono-IL-12-Fc.
  • the hair of the mouse was removed with a razor and CT26 HER2 / Neu cells (1 ⁇ 10 6 cells / mouse) were transplanted into the mouse subcutaneously diluted in 150 ⁇ L PBS.
  • additional 1 ⁇ g mono-IL-12-Fc of the measurement without the administration of the tumor twice a week, and the volume (V) of the tumor was calculated as V L x W 2/2.
  • bi-mIL-12-Fc was compared with mono-mIL-12-Fc in the number of spleen CD8 + T cells, the number of effect memory CD8 + T cells, and central memory CD8 in mice transplanted with tumors.
  • the effect of increasing the number of + T cells was observed to be low.
  • the effect CD8 + T cells are partially differentiated into memory CD8 + T cells through memory precursor effector cells (MPEC) and are mostly short-lived. It has been reported to die as short-lived effector cells (SLEC).
  • MPEC memory precursor effector cells
  • SLEC short-lived effector cells
  • FIG. 24 (E) shows memory precursor effect cells (KLRG1 - IL-7R + ) and short-lived effect cells (KLRG1) among CD8 + T cells present in the spleen after 3 days of the third administration of FIG. 21 (A). + IL-7R - an analysis of the rate of).
  • the mouse spleen was extracted on the 24th day of tumor transplantation, and then pulverized using a wire mesh in Petri dishes and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Splenocytes were stained at 4 ° C. for 30 minutes with antibodies recognizing CD3, CD8, KLRG1 and IL-7 receptor (IL-7R) bound to PE-cy5, PE, FITC or APC, and cold stained with PBS (pH 7.4).
  • IL-7R IL-7 receptor
  • bi-mIL-12-Fc did not increase the proportion of memory precursor cells compared to the control group, but rather increased the number of short-lived cells. Therefore, mono-mIL-12-Fc promoted the production of memory precursor cells more than bi-mIL-12-Fc and significantly increased the number of effect memory CD8 + T cells and memory CD8 + T cells, resulting in a higher tumor removal effect. Confirmed.
  • Example 20 Evaluation of the Effect of Mono-mIL-12-Fc on the Expression of Transcription Factors Related to Memory Cell Differentiation Induction
  • T-bet a transcription factor that differentiates CD8 + T cells into short-lived effect cells
  • Eomes eomesodermin
  • 25 (A) and 25 (B) show the percentage of CD8 + T cells with high expression of T-bet that inhibits the differentiation of memory cells in the spleen by killing mice 3 days after the third dose of FIG. 21 (A). This is the result of measuring the ratio of CD8 + T cells with high expression of Eomes and low expression of T-bet that promotes the differentiation of memory cells by flow cytometry.
  • the mouse spleen was extracted on the 24th day of tumor transplantation, and then pulverized using a wire mesh in Petri dishes and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Splenocytes were stained with PE-cy5 or FITC-coupled CD3 and CD8 antibodies for 30 minutes at 4 ° C, washed with cold PBS (pH 7.4), and stained with transcription factor Foxp3 / Transcription Factor.
  • the cells were fixed and permeabilized using Staining Buffer Set (Thermo Fisher Scientific), and then stained for 30 minutes at 4 ° C. with an antibody recognizing T-bet or Eomes bound to PE or efluor 660.
  • Permeabilization buffer was added and analyzed by flow cytometry FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to analyze the ratio of CD3 + CD8 + T-bet high cell population and CD3 + CD8 + Eomes + T-bet low cell population.
  • mono-mIL-12-Fc reduced the proportion of CD3 + CD8 + T-bet high cell populations in a tumor-dependent mouse compared to the control group, and decreased the concentration of CD3 + CD8 + Eomes + T-bet low cells. It was confirmed that the ratio was increased.
  • bi-mIL-12-Fc decreased the proportion of CD3 + CD8 + T-bet high cell population only in the group administered with 0.5 ⁇ g IL-12, and CD3 + CD8 + Eomes + T-bet low
  • the percentage of cell population was increased, and in the group administered with a molar concentration such as 1 ⁇ g IL-12, the percentage of CD3 + CD8 + T-bet high cell population was decreased or the percentage of CD3 + CD8 + Eomes + T-bet low cell population was decreased. There was no increasing effect.
  • mono-mIL-12-Fc reduces the proportion of CD3 + CD8 + T-bet high cell population and increases the ratio of CD3 + CD8 + Eomes + T-bet low cell population than bi-mIL-12-Fc.
  • the number of CD8 + T cells and memory CD8 + T cells was significantly increased to confirm that the tumor removal effect was high.
  • CD8 + T cells when stimulated with inflammatory cytokines such as IL-12 in the presence of T cell receptor and co-stimulatory signals, increase phosphorylation of STAT4, and phosphorylated STAT4 (pSTAT4) migrates inside the nucleus to the T-bet enhancer. It is known to bind to increase expression of T-bet. Therefore, when 8 ⁇ g IL-12 at a molar concentration of bi-mIL-12-Fc is administered, CD8 + T cells are differentiated into short-lived cells.
  • Fc administration increased the expression of pSTAT4 and T-bet when CD8 + T cells were activated in the groin lymph nodes, the tumor draining lymph nodes of mice transplanted with tumors than mono-mIL-12-Fc. Cognitive was measured.
  • Figure 25 (C) shows the molar concentrations of bi-mIL-12-Fc and mono-mIL-12-Fc at the same molar concentration of 1 ⁇ g rmIL-12 when the tumor size was 300 mm 3 in CT26 HER2 / Ne transplanted Balb / c mice.
  • the expression level of phosphorylated STAT4 in CD8 + T cells isolated from inguinal lymph nodes 24 hours after one intraperitoneal administration was measured by flow cytometry.
  • Inguinal lymph node cells were stained with PE-cy5 or FITC-coupled CD3 and CD8 antibodies for 30 minutes at 4 ° C, washed with cold PBS (pH 7.4), and fixed with cold methanol. The inguinal lymph node cells were then washed with cold PBS (pH 7.4), and then stained with cold PBS (pH 7.4) for 30 minutes at 4 ° C with an antibody recognizing pSTAT4 bound to APC, followed by FACS Calibur (flow cytometry). BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to compare the amount of pSTAT4 expressed in CD3 + CD8 + T cells. As a result, compared with mono-mIL-12-Fc, bi-mIL-12-Fc increased the expression of pSTAT4 when CD8 + T cells were activated in the groin lymph nodes of tumor-grafted mice.
  • Figure 25 (D) is a result of measuring the ratio of CD8 + T cells expressing T-bet inhibiting the differentiation of memory cells in the groin lymph nodes 72 hours after one intraperitoneal administration of Figure 25 (C) by flow cytometry.
  • Inguinal lymph node cells were stained with PE-cy5 or FITC-bound CD3 and CD8 antibody for 30 minutes at 4 ° C, washed with cold PBS (pH 7.4), and stained with transcription factor Foxp3 / Transcription.
  • Cells were fixed and permeabilized using a Factor Staining Buffer Set (Thermo Fisher Scientific). Afterwards, the antibody recognizes T-bet conjugated with PE or APC, and then stained at 4 ° C. for 30 minutes, and then added a permeabilization buffer to FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). ). Each sample was analyzed by dot plot to compare the proportion of CD3 + CD8 + T cells expressing T-bet.
  • bi-mIL-12-Fc increased the expression of T-bet when CD8 + T cells were activated in the groin lymph nodes of mice transplanted with tumors. Therefore, the differentiation of CD8 + T cells into monolithic cells when bi-mIL-12-Fc at the same molar concentration as 1 ⁇ g IL-12 was observed in the peritumor lymph nodes of mice transplanted with tumors than mono-mIL-12-Fc. It was confirmed that the expression of pSTAT4 and T-bet was increased when CD8 + T cells were activated in the inguinal lymph nodes (tumor draining lymph nodes).
  • spleen and groin lymph nodes were extracted from normal Balb / c mice, pulverized using a wire mesh in Petri dishes, and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Lymphocytes were stained for 30 minutes at 4 ° C with antibodies bound to PE-binding CD8, washed with cold PBS (pH 7.4), and then combined with anti-PE microbeads (Miltenyi Biotec) for 15 minutes. Miltenyi Biotec) was used to isolate CD8 + T cells.
  • pSTAT4 and T-bet were stained by the method of FIGS. 25 (C) and 25 (D), respectively, and analyzed by flow cytometry, FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to compare the amount of pSTAT4 or T-bet expressed in CD8 + T cells. As a result, when mono-mIL-12-Fc was cross-linked with an antibody that recognizes Fc, the CD8 + T cells were stimulated by two molecules of IL-12, similar to those treated with bi-mIL-12-Fc. As a result, the expression of pSTAT4 and T-bet was increased.
  • mono-mIL-12-Fc induces less expression of pSTAT4 and T-bet in CD8 + T cells than bi-mIL-12-Fc so that CD8 + T cells have a memory effect.
  • Differentiation into cells leads to differentiation into effect memory cells and central memory cells, so even at low concentrations (molar concentrations such as 0.5 ⁇ g IL-12), the tumors of mice transplanted with tumors can be removed to extend the lifespan of the mice. .
  • bi-mIL-12-Fc induces the expression of pSTAT4 and T-bet in CD8 + T cells to differentiate into short-lived effect cells and to prevent differentiation into memory cells, thus molar concentrations such as mono-mIL-12-Fc
  • molar concentrations such as mono-mIL-12-Fc
  • the tumor was not completely removed from the transplanted mouse, and cytotoxic CD8 + T in the effector phase was directly administered at a higher concentration (molar concentration such as 2 ⁇ g IL-12).
  • the cells must be expanded to remove the tumor.
  • Heterodimeric Fc-fused protein according to the antibody heavy chain constant region according to the present invention by forming a protein complex of two or more subunits to mimic the physiologically active protein forms in nature, It can maintain the activity as it exists in nature, and also has the advantage that the half-life in the body of the bioactive protein can be significantly extended.
  • heterodimeric-fusion protein heterodimeric Fc-fused protein
  • the heterodimeric-fusion protein form according to the present invention has the advantage that it is easy to prepare a monovalent heterodimer-fusion protein without further purification process optimization.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Endocrinology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Reproductive Health (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to a heterodimeric Fc-fused protein and a pharmaceutical composition comprising the heterodimeric Fc-fused protein, wherein in the heterodimeric Fc-fused protein comprising a first Fc region and a second Fc region of a Fc region pair of an immunoglobulin while a subunit of a biologically active protein is bound to at least one of N-terminal or C-terminal of the first Fc region and/or the second Fc region, the first Fc region and the second Fc region are CH3 domains modified to promote the formation of a heterodimer. In the heterodimeric Fc-fused protein according to the present invention, two or more subunits form a protein complex, so that a protein constituting a biologically active protein can be fused to Fc in an original shape and structure as the protein exists in nature and thus the protein can maintain an original activity as the protein exists in nature. The use of the heterodimeric Fc-fused protein according to the present invention remarkably increases the in vivo half-life of a biologically active protein contained in the heterodimeric Fc-fused protein, and thus various types of biological activities can be maintained for a long time in the body.

Description

항체 중쇄불변부위 이종이중체에 융합된 사이토카인 및 이를 포함하는 약제학적 조성물Cytokines fused to an antibody heavy chain constant region heterodimer and a pharmaceutical composition comprising the same
본 발명은 항체(immunoglobulin) 중쇄불변부위 (Fc) 쌍의 제1 Fc 영역 및 제2 Fc 영역을 포함하고, The present invention includes a first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
상기 제1 Fc 영역 및/또는 제2 Fc 영역의 N-말단 또는 C-말단 중 하나 이상의 말단에 생리활성 단백질의 서브유닛(subunit)이 결합되어 있는 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, Heterodimeric Fc-fused protein having a subunit of a bioactive protein bound to at least one of the N-terminus or C-terminus of the first Fc region and / or the second Fc region. ),
상기 제1 Fc 영역 및 제2 Fc 영역은 이종이중체(heterodimer)의 형성이 촉진되도록 CH3 도메인이 변이된 것임을 특징으로 하는 이종이중체-융합단백질(heterodimeric Fc-fused protein) 및 The first Fc region and the second Fc region are heterodimeric Fc-fused proteins, characterized in that the CH3 domain is mutated to promote the formation of heterodimers and
상기 이종이중체-융합단백질을 포함하는 약제학적 조성물에 대한 것이다. It relates to a pharmaceutical composition comprising the heterodimer-fusion protein.
본 발명에 따른 이종이중체-융합단백질(heterodimeric Fc-fused protein)은 2 이상의 서브유닛이 단백질 복합체(protein complex)를 형성하여 하나의 생리활성 단백질을 구성하는 단백질을 본래 자연계에 존재하는 형태 및 구조로 항체중쇄불변부위(Fc)에 융합할 수 있어, 자연계에 존재하는 그대로의 활성을 유지할 수 있는 장점이 있다.Heterodimeric Fc-fused protein according to the present invention is a form and structure in which two or more subunits form a protein complex so that a protein constituting one bioactive protein exists in nature. It can be fused to the antibody heavy chain constant region (Fc), there is an advantage that can maintain the activity as it exists in nature.
본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질을 이용할 경우, 상기 이종이중체-융합단백질에 포함된 생리활성 단백질의 체내 반감기가 현저하게 증가되어, 체내에서의 각종 생리활성이 오랜 시간 동안 지속될 수 있는 장점이 있다. When the antibody heavy chain constant region heterodimer-fusion protein according to the present invention is used, the half-life of the bioactive protein contained in the heterodimer-fusion protein is significantly increased, and various biological activities in the body are maintained for a long time. There is an advantage that can be sustained.
또한 본 발명에 따른 항체 중쇄불변부위 이종이중체 Fc의 N-말단 또는 C-말단에 생리활성 단백질의 서브유닛이 융합된 이종이중체-융합단백질은 야생형 Fc 기반 융합 단백질에 비해 발현 후, 정제과정이 용이하다.In addition, the heterodimer-fusion protein in which the subunit of the bioactive protein is fused to the N-terminus or C-terminus of the antibody heavy chain constant region heterodimer Fc according to the present invention is expressed after purification, compared with the wild type Fc-based fusion protein. This is easy.
자연계에 존재하는 인간 항체(Immunoglobulin G (IgG), IgM, IgD, IgE, IgA)는 동일한 아미노산 서열을 가진 두 개의 중쇄와 동일한 서열을 가진 두 개의 경쇄가 조합(assembly)된 형태로 존재한다. 이 때, 동일한 두 개의 중쇄 간의 동종이중화(homodimerization)는 항체의 불변영역(Constant Region)의 마지막 도메인(IgG, IgD, IgA의 경우 CH3 도메인, IgM의 경우 CH4 도메인, IgE의 경우 CH2 및 CH4 도메인) 간의 비공유 결합(non-covalent interaction) 및 힌지(hinge) 영역 간의 이황화 결합(disulfide bond)에 의해 유도된다. Human antibodies present in nature (Immunoglobulin G (IgG), IgM, IgD, IgE, IgA) exist in the form of two heavy chains having the same amino acid sequence and two light chains having the same sequence. In this case, homodimerization between two identical heavy chains is performed by the final domain of the constant region of the antibody (CH3 domain for IgG, IgD, IgA, CH4 domain for IgM, CH2 and CH4 domain for IgE). It is induced by non-covalent interactions between and disulfide bonds between hinge regions.
항체 유래 이종이중체 중쇄불변부위(heterodimeric Fc) 기술은 앞선 자연발생적 항체(IgG, IgM, IgA, IgD, IgE)의 동종이중화에 큰 기여를 하는 불변영역의 마지막 도메인(CH3 domain)간의 특정 비공유 결합을 통해 이종이중화가 선호되고, 동종이중화가 비선호되거나 배척되는 결합을 가지도록 엔지니어링함으로써 이종이중체 형태의 중쇄불변부위를 만드는 기술이다. 더 자세하게는 유전자 조작을 통해 각기 다른 두 개의 항체 중쇄의 CH3 도메인에 돌연변이를 유도하여, 두 개의 중쇄가 자연발생적 항체와 구조가 매우 유사하고, 서열에 있어 최소한의 편차를 가지면서 이종이중체를 형성하도록 유도하는 것이다(미국등록특허 제7,695,936호; 대한민국 등록특허 제1,522,954호). 상기의 이종이중체 중쇄불변부위 기술은 이중항체를 만들기 위한 기반기술로서, 지금까지 알려진 이종이중체 형성을 유도하는 CH3 도메인 돌연변이체는 대부분 CH3 도메인 상호작용면에 항체의 구조 기반 rational design에 의한 비대칭적 돌연변이 쌍을 도입함으로써 제조되었다 (Spreter Von Kreudenstein et al., 2014). 선구자적 연구로는 제넨텍사의 놉-인투-홀(knob-into-hole) 기술이 있고(Ridgway et al., 1996), 자임웍스사의 ZW1 (Von Kreudenstein et al., 2013), 젠코어사의 HA-TF(Moore GL et al., 2011), EMD세로노사의 SEEDbody(Davis JH et al., 2010)등 많은 다국적 제약회사에서 상기의 기반 기술을 개발 및 보고하였다. Antibody-derived heterodimeric heavy chain constant region (Hterodimeric Fc) technology allows specific non-covalent linkages between the last domain of the constant region (CH3 domain), which contributes significantly to the homologous duplication of earlier spontaneous antibodies (IgG, IgM, IgA, IgD, IgE). Heterogeneous redundancy is preferred, and it is a technique to create heterologous heavy chain invariant regions by engineering them to have a bond that is not preferred or rejected. More specifically, genetic modifications induce mutations in the CH3 domains of two different antibody heavy chains, forming two heterozygotes with very similar structures to naturally occurring antibodies and minimal variation in sequence. (US Patent No. 7,695,936; Korean Patent No. 1,522,954). The heterodimeric heavy chain constant region technology is a basic technology for making double antibodies. CH3 domain mutants known to induce heterodimers are known to be mostly asymmetric by structure-based rational design of the antibody on the surface of CH3 domain interaction. It was prepared by introducing a red pair of mutations (Spreter Von Kreudenstein et al., 2014). Pioneer research includes Genentech's knob-into-hole technology (Ridgway et al., 1996), Zymeworks ZW1 (Von Kreudenstein et al., 2013), Zencore's HA- Many multinational pharmaceutical companies have developed and reported these technologies, including TF (Moore GL et al., 2011) and EMD Serono's SEEDbody (Davis JH et al., 2010).
그 중에서도 본 발명에서 사용한 A107 돌연변이체는 효모 세포 표면 발현 시스템을 이용하여 제작된 인간 항체 이종이중체 중쇄불변부위 라이브러리로부터 선별된 고수율 이종이중체 중쇄불변부위로, CH3 도메인 상호작용면의 소수성 코어는 보존하고 전하를 띠는 아미노산에 돌연변이를 유도하여 공간보완적인 소수성 결합이 형성되게 하고 (K409WCH3A-D399V/F405TCH3B), 수소결합이 형성되도록 하여 (K370ECH3A-E357NCH3B) 이종이중체의 형성을 증진시킨 돌연변이체이다(Choi et al. 2016; 대한민국 특허출원 제2015-0142181호). Among them, the A107 mutant used in the present invention is a high yield heterodimeric heavy chain constant region selected from a human antibody heteroduplex heavy chain constant region library prepared using a yeast cell surface expression system, and a hydrophobic core of the CH3 domain interaction surface. Induces mutations in the conserved and charged amino acids to form complementary hydrophobic bonds (K409W CH3A -D399V / F405T CH3B ) and to form hydrogen bonds (K370E CH3A -E357N CH3B ) to form heterodimers (Choi et al. 2016; Korean Patent Application No. 2015-0142181).
상기의 A107 돌연변이체를 포함하여 현재까지 보고된 이종이중체 중쇄불변부위 돌연변이체(heterodimeric Fc variants)는 모두 인간 항체 동종형 중에서도 가장 많은 부분을 차지하는 IgG1을 기반으로 한 것이며, IgG1 이외의 다른 동종형(IgG2, IgG3, IgG4, IgA, IgM, IgE)에 대한 돌연변이체는 보고된 바가 없다. Heterodimeric Fc variants reported to date, including the A107 mutants described above, are all based on IgG1, which is the largest part of human antibody isoforms, and isotypes other than IgG1. No mutants for (IgG2, IgG3, IgG4, IgA, IgM, IgE) have been reported.
이는 미국 식약청(FDA)의 허가를 받아 시판되고 있는 치료용 항체의 대부분이 IgG1 동종형을 채택하고 있기 때문인데 (Irani et al. 2015), 최근에는 항체-의존성 세포 세포독성(antibody-dependent cellular cytotoxicity, ADCC)이나 보체-의존성 세포독성(complement-dependent cellular cytotoxicity, CDC)과 같은 항체 고유의 효과 기능(effector function)이 크게 필요하지 않은 면역조절 항체(immune-modulating antibody) 혹은 수용체 작용제(agonist) 융합 단백질의 경우, IgG1에 비해 이러한 효과 기능이 현저히 떨어지는 IgG2나 IgG4를 기반으로 한 치료용 단백질의 개발이 이루어지고 있는 실정이다. This is because most of the therapeutic antibodies marketed under the approval of the US Food and Drug Administration (FDA) adopt the IgG1 isotype (Irani et al. 2015), and recently antibody-dependent cellular cytotoxicity. , Immuno-modulating antibodies or receptor agonists, which do not require much of the antibody-specific effector functions, such as ADCC) or complement-dependent cellular cytotoxicity (CDC). In the case of proteins, the development of therapeutic proteins based on IgG2 or IgG4, which is significantly less effective than IgG1.
한편, 생리활성 단백질은 대부분 그 크기가 작은 경우가 많아, 체내에서의 반감기가 짧다는 문제점이 존재한다. 이러한 단점을 해결하기 위하여, PEG 등을 접합시키거나, 항체 유래 Fc(crystallizable fragment) 영역을 융합시키는 등의 시도가 있어왔지만, 아직까지 효율적으로 생리활성 단백질의 활성이 오랜 시간 동안 충분히 유지되는 형태의 개발은 이루어지지 못하고 있는 실정이다. On the other hand, most of the bioactive proteins are often small in size, there is a problem that the half-life in the body is short. In order to solve this drawback, attempts have been made to conjugate PEG or the like, or to fuse the antibody-derived crystallizable fragment (Fc) region, but the form of a form in which the activity of the physiologically active protein is sufficiently maintained for a long time efficiently. Development is not happening.
특히, 2 이상의 서브유닛이 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 단백질의 경우, 자연계에 존재하는 본연의 단백질 복합체 구조가 그대로 형성되어 본래 단백질의 활성을 제대로 나타낼 뿐 아니라, 그 활성이 오랜 시간 동안 충분히 유지될 수 있는 형태의 개발은 전무하다시피 한 상황이다. In particular, in the case of a protein in which two or more subunits form a protein complex and exhibit physiological activity, the native protein complex structure existing in nature is formed as it is, and the activity of the original protein is not only properly expressed. There is no such form of development that can be sustained for a long time.
이러한 기술적 배경하에서, 본 발명자들은 IgG1 뿐 아니라, 기존에 보고되지 않은 IgG2, IgG3, 그리고 IgG4와 같은 다른 동종형(isotype) 항체 유래의 Fc 영역을 포함하는 이종이중체(heterodimer) 변이체를 구축하고, 이를 이용하여 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서브유닛이 하나의 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 단백질의 하나 이상의 서브유닛을 상기 Fc 영역의 말단에 결합시킨 이종이중체-융합단백질(heterodimeric Fc-fused protein) 형태의 새로운 치료용 융합단백질을 개발함으로써 본 발명을 완성하였다. Under this technical background, we construct heterodimer variants comprising Fc regions from IgG1 as well as other previously reported isotype antibodies such as IgG2, IgG3, and IgG4, Using this, two or more different subunits are formed, and two or more subunits form one protein complex to bind one or more subunits of a protein showing physiological activity to the ends of the Fc region. The present invention was completed by developing a novel therapeutic fusion protein in the form of a heterodimeric Fc-fused protein.
특히, 본 발명에서의 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서브유닛이 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 단백질은 바람직하게는 인터루킨-12(Interleukin-12, IL-12)가 사용될 수 있다. In particular, a protein consisting of two or more different subunits in the present invention, wherein the two or more subunits form a protein complex and exhibit physiological activity is preferably Interleukin-12, IL-12) can be used.
인터루킨-12(IL-12)는 면역세포 중에서도 세포 독성 T 세포 (CTL; Cytotoxic T Lymphocytes), 자연 살해 세포 (NK; Natural Killer cells)와 같은 면역세포의 활성을 증가시켜 종양을 직접 죽이거나, 인터페론 감마(IFN-γ)와 같은 프로-염증성 사이토카인(pro-inflammatory cytokine)의 분비를 통해 면역 반응이 억제되어 있는 종양미세환경 (tumor microenvironment) 내 면역반응을 활성화시킴으로써, 종양유발(tumorigenesis)을 억제할 수 있다는 점에서 항암 사이토카인(anti-cancer cytokine)으로 많은 연구가 이루어져 왔다 (Lasek et al., 2014). 하지만 인터루킨-12를 이용한 치료법의 개발에 있어 사이토카인 자체의 짧은 반감기는 지속적인 투여를 요구하게 되고, 이는 곧 독성으로 이어질 수 있기 때문에, 이를 항체나 Fc와의 융합(fusion)을 통해 long-acting IL-12의 형태로 이용하고자 하는 연구들이 있었다 (Tugues et al., 2015). 하지만 상기 연구들은 CH3 도메인간의 상호작용을 통해 동종이중체를 이루는 야생형 항체의 융합으로 인해, 내생형 사이토카인 (endogenous form cytokine)과는 다르게 bivalency를 가지게 되고 이로 인해, 내생형 인터루킨 12 보다 떨어지는 생리활성을 보이거나, 인터루킨 12로 인한 원하지 않는 국소화 현상(localization)이 나타나는 등의 문제가 함께 발생되고 있는 상황이다(Tzeng et al., 2015; Dumont et al., 2006). Interleukin-12 (IL-12) increases the activity of immune cells, such as cytotoxic T Lymphocytes (CTL) and natural killer cells (NK), among other immune cells, to directly kill tumors or interferon Inhibits tumorigenesis by activating the immune response in the tumor microenvironment where the immune response is suppressed through the secretion of pro-inflammatory cytokine such as gamma (IFN-γ) Much research has been done on anti-cancer cytokine in that it can be done (Lasek et al., 2014). However, in the development of treatment with interleukin-12, the short half-life of the cytokine itself requires continuous administration, which can lead to toxicity, so long-acting IL- via fusion with antibodies or Fc. There have been studies to use in the form of 12 (Tugues et al., 2015). However, these studies have been shown to have bivalency, unlike endogenous form cytokine, due to the fusion of homozygous wild-type antibodies through interactions between CH3 domains, resulting in lower bioactivity than endogenous interleukin 12. Or problems such as undesired localization due to interleukin 12 (Tzeng et al., 2015; Dumont et al., 2006).
따라서, 야생형 항체 혹은 Fc 영역을 이용해 monovalency를 가지는 융합 단백질을 만들기 위한 노력으로 도 1의 (A)~(C)와 같이 하나의 Fc 영역의 C 말단에만 추가적인 정제를 위한 선택적인 태그 (tag)를 융합하거나 Fc 영역과 단백질을 따로 고순도로 정제하여 융합하는 등의 전략을 통해 융합단백질을 구축하는 방법을 이용해왔다. 하지만 이러한 형태는 많은 양의 단백질을 생산하는 데 있어 손실이 클 뿐만 아니라, 추가적인 정제 과정의 최적화를 위한 연구 또한 필요로 한다. Therefore, in an effort to produce a monovalency fusion protein using a wild-type antibody or an Fc region, an optional tag for further purification only at the C terminus of one Fc region, as shown in FIGS. The method of constructing a fusion protein has been used by fusion or by purifying the Fc region and protein separately in high purity. However, this form is not only costly in producing large amounts of protein, but also requires research to further optimize the purification process.
하지만, 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)의 형태를 이용하면 추가적인 정제 과정의 최적화 과정 없이 도 2와 같은 상기 monovalent 형태의 이종이중체-융합단백질을 용이하게 제조할 수 있다. However, using the form of the antibody heavy chain constant region heterodimer-fusion protein (heterodimeric Fc-fused protein) according to the present invention without the additional purification process optimization of the monovalent heterodimer-fusion protein as shown in Figure 2 It can be manufactured easily.
발명의 요약Summary of the Invention
본 발명에서 해결하고자 하는 과제는 2 이상의 서브유닛이 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 단백질의 활성이 오랜 시간 동안 충분히 유지될 수 있는 새로운 형태의 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)을 제공하는 것이다. The problem to be solved in the present invention is a heterozygote-fusion heterozygous form of a new type of antibody heavy chain constant region in which two or more subunits form a protein complex so that the activity of the protein showing physiological activity can be sufficiently maintained for a long time. It is to provide a protein (heterodimeric Fc-fused protein).
특히, 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)은 2 이상의 서브유닛이 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 단백질을 자연계에 존재하는 형태 그대로 최대한 모사함으로써, 자연계에 존재하는 그대로의 활성을 유지할 수 있는 형태이다. In particular, the antibody heavy chain constant region heterodimeric Fc-fused protein according to the present invention has two or more subunits that form a protein complex to form a protein complex that exhibits physiological activity as it exists in nature. By simulating as much as possible, it is a form which can maintain the activity as it exists in nature.
더불어 항체 중쇄불변부위 융합으로 융합단백질의 체내 반감기를 현저하게 증가되어, 체내에서의 각종 생리활성이 오랜 시간 동안 지속될 수 있는 장점이 있다.In addition, the half-life of the fusion protein is significantly increased by fusion of the antibody heavy chain constant region, so that various physiological activities in the body can be maintained for a long time.
또한, 본 발명은 상기 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)을 포함하는 약제학적 조성물 및 이를 이용한 질병, 특히 암의 치료를 위한 조성물 및 치료방법을 제공하는 것을 목적으로 한다. The present invention also provides a pharmaceutical composition comprising the antibody heavy chain constant region heterodimeric Fc-fused protein, and a composition and treatment method for the treatment of diseases, in particular cancer, using the same. do.
상기 과제를 해결하기 위하여, 본 발명은 항체(immunoglobulin) 중쇄불변부위 (Fc) 쌍의 제1 Fc 영역 및 제2 Fc 영역을 포함하고, In order to solve the above problems, the present invention includes a first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
상기 제1 Fc 영역 및/또는 제2 Fc 영역의 N-말단 또는 C-말단 중 하나 이상의 말단에 생리활성 단백질이 결합되어 있는 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, In a heterodimeric Fc-fused protein in which a bioactive protein is bound to at least one of the N-terminus or C-terminus of the first Fc region and / or the second Fc region,
상기 생리활성 단백질은 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서로 다른 서브유닛(subunit)은 하나의 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 것을 특징으로 하며,The physiologically active protein is composed of two or more different subunits (subunit), two or more different subunits (subunit) is characterized in that to form a protein complex (protein complex) to show the physiological activity,
상기 제1 Fc 영역 및 제2 Fc 영역은 이종이중체(heterodimeric Fc)의 형성이 촉진되도록 CH3 도메인이 변이된 것임을 특징으로 하는 이종이중체-융합단백질(heterodimeric Fc-fused protein)을 제공한다. The first Fc region and the second Fc region provide a heterodimeric Fc-fused protein, characterized in that the CH3 domain is mutated to promote formation of a heterodimeric Fc.
또한 본 발명은 상기 이종이종체-융합단백질을 포함하는 약제학적 조성물 및 이를 이용한 질병, 특히 암의 치료를 위한 조성물 및 치료방법을 제공한다. The present invention also provides a pharmaceutical composition comprising the heterologous-fusion protein and a composition and a method for the treatment of diseases, in particular cancer, using the same.
도 1(A) 내지 1(C)는 기존 야생형 인간 항체 중쇄불변부위를 이용해 단량체 및 이종이중체 형태의 융합단백질을 얻기 위한 전략을 나타낸 도면이다. 1 (A) to 1 (C) is a diagram showing a strategy for obtaining a fusion protein in the form of monomers and heterodimers using existing wild-type human antibody heavy chain constant region.
도 1(D)는 기존 문헌에서 놉-인투-홀 이종이중체 중쇄불변부위 (KiH heterodimeric Fc) 변이체가 포함된 IgG 형태의 항체에 단량체 사이토카인을 융합시켜 항체-사이토카인 (Immunocytokine)을 구축한 사례를 나타낸 도면이다. FIG. 1 (D) shows an antibody-cytokine (Immunocytokine) construct by fusing monomeric cytokines to an IgG-type antibody containing a KnoH-in-hole heterodimeric heavy chain constant region (KiH heterodimeric Fc) variant in the existing literature. It is a figure which shows an example.
도 2(A) 및 2(B)는 이종이중체 중쇄불변부위를 이용해 구축할 수 있는 단량체 및 이종이중체 융합단백질 형태를 제시한 도면이다. Figure 2 (A) and 2 (B) is a diagram showing the monomer and heterodimer fusion protein forms that can be constructed using heterodimeric heavy chain constant region.
도 2(C)는 이종이중체 중쇄불변부위가 포함된 IgG 형태의 인간 항체에 이종이중체 융합단백질 형태를 제시한 도면이다. Figure 2 (C) is a diagram showing a heterodimeric fusion protein form in a human antibody of the IgG form containing a heterodimeric heavy chain constant region.
도 3은 인간 항체 동종형별 이종이중체 형성을 위한 CH3 도메인 변이체의 제작을 위해 각 인간 항체 면역글로불린 G 동종형별 CH3 도메인의 서열을 나열하여 비교하고, 잠재적 돌연변이 위치를 선정한 결과를 나타낸 도면이다. Figure 3 is a view showing the results of selecting and comparing the sequence of the sequence of each human antibody immunoglobulin G isotype CH3 domain for the production of CH3 domain variants for heterodimer formation by human antibody isotype.
도 4는 도 3에서 선정된 위치에 돌연변이를 유도한 서열을 가지고 동종형별 이종이중체 중쇄불변부위 변이체의 구조 모델링을 진행, 그 결과로 얻어진 모델링 구조를 각각 야생형 IgG1 기반의 A107 돌연변이체와 비교 및 분석한 결과를 나타낸 도면이다. FIG. 4 is a structural modeling of heterologous heavy double chain constant region variants of the heterologous type having the mutation-induced sequence at the position selected in FIG. 3, and the resulting modeling structure is compared with the wild type IgG1 based A107 mutant and It is a figure which shows the result of an analysis.
도 5는 서열 및 구조 분석을 통해 구축된 동종형별 이종이중체 중쇄불변부위를 동물 세포에서 발현하기 위한 벡터의 모식도이다. 변이된 힌지 영역을 포함한 각 동종형별 이종이중체 중쇄불변부위 변이체는 제한효소 NotI/HindIII를 이용하여 벡터에 클로닝하였다. 5 is a schematic diagram of a vector for expressing heterologous heavy chain constant regions of homologous type constructed by sequence and structural analysis in animal cells. Heterozygous heavy chain constant region variants of each homotype, including the mutated hinge region, were cloned into the vector using the restriction enzyme NotI / HindIII.
도 6은 발현되는 단백질의 이중체 사이즈 차이로 이종이중체 중쇄불변부위 변이체가 어느 정도의 이종이중체 형성능을 가지는지 평가하기 위한 scFv-FcCH3A/FcCH3B 동시 발현 시스템을 간략하게 모식도로 나타낸 도면이다.FIG. 6 is a schematic diagram schematically illustrating a scFv-Fc CH3A / Fc CH3B co-expression system for evaluating the degree of heterodimer formation of heteroduplex heavy chain constant region variants due to the duplex size difference of the expressed protein. to be.
도 7은 도 6에서와 같이 CH3 돌연변이 쌍에 의한 항체 중쇄불변부위의 이종이중체의 형성 수율을 평가하기 위해 구축된 단일 사슬 항체 절편(scFv)이 융합된 scFv-Fc를 동물 세포 발현 벡터인 pcDNA3.1 벡터에 클로닝하기 위해 구축한 모식도이다. FIG. 7 shows a single-chain antibody fragment (scFv) fused with a single chain antibody fragment (scFv) constructed to evaluate the formation yield of heteroduplex of the antibody heavy chain constant region by the CH3 mutation pair as shown in FIG. 6. .1 Schematic diagram for cloning into vectors.
도 8은 도 5 및 도 7에 기재된 발현시스템에 따라 구축된 CH3 돌연변이 쌍이 도입된 동물세포 발현벡터를 도 6에서 기술한 이종이중체의 형성능 평가를 위해 HEK293F 세포에 공동형질 전환을 통해 일시적으로 발현 및 정제한 다음, 이종이중체 항체 형성능의 평가를 위해 5μg의 단백질을 비환원성 조건의 SDS-PAGE상에서 분리하고, Coomasie Blue 염색을 통해 크기 및 조합형태로 분석한 결과를 나타낸 도면이다. 이 때, 음성대조군으로 야생형 CH3가 이용된 야생형 Fc 을 이용하였다. Figure 8 is a transient expression of co-transformation in HEK293F cells for evaluating the formation ability of the heteroduplex described in Figure 6 animal cell expression vector introduced with a CH3 mutation pair constructed in accordance with the expression system described in Figures 5 and 7 And, after purification, 5 μg of protein was isolated on SDS-PAGE under non-reducing conditions for evaluation of heterodimer antibody formation ability, and analyzed by size and combination through Coomasie Blue staining. At this time, wild type Fc using wild type CH3 was used as a negative control.
도 9는 도 8과 같은 방법으로 SDS-PAGE로 단백질을 분리한 이후, AP 효소가 달린 항 인간 IgG 항체 (anti-human IgG-AP conjugated antibody)를 이용하여 웨스턴 블랏을 수행한 결과를 나타낸 도면이다. 9 is a diagram showing the result of Western blot using the anti-human IgG-AP conjugated antibody with AP enzyme after protein separation by SDS-PAGE in the same manner as in FIG. 8. .
도 10(A)는 본 발명의 대조군이 되는, 중쇄불변부위가 융합되지 않은 내생형의 인터루킨 12 사이토카인의 형태를 나타낸 모식도이다. Figure 10 (A) is a schematic diagram showing the form of the endogenous interleukin 12 cytokine unfused to the heavy chain constant region, which is a control of the present invention.
도 10(B)는 본 발명의 비교예로 아미노산 링커로 연결된 인터루킨 12 사이토카인을 야생형 IgG4 Fc에 융합한 bi-IL-12-Fc 융합단백질의 형태를 나타낸 모식도이다. FIG. 10 (B) is a schematic diagram showing the form of a bi-IL-12-Fc fusion protein in which an interleukin 12 cytokine linked with an amino acid linker is fused to a wild-type IgG4 Fc as a comparative example of the present invention.
도 10(C)는 인터루킨 12 사이토카인을 본 발명의 동종형별 이종이중체 중쇄불변부위 변이체 중 IgG4를 기반으로 만들어진 γ4-A107 변이체를 융합한 mono-IL-12-Fc 융합단백질의 형태를 나타낸 모식도이다.10 (C) is a schematic diagram showing the form of a mono-IL-12-Fc fusion protein in which the interleukin 12 cytokine is fused with a γ4-A107 variant made of IgG4 in a heterologous heavy double chain constant region variant of the present invention. to be.
도 11(A) 및 11(B)는 본 발명의 실시예(도 10c)의 융합단백질을 동물세포에서 발현 및 정제하기 위한 벡터의 모식도이다. 11 (A) and 11 (B) are schematic diagrams of vectors for expressing and purifying the fusion protein of the embodiment of the present invention (FIG. 10C) in animal cells.
도 12는 본 발명의 비교예(도 10(B)의 융합단백질을 동물세포에서 발현 및 정제하기 위한 벡터의 모식도이다. Figure 12 is a schematic diagram of a vector for the expression and purification of the fusion protein of the comparative example (Fig. 10 (B) in the animal cell of the present invention.
도 13은 인간 및 마우스의 인터루킨 유전자를 가지고 구축된 도 11(A) 및 (B)의 동물세포 발현벡터를 HEK293F 세포에 공동형질 전환을 통해 일시적으로 발현 및 정제한 다음, 5μg의 단백질을 비환원성 조건의 SDS-PAGE상에서 분리하고, Coomasie Blue 염색을 통해 크기 및 조합형태로 분석한 결과를 나타내는 도면이다.FIG. 13 transiently expresses and purifies the animal cell expression vectors of FIGS. 11 (A) and (B) constructed with human and mouse interleukin genes through cotransformation into HEK293F cells, and then amplifies 5 μg of protein. It is a diagram showing the results of the analysis on the size and combination form by separating on the SDS-PAGE of the condition, Coomasie Blue staining.
도 14는 도 13의 융합단백질들을 크기 배제 크로마토그래피를 이용해 분석한 결과를 나타낸 도면이다. FIG. 14 shows the results of analyzing the fusion proteins of FIG. 13 using size exclusion chromatography.
도 15는 IL-12에 대한 수용체를 갖지 않는 정상 말초혈액 면역세포 (normal PBMC)와 mitogen인 PHA(phytohaemagglutinin)를 처리하여 IL-12에 대한 수용체를 유도시킨 말초혈액 면역세포 (PHA-activated PBMC)에 대하여 구축된 mono-hIL-12-Fc와 야생형 bi-hIL-12-Fc의 결합능을 유세포 분석기 (FACS)로 확인한 결과를 나타내는 도면이다.FIG. 15 shows peripheral blood immune cells (PHA-activated PBMCs) that induced receptors for IL-12 by treating normal peripheral blood immune cells (normal PBMCs) having no receptor for IL-12 and phytohaemagglutinin (PHA). It is a figure which shows the result of having confirmed the binding ability of the mono-hIL-12-Fc and the wild type bi-hIL-12-Fc which were constructed about with the flow cytometer (FACS).
도 16은 Mitogen인 PHA를 처리하여 IL-12에 대한 수용체를 유도시킨 말초혈액 면역세포 (PHA-activated PBMC)에서 Fc (A107), recombinant human IL-12(rhIL-12), bi-hIL-12-Fc와 mono-hIL-12-Fc의 농도별 처리에 따른 세포증식을 WST-1 세포증식 어세이를 통해 측정한 결과를 나타내는 도면이다. FIG. 16 shows Fc (A107), recombinant human IL-12 (rhIL-12), and bi-hIL-12 in peripheral blood immune cells (PHA-activated PBMCs) inducing receptors for IL-12 by treating PHA, a mitogen. Figure showing the results of measuring the cell proliferation according to the concentration-specific treatment of -Fc and mono-hIL-12-Fc through the WST-1 cell proliferation assay.
도 17은 도 16에서 배양된 배양 상청액 내의 IFN-γ의 농도를 ELISA를 통해 측정한 결과를 나타내는 도면이다17 is a view showing the results of measuring the concentration of IFN-γ in the culture supernatant cultured in Figure 16 by ELISA
도 18은 마우스 IL-12가 마우스 IL-12 수용체뿐만 아니라 인간 IL-12 수용체에도 결합한다는 특성을 이용하여 구축된 mono-mIL-12-Fc와 bi-mIL-12-Fc의 결합능을 정상 말초혈액 면역세포 (normal PBMC)와 PHA라는 mitogen을 처리하여 IL-12에 대한 수용체를 유도시킨 말초혈액 면역세포 (PHA-activated PBMC)를 이용하여 유세포 분석기로 확인한 결과를 나타내는 도면이다 FIG. 18 shows normal peripheral blood binding ability of mono-mIL-12-Fc and bi-mIL-12-Fc constructed using the property that mouse IL-12 binds to human IL-12 receptor as well as mouse IL-12 receptor. A diagram showing the results confirmed by flow cytometry using peripheral blood immune cells (PHA-activated PBMCs) inducing receptors for IL-12 by treating immune cells (normal PBMC) and mitogen called PHA.
도 19는 PHA를 처리하여 IL-12에 대한 수용체를 유도시킨 말초혈액 면역세포 (PHA-activated PBMC)에서 Fc (A107), recombinant mouse IL-12(rmIL-12), bi-mIL-12-Fc와 mono-mIL-12-Fc의 농도 별 처리에 따른 세포증식을 WST-1 세포증식 어세이를 통해 측정한 결과를 나타내는 도면이다. FIG. 19 shows Fc (A107), recombinant mouse IL-12 (rmIL-12), bi-mIL-12-Fc in peripheral blood immune cells (PHA-activated PBMCs) inducing receptors for IL-12 by PHA treatment. And cell proliferation according to the concentration-specific treatment of mono-mIL-12-Fc is a diagram showing the results measured by the WST-1 cell proliferation assay.
도 20(A)는 CT26HER2 / Neu 암세포를 이식한 Balb/c 마우스에서 종양의 크기가 100 mm3일때 Fc (A107), rmIL-12, bi-mIL-12-Fc와 mono-mIL-12-Fc를 복강 투여하면서 측정한 종양 부피의 변화, 투여 끝에 마우스의 치사 후 종양의 크기의 확인결과를 나타내는 도면이다.Figure 20 (A) shows Fc (A107), rmIL-12, bi-mIL-12-Fc and mono-mIL-12-Fc when the tumor size is 100 mm3 in Balb / c mice transplanted with CT26 HER2 / Neu cancer cells It is a diagram showing the results of confirming the change in tumor volume measured during intraperitoneal administration and the size of tumor after lethality of mice at the end of administration.
도 20(B)는 상기 도 20(A)의 실험과정에서 주기적으로 측정된 마우스 체중 변화를 나타낸 그래프이다.20 (B) is a graph showing the weight change of the mouse periodically measured during the experiment of FIG. 20 (A).
도 21(A)는 CT26HER2 / Neu 이식 Balb/c 마우스에서 종양의 크기가 300 mm3일때 bi-mIL-12-Fc와 mono-mIL-12-Fc를 일주일에 두 번씩 농도 별로 복강 투여하면서 마우스의 종양 부피의 변화를 측정한 도면이다. 21 (A) shows bi-mIL-12-Fc and mono-mIL-12-Fc intraperitoneally administered twice a week at the tumor size of 300 mm 3 in CT26 HER2 / Neu transplanted Balb / c mice. It is a figure which measured the change of the tumor volume.
도 21(B)는 상기 도 21(A)의 실험과정에서 주기적으로 측정된 개별 마우스의 종양의 부피변화를 나타낸 그래프이다. Figure 21 (B) is a graph showing the change in the volume of the tumor of the individual mice periodically measured in the experimental procedure of Figure 21 (A).
도 21(C)는 상기 도 21(A)의 마지막 투여 후 3일째 마우스를 치사하여 종양의 크기의 확인한 결과를 나타내는 도면이다.Figure 21 (C) is a diagram showing the result of confirming the tumor size by killing the mouse 3 days after the last administration of Figure 21 (A).
도 21(D)는 상기 도 21(A)의 실험과정에서 주기적으로 측정된 마우스 체중 변화를 나타낸 그래프이다.Figure 21 (D) is a graph showing the weight change of the mouse periodically measured in the experimental procedure of Figure 21 (A).
도 21(E)는 상기 도 21(A)의 마지막 투여를 마친 후 1일째에 마우스의 얼굴 정맥에서 혈액을 채취하여 간 독성의 지표인 알라닌 아미노 전이효소 (ALT) 측정한 그래프이다.FIG. 21 (E) is a graph of alanine aminotransferase (ALT) which is an indicator of liver toxicity by collecting blood from the facial vein of the mouse on day 1 after the last administration of FIG. 21 (A).
도 22(A)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 CD4+ T세포, CD8+ T세포 및 NK세포 수의 증가를 측정한 그래프이다. Figure 22 (A) is a graph measuring the increase in the number of CD4 + T cells, CD8 + T cells and NK cells in the spleen after 3 days of the last administration of Figure 21 (A).
도 22(B)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 종양에 침윤되어 있는 총 면역세포, CD4+ T세포, CD8+ T세포의 수를 나타내는 도면이다.Figure 22 (B) is a view showing the total number of immune cells, CD4 + T cells, CD8 + T cells infiltrated into the tumor after 3 days of the third administration of Figure 21 (A).
도 23(A)는 상기 도 21(A)의 마지막 투여 24시간 후에 마우스의 얼굴 정맥에서 채혈하여 분리한 혈청내의 IFN-g 농도를 ELISA로 측정한 결과이다. Figure 23 (A) is the result of measuring the IFN-g concentration in the serum isolated by collecting blood from the facial vein of the mouse 24 hours after the last administration of Figure 21 (A) by ELISA.
도 23(B)는 CT26HER2 / Neu 암세포를 이식한 Balb/c 마우스에서 종양의 크기가 300 mm3일 때 1μg rmIL-12와 같은 몰 농도의 bi-mIL-12-Fc와 mono-mIL-12-Fc를 복강 투여한 후 1일, 3일 및 5일 후에 마우스의 얼굴 정맥에서 채혈하여 분리한 혈청내의 IFN-γ 농도를 ELISA로 측정한 그래프이다.FIG. 23 (B) shows bi-mIL-12-Fc and mono-mIL-12 at a molar concentration of 1 μg rmIL-12 when the tumor size was 300 mm 3 in Balb / c mice transplanted with CT26 HER2 / Neu cancer cells. IFN-γ concentration in serum isolated by blood collection from the facial veins of mice 1, 3 and 5 days after intraperitoneal administration of -Fc was measured by ELISA.
도 23(C)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 분리한 세포독성 T 세포의 CT26HER2 / Neu 암세포에 대한 세포독성효과를 측정한 그래프이다. Figure 23 (C) is a graph measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of cytotoxic T cells isolated from the spleen by killing the mouse 3 days after the last administration of Figure 21 (A).
도 23(D)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에서 분리한 세포독성 T 세포의 세포독성효과를 종양항원을 발현하는 CT26HER2 / Neu 암세포와 발현하지 않는 4T1세포를 이용하여 유세포 분석기로 분석한 도면이다. Figure 23 (D) shows the cytotoxic effect of cytotoxic T cells isolated from the spleen by killing mice 3 days after the third administration of Figure 21 (A) does not express CT26 HER2 / Neu cancer cells expressing tumor antigens It is a figure analyzed by the flow cytometer using 4T1 cells.
도 23(E)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에서 분리한 자연살해세포의 CT26HER2 / Neu 암세포에 대한 세포독성 효과를 측정한 그래프이다. Figure 23 (E) is a graph measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of natural killer cells isolated from the spleen by killing the mouse 3 days after the third administration of Figure 21 (A).
도 24(A)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 분리한 CD8+ 효과 T 세포의 수를 측정한 그래프이다. Figure 24 (A) is a graph measuring the number of CD8 + effect T cells isolated from the spleen by lethal mice three days after the last administration of Figure 21 (A).
도 24(B)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 분리한 CD8+ 효과기억 T 세포의 수를 측정한 그래프이다. Figure 24 (B) is a graph measuring the number of CD8 + effect memory T cells isolated from the spleen by lethal mice three days after the last administration of Figure 21 (A).
도 24(C)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 분리한 CD8+ 중심기억 T 세포의 수를 측정한 그래프이다. Figure 24 (C) is a graph measuring the number of CD8 + central memory T cells isolated from the spleen by killing the mouse three days after the last administration of Figure 21 (A).
도 24(D)는 상기 도 21(A)에서 1㎍ mono-IL-12-Fc의 투여 120일 후에 생존한 마우스와 같은 주령의 Balb/c 마우스에 CT26HER2 / Neu 암세포를 재이식하여 마우스의 종양 부피의 변화를 측정한 도면이다.FIG. 24 (D) shows CT26 HER2 / Neu in Balb / c mice of the same age as those surviving 120 days after administration of 1 μg mono-IL-12-Fc in FIG. 21 (A). Cancer cells are transplanted to measure the change in the tumor volume of the mouse.
도 24(E)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에 존재하는 CD8+ T세포 중 기억전구효과세포(KLRG1-IL-7R+)와 단명효과세포(KLRG1+IL-7R-)의 비율을 유세포 분석기(flow cytometry)로 분석한 도면이다. FIG. 24 (E) shows memory precursor effect cells (KLRG1 - IL-7R + ) and short-lived effect cells (KLRG1) among CD8 + T cells present in the spleen after 3 days of the third administration of FIG. 21 (A). + IL-7R -) is a diagram of analyzing the percentage of by flow cytometry (flow cytometry).
도 25(A)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사한 후 비장세포를 분리하여 기억세포의 분화를 억제하는 전사인자인 T-bet의 발현이 높은 CD8+ T세포의 비율을 유세포 분석기로 측정하여 분석한 그래프이다. FIG. 25 (A) shows the ratio of CD8 + T cells with high expression of T-bet, a transcription factor that inhibits the differentiation of memory cells by killing mice after 3 days of the third administration of FIG. 21 (A). It is a graph analyzed by measuring with a flow cytometer.
도 25(B)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사한 후 비장세포를 분리하여 기억세포의 분화를 촉진하는 Eomes의 발현이 높고 T-bet의 발현이 낮은 CD8+ T세포의 비율을 유세포 분석기로 측정하여 분석한 그래프이다. FIG. 25 (B) shows CD8 + T with high expression of Eomes and low expression of T-bet that promote the differentiation of memory cells by killing mice after 3 days of the third administration of FIG. 21 (A). It is a graph analyzed by measuring the percentage of cells by flow cytometry.
도 25(C)는 CT26HER2 / Neu 암세포를 이식한 Balb/c 마우스에서 종양의 크기가 300 mm3일 때 1 ㎍ rmIL-12와 같은 몰 농도의 bi-mIL-12-Fc와 mono-mIL-12-Fc를 1회 복강 투여하고 24시간 후에 사타구니 림프절에서 분리한 CD8+ T 세포에서 인산화된 STAT4의 발현 양을 유세포 분석기로 측정한 그래프이다. Figure 25 (C) shows CT26 HER2 / Neu Balb / c mice transplanted with cancer cells were intraperitoneally administered with bi-mIL-12-Fc and mono-mIL-12-Fc at a molar concentration of 1 μg rmIL-12 when the tumor size was 300 mm 3. After time, the expression level of phosphorylated STAT4 in CD8 + T cells isolated from inguinal lymph nodes was measured by flow cytometry.
도 25(D)는 도 25(C)의 1회 복강투여 72시간 후에 사타구니 림프절에서 기억세포의 분화를 억제하는 T-bet을 발현하는 CD8+ T 세포의 비율을 유세포 분석기로 측정한 그래프이다. Figure 25 (D) is a graph measuring the ratio of CD8 + T cells expressing T-bet inhibiting the differentiation of memory cells in the inguinal lymph nodes 72 hours after one intraperitoneal administration of Figure 25 (C) by flow cytometry.
도 25(E)는 정상 Balb/c 마우스의 비장과 사타구니 림프절에서 분리한 CD8+ T 세포를 Fc를 인지하는 항체로 교차 결합시킨 mono-mIL-12-Fc와 bi-mIL-12-Fc로 자극하였을 때 pSTAT4의 발현양을 유세포 분석기로 측정한 그래프이다. 25 (E) is stimulated with mono-mIL-12-Fc and bi-mIL-12-Fc cross-linked CD8 + T cells isolated from spleen and groin lymph nodes in normal Balb / c mice with Fc-recognized antibodies. When the expression level of pSTAT4 is measured by a flow cytometer.
도 25(F)는 정상 Balb/c 마우스의 비장과 사타구니 림프절에서 분리한 CD8+ T 세포를 Fc를 인지하는 항체로 교차 결합시킨 mono-mIL-12-Fc와 bi-mIL-12-Fc로 자극하였을 때 T-bet을 발현하는 CD8+ T 세포의 비율을 유세포 분석기로 측정한 그래프이다. FIG. 25 (F) shows that CD8 + T cells isolated from the spleen and inguinal lymph nodes of normal Balb / c mice were stimulated with mono-mIL-12-Fc and bi-mIL-12-Fc cross-linked with an antibody that recognizes Fc. When the ratio of CD8 + T cells expressing T-bet is a graph measured by flow cytometry.
도 26은 mono-mIL-12-Fc에 의한 기억전구효과세포와 기억세포의 분화 유도 기전 및 bi-mIL-12-Fc에 의한 단명효과세포의 분화 유도기전을 나타낸 전반적인 모식도이다. FIG. 26 is a schematic diagram showing the differentiation-induced mechanism of memory precursor effector and memory cells induced by mono-mIL-12-Fc and the differentiation-induced mechanism of short-lived effect cells induced by bi-mIL-12-Fc.
발명의 상세한 설명 및 바람직한 Detailed description of the invention and preferred 구현예Embodiment
다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로, 본 명세서에서 사용된 명명법은 본 기술분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In general, the nomenclature used herein is well known and commonly used in the art.
본 발명은 일 관점에서, 항체(immunoglobulin) 중쇄불변부위 (Fc) 쌍의 제1 Fc 영역 및 제2 Fc 영역을 포함하고, In one aspect, the present invention includes a first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
상기 제1 Fc 영역 및/또는 제2 Fc 영역의 N-말단 또는 C-말단 중 하나 이상의 말단에 생리활성 단백질이 결합되어 있는 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, In a heterodimeric Fc-fused protein in which a bioactive protein is bound to at least one of the N-terminus or C-terminus of the first Fc region and / or the second Fc region,
상기 생리활성 단백질은 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서로 다른 서브유닛(subunit)은 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 것을 특징으로 하며,The bioactive protein is composed of two or more different subunits (subunit), the two or more different subunits (subunit) is characterized in that the protein complex (protein complex) to form a physiological activity,
상기 제1 Fc 영역 및 제2 Fc 영역은 이종이중체(heterodimer)의 형성이 촉진되도록 CH3 도메인이 변이된 것임을 특징으로 하는 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 대한 것이다. The first Fc region and the second Fc region are for a heterodimeric Fc-fused protein, characterized in that the CH3 domain is mutated to promote formation of a heterodimer.
본 발명에서 있어서, “Fc 영역” 또는 “중쇄불변부위”는 항체 유래의 CH2 도메인, CH3 도메인 및 힌지 영역(hinge domain)을 포함하는 영역을 의미한다. 다만, IgE의 경우에는 CH2 도메인, CH3 도메인, CH4 도메인 및 힌지 영역(hinge domain)을 포함하는 영역을 의미한다. In the present invention, “Fc region” or “heavy chain constant region” means a region including a CH2 domain, a CH3 domain, and a hinge domain derived from an antibody. However, in the case of IgE, it means a region including a CH2 domain, a CH3 domain, a CH4 domain, and a hinge domain.
본 발명에서의 “제1 Fc 영역 및 제2 Fc 영역은 이종이중체(heterodimer)의 형성이 촉진되도록 변이”되었다는 표현은 자연계에 존재하는 항체는 2개의 Fc 영역이 서로 동일한 서열을 가지는 동종이중체(homodimer) 형태를 가지게 되는데, 이러한 Fc 영역의 일부 서열에 변이를 유발시킴으로써, 제1 Fc 영역 및 제2 Fc 영역 간의 특정 비공유 결합을 통해 이종이중체(heterodimer)의 형성이 촉진되며, 동종이중체의 형성이 감소되거나, 바람직하게는 거의 일어나지 않도록 변이되었다는 것을 의미한다. In the present invention, the expression “the first Fc region and the second Fc region are mutated to promote the formation of heterodimers” indicates that antibodies present in nature are homodimers in which two Fc regions have the same sequence. (homodimer) form, which causes mutations in some sequences of these Fc regions, thereby promoting the formation of heterodimers through specific non-covalent linkages between the first and second Fc regions and homodimers. It means that the formation of is reduced or preferably mutated so that it hardly occurs.
바람직하게는 본 발명에 따른 제1 Fc 영역 및 제2 Fc 영역의 이종이종체(heterodimer)의 형성이 촉진되도록 하는 변이는, 상기 항체 유래 제1 Fc 영역 및 제2 Fc 영역에 포함된 CH3 도메인 각각이 이종이중체(heterodimer)의 형성이 촉진되도록 하는 변이를 포함할 수 있다. Preferably, the mutations to facilitate the formation of a heterodimer of the first Fc region and the second Fc region according to the present invention are each of the CH3 domains included in the antibody-derived first and second Fc regions. May include mutations that facilitate the formation of this heterodimer.
본 발명에 있어서, “중쇄불변부위 이종이중체(heterodimeric Fc 또는 Fc heterodimer)”는 제1 Fc 영역 및 제2 Fc 영역을 포함하고, 상기 제1 Fc 영역 및 제2 Fc 영역은 이종이중체(heterodimer)의 형성이 촉진되도록 CH3 도메인이 변이된 것임을 특징으로 하는 이종이중체를 의미한다. In the present invention, “heaterodimeric Fc or Fc heterodimer” includes a first Fc region and a second Fc region, and the first Fc region and the second Fc region are heterodimers. It means a heterodimer, characterized in that the CH3 domain is modified to promote the formation of).
본 발명에서의 상기 제1 Fc 영역 및 제2 Fc 영역은 각각 인간 IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD 및 IgE로 이루어진 군으로부터 선택된 Fc 영역 유래인 것을 특징으로 할 수 있으며, 바람직하게는 상기 제1 Fc 영역 및 제2 Fc 영역은 각각 IgG1, IgG2, IgG3, 또는 IgG4 유래인 것을 특징으로 하고, The first Fc region and the second Fc region in the present invention may be derived from an Fc region selected from the group consisting of human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, and IgE, respectively, preferably Wherein the first Fc region and the second Fc region are derived from IgG1, IgG2, IgG3, or IgG4, respectively,
또한 상기 제1 Fc 영역 및 제2 Fc 영역은 동종형(isotype) 항체 유래인 것을 특징으로 할 수 있다. In addition, the first Fc region and the second Fc region may be characterized in that it is derived from an isotype antibody.
다른 관점에서, 상기 CH3 도메인의 변이는 다음의 군에서 선택된 하나 이상의 변이를 포함하는 것을 특징으로 할 수 있다. 본 발명에서의 모든 변이 위치는 EU index에 따른다. In another aspect, the mutation of the CH3 domain may be characterized in that it comprises one or more mutations selected from the following group. All mutation positions in the present invention are according to the EU index.
(1) 제1 Fc 영역의 CH3 도메인의 K370 위치에서의 아미노산 잔기의 치환; 및 제2 제2 Fc 영역의 CH3 도메인의 E357 및/또는 S364 위치에서의 아미노산 잔기의 치환; 및/또는 (1) substitution of the amino acid residue at position K370 of the CH3 domain of the first Fc region; And substitution of an amino acid residue at the E357 and / or S364 position of the CH3 domain of a second second Fc region; And / or
(2) 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 아미노산 잔기의 치환; 및 제2 Fc 영역의 CH3 도메인의 F405 및/또는 D399 위치에서의 아미노산 잔기의 치환.(2) substitution of the amino acid residue at position K409 of the CH3 domain of the first Fc region; And substitution of the amino acid residue at the F405 and / or D399 position of the CH3 domain of the second Fc region.
바람직하게는 상기 제1 Fc 영역의 CH3 도메인의 K370 위치에서의 아미노산 잔기의 치환은 K370E, K370R, K370M, K370D 또는 K370H이고, Preferably the substitution of the amino acid residue at the K370 position of the CH3 domain of the first Fc region is K370E, K370R, K370M, K370D or K370H,
상기 제2 Fc 영역의 CH3 도메인의 E357 위치에서의 아미노산 잔기의 치환은 E357N, E357D, E357A, E357I, E357G 또는 E357M이고, S364 위치에서의 아미노산 잔기의 치환은 S364T 또는 S364W인 것을 특징으로 할 수 있다. The substitution of the amino acid residue at the E357 position of the CH3 domain of the second Fc region may be E357N, E357D, E357A, E357I, E357G or E357M, and the substitution of the amino acid residue at the S364 position may be S364T or S364W. .
또한, 상기 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 아미노산 잔기의 치환은 K409W이고, 상기 제2 Fc 영역의 CH3 도메인의 F405 위치에서의 아미노산 잔기의 치환은 F405T이고, D399 위치에서의 아미노산의 치환은 D399V인 것을 특징으로 할 수 있다. The substitution of the amino acid residue at the K409 position in the CH3 domain of the first Fc region is K409W, and the substitution of the amino acid residue at the F405 position of the CH3 domain of the second Fc region is F405T, and the amino acid residue at the D399 position is The substitution may be characterized as being D399V.
상기 K370E와 같은 아미노산 잔기 변이는 370 번째 위치의 K가 E로 변이된 것을 의미하며, 본 발명에서의 모든 아미노산 잔기 변이는 이와 동일한 의미로 사용된다. An amino acid residue change such as K370E means that the K at position 370 is changed to E, and all amino acid residue changes in the present invention are used as the same meaning.
가장 바람직하게는 상기 제1 Fc 영역 또는 제2 Fc 영역의 CH3 도메인의 변이는 다음의 군에서 선택된 하나 이상의 변이를 포함하는 것을 특징으로 할 수 있다. (단, 변이 위치는 EU index에 따름)Most preferably, the mutation of the CH3 domain of the first Fc region or the second Fc region may include one or more mutations selected from the following group. (However, the mutation position is according to the EU index)
(1) 제1 Fc 영역의 CH3 도메인의 K370 위치에서의 K370E, K370R, K370M, K370D 또는 K370H의 아미노산 잔기의 치환; (1) substitution of the amino acid residue of K370E, K370R, K370M, K370D or K370H at the K370 position of the CH3 domain of the first Fc region;
(2) 제2 Fc 영역의 CH3 도메인의 E357 위치에서의 E357N, E357D, E357A, E357I, E357G 또는 E357M의 아미노산 잔기의 치환 및 S364 위치에서의 치환은 S364T 또는 S364W의 아미노산 잔기의 치환;(2) substitution of the amino acid residues of E357N, E357D, E357A, E357I, E357G, or E357M at the E357 position of the CH3 domain of the second Fc region and at the S364 position replaces the amino acid residue of S364T or S364W;
(3) 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 K409W의 아미노산 잔기의 치환; 및(3) substitution of the amino acid residue of K409W at the K409 position of the CH3 domain of the first Fc region; And
(4) 제2 Fc 영역의 CH3 도메인의 F405 위치에서의 F405T 아미노산 잔기의 치환 및 D399 위치에서의 D399V의 아미노산 잔기의 치환.(4) substitution of the F405T amino acid residue at the F405 position of the CH3 domain of the second Fc region and substitution of the amino acid residue of D399V at the D399 position;
상기와 같은 제1 Fc 영역 및 제2 Fc 영역의 CH3 도메인 내에는 하기의 결합을 추가로 포함하는 것을 특징으로 할 수 있다. The CH3 domains of the first Fc region and the second Fc region as described above may further include the following bonds.
(i) 제1 Fc 영역의 CH3 도메인 내 Y349 위치에 치환된 시스테인 (C); 및(i) cysteine (C) substituted at the Y349 position in the CH3 domain of the first Fc region; And
(ii) 제2 Fc 영역의 CH3 도메인 내 S354 위치에 치환된 시스테인 (C)의 결합.(ii) binding of cysteine (C) substituted at position S354 in the CH3 domain of the second Fc region.
또 다른 관점에서, 상기 CH3 도메인의 변이는 다음의 군에서 선택된 하나 이상의 변이를 포함하는 것을 특징으로 할 수 있다. In another aspect, the mutation of the CH3 domain may be characterized in that it comprises one or more mutations selected from the following group.
(1) 제1 Fc 영역의 CH3 도메인의 K360 위치에서의 아미노산 잔기의 치환; 및 제2 Fc 영역의 CH3 도메인의 E347 위치에서의 아미노산 잔기의 치환; 및/또는 (1) substitution of the amino acid residue at the K360 position of the CH3 domain of the first Fc region; And substitution of the amino acid residue at the E347 position of the CH3 domain of the second Fc region; And / or
(2) 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 아미노산 잔기의 치환; 및 제2 Fc 영역의 CH3 도메인의 F405 및 D399 위치에서의 아미노산 잔기의 치환. (2) substitution of the amino acid residue at position K409 of the CH3 domain of the first Fc region; And substitution of the amino acid residue at the F405 and D399 positions of the CH3 domain of the second Fc region.
바람직하게는 상기 제1 Fc 영역의 CH3 도메인의 K360 위치에서의 아미노산 잔기의 치환은 K360E이고, 상기 제2 Fc 영역의 CH3 도메인의 E347 위치에서의 아미노산 잔기의 치환은 E347R인 것을 특징으로 할 수 있으며, Preferably, the substitution of the amino acid residue at the K360 position of the CH3 domain of the first Fc region is K360E, and the substitution of the amino acid residue at the E347 position of the CH3 domain of the second Fc region may be E347R. ,
상기 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 아미노산 잔기의 치환은 K409W이고, 상기 제2 Fc 영역의 CH3 도메인의 F405 위치에서의 아미노산 잔기의 치환은 F405T이고, D399 위치에서의 아미노산의 치환은 D399V인 것을 특징으로 할 수 있다. Substitution of the amino acid residue at the K409 position of the CH3 domain of the first Fc region is K409W, Substitution of the amino acid residue at the F405 position of the CH3 domain of the second Fc region is F405T, Substitution of the amino acid at the D399 position is It may be characterized by the D399V.
가장 바람직하게는 상기 제1 Fc 영역 또는 제2 Fc 영역의 CH3 도메인의 변이는 다음의 군에서 선택된 하나 이상의 변이를 포함하는 것을 특징으로 할 수 있다. (단, 변이 위치는 EU index에 따름)Most preferably, the mutation of the CH3 domain of the first Fc region or the second Fc region may include one or more mutations selected from the following group. (However, the mutation position is according to the EU index)
(1) 제1 Fc 영역의 CH3 도메인의 K360 위치에서의 K360E의 아미노산 잔기의 치환; (1) substitution of the amino acid residue of K360E at the K360 position of the CH3 domain of the first Fc region;
(2) 제2 Fc 영역의 CH3 도메인의 E347 위치에서의 E347R의 아미노산 잔기의 치환; (2) substitution of the amino acid residue of E347R at the E347 position of the CH3 domain of the second Fc region;
(3) 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 K409W의 아미노산 잔기의 치환; (3) substitution of the amino acid residue of K409W at the K409 position of the CH3 domain of the first Fc region;
(4) 제2 Fc 영역의 CH3 도메인의 F405 위치에서의 F405T 아미노산 잔기의 치환 및 D399 위치에서의 D399V의 아미노산 잔기의 치환(4) substitution of the F405T amino acid residue at the F405 position of the CH3 domain of the second Fc region and at the D399 position for the amino acid residue of D399V
상기와 같은 제1 Fc 영역 및 제2 Fc 영역의 CH3 도메인 내에는 하기의 결합을 추가로 포함하는 것을 특징으로 할 수 있다. The CH3 domains of the first Fc region and the second Fc region as described above may further include the following bonds.
(i) 제1 Fc 영역의 CH3 도메인 내 Y349 위치에 치환된 시스테인 (C); 및(i) cysteine (C) substituted at the Y349 position in the CH3 domain of the first Fc region; And
(ii) 제2 Fc 영역의 CH3 도메인 내 S354 위치에 치환된 시스테인 (C)의 결합.(ii) binding of cysteine (C) substituted at position S354 in the CH3 domain of the second Fc region.
바람직하게는 본 발명에 따른 항체 유래 제1 Fc 영역 및 제2 Fc 영역에 포함된 CH3 도메인은 각각 Preferably, the CH3 domains included in the antibody-derived first and second Fc regions of the present invention are each
(1) 서열번호 1 및 서열번호 2;(1) SEQ ID NO: 1 and SEQ ID NO: 2;
(2) 서열번호 3 및 서열번호 4;(2) SEQ ID NO: 3 and SEQ ID NO: 4;
(3) 서열번호 5 및 서열번호 6; (3) SEQ ID NO: 5 and SEQ ID NO: 6;
(4) 서열번호 8 및 서열번호 9;(4) SEQ ID NO: 8 and SEQ ID NO: 9;
(5) 서열번호 11 및 서열번호 12; 및(5) SEQ ID NO: 11 and SEQ ID NO: 12; And
(6) 서열번호 14 및 서열번호 15; (6) SEQ ID NO: 14 and SEQ ID NO: 15;
의 서열번호로 표시되는 아미노산 서열로 구성된 군에서 선택된 서열을 가지는 것을 특징으로 할 수 있다. It may be characterized by having a sequence selected from the group consisting of the amino acid sequence represented by the sequence number of.
특히, 본 발명에 따른 항체 유래 제1 Fc 영역 및 제2 Fc 영역은 IgG4 유래의 표 1에 기재된 CH3 도메인의 서열을 가지는 것이 바람직하다. In particular, the antibody-derived first Fc region and the second Fc region preferably have a sequence of the CH3 domain described in Table 1 derived from IgG4.
Figure PCTKR2017008676-appb-T000001
Figure PCTKR2017008676-appb-T000001
본 발명에 따른 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, In the heterodimeric Fc-fused protein according to the present invention,
상기 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 생리활성 단백질의 서브유닛(subunit)이 결합되어 있을 수도 있고, The subunit of the bioactive protein may be bound only to one of the N-terminus or the C-terminus of the first Fc region or the second Fc region,
제1 Fc 영역 및 제2 Fc 영역의 N-말단 또는 C-말단 각각에 1 종의(하나의) 생리활성 단백질의 서로 다른 하나 이상의 서브유닛(subunit)이 각각 결합되어 있을 수도 있다(도 2(B) 및 도 2(C) 참조).One or more different subunits of one (one) bioactive protein may be respectively bound to the N-terminus or C-terminus of the first Fc region and the second Fc region (FIG. 2 ( B) and FIG. 2 (C)).
상기 “제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 생리활성 단백질의 서브유닛(subunit)이 결합”되어 있다는 의미는, 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단의 4개 말단 중에서 어느 하나의 말단에만 생리활성 단백질의 서브유닛(들) 중 하나가 결합되어 있고, 상기 생리활성 단백질의 나머지 서브유닛(들)이 링커-매개된 형태로 상기 1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에 결합된 생리활성 단백질의 서브유닛과 결합되어 있다는 의미이다. 상기 링커는 아미노산 링커인 것이 바람직하지만, 이에 한정되는 것은 아니다. The "subunit of the bioactive protein is bonded only to one terminal of either the N-terminus or the C-terminus of the first Fc region or the second Fc region", the first Fc region or the second Fc One of the subunit (s) of the bioactive protein is bound to either end of the N-terminal or C-terminal end of the region, and the remaining subunit (s) of the bioactive protein are linker-mediated. In the form of a bioactive protein bound to either the N-terminus or the C-terminus of the 1 Fc region or the second Fc region. The linker is preferably an amino acid linker, but is not limited thereto.
또한, 상기 “제1 Fc 영역 및 제2 Fc 영역의 N-말단 또는 C-말단 각각에 1 종의(하나의) 생리활성 단백질의 서로 다른 하나 이상의 서브유닛(subunit)이 각각 결합”되었다는 의미는 제1 Fc 영역 및 제2 Fc 영역 N-말단 모두에 생리활성 단백질의 서로 다른 하나 이상의 서브유닛(들)이 각각 결합되거나, 제1 Fc 영역 및 제2 Fc 영역의 C-말단 모두에 생리활성 단백질의 서로 다른 하나 이상의 서브유닛(들)이 각각 결합된 경우, In addition, "the one or more different subunits of one (one) bioactive protein is respectively bonded to each of the N-terminal or C-terminal of the first Fc region and the second Fc region" One or more different subunit (s) of the bioactive protein are bound to both the first Fc region and the second Fc region N-terminus, respectively, or the bioactive protein is bound to both the C-terminus of the first Fc region and the second Fc region. When each of one or more different subunit (s) of are combined,
또는 제1 Fc 영역 및 제2 Fc 영역의 N-말단 및 C-말단 모두에 생리활성 단백질의 서로 다른 하나 이상의 서브유닛이 각각 결합된 형태를 의미한다. Or one or more different subunits of the bioactive protein are respectively bound to both the N-terminus and the C-terminus of the first Fc region and the second Fc region.
본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질에 있어서, 상기 제1 Fc 영역 및/또는 제2 Fc 영역의 말단과 생리활성 단백질의 서브유닛(subunit)의 결합은 유전적 융합(genetic fusion)에 의해 융합된 것을 특징으로 할 수 있으며, In the antibody heavy chain constant region heterodimer-fusion protein according to the present invention, the binding of the terminal of the first Fc region and / or the second Fc region and the subunit of the bioactive protein is genetic fusion. It can be characterized by fused by),
또 다른 양태로 상기 제1 Fc 영역 및 제2 Fc 영역과 생리활성 단백질의 서브유닛(subunit)은 링커 매개된 형태로 결합된 것을 특징으로 할 수 있다. 상기 링커는 아미노산 링커인 것이 바람직하지만, 이에 한정되지는 않는다. In another embodiment, the first Fc region and the second Fc region and a subunit of the bioactive protein may be combined in a linker-mediated form. The linker is preferably an amino acid linker, but is not limited thereto.
또 다른 관점에서, 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, In another aspect, in the antibody heavy chain constant region heterodimeric-fusion protein (heterodimeric Fc-fused protein) according to the present invention,
상기 생리활성 단백질은 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서로 다른 서브유닛(subunit)은 (하나의) 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 것을 특징으로 한다.The bioactive protein is composed of two or more different subunits, and the two or more different subunits are characterized by forming a (one) protein complex to represent the biological activity.
“2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서로 다른 서브유닛(subunit)은 (하나의) 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 것”의 의미는 생리활성 단백질이 2개 이상의 서브유닛들이 단백질 복합체(protein complex)를 형성하는 경우에 목적하는 생리활성을 나타냄을 의미하는 것으로, The meaning of "consists of two or more different subunits, and two or more different subunits (physical) forms a (protein) protein complex to show the biological activity" means When two or more subunits form a protein complex (protein complex) means that the desired biological activity,
상기 생리활성 단백질은 인터루킨 12 (IL-12), 인터루킨 23 (IL-23), 인터루킨 27 (IL-27), 인터루킨 35 (IL-35)및 난포자극호르몬(FSH)로 이루어진 군으로부터 선택되는 것이 바람직하지만, 이에 한정되는 것은 아니며, 이외에도 본 발명의 목적에 부합하는 어떠한 생리활성 단백질도 사용될 수 있음은 통상의 기술자에게는 자명한 것이다. The bioactive protein is selected from the group consisting of interleukin 12 (IL-12), interleukin 23 (IL-23), interleukin 27 (IL-27), interleukin 35 (IL-35), and follicle stimulating hormone (FSH). Preferably, but not limited to this, it will be apparent to those skilled in the art that any physiologically active protein suitable for the purpose of the present invention can be used.
가장 바람직한 본 발명에 따른 생리활성 단백질은 인터루킨 12 (IL-12)이다. The most preferred bioactive protein according to the present invention is interleukin 12 (IL-12).
본 발명에 따른 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서로 다른 서브유닛(subunit)은 1 종의 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 단백질을, 본 발명에 따른 바람직한 생리활성 단백질인 인터루킨-12(IL-12)를 예로 들어 구체적으로 설명한다. Consists of two or more different subunits according to the present invention, two or more different subunits to form a protein complex (protein complex) to exhibit a physiological activity, according to the present invention Interleukin-12 (IL-12), which is a preferred bioactive protein, will be specifically described by way of example.
IL-12는 p35(IL-12A)와 p40(IL-12B)의 2개의 서브유닛으로 이루어져 있으며, 생리활성을 띠는 활성 형태는 상기 p35와 p40의 이종이중체(heterodimer)인 p70의 형태이다. 자연계에서는 IL-12가 활성을 가지기 위해서는 상기 p35와 p40의 이종이중체(heterodimer)인 p70의 형태로 존재하여야 한다. IL-12 consists of two subunits of p35 (IL-12A) and p40 (IL-12B), and its bioactive form is p70, a heterodimer of p35 and p40. . In nature, IL-12 must be present in the form of p70, a heterodimer of p35 and p40 in order to have activity.
본 발명에서는 자연계에서의 IL-12의 존재형태를 최대한 모사하기 위하여, 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질의 형태로 구현하였다. In the present invention, in order to maximally mimic the presence of IL-12 in nature, the antibody heavy chain constant region heterologous double-fusion protein according to the present invention was implemented.
구체적으로, 상기에서 설명한 바와 같이, 본 발명에 따른 제1 Fc 영역 및 제2 Fc 영역을 포함하고, 제1 Fc 영역 및 제2 Fc 영역 말단 중 어느 하나 이상의 말단에 생리활성 단백질의 하나 이상의 서브유닛(subunit)이 결합되어 있는 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, Specifically, as described above, at least one subunit of a bioactive protein comprising a first Fc region and a second Fc region according to the present invention, at one or more of the ends of the first and second Fc regions Heterodimeric Fc-fused protein in the antibody heavy chain constant region to which (subunit) is bound,
(i) 상기 제1 Fc 영역 및 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 하나의 생리활성 단백질을 구성하는 하나 이상의 서브유닛(subunit)(들)이 결합되고, 나머지 서브유닛(들)이 링커로 연결되어 결합되어 있을 수도 있고, (i) at least one subunit (s) constituting one bioactive protein is bound only at either end of the N-terminus or C-terminus of the first Fc region and the second Fc region, and the rest The subunit (s) may be linked by a linker,
(ii) 제1 Fc 영역 말단 및 제2 Fc 영역의 N-말단 및/또는 C-말단 각각에 1 종의 생리활성 단백질의 서로 다른 하나 이상의 서브유닛(subunit)(들)이 각각 결합되어 있을 수도 있다. (ii) one or more different subunit (s) of one bioactive protein may be bound to each of the N-terminus and / or C-terminus of the first Fc region and the second Fc region, respectively. have.
상기와 같은 경우에 있어, IL-12를 예로 들어 설명하면,In the above case, if IL-12 is described as an example,
(i)의 경우에는 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 p35 또는 p40 서브유닛이 결합되어 있고, 나머지 서브 유닛은 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에 결합된 p35 또는 p40의 서브유닛과 링커로 연결되어 이종이중체-융합단백질을 형성하도록 할 수 있다(도 2(B) 및 도 2C) 참조). In the case of (i), the p35 or p40 subunit is attached to only one terminal of either the N-terminus or the C-terminus of the first Fc region or the second Fc region, and the remaining subunits are the first Fc region or the second Fc region. The linker may be linked to a subunit of p35 or p40 that is bound to either the N-terminus or C-terminus of the Fc region to form a heterodimer-fusion protein (FIGS. 2B and 2C). ) Reference).
(ii)의 경우에는 제1 Fc 영역의 N-말단 또는 C-말단에 p35와 p40에서 선택된 어느 하나의 서브유닛이 결합되고, 제2 Fc 영역의 N-말단 또는 C-말단에는 다른 하나의 서브유닛이 결합된 형태를 가지는 이종이중체-융합단백질을 형성할 수 있다(도 2(B) 및 도 2C) 참조). In the case of (ii), any one subunit selected from p35 and p40 is coupled to the N-terminus or C-terminus of the first Fc region, and the other subunit is N-terminus or the C-terminus of the second Fc region. Units can form heterodimer-fusion proteins having a combined form (see FIGS. 2 (B) and 2C).
상기와 같은 형태로 인해, 자연계에 존재하는 본연의 이종이중체 형태를 유지하면서 기존 재조합 인터루킨 12 단백질과 유사한 생체 외 생리활성을 보이는 것을 확인하였다(도 2(B), 도 2(C) 및 도 10(C) 참조).Due to the above form, it was confirmed that the in vitro physiological activity similar to the existing recombinant interleukin 12 protein while maintaining the original heterodimeric form existing in nature (Fig. 2 (B), Fig. 2 (C) and Fig. 10 (C)).
이에 따라 본 발명에 따른 바람직한 항체 중쇄불변부위 이종이중체-융합단백질은 상기 생리활성 단백질이 인터루킨 12 (IL-12)이고, Accordingly, in the preferred antibody heavy chain constant region heterodimer-fusion protein according to the present invention, the physiologically active protein is interleukin 12 (IL-12),
상기 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 인터루킨 12 (IL-12)의 p35 또는 p40 서브유닛이 결합되어 있고, 나머지 서브유닛이 링커-매개된 형태로 상기 1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에 결합된 서브유닛과 결합되어 있거나, 또는The p35 or p40 subunit of interleukin 12 (IL-12) is bound to only one of the N-terminus or C-terminus of the first Fc region or the second Fc region, and the remaining subunits are linker-mediated. Form a subunit bound to either the N-terminus or C-terminus of the 1 Fc region or the second Fc region, or
제1 Fc 영역 및 제2 Fc 영역의 N-말단 또는 C-말단 각각에 인터루킨 12 (IL-12)의 p35 및 p40 서브유닛이 각각 결합되어 있는 것을 특징으로 한다. P35 and p40 subunits of interleukin 12 (IL-12) are respectively bound to the N-terminus or C-terminus of the first Fc region and the second Fc region.
또 다른 관점에서, 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, In another aspect, in the antibody heavy chain constant region heterodimeric-fusion protein (heterodimeric Fc-fused protein) according to the present invention,
상기 제1 Fc 영역 및 제2 Fc 영역 내 N-말단에 포함된 힌지(hinge) 영역은, 힌지 영역 내에 포함된 시스테인 잔기가 변이된 것을 특징으로 할 수 있다. The hinge region included in the N-terminus in the first Fc region and the second Fc region may be characterized by mutation of a cysteine residue included in the hinge region.
바람직하게는 상기 힌지 영역에서의 시스테인 잔기의 변이는 이종이중체(heterodimer) 형성을 위한 중심 힌지 영역(core hinge region) 내의 시스테인 잔기를 제외한 나머지 상부 힌지 영역 (upper hinge region)의 시스테인(cysteine) 잔기가 모두 세린(serine) 잔기로 치환된 것을 특징으로 할 수 있지만, 이에 한정되는 것은 아니다.Preferably the mutation of the cysteine residue in the hinge region is a cysteine residue of the upper hinge region except for the cysteine residue in the core hinge region for heterodimer formation. Are all substituted with a serine residue, but are not limited thereto.
또한, 본 발명에 있어서, 상기 제1 Fc 영역 및 제2 Fc 영역은 인간 IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD 및 IgE로 이루어진 완전한 항체(whole antibody) 형태에 포함되어 있는 것을 특징으로 할 수 있다. In addition, in the present invention, the first Fc region and the second Fc region are included in a whole antibody (whole antibody) form consisting of human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD and IgE. can do.
본 발명에 있어서, “완전한 항체(whole antibody) 형태”는 IgG, IgA 및 IgD의 경우에는 Fc 영역 내의 CH2 도메인, CH3 도메인 및 힌지 영역(IgE의 경우 CH4 도메인도 포함됨)에, 추가적으로 CH1 도메인, VH 도메인, CL 도메인 및 VL 도메인을 포함하는 온전한 형태의 항체를 의미한다. In the present invention, the "whole antibody form" means the CH2 domain, CH3 domain and hinge region (including CH4 domain in IgE) in the Fc region in the case of IgG, IgA and IgD, in addition to the CH1 domain, VH. By intact form is meant an antibody comprising a domain, a CL domain and a VL domain.
또 다른 관점에서, 본 발명은 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 약제학적 조성물에 대한 것이다. 본 발명에 따른 약학 조성물의 용도는 항체 중쇄불변부위 이종이중체-융합단백질 내에 포함된 생리활성 단백질의 용도에 따라 좌우되는 것을 특징으로 한다. In another aspect, the present invention relates to a pharmaceutical composition comprising the antibody heavy chain constant region heterodimer-fusion protein according to the present invention. The use of the pharmaceutical composition according to the invention is characterized in that it depends on the use of the bioactive protein contained in the antibody heavy chain constant heterodimer-fusion protein.
바람직하게는 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질 내에 포함된 생리활성 단백질은 IL-12 또는 이의 하나 이상의 서브유닛이며, 이에 따라 본 발명은 생리활성 단백질로 IL-12를 포함하는 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 암 치료를 위한 약제학적 조성물을 제공한다. Preferably, the bioactive protein contained in the antibody heavy chain constant region heterodimer-fusion protein according to the present invention is IL-12 or one or more subunits thereof, and thus the present invention includes IL-12 as a bioactive protein. It provides a pharmaceutical composition for the treatment of cancer comprising an antibody heavy chain constant region heterodimer-fusion protein.
상기 생리활성 단백질로 IL-12 또는 이의 하나 이상의 서브유닛을 포함하는 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 암 치료를 위한 약제학적 조성물로 치료가능한 암은 대장암, 흑색종, 유방암, 췌장암, 신장암, 전립선암, 난소암, 소장암, 식도암, 자궁경부암, 폐암, 림프종 및 혈액암으로 이루어진 군에서 선택될 수 있지만, 이에 한정되는 것은 아니다. Cancers treatable with a pharmaceutical composition for treating cancer comprising an antibody heavy chain constant region heterodimer-fusion protein comprising IL-12 or one or more subunits thereof as the bioactive protein include colorectal cancer, melanoma, breast cancer, Pancreatic cancer, kidney cancer, prostate cancer, ovarian cancer, small intestine cancer, esophageal cancer, cervical cancer, lung cancer, lymphoma and hematologic cancer may be selected from the group consisting of, but not limited to.
본 발명에 따른 약제학적 조성물에는 추가적으로 제약상 허용되는 담체가 포함될 수 있다. "제약상(약학적으로) 허용되는 담체"는 제제를 제제화하거나 또는 안정화시키는 것을 돕기 위해서 활성 성분에 추가될 수 있는 물질이고, 환자에게 유의한 해로운 독성 효과를 야기하지 않는다. Pharmaceutical compositions according to the invention may additionally include a pharmaceutically acceptable carrier. A “pharmaceutically acceptable carrier” is a substance that can be added to the active ingredient to help formulate or stabilize a formulation and does not cause significant deleterious toxic effects on the patient.
상기 담체는 환자를 자극하지 않고 본 발명에 따른 이종이중체-융합단백질의 생물학적 활성 및 특성을 저해하지 않는 담체 또는 희석제를 말한다. 액상 용액으로 제제화되는 조성물에 있어서 허용되는 약학적 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한, 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. 다른 담체는 예를 들어 문헌[Remington's Pharmaceutical Sciences (E. W. Martin)]에 기재되어 있다. The carrier refers to a carrier or diluent that does not irritate the patient and does not inhibit the biological activity and properties of the heterodimer-fusion protein according to the present invention. Acceptable pharmaceutical carriers in compositions formulated as liquid solutions are sterile and physiologically compatible, including saline, sterile water, Ringer's solution, buffered saline, albumin injectable solutions, dextrose solution, maltodextrin solution, glycerol, ethanol and One or more of these components may be mixed and used, and other conventional additives such as antioxidants, buffers and bacteriostatic agents may be added as necessary. In addition, diluents, dispersants, surfactants, binders and lubricants may be additionally added to formulate injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like. Other carriers are described, for example, in Remington's Pharmaceutical Sciences (E. W. Martin).
제약상 허용되는 담체는 멸균 주사가능한 용액제 또는 분산액제를 즉각 투여용(extemporaneous)으로 제조하기 위한 멸균 수용액 또는 분산액 및 멸균 분말을 포함한다. 제약 활성 물질을 위한 이러한 매질 및 작용제의 사용은 당업계에 공지되어 있다. 조성물은 바람직하게는 비경구 주사용으로 제제화된다. 조성물은 용액제, 마이크로에멀젼제, 리포좀제, 또는 높은 약물 농도에 적합한 기타 주문된 구조물로서 제제화될 수 있다. 담체는 예를 들어 물, 에탄올, 폴리올(예를 들어, 글리세롤, 프로필렌 글리콜 및 액체 폴리에틸렌 글리콜 등) 및 이것들의 적합한 혼합물을 함유하는 용매 또는 분산 매질일 수 있다. 일부 경우에, 조성물 중에 등장화제, 예를 들어 당, 폴리알콜, 예컨대 만니톨, 소르비톨 또는 염화나트륨을 포함시킬 수 있다. 멸균 주사가능한 용액제는 필요한 양의 이종이중체-융합단백질을 필요에 따라 상기 기재된 성분들 중 1 종 또는 이것들의 조합물과 함께 적절한 용매 중에 혼입시킨 후에 멸균 마이크로여과를 수행하여 제조될 수 있다. 일반적으로, 분산액제는 활성 화합물을 기본적인 분산 매질 및 상기 기재된 것들로부터의 기타 필요한 성분을 함유하는 멸균 비히클로 혼입시켜 제조된다. 멸균 주사가능한 용액제를 제조하기 위한 멸균 분말의 경우, 일부 제조 방법은 활성 성분 및 임의의 추가의 원하는 성분의 분말을 이것의 미리 멸균-여과시킨 용액으로부터 생성하는 진공 건조 및 냉동-건조(동결건조)이다.Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the preparation of sterile injectable solutions or dispersions for immediate administration. The use of such media and agents for pharmaceutically active substances is known in the art. The composition is preferably formulated for parenteral injection. The compositions may be formulated as solutions, microemulsions, liposomes, or other ordered structures suitable for high drug concentrations. The carrier can be, for example, a solvent or dispersion medium containing water, ethanol, polyols (eg glycerol, propylene glycol and liquid polyethylene glycols, etc.) and suitable mixtures thereof. In some cases, it is possible to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol or sodium chloride in the composition. Sterile injectable solutions can be prepared by incorporating the required amount of the heterodimer-fusion protein in an appropriate solvent with one or a combination of ingredients described above as required, followed by sterile microfiltration. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those described above. In the case of sterile powders for the preparation of sterile injectable solutions, some methods of preparation include vacuum drying and freeze-drying (freeze-drying), which produce a powder of the active ingredient and any further desired ingredients from its presterilized-filtered solution. )to be.
또한, 본 발명에 따른 약제학적 조성물은 고통받는 환자의 중증도에 따라 달라질 수 있는 투여량 및 빈도로 경구 또는 비경구 투여될 수 있다. 조성물은 필요에 따라 볼루스로서 또는 연속 주입에 의해 환자에게 투여될 수 있다. 또 다른 형태로, 본 발명에 따른 약제학적 조성물은 직장으로, 정맥으로(intravenously), 피하로(subcutaneously), 자궁내로(intrauterinely) 또는 뇌혈관내로(intracerebrovascularly) 투여될 수 있지만 이에 한정되는 것은 아니다. In addition, the pharmaceutical compositions according to the invention may be administered orally or parenterally at dosages and frequencies that may vary depending on the severity of the suffering patient. The composition may be administered to the patient as a bolus or by continuous infusion as needed. In another form, the pharmaceutical compositions according to the invention may be administered rectally, intravenously, subcutaneously, intrauterinely or intratracerebrovascularly, but are not limited thereto.
또한, 상기 IL-12를 포함하는 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 암 치료를 위한 약제학적 조성물은 다른 항암제와 병용 치료를 위한 용도로 사용될 수 있으며, 상기 다른 항암제는 세포독성 T 세포 및/또는 자연살해(NK: Natural Killer) 세포인 것이 바람직하지만, 이에 한정되는 것은 아니며, 다른 당해 기술분야에서 사용될 수 있는 모든 항암제가 병용 치료를 위해 사용될 수 있다. In addition, the pharmaceutical composition for treating cancer comprising the antibody heavy chain constant region heterodimer-fusion protein comprising IL-12 may be used for combination therapy with other anticancer agents, and the other anticancer agents may be cytotoxic T. Cells and / or Natural Killer (NK) cells are preferred, but are not limited to any of the other anticancer agents that can be used in the art and can be used for combination therapy.
특히 인터루킨 12을 포함하는 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 암 치료를 위한 약제학적 조성물을 세포독성 T 세포 및/또는 자연살해(NK: Natural Killer) 세포와 병용 치료를 위해 사용하는 경우, In particular, a pharmaceutical composition for treating cancer comprising an antibody heavy chain constant region heterodimer-fusion protein comprising interleukin 12 is used for combination treatment with cytotoxic T cells and / or Natural Killer (NK) cells. Occation,
(1) T세포 또는 자연살해(NK) 세포를 자극하여 사이토카인 분비의 증가;(1) stimulating T cells or natural killer (NK) cells to increase cytokine secretion;
(2) antibody-dependent cell-mediated cytotoxicity (ADCC) 또는 세포독성 T 림프구 (CTL)의 반응의 증가; (2) increased response of antibody-dependent cell-mediated cytotoxicity (ADCC) or cytotoxic T lymphocytes (CTL);
(3) 세포독성 T 림프구 (CTL) 및/또는 자연살해 세포 수의 증가; (3) an increase in the number of cytotoxic T lymphocytes (CTLs) and / or killer cells;
(4) 종양부근으로의 림프구 유입의 증가; 또는 (4) increased lymphocyte influx into the tumor area; or
(5) 생체 내에서 림프구의 IL-12R beta1과 IL-12R beta2 시그날의 증가;(5) increase of IL-12R beta1 and IL-12R beta2 signals in lymphocytes in vivo;
를 유도하는 것을 특징으로 할 수 있다. It may be characterized by deriving.
또 다른 관점에서, 본 발명은 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 약제학적 조성물을 치료를 요하는 환자에게 투여하는 단계를 포함하는 질병의 치료 또는 예방 방법을 제공한다. In another aspect, the present invention provides a method of treating or preventing a disease comprising administering to a patient in need thereof a pharmaceutical composition comprising an antibody heavy chain constant region heterodimer-fusion protein according to the present invention. .
상기 조성물의 경우와 마찬가지로, 치료 또는 예방 가능한 질병은 항체 중쇄불변부위 이종이중체-융합단백질 내에 포함된 생리활성 단백질의 용도에 따라 좌우되며, As with the composition, the treatable or preventable disease depends on the use of the bioactive protein contained in the antibody heavy chain constant heterodimer-fusion protein,
바람직하게는 본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질 내에 포함된 생리활성 단백질의 하나 이상의 서브유닛이 IL-12의 하나 이상의 서브유닛일 경우, 암 환자의 치료, 특히 대장암, 흑생종, 유방암, 췌장암, 신장암, 전립선암, 난소암, 소장암, 식도암, 자궁경부암, 폐암, 림프종 및 혈액암으로 이루어진 군에서 선택되는 암을 앓고 있는 환자의 치료 또는 예방 방법을 제공한다. Preferably, when at least one subunit of the physiologically active protein contained in the antibody heavy chain constant heterodimer-fusion protein according to the present invention is at least one subunit of IL-12, treatment of cancer patients, in particular colon cancer, melanoma The present invention provides a method of treating or preventing a patient suffering from cancer selected from the group consisting of breast cancer, pancreatic cancer, kidney cancer, prostate cancer, ovarian cancer, small intestine cancer, esophageal cancer, cervical cancer, lung cancer, lymphoma and hematologic cancer.
이하, 실시예를 통하여 본 발명을 더욱 상세히 설명하고자 한다. 이들 실시예는 오로지 본 발명을 예시하기 위한 것으로서, 본 발명의 범위가 이들 실시예에 의해 제한되는 것으로 해석되지 않는 것은 통상의 기술자에게는 자명한 것이다.Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are only for illustrating the present invention, and it is obvious to those skilled in the art that the scope of the present invention is not interpreted to be limited by these examples.
실시예Example
실시예 1: 인간 항체 동종형별 이종이중체 형성을 위한 항체 중쇄불변부위 CH3 도메인 변이체의 고안 (서열 분석)Example 1: Design of antibody heavy chain constant region CH3 domain variants for heterodimer formation by human antibody isotype (sequence analysis)
이종이중체 형성이 선호되는 CH3 도메인 돌연변이가 도입된 각 인간 항체 동종형(isotype)별 중쇄불변부위 이종이중체의 단편을 만들기 위해, 아래에 서술한대로 먼저 이중체 형성을 위한 상호작용에 주요하게 작용하는 CH3 도메인간의 아미노산 서열의 유사 정도를 분석하였다. 이 때, 이종의 CH3A:CH3B(본 발명에 있어, CH3A 및 CH3B는 각각 제1 Fc 영역의 CH3 영역 및 제2 Fc 영역의 CH3 영역을 의미한다)에 유도되는 돌연변이 쌍(A107)은 기존의 문헌 또는 특허에 공개된 중쇄불변부위 이종이중체 형성을 증진시키기 위한 전략으로 CH3A:CH3B 형성이 고수율로 형성되도록 한다(Choi et al. 2016; 대한민국 특허출원 제2015-0142181호). 도 3은 각 인간 항체 면역글로불린 G (IgG) 동종형별 CH3 도메인의 서열을 나열하여 비교한 것이다. 각 아미노산 서열은 International ImMunoGeneTics information system (IMGT ; URL: http://www.imgt.org/) 에서 확인하였다. 특히 IgG3의 경우 여러 다양한 Gm 동종이인자형(Gm allotype)중에서도 혈청 내 반감기가 다른 IgG 동종형과 유사한 정도로 유지되는 것으로 보고된 G3m(s,t)의 서열을 사용하였다(Stapleton NM et al., 2011). In order to make fragments of heavy chain constant region heterodimers for each human antibody isotype introduced with CH3 domain mutations in which heterodimer formation is preferred, it first plays a major role in the interaction for duplex formation as described below. The similarity of amino acid sequences between the CH3 domains was analyzed. In this case, mutation pairs (A107) induced in heterologous CH3A: CH3B (in the present invention, CH3A and CH3B mean CH3 region of the first Fc region and CH3 region of the second Fc region, respectively) Or to promote the formation of heavy chain constant region heterodimers disclosed in the patent to form a high yield of CH3A: CH3B (Choi et al. 2016; Korean Patent Application No. 2015-0142181). Figure 3 is a comparison of the sequence of each human antibody immunoglobulin G (IgG) isotype CH3 domains listed. Each amino acid sequence was identified in the International ImMunoGeneTics information system (IMGT; URL: http://www.imgt.org/). In particular, for IgG3, the sequence of G3m (s, t), which has been reported to maintain serum half-life similar to other IgG isotypes among various Gm allotypes, was used (Stapleton NM et al., 2011). ).
서열 분석 결과, A107 돌연변이가 도입되는 위치 중에서도 409번째 아미노산 서열이 IgG1, IgG2, IgG3와는 다르게 IgG4에서 아르기닌으로 다른 서열을 가지는 것 이외에는 다른 돌연변이의 위치에는 모든 동종형에서 보존되어 있는 서열을 가지는 것을 확인하였다. 이에 동일한 아미노산 서열 번호를 가지는 위치를 A107 돌연변이 쌍을 IgG1이 아닌 다른 동종형에 이식하기 위한 위치로 선정하였다. 본 발명에서의 모든 아미노산 위치 표기는 EU index (numbering)을 따른다. As a result of the sequence analysis, among the positions where the A107 mutation was introduced, the 409th amino acid sequence, unlike IgG1, IgG2, and IgG3, had a sequence conserved in all homotypes at the positions of the other mutations except that it had a different sequence from IgG4 to arginine. It was. Thus, the position having the same amino acid sequence number was selected as a position for transplanting the A107 mutant pair into an isoform other than IgG1. All amino acid position notations in the present invention follow the EU index (numbering).
실시예 2: 인간 항체 동종형별 이종이중체 형성을 위한 항체 중쇄불변부위 CH3 도메인 변이체의 고안 (구조 모델링)Example 2: Design of antibody heavy chain constant region CH3 domain variants for heterodimer formation by human antibody isotype (structural modeling)
실제 동종형별 CH3 도메인 변이체를 구축하기에 앞서 실시예 1에서 선정한 위치에 A107 돌연변이쌍이 도입되었을 때, 구조적으로도 돌연변이가 안정적으로 도입되어 이종이중체를 형성할 수 있을 지의 여부를 도 3에 각 돌연변이가 도입된 변이체의 서열을 이용, 구조 모델링을 통해 예측하였다. 구조 모델링은 이미 밝혀진 Immunoglobulin Fc heterodimer variant의 구조(PDB ID : 4X98)를 견본(template)으로 하여 온라인 모델링 서버 (URL : https://swissmodel.expasy.org/; Biasini M et al., 2014)를 통해 예측하였다. 얻어진 각각의 구조는 돌연변이가 도입된 이후 CH3 도메인의 구조적 변화 및 A107 돌연변이의 위치 등을 관찰하기 위하여 단백질의 구조를 볼 수 있는 Pymol 소프트웨어를 이용하여 중첩시켰다. 중첩 구조 상에서 기존 IgG1 동종형을 기반으로 하여 구축된 고수율로 CH3A:CH3B 중쇄불변부위 이종이중체를 형성하는 돌연변이체 A107의 모델링 구조와 비교하였을 때, 각 동종형에 A107 돌연변이가 도입되었을 때에도, 크게 구조가 달라지지 않고 유지되는 것을 확인하였고, 특히 이식된 A107 돌연변이 아미노산 잔기의 방향이 거의 일치하고 돌연변이 아미노산 간의 상호작용을 위한 거리 또한 비슷한 정도로 유지되고 있음을 확인하였다 (도 4 참조). When the A107 mutant pair was introduced at the position selected in Example 1 prior to constructing the actual homologous CH3 domain variants, each mutant shown in FIG. 3 shows whether the mutation can be stably introduced to form a heterodimer. Using the sequence of the introduced variant was predicted through structural modeling. Structural modeling uses an online modeling server (URL: https://swissmodel.expasy.org/; Biasini M et al., 2014) as a template of the structure of an already known Immunoglobulin Fc heterodimer variant (PDB ID: 4X98). Predicted through. Each of the structures obtained was superimposed using Pymol software to see the structure of the protein to observe the structural changes in the CH3 domain and the location of the A107 mutation after the mutations were introduced. When compared to the modeling structure of mutant A107, which forms a CH3A: CH3B heavy chain constant heterodimer with a high yield constructed on the basis of existing IgG1 isoforms on overlapping structures, even when A107 mutations were introduced into each isotype, It was confirmed that the structure remains largely unchanged, and in particular, the orientation of the transplanted A107 mutant amino acid residues is almost identical and the distance for interaction between the mutant amino acids is also maintained to a similar degree (see FIG. 4).
실시예 3: 인간 항체 동종형별 A107 중쇄불변부위 이종이중체 변이체 (A107 heterodimeric Fc isotype variants)의 구축Example 3: Construction of A107 heterodimeric Fc isotype variants by human antibody isotypes
실시예 1의 서열 분석과 실시예 2의 구조 분석을 통해 설계된 각 동종형별 A107 중쇄불변부위 이종이중체 돌연변이체는 합성 올리고뉴클레오티드 (Macrogen, Korea)를 이용해 통상의 기술자에 의해 행해지는 부위 지정 돌연변이 (Site-directed mutagenesis) 방법으로 동물세포 발현 벡터 pcDNA3.1(+) (Invitrogen, USA)에 signal sequence-hinge-CH2-CH3의 순서를 가지도록 인 프레임 (in-frame)으로 NotI/HindIII 제한효소를 이용하여 클로닝 하였다 (도 5 참조). The heterologous mutant A107 heavy chain constant region of each isotype designed through the sequence analysis of Example 1 and the structural analysis of Example 2 is a site-directed mutation performed by a person skilled in the art using synthetic oligonucleotides (Macrogen, Korea) Site-directed mutagenesis) was performed using the NotI / HindIII restriction enzyme in-frame to sequence the signal sequence-hinge-CH2-CH3 to the animal cell expression vector pcDNA3.1 (+) (Invitrogen, USA). Cloning was carried out (see FIG. 5).
이 때 사용된 힌지 영역은 이중체 형성을 위한 중심 힌지 영역 (core hinge region)내의 시스테인 잔기를 제외한 나머지 상부 힌지 영역 (upper hinge region)의 시스테인(cysteine) 잔기는 단백질 융합시, 원하지 않는 이황화 결합이 생기는 것을 막기 위해 세린(serine) 잔기로 치환하였다. 특히, IgG3의 경우 G3m(s,t) 동종이인자형이 가지는 힌지 영역 47개 아미노산 서열 중 중심 힌지 영역의 C 말단 15개 아미노산만으로도 IgG3가 가지는 높은 항체 고유의 효과 기능 (ADCC, CDC)이 유지되는 것을 문헌을 통해 확인하였고(Dall'Acqua WF et al., 2006), 이에 도 5에 나타낸 서열의 C 말단 15개 아미노산만을 사용하였다.The hinge region used here is a cysteine residue of the upper hinge region except for the cysteine residue in the core hinge region for duplex formation. Substituted with serine residues to prevent formation. In particular, in the case of IgG3, only 15 amino acids in the C-terminal region of the central hinge region among the 47 amino acid sequences of the G3m (s, t) allotypes maintain high antibody-specific effect functions (ADCC, CDC) of IgG3. This was confirmed through the literature (Dall'Acqua WF et al., 2006), whereby only 15 amino acids of the C terminus of the sequence shown in FIG. 5 were used.
표 2는 본 발명의 야생형 및 A107 중쇄불변부위 이종이중체 변이체쌍 내의 CH3 영역의 아미노산 서열 정보를 나타낸 것이다. Table 2 shows amino acid sequence information of the CH3 region in the wild type and A107 heavy chain constant heterodimeric variant pairs of the present invention.
Figure PCTKR2017008676-appb-T000002
Figure PCTKR2017008676-appb-T000002
Figure PCTKR2017008676-appb-I000001
Figure PCTKR2017008676-appb-I000001
실시예 4: 인간 항체 동종형별 A107 이종이중체 중쇄불변부위 변이체의 이종이중체 형성능 평가 방법Example 4 Method for Evaluating Heterodimer Formation Ability of A107 Heterodimeric Heavy Chain Constant Region Variants by Human Antigen Isotypes
상기 실시예 3에서 제작한 각 동종형별 A107 중쇄불변부위 이종이중체 돌연변이체가 실제로 야생형 A107 돌연변이체와 유사한 정도의 이종이중체 형성능을 가지는 지 확인하기 위하여 동종의 연구에서 중쇄불변부위 이종이중체 형성능 평가에 주요하게 사용되는 scFv-FcCH3A/FcCH3B 동시 발현 시스템을 사용하고자 하였다 (Choi et al., 2013). 도 6은 scFv-FcCH3A/FcCH3B 동시 발현 시스템을 나타낸 모식도이다. scFv-FcCH3A/FcCH3B 동시 발현 시스템에서 정제된 항체는 scFv-FcCH3A 동종이중체 (103 kDa)와 scFv-FcCH3A/FcCH3B 이종이중체 (78 kDa), FcCH3B 동종이중체 (53 kDa)의 분자량이 각각 다르게 나타나기 때문에, SDS-PAGE 상에서 이종이중체의 형성 정도를 비교할 수 있다. Evaluation of the heterodimer formation ability of the heavy chain constant region in the homogeneous study to confirm whether the A107 heavy chain constant region heterodimer mutant for each isotype prepared in Example 3 actually has a similar level of heterodimer formation ability as the wild type A107 mutant. We intended to use the scFv-Fc CH3A / Fc CH3B co-expression system, which is mainly used for (Choi et al., 2013). 6 is a schematic diagram showing a scFv-Fc CH3A / Fc CH3B co-expression system. Purified antibodies in the scFv-Fc CH3A / Fc CH3B co-expression system were scFv-Fc CH3A homodimer (103 kDa), scFv-Fc CH3A / Fc CH3B heterodimer (78 kDa), Fc CH3B homodimer (53 kDa) Since the molecular weights of) are different, it is possible to compare the degree of heterodimer formation on SDS-PAGE.
상기 FcCH3B 벡터는 실시예 3에서 구축된 벡터를 사용하였고, 추가적으로 FcCH3A의 N 말단에만 scFv를 도입하는 즉, pcDNA3.1(+)-scFv-hinge-CH2-CH3A (scFv-FcCH3A)의 포멧으로 발현되는 벡터를 클로닝하였다. 도 7은 상기 scFv-FcCH3A/FcCH3B 동시 발현 시스템에서 사용한 동물세포 발현벡터인 pcDNA3.1(+)-scFv-hinge-CH2-CH3A (scFv-FcCH3A) 벡터의 모식도를 나타낸 것이다. 사용한 scFv 항체는 DR4에 특이적으로 결합하는 인간화 항체 hAY4의 친화도가 향상된 버전인 hAY4a의 VH와 VL 부위를 연결한 항체이다(Lee, Park et al. 2010). NotI 제한 효소와 힌지 영역 바로 앞에 위치한 BsiWI 제한효소를 이용하여 클로닝 하였다. 변이체에 대한 대조군으로 야생형 Fc를 동일한 포멧 (scFv-Fc/Fc)으로 구축하였다. As the Fc CH3B vector, a vector constructed in Example 3 was used, and additionally, scFv was introduced only to the N terminus of Fc CH3A , that is, of pcDNA3.1 (+)-scFv-hinge-CH2-CH3A (scFv-Fc CH3A ). Vectors expressed in the format were cloned. Figure 7 shows a schematic diagram of the animal cell expression vector pcDNA3.1 (+)-scFv-hinge-CH2-CH3A (scFv-Fc CH3A ) vector used in the scFv-Fc CH3A / Fc CH3B co-expression system. The scFv antibody used is an antibody connecting the VH and VL regions of hAY4a, which is an enhanced version of the humanized antibody hAY4 that specifically binds DR4 (Lee, Park et al. 2010). Cloning was performed using NotI restriction enzyme and BsiWI restriction enzyme located immediately before the hinge region. Wild type Fc was constructed in the same format (scFv-Fc / Fc) as a control for the variants.
실시예 5: 인간 항체 동종형별 A107 이종이중체 중쇄불변부위 변이체를 포함한 항체의 발현 및 정제Example 5 Expression and Purification of Antibodies Containing A107 Heterodimeric Heavy Chain Constant Region Variants by Human Antibody Homotype
상기 구축된 scFv-FcCH3A 및 FcCH3B의 동시 발현은 각각의 발현 벡터(1:1 비율)와 폴리에틸렌이민 (Polyethylenimine, PEI) (Polyscience)의 혼합물을 HEK293-F(Invitrogen) 세포에 일시적 트랜스펙션(transient transfection)하여 무혈청 FreeStyle 293 발현 배지(Invitrogen)가 들어있는 진탕 플라스크에서 배양함으로써 이루어진다. 자세한 방법은 다음과 같다. ScFv -Fc CH3A constructed above Simultaneous expression of Fc CH3B was performed by transient transfection of HEK293-F (Invitrogen) cells with a mixture of respective expression vectors (1: 1 ratio) and polyethyleneimine (Polyethylenimine, PEI) (Polyscience). By culturing in a shake flask containing FreeStyle 293 expression medium (Invitrogen). The detailed method is as follows.
진탕 플라스크(Corning)에 200 mL 트랜스펙션 시, HEK293-F 세포를 2.0×106 세포/ml의 밀도로 배지 100 ml에 파종하여, 150 rpm, 8 % CO2에서 배양하였다. 각각의 인간화 항체를 생산하기 위해 그에 따른 중쇄 및 경쇄 플라스미드를 10 ml FreeStyle 293 발현 배지 (Invitrogen)에 중쇄 125μg, 경쇄 125μg 총 250μg (2.5μg/ml)으로 희석하여, PEI 750μg을 희석한 10 ml의 배지 (7.5 μg/ml)와 혼합하여 실온에서 10분 동안 반응시켰다. 그 후, 반응시킨 혼합배지를 앞서 100 ml로 파종한 세포에 넣어 4시간 동안 150 rpm, 8% CO2에서 배양 후, 나머지 100 ml의 FreeStyle 293 발현 배지를 추가하여 짧게는 5일에서 길게는 7일동안 배양하게 되면, 세포가 생산한 단백질 즉, 중쇄불변부위 변이체를 포함한 항체는 세포에 의해 세포 바깥으로 분비되어 배지에 쌓이게 된다. 때문에 단백질은 세포 배양 후 2500 rpm에서 20 분간 원심분리하여 채취한 세포 배양 상등액으로부터 단백질 A 세파로오스 컬럼 (protein A Sepharose column, GE healthcare)을 이용하여 정제하였다. 이 때, 정제방법은 단백질 A 컬럼 회사에서 제공하는 표준 프로토콜을 참조하였으며, 정제된 단백질은 BCA protein assay kit (Thermo)내 용액을 이용하여 562 nm 파장에서 흡광도를 측정하고 그려진 표준 곡선에 따라 그 양을 정량하였다. Upon 200 mL transfection in a shake flask, HEK293-F cells were seeded in 100 ml of medium at a density of 2.0 × 10 6 cells / ml and incubated at 150 rpm, 8% CO 2 . To produce the respective humanized antibodies, the resulting heavy and light chain plasmids were diluted in 10 ml FreeStyle 293 expression medium (Invitrogen) with 125 μg of heavy chain and 125 μg of light chain in total, 250 μg (2.5 μg / ml), with 10 ml of dilute 750 μg of PEI. The mixture was mixed with the medium (7.5 μg / ml) and reacted for 10 minutes at room temperature. Thereafter, the reacted mixed medium was put in the cells seeded with 100 ml in advance, incubated at 150 rpm and 8% CO 2 for 4 hours, and then the remaining 100 ml of FreeStyle 293 expression medium was added. After one day of incubation, the proteins produced by the cells, ie antibodies containing heavy chain constant region variants, are secreted out of the cells by the cells and accumulated in the medium. Therefore, the protein was purified using a protein A Sepharose column (GE healthcare) from the cell culture supernatant collected by centrifugation at 2500 rpm for 20 minutes after cell culture. At this time, the purification method referred to the standard protocol provided by the Protein A column company, and the purified protein was measured by absorbance at a wavelength of 562 nm using a solution in a BCA protein assay kit (Thermo), and the amount was determined according to the drawn standard curve. Was quantified.
실시예 6: 인간 항체 동종형별 A107 이종이중체 중쇄불변부위 변이체의 이종이중체 형성능 평가 Example 6 Evaluation of Heterodimer Formation Ability of A107 Heterodimeric Heavy Chain Constant Region Variants by Human Antigen Isotypes
상기 실시예 5에서 정제된 동종형별 A107 중쇄불변부위 이종이중체 변이체를 포함한 항체 5 μg을 12% 비환원성 조건으로 SDS-PAGE 상에서 분석하였다(도 8). CH3A 변이체의 동종이중체는 103 kD, CH3B 변이체의 동종이중체는 53kD, CH3B 변이체의 단량체는 25 kD에서 관찰되었으며, CH3A 변이체와 CH3B 변이체의 이종이중체는 78 kD에서 관찰되었다. 더 정교한 동종이중체의 정도를 파악하기 위해 웨스턴 블랏(Westerm blot)을 함께 수행하였다. 웨스턴 블랏은 SDS-PAGE 분석보다 적은 양인 0.1 μg의 단백질을 12% 비환원성 조건으로 분리한 이후, 통상의 기술자들이 하는 방법을 이용해 anti-human IgG-AP conjugated antibody (Sigma)을 처리하여 수행하였다(도 9). 5 μg of the antibody including the heterologous heterologous heterologous heterologous variant A107 heavy chain in Example 5 was analyzed on SDS-PAGE under 12% non-reducing conditions (FIG. 8). The homodimer of the CH3A variant was 103 kD, the homodimer of the CH3B variant was 53 kD, the monomer of the CH3B variant was 25 kD, and the heterodimer of the CH3A and CH3B variants was 78 kD. Western blots were performed together to determine the extent of more sophisticated homodimers. Western blot was performed by separating anti-human IgG-AP conjugated antibody (Sigma) using a method performed by those skilled in the art after separating 0.1 μg of protein, which is less than SDS-PAGE analysis, in 12% non-reducing conditions ( 9).
도 8 및 도 9에서 대조군인 야생형 CH3 도메인이 도입된 IgG1 이종이중체는 SDS-PAGE상에서 CH3A/CH3B 각각의 동종이중체 및 CH3A:CH3B 이종이중체가 모두 관찰된 반면, IgG1를 제외한 IgG2, IgG3, IgG4에 A107 이종이중체 형성 돌연변이를 도입한 인간 항체 동종형별 A107 중쇄불변부위 이종이중체 변이체는 모두 기존에 보고된 IgG1 기반 A107 돌연변이체와 유사하거나 더 높은 수율로 이종이중체를 형성하는 것을 확인하였다. 이 때, IgG4 변이체의 경우, CH3A 혹은 CH3B가 포함된 Fc 모노머 (Half Fc)도 함께 관찰되었는데, 이는 자연적으로 일어나는 IgG4의 특징 중의 하나로, 혈액 내에서 Fab-arm exchange가 일어나기에 앞서 힌지 영역 (특히, 중심 힌지 영역 내 228번째 세린)을 중심으로 Half Fc를 형성하는 특징에 기인한 결과이다 (Liu H et al., 2012). In FIG. 8 and FIG. 9, the IgG1 heterodimer into which the wild-type CH3 domain was introduced as a control group showed all homodimers of CH3A / CH3B and CH3A: CH3B heterodimers on SDS-PAGE, whereas IgG2, IgG3, Human antibody isotype A107 heavy chain constant region heterodimer variants incorporating A107 heterodimerization mutations into IgG4 were found to form heterodimers with similar or higher yields than previously reported IgG1-based A107 mutants. . At this time, in the case of the IgG4 variant, the Fc monomer (Half Fc) containing CH3A or CH3B was also observed, which is one of the characteristics of naturally occurring IgG4, and the hinge region (particularly, before the Fab-arm exchange in the blood) occurs. , 228 th serine in the central hinge region). This is due to the characteristic of forming half Fc (Liu H et al., 2012).
실시예 7: 인간/마우스 IL-12 fusion protein 구축 Example 7: Construction of human / mouse IL-12 fusion protein
이종이중체 형성능이 기존에 보고된 IgG1 기반 A107 중쇄불변부위 이종이중체 변이체와 유사한 정도로 유지되는 것이 확인된 상기 실시예 1-6의 동종형별 변이체 중에서도 IgG4를 기반으로 한 변이체 (γ4-A107)를 이용하여 지속성 인터루킨 12 (Interleukin 12) 융합단백질을 구축하였다. 자연계에 존재하는 인터루킨 12는 p35 단위체 (p35; IL-12A) 과 p40 단위체 (p40; IL-12B) 서로 다른 두 개의 단위체로 이루어져있고, 상기 두 개의 단위체가 상호작용하여 이종이중체를 형성함으로써 활성을 가진다. 이러한 이종이중체의 형성은 두 개의 단위체 사이에 존재하는 하나의 이황화 결합에 의해 더욱 단단하게 안정적으로 결합함으로써 이루어진다. 이에 각각의 단위체를 서로 다른 이종이중체 Fc 변이체 (CH3A 혹은 CH3B)에 연결함으로써 자연계에 존재하는 사이토카인의 이종이중체 형태를 유지하고자 하였다. Among the heterologous variants of Example 1-6, wherein the heterodimer-forming ability was maintained at a similar level to the previously reported IgG1-based A107 heavy chain constant region heterodimer variants, the IgG4-based variant (γ4-A107) was used. Persistent Interleukin 12 fusion protein was constructed. Interleukin 12 in nature consists of two different units of p35 monomer (p35; IL-12A) and p40 monomer (p40; IL-12B), which are active by interacting with each other to form heterodimers. Has Formation of such heterodimers is achieved by binding more stably with one disulfide bond present between two units. Therefore, we tried to maintain the heterodimer form of cytokines in nature by connecting each monomer to different heterodimeric Fc variants (CH3A or CH3B).
융합단백질의 구축을 위한 중쇄불변부위 이종이중체 변이체는 IgG4를 기반으로 하여 A107 돌연변이가 도입되어 이종이중체를 형성하는 γ4-A107을 사용하였다. 기존 보고에 따르면, 항체와 사이토카인을 융합한 형태인 이뮤노사이토카인 (Immunocytokine)의 구축에 있어 IgG1이 가지는 ADCC/CDC와 같은 항체 고유 기능이 오히려 생체 내 제거현상(clearance)을 촉진한다. 따라서 ADCC/CDC의 기능이 IgG1과 비교하였을 때 거의 나타나지 않는 IgG4 동종형을 사용하여 융합단백질을 구축하였다 (Gillies SD et al., 1999). The heavy chain constant region heterodimeric variant for the construction of the fusion protein was used γ4-A107 to introduce a heterodimer by introducing an A107 mutation based on IgG4. According to the existing reports, intrinsic functions such as ADCC / CDC of IgG1 in the construction of immunocytokine, which is a fusion form of antibody and cytokine, promote the clearance in vivo. Thus, the fusion protein was constructed using an IgG4 isotype that shows little function of ADCC / CDC compared to IgG1 (Gillies SD et al., 1999).
도 10은 본 발명에 사용된 IL-12의 재조합 단백질, γ4-A107를 이용한 Monovalent IL-12 융합단백질(mono-IL-12-Fc) 그리고 야생형 Fc를 이용한 bivalent IL-12 융합단백질(bi-IL-12-Fc)의 모식도를 나타낸 것이다. 그 중에서도 (C)는 본 발명에서 제작한 CH3 돌연변이체 쌍이 도입된 융합단백질이다. 인간 인터루킨 12 (hIL-12, Uniprot entry name P29460, P29459; 서열번호 17-18) 및 마우스 인터루킨 12(mIL-12, Uniprot entry name P43432, P43431; 서열번호 19-20)는 모두 signal sequence를 제외한 mature form을 코딩하는 DNA서열만을 증폭하여 γ4-A107 변이체가 들어가있는 동물세포 발현 벡터에 도 11(A) 및 (B)와 같은 인 프레임 (in-frame)으로 NotI/BsiWI 제한효소를 이용하여 클로닝 하였고 각각 mono-hIL-12-Fc와 mono-mIL-12-Fc로 명명하였다. 특히 인간/마우스 p35 단위체는 p40 단위체와의 충분한 상호작용이 가능하도록 p35 단위체와 힌지 영역 사이에 15 개의 유연성이 있는 펩타이드 링커 (flexible (G4S)3 Linker)를 부여하였다. (C)에 대한 비교예로 인간 인터루킨 12(hIL-12)와 마우스 인터루킨 12(mIL-12)를 야생형 IgG4 Fc(wt IgG4)를 융합한 bi-hIL-12-Fc와 bi-mIL-12-Fc를 구축하였다. 이종이중체를 이루어야만 활성을 가지는 IL-12를 하나의 Fc에 융합시키기 위해 두 개의 단위체를 상기 15개 펩타이드 링커로 연결해준 다음, γ4-A107 변이체가 들어가있는 동물세포 발현 벡터에 도 12와 같은 인-프레임 (in-frame)으로 NotI/BsiWI 제한효소를 이용하여 클로닝 하였다. 상기 비교예는 IL-12 융합단백질을 만들기 위한 기존의 연구에서 사용된 형태의 융합단백질이다 (Lisan S. Peng et al., 1999). 10 is a recombinant protein of IL-12, monovalent IL-12 fusion protein using γ4-A107 (mono-IL-12-Fc) and bivalent IL-12 fusion protein using wild type Fc used in the present invention (bi-IL) The schematic diagram of -12-Fc) is shown. Among them, (C) is a fusion protein to which the CH3 mutant pair produced in the present invention is introduced. Human interleukin 12 (hIL-12, Uniprot entry name P29460, P29459; SEQ ID NO: 17-18) and mouse interleukin 12 (mIL-12, Uniprot entry name P43432, P43431; SEQ ID NO: 19-20) are both mature except signal sequence Only the DNA sequence encoding the form was amplified and cloned using an NotI / BsiWI restriction enzyme in an in-frame as shown in FIGS. 11 (A) and (B) in an animal cell expression vector containing a γ4-A107 variant. They were named mono-hIL-12-Fc and mono-mIL-12-Fc, respectively. In particular, the human / mouse p35 monomer was assigned 15 flexible peptide linkers (G 4 S) 3 Linker between the p35 monomer and the hinge region to allow sufficient interaction with the p40 monomer. As a comparative example for (C), bi-hIL-12-Fc and bi-mIL-12- fused with human interleukin 12 (hIL-12) and mouse interleukin 12 (mIL-12) were fused with wild-type IgG4 Fc (wt IgG4). Fc was constructed. In order to fuse IL-12, which is active only by heteroduplexing, to one Fc, two units are connected to the 15 peptide linkers, and then the animal cell expression vector containing the γ4-A107 variant is shown in FIG. 12. It was cloned in-frame using NotI / BsiWI restriction enzyme. The comparative example is a fusion protein of the type used in previous studies to make IL-12 fusion proteins (Lisan S. Peng et al., 1999).
표 3은 상기 융합단백질의 구축에 사용된 인간 및 마우스 인터루킨 12의 단위체의 mature form에 대한 아미노산 서열이다. Table 3 shows the amino acid sequences for the mature forms of the monomers of human and mouse interleukin 12 used to construct the fusion protein.
Figure PCTKR2017008676-appb-T000003
Figure PCTKR2017008676-appb-T000003
실시예 8: IL-12 fusion protein 발현/정제Example 8: IL-12 fusion protein expression / purification
도 10 (C)의 mono-IL-12-Fc 융합단백질은 human/mouse IL-12.p40-γ4-A107A 및 human/mouse IL-12.p35-γ4-A107B의 발현 벡터를 1:1 비율로 하여 실시예 5와 같은 방법으로 발현/정제하였다. 도 10 (B)의 bi-IL-12-Fc 융합단백질은 human/mouse scIL-12-IgG4 Fc(wt) 발현 벡터의 단일 트랜스팩션을 통해 발현/정제하였다. 모든 융합단백질은 100 ml HEK293F cell culture 당 12-13 mg의 유사한 정도로 발현/정제되었다. The mono-IL-12-Fc fusion protein of FIG. 10C shows 1: 1 ratio of expression vectors of human / mouse IL-12.p40-γ4-A107A and human / mouse IL-12.p35-γ4-A107B. It was expressed / purified in the same manner as in Example 5. The bi-IL-12-Fc fusion protein of FIG. 10 (B) was expressed / purified via a single transfection of the human / mouse scIL-12-IgG4 Fc (wt) expression vector. All fusion proteins were expressed / purified at a similar level of 12-13 mg per 100 ml HEK293F cell culture.
상기 정제된 mono-IL-12-Fc 및 bi-IL-12-Fc 융합단백질 5 μg을 12% 비환원성 조건으로 SDS-PAGE 상에서 분석하였다 (도 13). IL-12.p40-CH3A 변이체의 모노머는 60 kD, 동종이중체는 120 kD, IL-12.p35-CH3B 변이체의 모노머는 50 kD 동종이중체는 100 kD에서 관찰되었으며, IL-12.p40-CH3A 변이체와 IL-12.p35-CH3B 변이체의 이종이중체는 110 kD에서 관찰되었다. 다만 인간과 마우스의 인터루킨 단위체가 연결된 단백질은 다소 다른 사이즈에서 밴드가 관찰되었는데, 이는 서로 다른 글리코실화(glycosylation) 패턴에 의한 것임을 참고문헌을 통해 확인하였다 (Lo et al., 2007). 또한 앞선 실시예 6에 서술한 것과 마찬가지로 IgG4를 기반으로 한 모든 IL-12 융합단백질에서 모노머가 관찰되었다. p35 단위체는 p40 단위체의 도움 없이는 자연적으로도 모노머 형태로 발현되지 않는다는 기존의 보고와 유사하게 이종이중체 중쇄불변부위 변이체를 이용한 mono-IL-12-Fc 융합단백질에서는 p40 단위체가 연결된 CH3A 모노머만이 관찰되었다 (Gillies et al., 1998b). 5 μg of the purified mono-IL-12-Fc and bi-IL-12-Fc fusion proteins were analyzed on SDS-PAGE under 12% non-reducing conditions (FIG. 13). The monomer of IL-12.p40-CH3A variant was 60 kD, the homodimer was 120 kD, the monomer of IL-12.p35-CH3B variant was 50 kD and the homodimer was 100 kD, and IL-12.p40- Heterodimers of the CH3A and IL-12.p35-CH3B variants were observed at 110 kD. However, it was confirmed through reference that the protein linked to the interleukin monomers of humans and mice was slightly different in size, due to different glycosylation patterns (Lo et al., 2007). In addition, as described in Example 6, monomers were observed in all IL-12 fusion proteins based on IgG4. Similar to previous reports that p35 monomers are not naturally expressed in monomeric form without the help of p40 monomers, in mono-IL-12-Fc fusion proteins using heteroduplex heavy chain constant region variants, only CH3A monomers to which p40 monomers are linked Was observed (Gillies et al., 1998b).
도 14는 상기 융합단백질들의 크기 별 배제 크로마토그래피 (SEC; Size-Exclusion Chromatography) 결과이다. Mono-hIL-12-Fc 융합단백질에서 일부 중합체가 관찰되었다. 14 is a result of size-exclusion chromatography (SEC) of the fusion proteins. Some polymers were observed in the Mono-hIL-12-Fc fusion protein.
실시예 9: Mono-hIL-12-Fc 융합단백질의 IL-12 수용체에 대한 결합능 평가Example 9 Evaluation of Binding Capacity of Mono-hIL-12-Fc Fusion Protein to IL-12 Receptor
실시예 8에서 발현, 정제한 mono-hIL-12-Fc의 IL-12 수용체에 대한 결합능을 bi-hIL-12-Fc와 비교 분석하였다. The binding ability of the mono-hIL-12-Fc expressed and purified in Example 8 to the IL-12 receptor was compared with bi-hIL-12-Fc.
도 15는 bi-hIL-12-Fc와 비교하여 구축된 mono-hIL-12-Fc이 IL-12 수용체에 결합능을 보이는 것을 유세포 분석기기인 FACS Calibur (BD Biosciences)로 확인한 결과이다. FIG. 15 shows the result of confirming that the mono-hIL-12-Fc constructed as compared with bi-hIL-12-Fc showed binding ability to the IL-12 receptor by FACS Calibur (BD Biosciences).
구체적으로는, 인간의 말초혈액에서 면역세포 (PBMC)를 분리하기 위하여 15 ml 시험관에 Ficoll (GE Healthcare) 5 ml을 채워 넣었다. 채혈한 혈액은 PBS (pH 7.4)와 1:1로 섞은 후 흔들어 준 다음 10 ml 만큼 취해서 Ficoll이 담긴 시험관에 Ficoll과 섞이지 않게 넣고 750 g에서 20분간 no break 상태로 원심 분리하였다. 그 후 Ficoll 위에 형성된 buffy coat를 회수하여 PBS (pH 7.4)로 2번 세척한 후 T세포, B세포, NK 세포 및 단핵구를 포함하는 PBMC를 얻었다. 분리된 정상의 PBMC는 IL-12의 binding을 볼 수 있을 만큼 많은 양의 IL-12R 를 발현하고 있지 않다. 때문에 PHA (Sigma-Aldrich) 라는 mitogen을 72 h 동안 처리하여 T세포와 NK세포가 활성화 할 수 있도록 자극을 주었다. PHA를 처리하게 되면 면역세포가 분열하면서 T세포와 NK세포에 IL-12 수용체가 발현된다는 보고가 있다. 10% FBS가 포함된 RPMI1640 배지에 1×106 cells/ml의 PBMC를 넣고 mitogen으로 PHA를 10μg/ml 농도로 첨가한 후 37도, 5% CO2 배양기에서 배양 72 시간 동안 배양하였다. 정상 PBMC와 PHA로 활성화된 PBMC는 차가운 PBS(pH 7.4)로 세척하고 각 샘플당 5×105 개의 세포를 준비하였다. Fc (A107), bi-hIL-12-Fc 및 mono-hIL-12-Fc를 1 μM 농도로 첨가하여 4℃에서 30분간 반응시킨 후 차가운 PBS (pH 7.4)로 세척하였다. 인간 IgG4를 인지하는 FITC 형광이 연결된 이차항체(Sigma-Aldrich)를 4℃에서 30분간 반응시키고 PBS(pH 7.4)로 세척 후, 유세포 분석기기인 FACS Calibur (BD Bioscience)로 분석하였다. 분석 후, 각각의 샘플에 대한 히스토그램 그래프를 얻어 mono-hIL-12-Fc의 IL-12 수용체에 대한 결합능을 평가하였다. Specifically, 5 ml of Ficoll (GE Healthcare) was charged into a 15 ml test tube in order to separate immune cells (PBMC) from human peripheral blood. The collected blood was mixed 1: 1 with PBS (pH 7.4), shaken, and then taken up to 10 ml, mixed with Ficoll in a test tube containing Ficoll, and centrifuged at 750 g for 20 minutes with no break. Thereafter, the buffy coat formed on Ficoll was recovered and washed twice with PBS (pH 7.4) to obtain PBMCs including T cells, B cells, NK cells and monocytes. The isolated normal PBMC does not express a large amount of IL-12R to see the binding of IL-12. Thus, PHA (Sigma-Aldrich) was treated for 72 h to stimulate T and NK cells. PHA treatment has been reported to express IL-12 receptors on T cells and NK cells as the immune cells divide. 1 × 10 6 cells / ml of PBMC was added to RPMI1640 medium containing 10% FBS, and PHA was added at a concentration of 10 μg / ml as a mitogen, followed by incubation for 72 hours at 37 degrees and 5% CO 2 incubator. Normal PBMC and PHA activated PBMC were washed with cold PBS (pH 7.4) and 5 × 10 5 cells were prepared for each sample. Fc (A107), bi-hIL-12-Fc and mono-hIL-12-Fc were added at a concentration of 1 μM, reacted at 4 ° C. for 30 minutes, and washed with cold PBS (pH 7.4). FITC fluorescence-linked secondary antibody (Sigma-Aldrich) that recognizes human IgG4 was reacted at 4 ° C. for 30 minutes, washed with PBS (pH 7.4), and analyzed by FACS Calibur (BD Bioscience), a flow cytometer. After analysis, histogram graphs for each sample were obtained to assess the binding capacity of mono-hIL-12-Fc to the IL-12 receptor.
분석결과, bi-hIL-12-Fc와 mono-hIL-12-Fc은 IL-12 수용체를 발현하지 않는 정상 PBMC에는 결합하지 않고 PHA로 활성화되어 IL-12 수용체를 발현하는 PBMC에만 결합하였다. 따라서 mono-hIL-12-Fc의 IL-12 수용체에 대한 결합능은 bi-hIL-12-Fc와 동일한 것으로 확인되었다. As a result, bi-hIL-12-Fc and mono-hIL-12-Fc did not bind to normal PBMCs that did not express IL-12 receptors, but only to PBMCs that were activated by PHA and expressed IL-12 receptors. Therefore, the binding capacity of mono-hIL-12-Fc to IL-12 receptor was confirmed to be the same as bi-hIL-12-Fc.
실시예 10: Mono-hIL-12-Fc 융합단백질의 PBMC 증식 유도 능력 평가Example 10 Evaluation of PBMC Proliferation Induction Capacity of Mono-hIL-12-Fc Fusion Protein
IL-12 fusion protein내 IL-12 moiety가 IL-12 수용체에 결합하여 실제 recombinant IL-12(rIL-12) 만큼의 생리활성을 유지하는지를 recombinant human IL-12 (rhIL-12, Thermo Fisher Scientific)을 대조 군으로 하여 확인하였다.Whether the IL-12 moiety in the IL-12 fusion protein binds to the IL-12 receptor and maintains the biological activity of the actual recombinant IL-12 (rIL-12) is determined by recombinant human IL-12 (rhIL-12, Thermo Fisher Scientific). It confirmed as a control group.
도 16은 PHA로 활성화된 PBMC에서 Fc (A107), rhIL-12, bi-hIL-12-Fc와 mono-hIL-12-Fc에 의한 세포증식 능을 확인한 WST-1 세포증식 시험 결과이다. FIG. 16 shows WST-1 cell proliferation test results confirming cell proliferation ability by Fc (A107), rhIL-12, bi-hIL-12-Fc and mono-hIL-12-Fc in PHA-activated PBMCs.
구체적으로는, 실시예 9에서와 동일하게 PHA로 활성화된 PBMC (2 x 104, 50 μl)를 96웰 플레이트(SPL, Korea)에 첨가하고 10 % FBS가 포함된 RPMI1640 배지로 연속적으로 희석된 50-0.4 pM의 Fc (A107), rhIL-12, bi-hIL-12-Fc와 mono-hIL-12-Fc를 50 μl씩 첨가한 다음 72 시간 동안 5 % CO2, 37 ℃ 조건에서 배양 후, 세포증식시험을 위해 WST-1 (Water-soluble Tetrazolium salts, Sigma-aldrich) 시약을 10 μl씩 각 웰에 첨가한 후 37 ℃에서 4시간 반응시키고 570 nm에서 흡광도를 마이크로플레이트 리더(Molecular Devices)를 이용하여 측정하였다.Specifically, PBMC (2 × 10 4, 50 μl) activated with PHA was added to 96-well plate (SPL, Korea) and serially diluted with RPMI1640 medium containing 10% FBS as in Example 9. 50 μl of -0.4 pM Fc (A107), rhIL-12, bi-hIL-12-Fc, and mono-hIL-12-Fc were added, followed by incubation at 5% CO 2 , 37 ° C for 72 hours. For cell proliferation test, 10 μl of water-soluble Tetrazolium salts (Sigma-aldrich) reagent was added to each well, followed by 4 hours of reaction at 37 ° C. and absorbance at 570 nm using a microplate reader (Molecular Devices). It measured using.
분석 결과, mono-hIL-12-Fc는 rhIL-12와 유사하거나 더 높은 PBMC 증식능을 가지는 것을 확인하였다. As a result, it was confirmed that mono-hIL-12-Fc had a PBMC proliferation similar to or higher than that of rhIL-12.
실시예 11: Mono-hIL-12-Fc 융합단백질의 PBMC에 대한 IFN-γ 분비 유도 능력 평가Example 11: Evaluation of the ability to induce IFN-γ secretion of mono-hIL-12-Fc fusion protein against PBMC
도 17은 PHA로 활성화된 PBMC에서 Fc (A107), rhIL-12, bi-hIL-12-Fc와 mono-hIL-12-Fc에 의한 IFN-γ 분비 양을 측정한 ELISA 결과이다. 17 shows ELISA results of measuring the amount of IFN-γ secretion by Fc (A107), rhIL-12, bi-hIL-12-Fc and mono-hIL-12-Fc in PHA activated PBMC.
구체적으로는, 실시예 10에서 72시간 동안 배양된 세포배양액 내 IFN-γ 농도를 측정하기 위하여 ELISA용 96-웰 플레이트 (Thermo Fisher Scientific, Korea)에 인간 IFN-γ 포획 항체 (Thermo Fisher Scientific)를 12시간 동안 코팅한 후, PBST (PBS with 0.1% Tween-20)으로 세척 후, 1% BSA (PBS with 1% bovine serum albumin)를 넣고 1시간 동안 실온에서 blocking하였다. PBST (PBS with 0.1% Tween-20)으로 세척 후, 실시 예 2에서 배양된 배양액을 1% BSA로 5배 희석하여 100 μl씩 첨가한 다음 실온에서 2시간 동안 반응시켰다. PBST (PBS with 0.1% Tween-20)으로 세척 후, biotin이 결합된 IFN-γ 검출 항체(Thermo Fisher Scientific)를 실온에서 1시간 동안 결합시켰다. PBST (PBS with 0.1% Tween-20)으로 세척 후, avidin이 결합된 horse radish peroxidase (HRP) (Thermo Fisher Scientific)를 실온에서 30분간 결합시킨 다음 PBST (PBS with 0.1% Tween-20)으로 세척 후, 3,3′,5,5′-Tetramethylbenzidine (TMB, sigma-aldrich) 기질을 넣어 마이크로플레이트 리더를 이용하여 405 nm에서의 흡광도를 측정하였다.Specifically, in order to measure IFN-γ concentration in cell culture cultured for 72 hours in Example 10, a human IFN-γ capture antibody (Thermo Fisher Scientific) was placed in a 96-well plate (Thermo Fisher Scientific, Korea) for ELISA. After coating for 12 hours, washed with PBST (PBS with 0.1% Tween-20), 1% BSA (PBS with 1% bovine serum albumin) was added and blocked for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), the culture cultured in Example 2 was diluted 5 times with 1% BSA, added 100 μl, and then reacted at room temperature for 2 hours. After washing with PBST (PBS with 0.1% Tween-20), the biotin-bound IFN-γ detection antibody (Thermo Fisher Scientific) was bound for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), avidin-coupled horse radish peroxidase (HRP) (Thermo Fisher Scientific) was combined for 30 minutes at room temperature, followed by washing with PBST (PBS with 0.1% Tween-20). , 3,3 ', 5,5'-Tetramethylbenzidine (TMB, sigma-aldrich) substrate was added to measure the absorbance at 405 nm using a microplate reader.
분석 결과, mono-hIL-12-Fc은 PBMC에 대한 IFN-γ 분비 유도능이 rhIL-12와 유사거나 더 높은 것을 확인하였다. As a result, mono-hIL-12-Fc showed that IFN-γ secretion ability to PBMC is similar or higher than rhIL-12.
실시예 12: Mono-mIL-12-Fc의 IL-12 수용체에 대한 결합능 평가.Example 12 Evaluation of the Avidity of Mono-mIL-12-Fc to IL-12 Receptor.
실시예 8에서 발현, 정제한 mono-mIL-12-Fc의 IL-12 수용체에 대한 결합능을 bi-mIL-12-Fc와 비교 분석하였다. The binding ability of the mono-mIL-12-Fc expressed and purified in Example 8 to the IL-12 receptor was compared with bi-mIL-12-Fc.
도 18은 bi-mIL-12-Fc과 비교하여 구축된 mono-mIL-12-Fc이 IL-12 수용체에 결합능을 보이는 것을 확인한 Flow cytometry 결과이다. FIG. 18 is a flow cytometry result confirming that mono-mIL-12-Fc constructed as compared with bi-mIL-12-Fc shows binding ability to IL-12 receptor.
구체적으로는, 마우스 IL-12는 마우스 IL-12 수용체뿐만 아니라 인간 IL-12 수용체에도 결합한다고 보고되어 실시예 9에서와 같은 방법으로 분석하였다. 분석결과, bi-mIL-12-Fc과 mono-mIL-12-Fc은 IL-12 수용체를 발현하지 않는 정상 PBMC에는 결합하지 않고 PHA로 활성화되어 IL-12 수용체를 발현하는 PBMC에만 결합하였다. 따라서 mono-mIL-12-Fc의 IL-12 수용체에 대한 결합능은 bi-mIL-12-Fc와 동일한 것으로 확인되었다. Specifically, mouse IL-12 was reported to bind not only mouse IL-12 receptor but also human IL-12 receptor, and analyzed in the same manner as in Example 9. As a result, bi-mIL-12-Fc and mono-mIL-12-Fc did not bind to normal PBMCs that did not express IL-12 receptors, but only to PBMCs that were activated by PHA and expressed IL-12 receptors. Therefore, the binding capacity of mono-mIL-12-Fc to IL-12 receptor was confirmed to be the same as bi-mIL-12-Fc.
실시예 13: Mono-mIL-12-Fc의 PBMC 증식 유도 능력 평가Example 13: Evaluation of PBMC proliferation ability of Mono-mIL-12-Fc
도 19는 PHA로 활성화된 PBMC에서 Fc (A107), recombinant mouse IL-12(rmIL-12), bi-mIL-12-Fc과 mono-mIL-12-Fc에 의한 세포증식 능을 확인한 WST-1 세포증식 어세이 결과이다. Figure 19 WST-1 confirmed the cell proliferation ability by Fc (A107), recombinant mouse IL-12 (rmIL-12), bi-mIL-12-Fc and mono-mIL-12-Fc in PHA activated PBMC Cell proliferation assay results.
구체적으로는, 실시예 9에서와 동일하게 PHA로 활성화된 PBMC (2×104, 50 μl)를 96웰 플레이트에 첨가하고 10 % FBS가 포함된 RPMI1640 배지로 연속적으로 희석된 50-0.4 pM의 Fc (A107), rmIL-12, bi-mIL-12-Fc과 mono-mIL-12-Fc를 50 μl씩 첨가한 다음 72시간동안 5 % CO2, 37℃ 조건에서 배양 후, 실시예 10과 같은 방법으로 WST 어세이를 수행하였다. 분석 결과, mono-mIL-12-Fc은 rmIL-12와 유사하게 PBMC 증식을 유도하는 능력을 가지는 것을 확인하였다. Specifically, 50-0.4 pM of PHA-activated PBMC (2 × 10 4 , 50 μl) was added to a 96 well plate and serially diluted with RPMI1640 medium containing 10% FBS as in Example 9. 50 μl of Fc (A107), rmIL-12, bi-mIL-12-Fc and mono-mIL-12-Fc were added thereto, followed by incubation at 5% CO 2 , 37 ° C. for 72 hours. The WST assay was performed in the same way. As a result, mono-mIL-12-Fc was found to have the ability to induce PBMC proliferation similar to rmIL-12.
실시예 14: Mono-mIL-12-Fc의 생체 내 종양성장 억제능 평가Example 14 Evaluation of Tumor Growth Inhibitory Activity of Mono-mIL-12-Fc in Vivo
실시예 13에서 mono-mIL-12-Fc의 PHA로 활성화된 PBMC에서의 세포증식능을 확인하였다. 생체 내에서도 mono-mIL-12-Fc의 효과가 동일하게 나타나는지 확인하였다. In Example 13, cell proliferation in PBMCs activated with PHA of mono-mIL-12-Fc was confirmed. It was confirmed whether the effect of mono-mIL-12-Fc is the same in vivo.
도 20(A)와 20(B)는 mono-mIL-12-Fc의 마우스 생체 내에서 종양 성장 억제 활성을 100 mm3 크기의 종양에서 측정한 결과이다. 20 (A) and 20 (B) show the results of measuring tumor growth inhibitory activity of mono-mIL-12-Fc in 100 mm 3 tumors.
구체적으로, 4 주령의 암컷 Balb/c 마우스 (NARA Biotech, Korea)의 털을 면도기로 제거하고 CT26HER2 / Neu 대장암세포 (1×106 세포/마우스)를 150 μL PBS에 희석하여 마우스 피하에 이식하였다. 유사한 크기의 종양 (평균 부피 100~120 mm3)을 가진 마우스를 처리집단으로 무작위로 배정하고, Fc (A107), rmIL-12 (Thermo Fisher Scientific), bi-mIL-12-Fc과 mono-mIL-12-Fc을 마우스 한 마리당 1 μg 분자당량의 IL-12를 1주일에 2회씩 총 6회 복강으로 주사하였다. 종양은 일주일에 2회씩 측정하고, 종양의 부피 (V)는 V=길이x폭2/2 로 계산하였다. Specifically, hair of 4 week old female Balb / c mice (NARA Biotech, Korea) was removed with a razor and CT26 HER2 / Neu colorectal cancer cells (1 × 10 6 cells / mouse) were diluted in 150 μL PBS and transplanted into the mouse subcutaneous. It was. Mice with similarly sized tumors (mean volume 100-120 mm 3 ) were randomly assigned to treatment groups, Fc (A107), rmIL-12 (Thermo Fisher Scientific), bi-mIL-12-Fc and mono-mIL -12-Fc was injected intraperitoneally with 1 μg molecular equivalent of IL-12 per mouse twice a week. Tumors are measured two times a week, and the volume (V) of the tumor was calculated as V = L x W 2/2.
도 20(A)에 나타난 바와 같이, 대조군에 비해 1 ㎍의 rmIL-12 투여는 종양 성장 억제효과가 없었으나 같은 몰 농도의 mono-mIL-12-Fc과 bi-mIL-12-Fc는 종양 성장을 억제하였다. 또한, 도 20(B)에서 대조군과 비교하여 mono-mIL-12-Fc과 bi-mIL-12-Fc의 투여 시 마우스의 체중 변화가 거의 없는 것을 확인하였으며, 이에 따라 독성은 없는 것으로 판단되었다. As shown in FIG. 20 (A), 1 μg of rmIL-12 had no tumor growth inhibitory effect compared to the control group, but mono-mIL-12-Fc and bi-mIL-12-Fc at the same molar concentration showed tumor growth. Was suppressed. In addition, in FIG. 20 (B), it was confirmed that the weight of the mice was little changed when the mono-mIL-12-Fc and the bi-mIL-12-Fc were administered compared to the control group, and thus, the toxicity was not determined.
도 21(A), 21(B)와 21(C)는 농도별로 투여한 mono-mIL-12-Fc의 마우스 생체 내에서 종양 성장 억제 활성을 300 mm3 크기의 종양을 가진 마우스에서 측정한 결과이다. Figure 21 (A), 21 (B) and 21 (C) is the result of measuring the tumor growth inhibitory activity in mice in vivo of mono-mIL-12-Fc administered at different concentrations in mice with 300 mm 3 tumor size to be.
구체적으로, 4 주령의 암컷 Balb/c 마우스 (NARA Biotech, Korea)의 털을 면도기로 제거하고 CT26HER2 / Neu 대장암세포 (1×106 세포/마우스)를 150 μL PBS에 희석하여 마우스 피하에 이식하였다. 유사한 크기의 종양 (평균 부피 300 mm3)을 가진 마우스를 처리집단으로 무작위로 배정하고, 마우스 한 마리당 0.1~2 μg rmIL-12와 몰 농도가 같은 bi-mIL-12-Fc과 mono-mIL-12-Fc를 1주일에 2회씩 총 6회 복강으로 주사하였다. 종양은 일주일에 2회씩 측정하고, 종양의 부피 (V)는 V=길이x폭2/2 로 계산하였다. Specifically, hair of 4 week old female Balb / c mice (NARA Biotech, Korea) was removed with a razor and CT26 HER2 / Neu colorectal cancer cells (1 × 10 6 cells / mouse) were diluted in 150 μL PBS and transplanted into the mouse subcutaneous. It was. Mice with similarly sized tumors (mean volume 300 mm 3 ) were randomly assigned to treatment groups and bi-mIL-12-Fc and mono-mIL- with the same molar concentration of 0.1 to 2 μg rmIL-12 per mouse. 12-Fc was injected intraperitoneally, twice a week. Tumors are measured two times a week, and the volume (V) of the tumor was calculated as V = L x W 2/2.
도 21(A)와 21(B) 및 21(C)에 나타낸 바와 같이 크기가 큰 종양에 대해서도 1 μg IL-12 이하의 몰 농도에서 mono-mIL-12-Fc가 bi-IL-12-Fc에 비해 유의적으로 종양성장을 억제하는 효과가 높았다. 0.25 μg IL-12와 같은 몰 농도의 bi-mIL-12-Fc는 종양의 성장을 억제하는 효과는 보였지만 종양을 제거하지는 못했다. 그러나 같은 몰 농도의 mono-mIL-12-Fc는 40%의 마우스에서 종양을 제거하는 효과를 보였다. 또한 bi-mIL-12-Fc가 종양을 제거하지 못한 0.5 μg IL-12 농도에서 mono-mIL-12-Fc를 5회만 투여하더라도 73%의 마우스의 종양이 제거되었다. As shown in FIGS. 21 (A), 21 (B) and 21 (C), bi-IL-12-Fc was mono-mIL-12-Fc at a molar concentration of 1 μg IL-12 or less even for large tumors. Compared with that, the effect of inhibiting tumor growth was significantly higher. A molar concentration of bi-mIL-12-Fc, such as 0.25 μg IL-12, was shown to inhibit tumor growth but did not remove the tumor. However, the same molar concentration of mono-mIL-12-Fc was effective in removing tumors in 40% of mice. In addition, 73% of the tumors were cleared by only 5 doses of mono-mIL-12-Fc at a concentration of 0.5 μg IL-12 where bi-mIL-12-Fc did not remove tumors.
실시예 15: Mono-mIL-12-Fc의 생체 내 독성 평가Example 15 In Vivo Toxicity Evaluation of Mono-mIL-12-Fc
도 21(D)은 농도 별로 투여한 mono-mIL-12-Fc의 생체 내에서의 독성을 체중변화로 측정한 결과이다. Figure 21 (D) is the result of measuring the weight change in vivo toxicity of mono-mIL-12-Fc administered by concentration.
구체적으로, 도 21(A)와 같이 투여된 마우스의 체중을 1주일에 두 번씩 측정하여 체중감소 여부를 관찰하였다. 대조군의 경우 종양의 크기가 커지면서 체중이 증가하였으나, 모든 농도의 bi-mIL-12-Fc과 mono-mIL-12-Fc를 투여한 마우스에서 투여 전에 비해 체중 감소가 없는 것을 확인하였다. 따라서 mono-mIL-12-Fc는 체중감소를 유도하는 생체 내 독성은 없는 것으로 판단되었다. Specifically, the weight of the mice administered as shown in Figure 21 (A) was measured twice a week to observe the weight loss. In the case of the control group, the weight of the tumor was increased as the tumor size increased, but it was confirmed that there was no weight loss in the mice administered with all concentrations of bi-mIL-12-Fc and mono-mIL-12-Fc. Therefore, mono-mIL-12-Fc was not considered to be in vivo toxic to induce weight loss.
도 21(D)은 간 독성의 지표인 알라닌 아미노 전이효소 (ALT) 측정한 결과이다. Figure 21 (D) is the result of measuring alanine aminotransferase (ALT) which is an indicator of liver toxicity.
구체적으로, 도 21(A)의 마우스에서 마지막 투여 24시간째에 얼굴정맥에서 채혈하였다. 혈액은 2시간 동안 실온에 방치하여 혈액응고를 유도한 후 8000 rpm에서 10분간 원심 분리하여 상층의 혈청을 회수하였다. 혈청 내 ALT의 농도를 측정하기 위해 마지막 IL-12-Fc 융합단백질 투여 24시간 뒤, 마우스의 얼굴 정맥에서 채혈하였다. 혈액은 2시간 동안 실온에 방치 하여 혈액응고를 유도한 후 8000 rpm에서 10분간 원심 분리하여 상층의 혈청을 회수하였다. 혈청 내 ALT 농도를 측정하기 위해서 ALT 측정용 기질액 (alanine과 α-ketoglutarate 혼합액)을 15 ml 시험관에 취해 37℃ 항온수조에서 5분간 배양하였다. 여기에 종양이 이식된 마우스에 bi-mIL-12-Fc과 mono-mIL-12-Fc를 투여한 다음 혈액을 채취하여 분리한 혈청을 10배 희석하여 200 μl씩 첨가한 후 진탕하여 37℃ 항온수조에서 30분간 배양하였다. 항온수조에서 꺼낸 시험관에 정색시액 (2,4-dinitrophenyl-1-hydrazone)을 1 ml 가해 섞은 후 실온에서 20분간 방치하였다. 그 후, 0.4 N 수산화나트륨 용액을 10 ml 가하여 혼합한 후 실온에 10분 방치하였다. 광전분광광도계(GeneQuant100, GE Healthcare)를 사용하여 505 nm의 파장에서 흡광도를 측정하였다. 혈청대신 표준곡선시액을 첨가하여 작성한 표준곡선에 의해 ALT의 단위를 환산하여 구하였다. Bi-mIL-12-Fc과 mono-mIL-12-Fc를 투여한 마우스에서 채혈하여 얻은 혈청은 대조군이나 정상 Balb/c 마우스에서 채혈하여 얻은 혈청과 비슷한 ALT 활성을 나타내어 bi-mIL-12-Fc과 mono-mIL-12-Fc를 종양이식마우스에 0.5 μg이나 1 μg IL-12와 같은 몰 농도를 투여하였을 때 간 독성을 유도하지 않는 것으로 확인되었다. Specifically, blood was collected from the facial vein at 24 hours of the last administration in the mouse of FIG. 21 (A). Blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer. To measure the concentration of ALT in the serum, blood was collected from the facial vein of the mouse 24 hours after the last IL-12-Fc fusion protein administration. The blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer. In order to measure serum ALT concentration, ALT measurement substrate solution (alanine and α-ketoglutarate mixed solution) was taken in a 15 ml test tube and incubated in a 37 ° C constant temperature water bath for 5 minutes. Here, the mice transplanted with the tumor were treated with bi-mIL-12-Fc and mono-mIL-12-Fc, and blood samples were collected by diluting the serum 10-fold and adding 200 μl and shaking. Incubate for 30 minutes in a water bath. 1 ml of color solution (2,4-dinitrophenyl-1-hydrazone) was added to the test tube taken out of the constant temperature water bath, followed by mixing at room temperature for 20 minutes. Thereafter, 10 ml of 0.4 N sodium hydroxide solution was added thereto, mixed, and left to stand at room temperature for 10 minutes. Absorbance was measured at a wavelength of 505 nm using a photospectrophotometer (GeneQuant100, GE Healthcare). The standard curve prepared by adding the standard curve solution instead of the serum was used to calculate the unit of ALT. Serum obtained from blood samples from bi-mIL-12-Fc and mono-mIL-12-Fc mice showed ALT activity similar to that obtained from control or normal Balb / c mice, resulting in bi-mIL-12-Fc. And mono-mIL-12-Fc did not induce hepatotoxicity when administered to molar concentrations such as 0.5 μg or 1 μg IL-12.
실시예 16: 생체 내에서 mono-mIL-12-Fc의 면역세포 증식 유도능 평가Example 16: Evaluation of immune cell proliferation induction capacity of mono-mIL-12-Fc in vivo
실시예 15에서 2 μg IL-12와 같은 몰 농도를 투여하였을 때는 bi-mIL-12-Fc과 mono-mIL-12-Fc 모두 종양을 제거하였으나 1 μg IL-12 보다 낮은 몰 농도에서는 mono-mIL-12-Fc가 bi-mIL-12-Fc 보다 유의적으로 종양성장 억제효과가 높았다. 실제로, mono-mIL-12-Fc의 높은 종양성장 억제효과가 IL-12의 수용체를 가진 NK세포, CD4+ T세포 및 CD8+ T세포와 같은 고유의 효과세포의 수의 증가와 관련이 있는지를 확인하였다. When the molar concentration of 2 μg IL-12 was administered in Example 15, both bi-mIL-12-Fc and mono-mIL-12-Fc removed tumors, but mono-mIL at a molar concentration lower than 1 μg IL-12. -12-Fc showed significantly higher tumor growth inhibitory effect than bi-mIL-12-Fc. Indeed, whether the high tumor growth inhibitory effect of mono-mIL-12-Fc is associated with an increase in the number of inherent effector cells such as NK cells, CD4 + T cells and CD8 + T cells with IL-12 receptors. Confirmed.
도 22(A)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 CD4+ T세포, CD8+ T세포 및 NK세포 수의 증가를 확인한 결과이다. Figure 22 (A) is the result of confirming the increase in the number of CD4 + T cells, CD8 + T cells and NK cells in the spleen after 3 days of the last administration of Figure 21 (A).
구체적으로, 21(A)과 같이 처리한 후 종양이식 34일째에 마우스의 비장을 적출하여 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml로 세척하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈시킨 다음 PBS로 세척하여 비장세포 혼탁액을 준비하고 헤모사이토미터로 세포의 수를 계수하였다. 비장림프구를 APC, FITC, PE 또는 PE-cy5가 결합된 CD45, CD3, CD4, CD8 및 CD49b를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 CD45+CD3+CD4+ 세포군, CD45+CD3+CD8+ 세포군 및 CD45+CD3-CD49b+ 세포군을 각각 CD4+ T세포, CD8+ T세포, NK세포로 정의하여 전체 비장세포에 대한 비율을 구하고 헤모사이토미터로 계수된 세포의 수를 곱하여 mono-mIL-12-Fc 투여 후 증가된 CD4+ T세포, CD8+ T세포 및 NK세포의 수를 분석하였다. Specifically, after treatment as shown in 21 (A), the mouse spleen was extracted at 34 days after tumor transplantation, pulverized with a wire mesh in Petri dishes and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to erythrocytes, and red blood cells were lysed and washed with PBS to prepare splenocyte turbidity, and the number of cells was counted with a hemocytometer. Spleen lymphocytes were stained at 4 ° C. for 30 minutes with antibodies recognizing CD45, CD3, CD4, CD8 and CD49b bound to APC, FITC, PE or PE-cy5 and washed with cold PBS (pH 7.4). Analysis was performed by FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed with a dot plot to identify CD45 + CD3 + CD4 + cell populations, CD45 + CD3 + CD8 + cell populations, and CD45 + CD3 - CD49b + cell populations, respectively, as CD4 + T cells, CD8 + T cells, and NK cells. The ratio of total splenocytes was calculated and the number of CD4 + T cells, CD8 + T cells and NK cells increased after mono-mIL-12-Fc administration by multiplying the number of cells counted with hemocytometer. .
그 결과, 대조군에 비해 mono-mIL-12-Fc는 종양이 이식된 마우스에서 농도의존적으로 CD4+ T세포, CD8+ T세포의 수를 증가시켰음을 확인할 수 있었다. 반면, bi-mIL-12-Fc는 0.5 μg IL-12와 같은 몰 농도를 투여한 군에서만 CD8+ T세포의 수를 증가시켰고, 1 μg IL-12와 같은 몰 농도를 투여한 군에서는 CD4+ T세포와 CD8+ T세포의 수를 증가시키지 못했다. NK세포는 종양이식 마우스에서 기억세포를 형성하지 못한다는 선행연구결과 (Cerwenka and Lanier, 2016; Schreiber et al., 2011)와 일치하게 종양이식 34일째에는 mono-mIL-12-Fc와 bi-mIL-12-Fc투여군 모두에서 NK세포 수가 대조군과 비슷함을 확인하였다. 따라서 mono-mIL-12-Fc는 bi-mIL-12-Fc보다 CD4+ T세포, CD8+ T세포의 수를 유의적으로 증가시켜 종양형성 억제효과가 높았음을 확인하였다. As a result, it was confirmed that mono-mIL-12-Fc increased the number of CD4 + T cells and CD8 + T cells in a concentration-dependent manner compared to the control mice. On the other hand, bi-mIL-12-Fc increased the number of CD8 + T cells only in the molar concentrations such as 0.5 μg IL-12 and in the molar concentrations such as 1 μg IL-12, CD4 + It did not increase the number of T cells and CD8 + T cells. Consistent with previous studies showing that NK cells do not form memory cells in tumor-grafted mice (Cerwenka and Lanier, 2016; Schreiber et al., 2011), mono-mIL-12-Fc and bi-mIL on day 34 of tumor transplantation It was confirmed that the number of NK cells in all -12-Fc administration group was similar to the control group. Therefore, mono-mIL-12-Fc significantly increased the number of CD4 + T cells and CD8 + T cells than bi-mIL-12-Fc, which was confirmed to have a higher tumor suppression effect.
종양성장을 억제하기 위해서는 종양에 침윤된 적응 면역세포인 CD4+ T세포 와 CD8+ T세포 수의 증가가 중요하다는 보고(Schreiber et al., 2011)를 근거로 mono-mIL-12-Fc가 종양에 침윤된 적응 면역세포의 수를 증가시켰는지를 분석하였다. Mono-mIL-12-Fc를 6번의 투여하였을 때 종양이 없는 마우스가 많았으므로 3번 투여 후 마우스의 종양에 침윤된 면역세포의 수를 분석하였다. To suppress tumor growth, mono-mIL-12-Fc infiltrated tumors based on the report that the increase in the number of CD4 + T cells and CD8 + T cells, which are adaptive immune cells infiltrated into tumors, is important (Schreiber et al., 2011). It was analyzed whether the number of adaptive immune cells increased. When 6 mono-mIL-12-Fc were administered to a large number of mice without tumors, the number of immune cells infiltrated into the tumors of mice after 3 administrations was analyzed.
도 22(B)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 종양에 침윤되어 있는 총 면역세포, CD4+ T세포 및 CD8+ T세포의 수를 나타낸 결과이다. Figure 22 (B) is the result showing the total number of immune cells, CD4 + T cells and CD8 + T cells infiltrated into the tumor by 3 days after the third administration of the 21 (A).
구체적으로, 21(A)과 같이 처리한 후 종양이식 24일째에 마우스의 종양을 적출하여 무게를 측정한 후, 페트리디쉬에서 철망과 collagenase (100 μg/ml)을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml를 넣어 50 g에서 5분간 원심분리하여 실질조직을 제거하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈시킨 다음 PBS로 세척하여 세포 혼탁액을 준비하고 헤모사이토미터로 세포의 수를 계수하였다. 종양에서 분리한 세포를 APC, FITC 또는 PE-cy5가 결합된 CD45, CD3, CD4 및 CD8를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석한 후 CD45+ 세포군, CD45+CD3+CD4+ 세포군 및 CD45+CD3+CD8+ 세포군 및 CD45+CD3-CD49b+ 세포군을 각각 총 종양침윤면역세포, 종양침윤 CD4+ T세포 및 종양침윤 CD8+ T세포로 정의하였다. 전체 종양에서 분리한 세포에 대한 이들 세포의 비율을 구하여 헤모사이토미터로 계수된 세포의 수를 곱한 다음 mono-mIL-12-Fc 투여 후 증가된 총 종양침윤면역세포, 종양침윤 CD4+ T세포 및 종양침윤 CD8+ T세포의 수를 분석하였다. Specifically, after treatment as 21 (A), the tumor of the mouse was extracted and weighed on the 24th day of tumor transplantation, and then pulverized using a wire mesh and collagenase (100 μg / ml) in Petri dish, followed by 2% FBS. 10 ml of the medium containing the was added and centrifuged at 50 g for 5 minutes to remove the parenchyma. Thereafter, 1 ml of red blood cell lysis buffer was added to erythrocytes, and red blood cells were lysed, washed with PBS to prepare a cell suspension, and the number of cells was counted with a hemocytometer. Cells isolated from tumors were stained at 4 ° C. for 30 minutes with antibodies recognizing CD45, CD3, CD4 and CD8 bound with APC, FITC or PE-cy5, washed with cold PBS (pH 7.4) and FACS, a flow cytometer. Analyzes were performed with Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by a dot plot, and then the CD45 + cell group, CD45 + CD3 + CD4 + cell group, and CD45 + CD3 + CD8 + cell group and CD45 + CD3 - CD49b + cell group were measured as total tumor infiltrating immune cells and tumor infiltrates, respectively. CD4 + T cells and tumor infiltrating CD8 + T cells were defined. Calculate the ratio of these cells to the cells isolated from the total tumor, multiply the number of cells counted with hemocytometer, and then increase total tumor infiltrating immune cells, tumor infiltrating CD4 + T cells after mono-mIL-12-Fc administration, and The number of tumor infiltrating CD8 + T cells was analyzed.
그 결과, 대조군에 비해 bi-mIL-12-Fc와 mono-mIL-12-Fc는 종양이 이식된 마우스에서 농도의존적으로 종양에 침윤된 총 면역세포와 CD4+ T세포, CD8+ T세포의 수를 증가시켰음을 확인할 수 있었다. 같은 몰 농도의 투여군에서 mono-mIL-12-Fc는 bi-mIL-12-Fc보다 유의적으로 종양에 침윤된 총 면역세포와 CD4+ T세포, CD8+ T세포의 수를 증가시켰다. 따라서 mono-mIL-12-Fc는 bi-mIL-12-Fc보다 종양에 침윤된 CD4+ T세포, CD8+ T세포의 수를 유의적으로 증가시켜 종양형성 억제효과가 높았음을 확인하였다. As a result, compared with the control group, bi-mIL-12-Fc and mono-mIL-12-Fc showed the number of total immune cells, CD4 + T cells, and CD8 + T cells infiltrating tumors in tumor-transplanted mice. It can be confirmed that increased. Mono-mIL-12-Fc increased the number of total immune cells, CD4 + T cells, and CD8 + T cells infiltrated tumors more significantly than bi-mIL-12-Fc. Therefore, mono-mIL-12-Fc significantly increased the number of CD4 + T cells and CD8 + T cells infiltrating tumors than bi-mIL-12-Fc.
실시예 17: Mono-mIL-12-Fc의 생체 내 면역세포의 사이토카인 분비 및 세포독성증진 기능 평가Example 17: Evaluation of cytokine secretion and cytotoxicity function of mono-mIL-12-Fc in vivo immune cells
IL-12는 T 세포와 NK 세포의 IFN-g 분비를 증가시켜 암세포의 성장을 억제시키는 것으로 알려져 있다 (Trinchieri, 2003). 또한 IL-12는 세포독성 T세포와 자연살해세포의 암세포에 대한 직접적인 세포독성효과를 증진시켜 항암효과를 나타낸다. 따라서 Mono-IL-12-Fc의 높은 항암효과가 종양을 이식한 마우스의 혈중 IFN-g 농도 증가와 세포독성 T세포 및 자연살해세포의 암세포에 대한 직접적인 세포독성효과 증진에 의한 것인지를 분석하였다. IL-12 is known to inhibit the growth of cancer cells by increasing IFN-g secretion of T cells and NK cells (Trinchieri, 2003). IL-12 also has an anticancer effect by enhancing direct cytotoxic effects on cancer cells of cytotoxic T cells and natural killer cells. Therefore, we analyzed whether the anti-cancer effect of Mono-IL-12-Fc was due to the increase of serum IFN-g concentration and the direct cytotoxic effect of cytotoxic T cells and natural killer cells on cancer cells.
도 23(A)는 상기 도 21(A)의 마지막 투여 24시간 후에 마우스의 얼굴 정맥에서 채혈하여 분리한 혈청내의 IFN-g 농도를 ELISA로 측정한 결과이다. Figure 23 (A) is the result of measuring the IFN-g concentration in the serum isolated by collecting blood from the facial vein of the mouse 24 hours after the last administration of Figure 21 (A) by ELISA.
구체적으로는, 도 20(A)에서의 마지막 mIL-12-Fc 융합단백질 투여 24시간 뒤, 마우스의 얼굴 정맥에서 채혈하였다. 혈액은 2시간 동안 실온에 방치 하여 혈액응고를 유도한 후 8000 rpm에서 10분간 원심 분리하여 상층의 혈청을 회수하였다. 혈청 내 IFN-γ 농도를 측정하기 위하여 ELISA용 96-웰 플레이트(Thermo Fisher Scientific)에 마우스 IFN-γ 포획 항체를 12시간 동안 코팅한 후, PBST (PBS with 0.1% Tween-20)으로 세척 후, 1% BSA (PBS with 1% bovine serum albumin)를 넣고 1시간 동안 실온에서 blocking하였다. PBST (PBS with 0.1% Tween-20)으로 세척 후, 혈청을 1% BSA로 10배 희석하여 실온에서 2시간 동안 반응시켰다. PBST (PBS with 0.1% Tween-20)으로 세척 후, biotin이 결합된 마우스 IFN-γ 검출 항체(Thermo Fisher Scientific)를 실온에서 1시간 동안 결합시켰다. PBST (PBS with 0.1% Tween-20)으로 세척 후, avidin이 결합된 HRP (Thermo Fisher Scientific)를 실온에서 30분간 결합시킨 다음 PBST (PBS with 0.1% Tween-20)로 세척 후, TMB (Sigma-Aldrich Korea)기질을 넣어 마이크로플레이트 리더를 이용하여 450 nm에서의 흡광도를 측정하였다. 도 23(A)에 나타낸 바와 같이 bi-mIL-12-Fc를 투여한 마우스의 혈청 내 IFN-γ 농도는 대조군에 비해 증가되지 않았다. 그러나 1 μg IL-12와 같은 몰 농도까지 mono-mIL-12-Fc를 투여한 마우스의 혈청 내 IFN-γ 농도는 대조군에 비해 농도의존적으로 증가됨을 관찰하였고 mono-mIL-12-Fc의 종양생성 억제효과는 일부 암세포 증식억제 효과를 가지는 것으로 알려진 IFN-γ의 분비를 증가시켰기 때문임을 확인하였다. Specifically, blood was collected from the facial vein of the mouse 24 hours after the last mIL-12-Fc fusion protein administration in FIG. 20 (A). The blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer. To measure serum IFN-γ concentration, a mouse IFN-γ capture antibody was coated in a 96-well plate for ELISA (Thermo Fisher Scientific) for 12 hours, followed by washing with PBST (PBS with 0.1% Tween-20). 1% BSA (PBS with 1% bovine serum albumin) was added and blocked for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), the serum was diluted 10-fold with 1% BSA and reacted at room temperature for 2 hours. After washing with PBST (PBS with 0.1% Tween-20), biotin-coupled mouse IFN-γ detection antibody (Thermo Fisher Scientific) was bound for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), avidin-bound HRP (Thermo Fisher Scientific) was bound for 30 minutes at room temperature, followed by washing with PBST (PBS with 0.1% Tween-20), followed by TMB (Sigma- Aldrich Korea) was used to measure the absorbance at 450 nm using a microplate reader. As shown in FIG. 23 (A), serum IFN-γ levels of mice administered bi-mIL-12-Fc were not increased compared to the control group. However, the concentration of IFN-γ in the serum of mice treated with mono-mIL-12-Fc up to a molar concentration such as 1 μg IL-12 was found to be increased in a concentration-dependent manner compared with the control group. Tumor formation of mono-mIL-12-Fc was observed. It was confirmed that the inhibitory effect was due to increased secretion of IFN-γ, which is known to have some cancer cell proliferation inhibitory effects.
종양을 이식한 마우스에서 bi-mIL-12-Fc가 NK세포와 T세포에 의한 IFN-γ의 분비를 유도하는 능력이 낮아서 도 23(A)에서 bi-mIL-12-Fc를 투여한 마우스의 혈중 IFN-γ의 농도가 낮았는지를 규명하기 위해 mono-mIL-12-Fc와 bi-mIL-12-Fc를 한 번만 투여한 후 시간 별로 혈중 IFN-γ의 농도를 측정하였다. In mice transplanted with tumors, bi-mIL-12-Fc was not capable of inducing IFN-γ secretion by NK cells and T cells, and thus, bi-mIL-12-Fc was administered in FIG. 23 (A). To determine if the level of IFN-γ in the blood was low, mono-mIL-12-Fc and bi-mIL-12-Fc were administered only once, and then blood IFN-γ levels were measured over time.
도 23(B)는 CT26HER2 / Neu 대장암 세포를 이식한 Balb/c 마우스에 bi-mIL-12-Fc와 mono-mIL-12-Fc를 1회 복강 투여한 후 시간 별로 혈청내의 IFN-γ 농도를 ELISA로 측정한 결과이다. FIG. 23 (B) shows IFN-γ in serum by time after intraperitoneal administration of bi-mIL-12-Fc and mono-mIL-12-Fc to Balb / c mice transplanted with CT26 HER2 / Ne colorectal cancer cells. Concentration was measured by ELISA.
구체적으로는, CT26HER2 / Neu 대장암 세포를 이식한 Balb/c 마우스에서 종양의 크기가 300 mm3일 때 1 ㎍ rmIL-12와 같은 몰 농도의 bi-mIL-12-Fc와 mono-mIL-12-Fc를 복강 투여하였다. 1일, 3일 및 5일 후에 마우스의 얼굴 정맥에서 채혈하였다. 혈액은 2시간 동안 실온에 방치 하여 혈액응고를 유도한 후 8000 rpm에서 10분간 원심 분리하여 상층의 혈청을 회수하였다. 혈청 내 IFN-γ 농도를 측정하기 위하여 ELISA용 96-웰 플레이트(Thermo Fisher Scientific)에 마우스 IFN-γ 포획 항체를 12시간 동안 코팅한 후, PBST (PBS with 0.1% Tween-20)으로 세척 후, 1% BSA (PBS with 1% bovine serum albumin)를 넣고 1시간 동안 실온에서 blocking하였다. PBST (PBS with 0.1% Tween-20)으로 세척 후, 혈청을 1% BSA로 10배 희석하여 실온에서 2시간 동안 반응시켰다. PBST (PBS with 0.1% Tween-20)으로 세척 후, biotin이 결합된 마우스 IFN-γ 검출 항체(Thermo Fisher Scientific)를 실온에서 1시간 동안 결합시켰다. PBST (PBS with 0.1% Tween-20)으로 세척 후, avidin이 결합된 HRP (Thermo Fisher Scientific)를 실온에서 30분간 결합시킨 다음 PBST (PBS with 0.1% Tween-20)로 세척 후, TMB (Sigma-Aldrich Korea)기질을 넣어 마이크로플레이트 리더를 이용하여 450 nm에서의 흡광도를 측정하였다. 도 23(B)에 나타낸 바와 같이 종양을 이식한 마우스에서 bi-mIL-12-Fc 투여군은 5일째까지 mono-mIL-12-Fc 투여군과 비슷한 혈중 IFN-γ의 농도를 보여 bi-mIL-12-Fc가 효과세포에서 IFN-γ의 분비를 유도하는 능력에 대한 본질적 결함은 없는 것으로 나타났다. Specifically, in a Balb / c mouse transplanted with CT26 HER2 / Neu colorectal cancer cells, when the tumor size was 300 mm 3 , bi-mIL-12-Fc and mono-mIL- at a molar concentration of 1 μg rmIL-12 12-Fc was administered intraperitoneally. Blood was collected from the facial veins of mice after 1, 3 and 5 days. The blood was left at room temperature for 2 hours to induce coagulation, and then centrifuged at 8000 rpm for 10 minutes to recover serum from the upper layer. To measure serum IFN-γ concentration, a mouse IFN-γ capture antibody was coated in a 96-well plate for ELISA (Thermo Fisher Scientific) for 12 hours, followed by washing with PBST (PBS with 0.1% Tween-20). 1% BSA (PBS with 1% bovine serum albumin) was added and blocked for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), the serum was diluted 10-fold with 1% BSA and reacted at room temperature for 2 hours. After washing with PBST (PBS with 0.1% Tween-20), biotin-coupled mouse IFN-γ detection antibody (Thermo Fisher Scientific) was bound for 1 hour at room temperature. After washing with PBST (PBS with 0.1% Tween-20), avidin-bound HRP (Thermo Fisher Scientific) was bound for 30 minutes at room temperature, followed by washing with PBST (PBS with 0.1% Tween-20), followed by TMB (Sigma- Aldrich Korea) was used to measure the absorbance at 450 nm using a microplate reader. As shown in FIG. 23 (B), the bi-mIL-12-Fc-administered group showed a similar concentration of IFN-γ in the blood to the mono-mIL-12-Fc-treated group until day 5 in the tumor-grafted mice. There was no inherent deficiency in the ability of -Fc to induce the secretion of IFN-γ in effector cells.
도 23(C)는 상기 도 21(A)의 마지막 투여 3일 후에 마우스를 치사하여 비장에서 분리한 세포독성 T 세포의 CT26HER2 / Neu 암세포에 대한 세포독성효과를 측정한 그래프이다. Figure 23 (C) is a graph measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of cytotoxic T cells isolated from the spleen by killing the mouse 3 days after the last administration of Figure 21 (A).
구체적으로는, 도 21(A)에서의 마지막 사이토카인 투여 후 72시간 뒤, 마우스를 치사하고 비장을 적출한 다음 70 마이크론 메쉬를 넣은 60 mm 접시에 비장과 PBS를 넣고 분쇄하였다. 원심분리 하여 얻은 세포에 적혈구 용혈 완충액을 넣어 적혈구를 용혈 시킨 후 PBS로 세척하고 CD3를 인지하는 APC가 결합된 항체 (Thermo Fisher Scientific), CD8을 인지하는 PE가 결합된 항체 (Thermo Fisher Scientific)를 넣어 4℃에서 30 분간 반응시켰다. PBS로 세척 후, 세포독성 T 세포 (CD3+CD8+)를 FACS Aria III (BD biosciences, Korea)로 분리하였다. 세포독성 T 세포의 표적세포인 CT26HER2 / Neu 암세포에 대한 세포독성효과를 측정하기 위하여 CT26HER2 / Neu 암세포는 calcein AM (Thermo Fisher Scientific Inc., 10 μM)으로 염색하였다. CT26HER2/Neu 암세포(2×106)를 2 ml의 DPBS에 현탁 시킨 후 calcein AM (10 mM) 2 μl를 넣고 섞은 다음 5 % CO2, 37℃ 조건에서 45 분간 반응시켰다. 10% FBS가 첨가된 RPMI1640을 10 ml 넣어 3번 세척한 다음 96웰 플레이트에 각 웰 당 2×104 개의 세포를 넣고 세포독성 T 세포 (1×105/100 μl/well)를 각각 넣어 5 % CO2, 37℃ 배양기에서 4시간 동안 배양하였다. 녹색 형광을 내는 살아있는 CT26HER2 / Neu 암세포와 녹색 형광을 내지 않는 죽은 CT26HER2 / Neu 암세포를 유세포 분석기로 분석하여 세포독성 T세포의 세포독성 효과를 백분율로 표시하였다. Mono-mIL-12-Fc를 투여한 종양이식 마우스에서 분리한 세포독성 T세포는 bi-mIL-12-Fc 를 투여한 종양이식 마우스에서 분리한 세포독성 T세포나 대조군에서 분리한 세포독성 T세포보다 표적세포인 CT26HER2/Neu 암세포에 대한 세포독성효과가 높았으며 mono-mIL-12-Fc의 종양생성 억제효과는 일부 세포독성 T세포에 의한 암세포의 직접적인 세포독성효과임을 확인하였다.Specifically, 72 hours after the last cytokine administration in FIG. 21 (A), mice were killed and spleens were removed and spleens and PBS were put into a 60 mm dish containing 70 micron mesh and ground. The cells obtained by centrifugation were lysed with red blood cell hemolysis buffer, and then washed with PBS and washed with PBS, an APC conjugated antibody that recognizes CD3 (Thermo Fisher Scientific), and a PE conjugated antibody that recognizes CD8 (Thermo Fisher Scientific). The reaction was carried out for 30 minutes at 4 ℃. After washing with PBS, cytotoxic T cells (CD3 + CD8 + ) were isolated by FACS Aria III (BD biosciences, Korea). To determine the cytotoxic effect on target cells in CT26 HER2 / Neu cancer cells of a cytotoxic T cell CT26 HER2 / Neu Cancer cells were stained with calcein AM (Thermo Fisher Scientific Inc., 10 μM). After CT26 HER2 / Neu cancer cells (2 × 10 6 ) were suspended in 2 ml of DPBS, 2 μl of calcein AM (10 mM) was added and mixed, followed by reaction for 45 minutes at 5% CO 2 and 37 ° C. A RPMI1640 with 10% FBS was added 10 ml into three times, washed, and then put into a 2 × 10 4 cells per well in a 96-well plate cytotoxic T cells (1 × 10 5/100 μl / well) into each 5 % CO 2 , incubated for 4 hours at 37 ℃ incubator. Live CT26 HER2 / Neu cancer cells that fluoresce green and dead CT26 HER2 / Neu cancer cells that do not fluoresce green were analyzed by flow cytometry to express the cytotoxic effect of cytotoxic T cells in percentage. Cytotoxic T cells isolated from tumor-grafted mice treated with Mono-mIL-12-Fc were cytotoxic T cells isolated from tumor-grafted mice administered bi-mIL-12-Fc or cytotoxic T cells isolated from the control group. The cytotoxic effect on CT26 HER2 / Neu cancer cells, which are more target cells, was higher, and the tumor suppression effect of mono-mIL-12-Fc was a direct cytotoxic effect of cancer cells by some cytotoxic T cells.
도 23(D)는 종양이식 마우스에서 mono-IL-12-Fc의 투여에 의해 증진된 세포독성 T 세포의 세포독성효과가 암 항원특이적인 것인지를 측정하기 위해 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에서 분리한 세포독성 T 세포의 세포독성효과를 종양항원을 발현하는 CT26HER2 / Neu 암세포와 발현하지 않는 4T1세포를 이용하여 측정한 결과이다. Figure 23 (D) is the third dose of Figure 21 (A) to determine whether the cytotoxic effect of cytotoxic T cells enhanced by administration of mono-IL-12-Fc in tumor-grafted mice is cancer antigen specific. Three days later, the cytotoxic effect of cytotoxic T cells isolated from the spleen by lethal mice was measured using CT26 HER2 / Neu cancer cells expressing tumor antigen and 4T1 cells not expressing tumor antigen.
구체적으로는, 도 20(A)에서 mono-IL-12-Fc의 3회 투여 72시간 뒤, 마우스를 치사하고 비장을 적출한 다음 70 마이크론 메쉬를 넣은 60 mm 접시에 비장과 PBS를 넣고 비장을 분쇄하였다. 세포독성 T 세포의 표적세포인 CT26HER2 / Neu 암세포와 세포독성 T세포의 표적세포가 아닌 4T1에 대한 세포독성효과를 측정하기 위하여 도 21(C)와 같은 방법으로 CT26HER2 / Neu 암세포와 4T1 암세포를 calcein AM (Thermo Fisher Scientific Inc., 10 μM)으로 염색하였다. 10% FBS가 첨가된 RPMI1640을 10 ml 넣어 3번 세척한 다음 96웰 플레이트에 각 웰 당 2×104 개의 세포를 넣고 세포독성 T 세포 (1×105/100 μl/well)를 각각 넣어 5 % CO2, 37℃ 배양기에서 4시간 동안 배양하였다. 녹색 형광을 내는 살아있는 CT26HER2 / Neu 암세포 또는 4T1 암세포와 녹색 형광을 내지 않는 죽은 CT26HER2 / Neu 암세포 또는 4T1 암세포를 유세포 분석기로 분석하여 세포독성 T 세포의 세포독성 효과를 백분율로 표시하였다. 분석결과 mono-mIL-12-Fc의 투여에 의해 증진된 세포독성 T세포의 세포독성 효과는 표적세포 특이적임을 확인하였다.Specifically, 72 hours after the three-time administration of mono-IL-12-Fc in FIG. 20 (A), mice were killed and spleens were removed, and spleen and PBS were placed in a 60 mm dish containing 70 micron mesh. Pulverized. Cytotoxic T target cells in CT26 HER2 / Neu tumor cells and cytotoxic T Target cells 4T1 cytotoxic effect in the same way as FIG. 21 (C) to measure CT26 HER2 / Neu cancer with 4T1 tumor cells to a non-cellular in the cell Were stained with calcein AM (Thermo Fisher Scientific Inc., 10 μΜ). The RPMI1640 with 10% FBS was added 10 ml into three times, washed, and then put into a 2 × 10 4 cells per well in a 96-well plate cytotoxic T cells (1 × 10 5/100 μl / well) into each 5 % CO 2 , incubated for 4 hours at 37 ℃ incubator. Green Fluorescent Live CT26 HER2 / Neu Dead CT26 HER2 / Neu with no green fluorescence with cancer cells or 4T1 cancer cells Cancer cells or 4T1 cancer cells were analyzed by flow cytometry to express the cytotoxic effects of cytotoxic T cells in percentage. As a result, it was confirmed that the cytotoxic effect of the cytotoxic T cells enhanced by the administration of mono-mIL-12-Fc was target cell specific.
도 23(E)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에서 분리한 자연살해세포의 CT26HER2/Neu 암세포에 대한 세포독성 효과를 측정한 결과이다. Figure 23 (E) is a result of measuring the cytotoxic effect on CT26 HER2 / Neu cancer cells of natural killer cells isolated from the spleen by killing the mouse 3 days after the third administration of Figure 21 (A).
구체적으로는, 도 21(A)에서의 3번째 투여 3일 후에, 마우스를 치사하고 비장을 적출한 다음 70 마이크론 메쉬를 넣은 60 mm 접시에 비장과 PBS를 넣고 분쇄하였다. 원심분리 하여 얻은 세포에 적혈구 용혈 완충액을 넣어 적혈구를 용혈 시킨 후 PBS로 세척하고 CD3를 인지하는 APC가 결합된 항체 (Thermo Fisher Scientific), CD49b을 인지하는 PE가 결합된 항체 (Thermo Fisher Scientific)를 넣어 4℃에서 30 분간 반응시켰다. PBS로 세척 후, 자연살해 세포 (CD3-CD49b+)를 FACS Aria III (BD biosciences, Korea)로 분리하였다. 자연살해 세포의 표적세포인 CT26HER2 / Neu 암세포에 대한 세포독성효과를 측정하기 위하여 CT26HER2 / Neu 암세포는 calcein AM (Thermo Fisher Scientific Inc., 10 μM)으로 염색하였다. CT26HER2 / Neu 암세포(2× 106)를 2 ml의 DPBS에 현탁 시킨 후 calcein AM (10 mM) 2 μl를 넣고 섞은 다음 5 % CO2, 37℃ 조건에서 45 분간 반응시켰다. 10% FBS가 첨가된 RPMI1640을 10 ml 넣어 3번 세척한 다음 96웰 플레이트에 각 웰 당 2×104 개의 세포를 넣고 자연살해 세포 (1×105/100 μl/well)를 각각 넣어 5 % CO2, 37℃ 배양기에서 4시간 동안 배양하였다. 녹색 형광을 내는 살아있는 CT26HER2 / Neu 암세포와 녹색 형광을 내지 않는 죽은 CT26HER2 / Neu 암세포를 유세포 분석기로 분석하여 자연살해 세포의 세포독성 효과를 백분율로 표시하였다. Mono-mIL-12-Fc를 투여한 종양이식 마우스에서 분리한 자연살해 세포는 bi-mIL-12-Fc 를 투여한 종양이식 마우스에서 분리한 자연살해 세포나 대조군에서 분리한 세포독성 T세포보다 표적세포인 CT26HER2 / Neu 암세포에 대한 세포독성효과가 높았으며 mono-mIL-12-Fc의 종양생성 억제효과는 일부 자연살해 세포에 의한 암세포의 직접적인 세포독성효과임을 확인하였다.Specifically, 3 days after the third dose in FIG. 21 (A), mice were killed and spleens were removed, and spleens and PBS were put in a 60 mm dish containing 70 micron mesh and ground. The cells obtained by centrifugation were lysed with red blood cell hemolysis buffer, washed with PBS, and washed with PBS, and APC-bound antibody (Thermo Fisher Scientific) that recognizes CD3 and PE-bound antibody (Thermo Fisher Scientific) that recognizes CD49b. The reaction was carried out for 30 minutes at 4 ℃. After washing with PBS, natural killer cells (CD3 - CD49b + ) were separated by FACS Aria III (BD biosciences, Korea). CT26 HER2 / Neu Cancer cells were stained with calcein AM (Thermo Fisher Scientific Inc., 10 μM) to measure the cytotoxicity of the target cells in CT26 HER2 / Neu cancer cells of NK cells. CT26 HER2 / Neu cancer cells (2 × 10 6 ) were suspended in 2 ml of DPBS, 2 μl of calcein AM (10 mM) were mixed and allowed to react for 45 minutes at 5% CO 2 , 37 ° C. A RPMI1640 with 10% FBS was added 10 ml into three by three times, and then each well in a 96-well plate into a 2 × 10 4 cells into natural killer cells (1 × 10 5/100 μl / well) , respectively 5% Incubated for 4 hours in a CO 2 , 37 ℃ incubator. Live CT26 HER2 / Neu cancer cells that fluoresce green and dead CT26 HER2 / Neu cancer cells that do not fluoresce green were analyzed by flow cytometry to express the cytotoxic effect of natural killer cells in percentage. Natural killer cells isolated from tumor-grafted mice treated with Mono-mIL-12-Fc were more targeted than natural killer cells isolated from bi-mIL-12-Fc-treated mice or with cytotoxic T cells isolated from the control group. The cytotoxic effect on CT26 HER2 / Neu cancer cells, which are the cells, was high, and the tumor suppression effect of mono-mIL-12-Fc was a direct cytotoxic effect of cancer cells by some natural killer cells.
실시예 18: 생체 내에서 mono-mIL-12-Fc의 효과 CD8+ T세포 및 기억 CD8+ T세포 형성능 평가Example 18 Effect of mono-mIL-12-Fc in Vivo Evaluation of CD8 + T Cells and Memory CD8 + T Cell Formability
종양을 이식한 마우스에서 적응면역의 생성은 효과 기억 CD8+ T세포 및 기억 CD8+ T세포 형성여부로 평가된다. Mono-mIL-12-Fc에 의한 종양제거효과가 효과 기억 CD8+ T세포 및 기억 CD8+ T세포 형성에 의한 것인지를 측정하였다. The generation of adaptive immunity in tumor-grafted mice is assessed by effector memory CD8 + T cells and memory CD8 + T cell formation. Tumor elimination effect by Mono-mIL-12-Fc was determined by the effect memory CD8 + T cells and memory CD8 + T cell formation.
도 24(A), 24(B) 및 24(C)는 각각 종양을 가진 마우스에 mono-mIL-12-Fc를 투여 하였을 때 생성된 효과 CD8+ T세포, 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 측정한 결과이다. 24 (A), 24 (B) and 24 (C) show effect CD8 + T cells, effect memory CD8 + T cells and memory CD8 generated when mono-mIL-12-Fc was administered to mice with tumors, respectively. + The result of measuring the number of T cells.
구체적으로, 21(A)과 같이 처리한 후 종양이식 34일째에 마우스의 비장을 적출하여 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml로 세척한 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈시킨 다음 PBS로 세척하여 비장세포 혼탁액을 준비하고 헤모사이토미터로 세포의 수를 계수하였다. 비장세포를 PE-cy5, PE, FITC 또는 APC가 결합된 CD3, CD8, CD62L 및 IL-7 receptor(IL-7R)를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 CD3+CD8+CD62LlowIL-7Rlow 세포군, CD3+CD8+CD62LlowIL-7Rhi 세포군 및 CD3+CD8+CD62LhiIL-7Rhi 세포군을 각각 효과 CD8+ T세포, 효과 기억 CD8+ T세포 및 기억 CD8+ T세포로 정의하여 전체 비장세포에 대한 비율을 구하고 헤모사이토미터로 계수된 세포의 수를 곱하여 mono-mIL-12-Fc 투여 후 증가된 효과 CD8+ T세포, 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 분석하였다. 그 결과, 대조군에 비해 mono-mIL-12-Fc는 종양이 이식된 마우스에서 농도의존적으로 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 증가시켰음을 확인할 수 있었다. 반면, bi-mIL-12-Fc는 0.5 μg IL-12와 같은 몰 농도를 투여한 군에서만 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 증가시켰고, 1 μg IL-12와 같은 몰 농도를 투여한 군에서는 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 증가시키지 못했다. 따라서 mono-mIL-12-Fc는 bi-mIL-12-Fc보다 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 유의적으로 증가시켜 종양제거효과가 높았음을 확인하였다. Specifically, after treatment as 21 (A), the mouse spleen was extracted at 34 days after tumor transplantation, pulverized using a wire mesh in Petri dishes, washed with 10 ml of medium containing 2% FBS, and then erythrocyte hemolysis buffer 1 ml of red blood cell lysis buffer was added to erythrocytes, and then washed with PBS to prepare a splenocyte suspension, and the number of cells was counted with a hemocytometer. Splenocytes were stained at 4 ° C. for 30 minutes with antibodies recognizing CD3, CD8, CD62L and IL-7 receptor (IL-7R) bound to PE-cy5, PE, FITC or APC, and cold stained with PBS (pH 7.4). After washing, they were analyzed by flow cytometry, FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to determine CD3 + CD8 + CD62L low IL-7R low Cell groups, CD3 + CD8 + CD62L low IL-7R hi cell populations and CD3 + CD8 + CD62L hi IL-7R hi cell populations were defined as effect CD8 + T cells, effect memory CD8 + T cells and memory CD8 + T cells, respectively. The number of effect CD8 + T cells, effect memory CD8 + T cells, and memory CD8 + T cells after mono-mIL-12-Fc administration was analyzed by multiplying the number of cells counted by hemocytometer. . As a result, it was confirmed that mono-mIL-12-Fc increased the number of effect memory CD8 + T cells and memory CD8 + T cells in a tumor-grafted mouse compared with the control group. On the other hand, bi-mIL-12-Fc increased the number of effect memory CD8 + T cells and memory CD8 + T cells only in the group administered with a molar concentration such as 0.5 μg IL-12, and the molar like 1 μg IL-12. The concentration group did not increase the number of effect memory CD8 + T cells and memory CD8 + T cells. Therefore, mono-mIL-12-Fc significantly increased the number of effect memory CD8 + T cells and memory CD8 + T cells than bi-mIL-12-Fc.
도 24(D)는 상기 도 21(A)에서 1㎍ mono-IL-12-Fc의 투여 120일 후에 생존한 마우스에 CT26HER2 / Neu 암세포를 재이식하여 마우스의 종양 부피의 변화를 측정한 결과이다.Figure 24 (D) is in the mice survived 120 days after the administration in 21 (A) to FIG 1㎍ mono-IL-12-Fc CT26 HER2 / Neu Cancer cells are transplanted to measure the change in the tumor volume of the mouse.
구체적으로, 도 21(A)에서 1㎍ mono-IL-12-Fc의 투여 후 생존한 마우스와 주령이 일치하는 암컷 Balb/c 마우스(NARA Biotech, Korea)에 마지막 1㎍ mono-IL-12-Fc를 투여한지 120일째에 마우스의 털을 면도기로 제거하고 CT26HER2 / Neu 세포 (1×106 세포/마우스)를 150 μL PBS에 희석하여 마우스 피하에 이식하였다. 그 후, 추가적인 1㎍ mono-IL-12-Fc의 투여 없이 종양을 일주일에 2회씩 측정하고, 종양의 부피 (V)는 V=길이x폭2/2 로 계산하였다. 그 결과, 대조군에 비해 1㎍ mono-mIL-12-Fc의 투여 후 살아남은 마우스는 11일째부터 종양이 제거된 것을 확인할 수 있었다. 따라서 종양이식 마우스에 mono-mIL-12-Fc를 투여하면 효과 기억 CD8+ T세포 및 기억 CD8+ T세포를 생성하여 다시 같은 종양을 재이식하더라도 종양을 제거하는 것으로 확인되었다. Specifically, in FIG. 21 (A), the last 1 μg mono-IL-12- was applied to female Balb / c mice (NARA Biotech, Korea) that matched the age of the surviving mice after administration of 1 μg mono-IL-12-Fc. On day 120 after Fc administration, the hair of the mouse was removed with a razor and CT26 HER2 / Neu cells (1 × 10 6 cells / mouse) were transplanted into the mouse subcutaneously diluted in 150 μL PBS. Thereafter, additional 1㎍ mono-IL-12-Fc of the measurement without the administration of the tumor twice a week, and the volume (V) of the tumor was calculated as V = L x W 2/2. As a result, compared with the control group, the mice survived after the administration of 1 μg mono-mIL-12-Fc was confirmed that the tumor was removed from day 11. Therefore, it was confirmed that administration of mono-mIL-12-Fc to tumor-grafted mice produces effect memory CD8 + T cells and memory CD8 + T cells, even if the same tumor is transplanted again.
실시예 19: 생체 내에서 mono-mIL-12-Fc의 기억전구 효과 CD8+ T세포 형성능력 평가Example 19 Evaluation of Memory Bulb Effect of Mono-mIL-12-Fc in Vivo CD8 + T Cell Formation Capacity
실시예 16과 실시예18에서 bi-mIL-12-Fc는 mono-mIL-12-Fc에 비해 종양을 이식한 마우스에서 비장 CD8+ T세포의 수, 효과 기억 CD8+ T세포의 수 및 중심 기억 CD8+ T세포의 수를 증가시키는 효과가 낮음을 관찰하였다. 활성화된 CD8+ T세포들이 종양세포를 직접 파괴하는 과정(effector phase)이 지나면 효과 CD8+ T세포는 일부만 기억전구효과세포(memory precursor effector cells, MPEC)를 거쳐 기억 CD8+ T세포로 분화되고 대부분 단명효과세포(short-lived effector cells, SLEC)로서 죽는 것으로 보고되어 있다. 따라서 bi-mIL-12-Fc의 투여로 활성화된 CD8+ T세포들이 단명효과세포로 분화되었기 때문에 기억 CD8+ T세포의 수가 적게 생성되어 종양을 제거하지 못하였는지를 측정하였다. In Examples 16 and 18, bi-mIL-12-Fc was compared with mono-mIL-12-Fc in the number of spleen CD8 + T cells, the number of effect memory CD8 + T cells, and central memory CD8 in mice transplanted with tumors. The effect of increasing the number of + T cells was observed to be low. After activated CD8 + T cells directly destroy tumor cells, the effect CD8 + T cells are partially differentiated into memory CD8 + T cells through memory precursor effector cells (MPEC) and are mostly short-lived. It has been reported to die as short-lived effector cells (SLEC). Thus, the CD8 + T cell activation by the administration of a bi-mIL-12-Fc are generated less the number of memory CD8 + T cells differentiate into short-lived because the effect was determined whether the cells do not remove the tumor.
도 24(E)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에 존재하는 CD8+ T세포 중 기억전구효과세포(KLRG1-IL-7R+)와 단명효과세포(KLRG1+IL-7R-)의 비율을 분석한 결과이다. FIG. 24 (E) shows memory precursor effect cells (KLRG1 - IL-7R + ) and short-lived effect cells (KLRG1) among CD8 + T cells present in the spleen after 3 days of the third administration of FIG. 21 (A). + IL-7R - an analysis of the rate of).
구체적으로, 21(A)과 같이 처리한 후 종양이식 24일째에 마우스의 비장을 적출한 후, 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml를 넣어 세척하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈시킨 다음 PBS로 세척하여 세포 혼탁액을 준비하였다. 비장세포를 PE-cy5, PE, FITC 또는 APC가 결합된 CD3, CD8, KLRG1 및 IL-7 receptor(IL-7R)를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 CD3+CD8+KLRG1-IL-7R+ 세포군과 CD3+CD8+KLRG1_+IL-7R- 세포군을 각각 기억전구효과세포와 단명효과세포로 정의하여 전체 비장세포에 대한 비율을 분석하였다. 그 결과, 대조군에 비해 mono-mIL-12-Fc는 종양이 이식된 마우스에서 농도의존적으로 기억전구효과세포의 비율을 증가시켰음을 확인할 수 있었다. 반면, bi-mIL-12-Fc의 투여는 대조군에 비해 기억전구효과세포의 비율은 증가시키지 못했고 오히려 단명효과 세포의 수를 증가시켰다. 따라서 mono-mIL-12-Fc는 bi-mIL-12-Fc보다 기억전구세포의 생성을 촉진하여 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 유의적으로 증가시켜 종양제거효과가 높았음을 확인하였다. Specifically, after treatment as 21 (A), the mouse spleen was extracted on the 24th day of tumor transplantation, and then pulverized using a wire mesh in Petri dishes and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Splenocytes were stained at 4 ° C. for 30 minutes with antibodies recognizing CD3, CD8, KLRG1 and IL-7 receptor (IL-7R) bound to PE-cy5, PE, FITC or APC, and cold stained with PBS (pH 7.4). After washing, they were analyzed by flow cytometry, FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to define CD3 + CD8 + KLRG1 - IL-7R + cell group and CD3 + CD8 + KLRG1_ + IL-7R - cell group as memory precursor cells and short-lived effect cells, respectively. The ratio for splenocytes was analyzed. As a result, it was confirmed that mono-mIL-12-Fc increased the percentage of memory precursor effector cells in a concentration-dependent manner compared with the control group. On the other hand, the administration of bi-mIL-12-Fc did not increase the proportion of memory precursor cells compared to the control group, but rather increased the number of short-lived cells. Therefore, mono-mIL-12-Fc promoted the production of memory precursor cells more than bi-mIL-12-Fc and significantly increased the number of effect memory CD8 + T cells and memory CD8 + T cells, resulting in a higher tumor removal effect. Confirmed.
실시예 20: Mono-mIL-12-Fc가 기억세포 분화유도에 관계된 전사인자의 발현에 미치는 영향 평가Example 20: Evaluation of the Effect of Mono-mIL-12-Fc on the Expression of Transcription Factors Related to Memory Cell Differentiation Induction
CD8+ T세포에 높은 농도의 IL-12를 투여하거나 2일이상 지속적으로 IL-12를 주면서 활성화시키면 CD8+ T세포를 단명효과세포로 분화시키는 전사인자인 T-bet의 발현이 높아지고 기억전구효과세포로 분화시키는 전사인자인 eomesodermin(Eomes)의 발현이 감소되는 것으로 보고되어 있다. 따라서 mono-mIL-12-Fc와 bi-mIL-12-Fc가 CD8+ T세포에서 T-bet과 Eomes의 발현을 다르게 조절하여 단명효과세포로 분화되는 비율이 달라졌는지를 측정하였다. Administration of high concentrations of IL-12 to CD8 + T cells or continuous activation of IL-12 for more than two days increases expression of T-bet, a transcription factor that differentiates CD8 + T cells into short-lived effect cells, Has been reported to reduce the expression of eomesodermin (Eomes), a transcription factor that differentiates to. Therefore, we measured whether mono-mIL-12-Fc and bi-mIL-12-Fc differentiated T-bet and Eomes in CD8 + T cells and differentiated them into short-lived effect cells.
도 25(A)와 25(B)는 상기 도 21(A)의 3번째 투여 3일 후에 마우스를 치사하여 비장에서 기억세포의 분화를 억제하는 T-bet의 발현이 높은 CD8+ T세포의 비율과 기억세포의 분화를 촉진하는 Eomes의 발현이 높고 T-bet의 발현이 낮은 CD8+ T세포의 비율을 유세포 분석기로 측정하여 분석한 결과이다. 25 (A) and 25 (B) show the percentage of CD8 + T cells with high expression of T-bet that inhibits the differentiation of memory cells in the spleen by killing mice 3 days after the third dose of FIG. 21 (A). This is the result of measuring the ratio of CD8 + T cells with high expression of Eomes and low expression of T-bet that promotes the differentiation of memory cells by flow cytometry.
구체적으로, 21(A)과 같이 처리한 후 종양이식 24일째에 마우스의 비장을 적출한 후, 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml를 넣어 세척하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈 시킨 다음 PBS로 세척하여 세포 혼탁액을 준비하였다. 비장세포를 PE-cy5 또는 FITC가 결합된 CD3와 CD8를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 핵내에 전사인자를 염색하는 시약인 Foxp3/Transcription Factor Staining Buffer Set (Thermo Fisher Scientific)을 이용하여 세포를 고정(fixation)하고 투과(permeabilization)시킨 다음 PE 또는 efluor 660이 결합된 T-bet 또는 Eomes를 인지하는 항체를 넣어 4℃에서 30분간 염색한 다음 투과버퍼(permeabilization buffer)를 넣어 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 CD3+CD8+T-bethigh 세포군과 CD3+CD8+Eomes+T-betlow 세포군의 비율을 분석하였다. 그 결과, 대조군에 비해 mono-mIL-12-Fc는 종양이 이식된 마우스에서 농도의존적으로 CD3+CD8+T-bethigh 세포군의 비율은 감소시키고, CD3+CD8+Eomes+T-betlow 세포군의 비율을 증가시켰음을 확인할 수 있었다. 반면 bi-mIL-12-Fc는 0.5 μg IL-12와 같은 몰 농도를 투여한 군에서만 CD3+CD8+T-bethigh 세포군의 비율은 감소시키고, CD3+CD8+Eomes+T-betlow 세포군의 비율을 증가시켰고, 1 μg IL-12와 같은 몰 농도를 투여한 군에서는 CD3+CD8+T-bethigh 세포군의 비율은 감소시키거나 CD3+CD8+Eomes+T-betlow 세포군의 비율을 증가시키는 효과가 없었다. 따라서 mono-mIL-12-Fc는 bi-mIL-12-Fc보다 CD3+CD8+T-bethigh 세포군의 비율은 감소시키고, CD3+CD8+Eomes+T-betlow 세포군의 비율을 증가시켜 효과 기억 CD8+ T세포 및 기억 CD8+ T세포의 수를 유의적으로 증가시켜 종양제거효과가 높았음을 확인하였다. Specifically, after treatment as 21 (A), the mouse spleen was extracted on the 24th day of tumor transplantation, and then pulverized using a wire mesh in Petri dishes and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Splenocytes were stained with PE-cy5 or FITC-coupled CD3 and CD8 antibodies for 30 minutes at 4 ° C, washed with cold PBS (pH 7.4), and stained with transcription factor Foxp3 / Transcription Factor. The cells were fixed and permeabilized using Staining Buffer Set (Thermo Fisher Scientific), and then stained for 30 minutes at 4 ° C. with an antibody recognizing T-bet or Eomes bound to PE or efluor 660. Permeabilization buffer was added and analyzed by flow cytometry FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to analyze the ratio of CD3 + CD8 + T-bet high cell population and CD3 + CD8 + Eomes + T-bet low cell population. As a result, mono-mIL-12-Fc reduced the proportion of CD3 + CD8 + T-bet high cell populations in a tumor-dependent mouse compared to the control group, and decreased the concentration of CD3 + CD8 + Eomes + T-bet low cells. It was confirmed that the ratio was increased. On the other hand, bi-mIL-12-Fc decreased the proportion of CD3 + CD8 + T-bet high cell population only in the group administered with 0.5 μg IL-12, and CD3 + CD8 + Eomes + T-bet low The percentage of cell population was increased, and in the group administered with a molar concentration such as 1 μg IL-12, the percentage of CD3 + CD8 + T-bet high cell population was decreased or the percentage of CD3 + CD8 + Eomes + T-bet low cell population was decreased. There was no increasing effect. Therefore, mono-mIL-12-Fc reduces the proportion of CD3 + CD8 + T-bet high cell population and increases the ratio of CD3 + CD8 + Eomes + T-bet low cell population than bi-mIL-12-Fc. The number of CD8 + T cells and memory CD8 + T cells was significantly increased to confirm that the tumor removal effect was high.
CD8+ T 세포는 T 세포 수용체 신호와 공동자극신호의 존재 하에서 IL-12와 같은 염증성 사이토카인으로 자극하면 STAT4의 인산화가 증가되고 인산화된 STAT4 (pSTAT4)는 핵 내부로 이동하여 T-bet enhancer에 결합하여 T-bet의 발현을 증가시키는 것으로 알려져 있다. 따라서 1 μg IL-12와 같은 몰 농도의 bi-mIL-12-Fc를 투여하였을 때 CD8+ T세포가 단명화세포로 분화되는 것이 1 μg IL-12와 같은 몰 농도의 bi-mIL-12-Fc를 투여하면 mono-mIL-12-Fc보다 종양을 이식한 마우스의 종양주위 림프절(tumor draining lymph node)인 사타구니림프절에서 CD8+ T세포가 활성화될 때 pSTAT4와 T-bet의 발현을 증가시켰기 때문인지를 측정하였다.CD8 + T cells, when stimulated with inflammatory cytokines such as IL-12 in the presence of T cell receptor and co-stimulatory signals, increase phosphorylation of STAT4, and phosphorylated STAT4 (pSTAT4) migrates inside the nucleus to the T-bet enhancer. It is known to bind to increase expression of T-bet. Therefore, when 8 μg IL-12 at a molar concentration of bi-mIL-12-Fc is administered, CD8 + T cells are differentiated into short-lived cells. Fc administration increased the expression of pSTAT4 and T-bet when CD8 + T cells were activated in the groin lymph nodes, the tumor draining lymph nodes of mice transplanted with tumors than mono-mIL-12-Fc. Cognitive was measured.
도 25(C)는 CT26HER2 / Neu 이식 Balb/c 마우스에서 종양의 크기가 300 mm3일 때 1μg rmIL-12와 같은 몰 농도의 bi-mIL-12-Fc와 mono-mIL-12-Fc의 1회 복강 투여 24시간 후에 사타구니 림프절에서 분리한 CD8+ T 세포에서 인산화된 STAT4의 발현양을 유세포 분석기로 측정한 결과이다. Figure 25 (C) shows the molar concentrations of bi-mIL-12-Fc and mono-mIL-12-Fc at the same molar concentration of 1 μg rmIL-12 when the tumor size was 300 mm 3 in CT26 HER2 / Ne transplanted Balb / c mice. The expression level of phosphorylated STAT4 in CD8 + T cells isolated from inguinal lymph nodes 24 hours after one intraperitoneal administration was measured by flow cytometry.
구체적으로, 도 23(B)와 같이 CT26HER2 / Neu 대장암세포를 이식한 Balb/c 마우스에서 종양의 크기가 300 mm3일 때 1μg rmIL-12와 같은 몰 농도의 bi-mIL-12-Fc와 mono-mIL-12-Fc를 복강 투여하였다. 24시간 후에 마우스의 사타구니 림프절을 적출한 후, 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml를 넣어 세척하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈 시킨 다음 PBS로 세척하여 세포 혼탁액을 준비하였다. 사타구니 림프절세포를 PE-cy5 또는 FITC가 결합된 CD3와 CD8를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 차가운 메탄올을 넣어 고정(fixation)하였다. 그 후 사타구니 림프절 세포를 차가운 PBS(pH 7.4)로 세척한 다음 APC가 결합된 pSTAT4를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 후 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 CD3+CD8+T 세포에 발현된 pSTAT4의 양을 비교하였다. 그 결과, mono-mIL-12-Fc에 비해 bi-mIL-12-Fc는 종양이 이식된 마우스의 사타구니 림프절에서 CD8+T 세포가 활성화될 때 pSTAT4의 발현을 증가시키는 효과를 보였다.Specifically, in the Balb / c mouse transplanted with CT26 HER2 / Neu colorectal cancer cells as shown in FIG. 23 (B), when the tumor size is 300 mm 3 , bi-mIL-12-Fc with a molar concentration of 1 μg rmIL-12 and mono-mIL-12-Fc was intraperitoneally administered. After 24 hours, the groin lymph nodes of the mouse were extracted, pulverized with a wire mesh in Petri dishes, and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Inguinal lymph node cells were stained with PE-cy5 or FITC-coupled CD3 and CD8 antibodies for 30 minutes at 4 ° C, washed with cold PBS (pH 7.4), and fixed with cold methanol. The inguinal lymph node cells were then washed with cold PBS (pH 7.4), and then stained with cold PBS (pH 7.4) for 30 minutes at 4 ° C with an antibody recognizing pSTAT4 bound to APC, followed by FACS Calibur (flow cytometry). BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to compare the amount of pSTAT4 expressed in CD3 + CD8 + T cells. As a result, compared with mono-mIL-12-Fc, bi-mIL-12-Fc increased the expression of pSTAT4 when CD8 + T cells were activated in the groin lymph nodes of tumor-grafted mice.
도 25(D)는 도 25(C)의 1회 복강투여 72시간 후에 사타구니 림프절에서 기억세포의 분화를 억제하는 T-bet을 발현하는 CD8+ T 세포의 비율을 유세포 분석기로 측정한 결과이다. Figure 25 (D) is a result of measuring the ratio of CD8 + T cells expressing T-bet inhibiting the differentiation of memory cells in the groin lymph nodes 72 hours after one intraperitoneal administration of Figure 25 (C) by flow cytometry.
구체적으로, 도 23(B)와 같이 CT26HER2 / Neu 대장암세포를 이식한 Balb/c 마우스에서 종양의 크기가 300 mm3일 때 1μg rmIL-12와 같은 몰 농도의 bi-mIL-12-Fc와 mono-mIL-12-Fc를 복강 투여하였다. 72시간 후에 마우스의 사타구니 림프절을 적출한 후, 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml를 넣어 세척하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈 시킨 다음 PBS로 세척하여 세포 혼탁액을 준비하였다. 사타구니 림프절세포를 PE-cy5 또는 FITC가 결합된 CD3와 CD8를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 핵내에 전사인자를 염색하는 시약인 Foxp3/Transcription Factor Staining Buffer Set (Thermo Fisher Scientific)을 이용하여 세포를 고정(fixation)하고 투과(permeabilization)시켰다. 그 후, PE 또는 APC가 결합된 T-bet을 인지하는 항체를 넣어 4℃에서 30분간 염색한 다음 투과버퍼(permeabilization buffer)를 넣어 유세포 분석기인 FACS Calibur (BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 T-bet을 발현하는 CD3+CD8+T 세포의 비율을 비교하였다. 그 결과, mono-mIL-12-Fc에 비해 bi-mIL-12-Fc는 종양이 이식된 마우스의 사타구니 림프절에서 CD8+T 세포가 활성화될 때 T-bet의 발현을 증가시키는 효과를 보였다. 따라서 1 μg IL-12와 같은 몰 농도의 bi-mIL-12-Fc를 투여하였을 때 CD8+ T세포가 단명화세포로 분화되는 것은 mono-mIL-12-Fc보다 종양을 이식한 마우스의 종양주위 림프절(tumor draining lymph node)인 사타구니림프절에서 CD8+ T세포가 활성화될 때 pSTAT4와 T-bet의 발현을 증가시켰기 때문임을 확인하였다. Specifically, in the Balb / c mouse transplanted with CT26 HER2 / Neu colorectal cancer cells as shown in FIG. 23 (B), when the tumor size is 300 mm 3 , bi-mIL-12-Fc with a molar concentration of 1 μg rmIL-12 and mono-mIL-12-Fc was intraperitoneally administered. After 72 hours, the groin lymph nodes of the mice were removed, pulverized with a wire mesh in Petri dishes, and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Inguinal lymph node cells were stained with PE-cy5 or FITC-bound CD3 and CD8 antibody for 30 minutes at 4 ° C, washed with cold PBS (pH 7.4), and stained with transcription factor Foxp3 / Transcription. Cells were fixed and permeabilized using a Factor Staining Buffer Set (Thermo Fisher Scientific). Afterwards, the antibody recognizes T-bet conjugated with PE or APC, and then stained at 4 ° C. for 30 minutes, and then added a permeabilization buffer to FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). ). Each sample was analyzed by dot plot to compare the proportion of CD3 + CD8 + T cells expressing T-bet. As a result, compared with mono-mIL-12-Fc, bi-mIL-12-Fc increased the expression of T-bet when CD8 + T cells were activated in the groin lymph nodes of mice transplanted with tumors. Therefore, the differentiation of CD8 + T cells into monolithic cells when bi-mIL-12-Fc at the same molar concentration as 1 μg IL-12 was observed in the peritumor lymph nodes of mice transplanted with tumors than mono-mIL-12-Fc. It was confirmed that the expression of pSTAT4 and T-bet was increased when CD8 + T cells were activated in the inguinal lymph nodes (tumor draining lymph nodes).
도 25(E)와 도 25(F)는 한 분자에서 2분자의 IL-12를 발현하는 bi-mIL-12-Fc 처럼 mono-mIL-12-Fc를 Fc를 인지하는 항체로 교차결합시켜 2분자의 IL-12에 의해 CD8+ T세포가 자극을 받도록 하면 bi-mIL-12-Fc를 처리했을 때와 비슷한 수준으로 pSTAT4와 T-bet의 발현이 증가되는지를 측정한 결과이다. 25 (E) and 25 (F) cross-link mono-mIL-12-Fc with an antibody that recognizes Fc, such as bi-mIL-12-Fc, which expresses two molecules of IL-12 in one molecule. When CD8 + T cells were stimulated by IL-12 of the molecule, we measured whether pSTAT4 and T-bet expression was increased to a level similar to that of bi-mIL-12-Fc treatment.
구체적으로는, 정상 Balb/c 마우스에서 비장과 사타구니 림프절을 적출한 후, 페트리디쉬에서 철망을 이용해 분쇄한 다음 2%의 FBS가 포함된 배지 10 ml를 넣어 세척하였다. 그 후, 적혈구 용혈버퍼 (red blood cell lysis buffer)를 1 ml 넣어 적혈구를 용혈 시킨 다음 PBS로 세척하여 세포 혼탁액을 준비하였다. 림프구세포를 PE 가 결합된 CD8를 인지하는 항체를 넣어 4℃에서 30분간 염색하고 차가운 PBS(pH 7.4)로 세척한 다음 anti-PE microbeads(Miltenyi Biotec)를 15분간 결합시켜 MACS separator와 LS column(Miltenyi Biotec)을 이용하여 CD8+ T 세포를 분리하였다. 0.5 μg/ml의 CD3를 인지하는 항체를 100μl씩 넣어 4℃에서 12시간 배양한 96-well round bottom plate를 PBS로 세척하여 플레이트에 부착되지 않은 CD3를 인지하는 항체를 제거하고 2 μg/ml의 CD28을 인지하는 항체를 50 μl씩 넣었다. 그 후 mono-mIL-12-Fc와 bi-mIL-12-Fc를 여러 농도의 Fc를 인지하는 항체와 4℃에서 30분간 반응시킨 다음 20 pM IL-12와 같은 농도가 되도록 첨가하였다. 그 후, CD8+ T 세포(4×104/well)를 넣고 37℃의 세포배양기에서 pSTAT4의 발현을 측정하기 위해서는 3시간, T-bet의 발현을 측정하기 위해서는 3일간 배양하였다. pSTAT4와 T-bet의 발현은 각각 도 25(C)와 도25(D)의 방법으로 염색한 다음 유세포 분석기인 FACS Calibur(BD Bioscience)와 Flow jo(Thermo Fisher Scientific)로 분석하였다. 각각의 샘플을 닷플랏(dot plot)으로 분석하여 CD8+ T 세포에 발현된 pSTAT4 또는 T-bet의 양을 비교하였다. 그 결과, mono-mIL-12-Fc를 Fc를 인지하는 항체로 교차결합시켜 2분자의 IL-12에 의해 CD8+ T세포가 자극을 받도록 하면 bi-mIL-12-Fc를 처리했을 때와 비슷한 수준으로 pSTAT4와 T-bet의 발현이 증가됨을 확인하였다. Specifically, spleen and groin lymph nodes were extracted from normal Balb / c mice, pulverized using a wire mesh in Petri dishes, and washed with 10 ml of medium containing 2% FBS. Thereafter, 1 ml of red blood cell lysis buffer was added to lyse red blood cells, and then washed with PBS to prepare a cell suspension. Lymphocytes were stained for 30 minutes at 4 ° C with antibodies bound to PE-binding CD8, washed with cold PBS (pH 7.4), and then combined with anti-PE microbeads (Miltenyi Biotec) for 15 minutes. Miltenyi Biotec) was used to isolate CD8 + T cells. Put 100 μl of antibody that recognizes 0.5 μg / ml of CD3, and wash 96-well round bottom plate incubated for 12 hours at 4 ° C with PBS to remove the antibody that recognizes CD3 not attached to the plate. 50 μl of antibody recognizing CD28 was added. Then, mono-mIL-12-Fc and bi-mIL-12-Fc were reacted for 30 minutes at 4 ° C. with an antibody that recognizes various concentrations of Fc, and then added to the same concentration as 20 pM IL-12. Thereafter, CD8 + T cells (4 × 10 4 / well) were added thereto, and cultured at 37 ° C. for 3 hours to measure pSTAT4 expression, and 3 days to measure T-bet expression. Expression of pSTAT4 and T-bet were stained by the method of FIGS. 25 (C) and 25 (D), respectively, and analyzed by flow cytometry, FACS Calibur (BD Bioscience) and Flow jo (Thermo Fisher Scientific). Each sample was analyzed by dot plot to compare the amount of pSTAT4 or T-bet expressed in CD8 + T cells. As a result, when mono-mIL-12-Fc was cross-linked with an antibody that recognizes Fc, the CD8 + T cells were stimulated by two molecules of IL-12, similar to those treated with bi-mIL-12-Fc. As a result, the expression of pSTAT4 and T-bet was increased.
결론적으로, 도 26에 묘사한 바와 같이 mono-mIL-12-Fc는 bi-mIL-12-Fc에 비해 CD8+ T세포에서 pSTAT4와 T-bet의 발현을 적게 유도하여 CD8+ T세포가 기억전구효과세포로의 분화를 거쳐 효과기억세포 및 중심기억세포로 분화되도록 유도하므로 낮은 농도(0.5 μg IL-12와 같은 몰농도)에서도 종양을 이식한 마우스의 종양을 제거하여 마우스의 수명을 연장시킬 수 있다. 반면 bi-mIL-12-Fc는 CD8+ T세포에서 pSTAT4와 T-bet의 발현을 많이 유도하여 단명효과세포로 분화되도록 하고 기억세포로 분화되지 못하게 하므로 mono-mIL-12-Fc와 같은 몰농도로 투여할 경우 종양을 이식한 마우스에서 종양이 완전히 제거되지 않으며 더 높은 농도(2 μg IL-12와 같은 몰농도)를 투여하여 종양세포를 직접 파괴하는 과정(effector phase)에서 세포독성 CD8+ T세포들을 증식(expansion)시켜야 종양을 제거할 수 있다.In conclusion, as illustrated in FIG. 26, mono-mIL-12-Fc induces less expression of pSTAT4 and T-bet in CD8 + T cells than bi-mIL-12-Fc so that CD8 + T cells have a memory effect. Differentiation into cells leads to differentiation into effect memory cells and central memory cells, so even at low concentrations (molar concentrations such as 0.5 μg IL-12), the tumors of mice transplanted with tumors can be removed to extend the lifespan of the mice. . On the other hand, bi-mIL-12-Fc induces the expression of pSTAT4 and T-bet in CD8 + T cells to differentiate into short-lived effect cells and to prevent differentiation into memory cells, thus molar concentrations such as mono-mIL-12-Fc When administered to the mouse, the tumor was not completely removed from the transplanted mouse, and cytotoxic CD8 + T in the effector phase was directly administered at a higher concentration (molar concentration such as 2 μg IL-12). The cells must be expanded to remove the tumor.
본 발명에 따른 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)은 2 이상의 서브유닛이 단백질 복합체(protein complex)를 형성하여 생리활성 단백질을 자연계에 존재하는 형태를 최대한 모사함으로써, 자연계에 존재하는 그대로의 활성을 유지할 수 있으며, 또한, 상기 생리활성 단백질의 체내에서의 반감기가 현저하게 연장될 수 있는 장점이 있다. Heterodimeric Fc-fused protein according to the antibody heavy chain constant region according to the present invention by forming a protein complex of two or more subunits to mimic the physiologically active protein forms in nature, It can maintain the activity as it exists in nature, and also has the advantage that the half-life in the body of the bioactive protein can be significantly extended.
또한, 본 발명에 따른 이종이중체-융합단백질(heterodimeric Fc-fused protein) 형태는 추가적인 정제 과정의 최적화 과정 없이 monovalent 형태의 이종이중체-융합단백질의 제조가 용이하다는 장점도 가진다.In addition, the heterodimeric-fusion protein (heterodimeric Fc-fused protein) form according to the present invention has the advantage that it is easy to prepare a monovalent heterodimer-fusion protein without further purification process optimization.
이상으로 본 발명의 내용의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.As described above in detail a specific part of the content of the present invention, for those skilled in the art, such a specific description is only a preferred embodiment, which is not limited by the scope of the present invention Will be obvious. Thus, the substantial scope of the present invention will be defined by the appended claims and their equivalents.
전자파일 첨부하였음.Electronic file attached.

Claims (17)

  1. 항체(immunoglobulin) 중쇄불변부위 (Fc) 쌍의 제1 Fc 영역 및 제2 Fc 영역을 포함하고, A first Fc region and a second Fc region of an antibody (immunoglobulin) heavy chain constant region (Fc) pair,
    상기 제1 Fc 영역 및/또는 제2 Fc 영역의 N-말단 또는 C-말단 중 하나 이상의 말단에 생리활성 단백질의 서브유닛이 결합되어 있는 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein)에 있어서, Heteroodimeric Fc-fused antibody heavy chain constant region, in which a subunit of a physiologically active protein is bound to one or more of the N-terminus or C-terminus of the first Fc region and / or the second Fc region. protein)
    상기 생리활성 단백질은 2 이상의 서로 다른 서브유닛(subunit)으로 이루어져 있으며, 2 이상의 서로 다른 서브유닛(subunit)은 단백질 복합체(protein complex)를 형성하여 생리활성을 나타내는 것을 특징으로 하며,The bioactive protein is composed of two or more different subunits (subunit), the two or more different subunits (subunit) is characterized in that the protein complex (protein complex) to form a physiological activity,
    상기 제1 Fc 영역 및 제2 Fc 영역은 이종이중체(heterodimeric Fc)의 형성이 촉진되도록 CH3 도메인이 변이된 것임을 특징으로 하는 항체 중쇄불변부위 이종이중체-융합단백질(heterodimeric Fc-fused protein).The first Fc region and the second Fc region are heterologous antibody-heteroodimeric Fc-fused proteins, characterized in that the CH3 domain is mutated to promote the formation of heterodimeric Fc.
  2. 제1항에 있어서, The method of claim 1,
    상기 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 생리활성 단백질의 서브유닛 중 하나가 결합되어 있고, 상기 생리활성 단백질의 나머지 서브유닛이 링커-매개된 형태로 상기 1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에 결합된 생리활성 단백질의 서브유닛과 결합되어 있는 항체 중쇄불변부위 이종이중체-융합단백질.One of the subunits of the bioactive protein is bound to either the N-terminus or the C-terminus of the first Fc region or the second Fc region, and the remaining subunits of the bioactive protein are linker-mediated. The antibody heavy chain constant heterodimer-fusion protein, which is bound to a subunit of a bioactive protein bound to either the N-terminus or the C-terminus of the 1 Fc region or the second Fc region in the form.
  3. 제1항에 있어서, The method of claim 1,
    제1 Fc 영역 및 제2 Fc 영역의 N-말단 또는 C-말단 각각에 1 종의 생리활성 단백질을 구성하는 서로 다른 하나 이상의 서브유닛(subunit)이 각각 결합되어 있는 항체 중쇄불변부위 이종이중체-융합단백질.The antibody heavy chain constant region heterodimer, in which one or more different subunits constituting one bioactive protein is respectively bound to the N-terminus or C-terminus of the first Fc region and the second Fc region, respectively. Fusion proteins.
  4. 제1항에 있어서, The method of claim 1,
    서로 다른 서브유닛이 1 종의 단백질 복합체(protein complex)를 형성하여 활성을 나타내는 생리활성 단백질은 인터루킨 12 (IL-12), 인터루킨 23 (IL-23), 인터루킨 27 (IL-27), 인터루킨 35 (IL-35) 및 난포자극호르몬(FSH)로 이루어진 군으로부터 선택되는 것을 특징으로 하는 이종이중체-융합단백질.Physiologically active proteins in which different subunits form one protein complex and exhibit activity are interleukin 12 (IL-12), interleukin 23 (IL-23), interleukin 27 (IL-27), and interleukin 35 (IL-35) and follicle stimulating hormone (FSH) heterodimer-fusion protein, characterized in that it is selected from the group consisting of.
  5. 제4항에 있어서, The method of claim 4, wherein
    서로 다른 서브유닛이 1 종의 단백질 복합체(protein complex)를 형성하여 활성을 나타내는 생리활성 단백질은 인터루킨 12 (IL-12)인 것을 특징으로 하는 항체 중쇄불변부위 이종이중체-융합단백질.Biologically active heterozygote-fusion protein, characterized in that the physiologically active protein showing the activity by different subunits forming one protein complex (interleukin 12) (IL-12).
  6. 제5항에 있어서, The method of claim 5,
    상기 제1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에만 인터루킨 12 (IL-12)의 p35 또는 p40 서브유닛이 결합되어 있고, 나머지 서브유닛이 링커-매개된 형태로 상기 1 Fc 영역 또는 제2 Fc 영역의 N-말단 또는 C-말단 중 어느 하나의 말단에 결합된 서브유닛과 결합되어 있는 항체 중쇄불변부위 이종이중체-융합단백질.The p35 or p40 subunit of interleukin 12 (IL-12) is bound to only one of the N-terminus or C-terminus of the first Fc region or the second Fc region, and the remaining subunits are linker-mediated. The antibody heavy chain constant heterodimer-fusion protein, which is bound to a subunit bound to either N-terminus or C-terminus of the 1 Fc region or the second Fc region in the form.
  7. 제5항에 있어서, The method of claim 5,
    제1 Fc 영역 및 제2 Fc 영역의 N-말단 또는 C-말단 각각에 인터루킨 12 (IL-12)의 p35 및 p40 서브유닛이 각각 결합되어 있는 항체 중쇄불변부위 이종이중체-융합단백질.An antibody heavy chain constant heterodimer-fusion protein wherein p35 and p40 subunits of interleukin 12 (IL-12) are respectively bound to the N-terminus or C-terminus of the first Fc region and the second Fc region.
  8. 제1항에 있어서, The method of claim 1,
    상기 제1 Fc 영역 및 제2 Fc 영역은 각각 인간 IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD 및 IgE로 이루어진 군으로부터 선택된 Fc 영역 유래인 것을 특징으로 하는 항체 중쇄불변부위 이종이중체-융합단백질.Wherein the first Fc region and the second Fc region are derived from an Fc region selected from the group consisting of human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, and IgE, respectively. protein.
  9. 제1항에 있어서, The method of claim 1,
    상기 제1 Fc 영역 및 제2 Fc 영역은 인간 IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD 및 IgE로 이루어진 완전한 항체(whole antibody) 형태에 포함되어 있는 것을 특징으로 하는 항체 중쇄불변부위 이종이중체-융합단백질.Wherein the first Fc region and the second Fc region are contained in a whole antibody form consisting of human IgG1, IgG2, IgG3, IgG4, IgM, IgA, IgD, and IgE. Sieve-fusion protein.
  10. 제1항에 있어서, The method of claim 1,
    상기 제1 Fc 영역 또는 제2 Fc 영역의 CH3 도메인의 변이는 다음의 군에서 선택된 하나 이상의 변이를 포함하는 것을 특징으로 하는 항체 중쇄불변부위 이종이중체-융합단백질. (단, 변이 위치는 EU index에 따름)The mutation of the CH3 domain of the first Fc region or the second Fc region comprises at least one mutation selected from the group of the antibody heavy chain constant heterodimer-fusion protein. (However, the mutation position is according to the EU index)
    (1) 제1 Fc 영역의 CH3 도메인의 K370 위치에서의 K370E, K370R, K370M, K370D 또는 K370H의 아미노산 잔기의 치환; (1) substitution of the amino acid residue of K370E, K370R, K370M, K370D or K370H at the K370 position of the CH3 domain of the first Fc region;
    (2) 제2 Fc 영역의 CH3 도메인의 E357 위치에서의 E357N, E357D, E357A, E357I, E357G 또는 E357M의 아미노산 잔기의 치환, 및 S364 위치에서의 치환은 S364T 또는 S364W의 아미노산 잔기의 치환;(2) substitution of the amino acid residues of E357N, E357D, E357A, E357I, E357G, or E357M at the E357 position of the CH3 domain of the second Fc region, and the substitution at the S364 position comprises a substitution of an amino acid residue of S364T or S364W;
    (3) 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 K409W의 아미노산 잔기의 치환; 및(3) substitution of the amino acid residue of K409W at the K409 position of the CH3 domain of the first Fc region; And
    (4) 제2 Fc 영역의 CH3 도메인의 F405 위치에서의 F405T 아미노산 잔기의 치환, 및 D399 위치에서의 D399V의 아미노산 잔기의 치환. (4) substitution of the F405T amino acid residue at the F405 position of the CH3 domain of the second Fc region, and substitution of the amino acid residue of D399V at the D399 position.
  11. 제1항에 있어서, The method of claim 1,
    상기 제1 Fc 영역 또는 제2 Fc 영역의 CH3 도메인의 변이는 다음의 군에서 선택된 하나 이상의 변이를 포함하는 것을 특징으로 하는 항체 중쇄불변부위 이종이중체-융합단백질. (단, 변이 위치는 EU index에 따름)The mutation of the CH3 domain of the first Fc region or the second Fc region comprises at least one mutation selected from the group of the antibody heavy chain constant heterodimer-fusion protein. (However, the mutation position is according to the EU index)
    (1) 제1 Fc 영역의 CH3 도메인의 K360 위치에서의 K360E의 아미노산 잔기의 치환; (1) substitution of the amino acid residue of K360E at the K360 position of the CH3 domain of the first Fc region;
    (2) 제2 Fc 영역의 CH3 도메인의 E347 위치에서의 E347R의 아미노산 잔기의 치환; (2) substitution of the amino acid residue of E347R at the E347 position of the CH3 domain of the second Fc region;
    (3) 제1 Fc 영역의 CH3 도메인의 K409 위치에서의 K409W의 아미노산 잔기의 치환; (3) substitution of the amino acid residue of K409W at the K409 position of the CH3 domain of the first Fc region;
    (4) 제2 Fc 영역의 CH3 도메인의 F405 위치에서의 F405T 아미노산 잔기의 치환, 및 D399 위치에서의 D399V의 아미노산 잔기의 치환. (4) substitution of the F405T amino acid residue at the F405 position of the CH3 domain of the second Fc region, and substitution of the amino acid residue of D399V at the D399 position.
  12. 제10항 또는 제11항에 있어서, The method according to claim 10 or 11, wherein
    상기 제1 Fc 영역 및 제2 Fc 영역의 CH3 도메인 내에 하기의 결합을 추가로 포함하는 것을 특징으로 하는 이종이중체-융합단백질. (단, 상기 위치는 EU index에 따름)The heterodimer-fusion protein of claim 1, further comprising the following bonds in the CH3 domains of the first and second Fc regions. (However, the position is according to the EU index)
    (1) 제1 Fc 영역의 CH3 도메인 내 Y349 위치에 치환된 시스테인 (C); 및(1) cysteine (C) substituted at the Y349 position in the CH3 domain of the first Fc region; And
    (2) 제2 Fc 영역의 CH3 도메인 내 S354 위치에 치환된 시스테인 (C)의 결합.(2) binding of cysteine (C) substituted at the S354 position in the CH3 domain of the second Fc region.
  13. 제1항 내지 제12항 중 어느 한 항에 따른 항체 중쇄불변부위 이종이중체-융합단백질을 포함하는 약제학적 조성물. A pharmaceutical composition comprising the antibody heavy chain constant region heterodimer-fusion protein according to any one of claims 1 to 12.
  14. 제13항에 있어서, The method of claim 13,
    상기 항체 중쇄불변부위 이종이중체-융합단백질 내에 포함된 생리활성 단백질은 인터루킨 12 (IL-12)인 것을 특징으로 하는 약제학적 조성물. Pharmaceutical composition, characterized in that the bioactive protein contained in the antibody heavy chain constant region heterodimer-fusion protein is interleukin 12 (IL-12).
  15. 제14항에 있어서, The method of claim 14,
    암 치료를 위한 약제학적 조성물. Pharmaceutical compositions for the treatment of cancer.
  16. 제15항에 있어서, The method of claim 15,
    상기 암은 대장암, 흑생종, 유방암, 췌장암, 신장암, 전립선암, 난소암, 소장암, 식도암, 자궁경부암, 폐암, 림프종 및 혈액암으로 이루어진 군에서 선택되는 것을 특징으로 하는 약제학적 조성물. The cancer is a pharmaceutical composition, characterized in that selected from the group consisting of colon cancer, melanoma, breast cancer, pancreatic cancer, kidney cancer, prostate cancer, ovarian cancer, small intestine cancer, esophageal cancer, cervical cancer, lung cancer, lymphoma and hematologic cancer.
  17. 제15항에 있어서, The method of claim 15,
    다른 항암제와 병용 치료를 위한 약제학적 조성물. Pharmaceutical compositions for the combination treatment with other anticancer agents.
PCT/KR2017/008676 2016-08-10 2017-08-10 Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same WO2018030806A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
CN201780062851.0A CN110267977A (en) 2016-08-10 2017-08-10 Cell factor immunoglobulin Fc merges heterodimer and the pharmaceutical composition comprising it
SG11201901071TA SG11201901071TA (en) 2016-08-10 2017-08-10 Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same
US16/323,839 US10696722B2 (en) 2016-08-10 2017-08-10 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
EP17839824.4A EP3511340A4 (en) 2016-08-10 2017-08-10 Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same
MX2019001651A MX2019001651A (en) 2016-08-10 2017-08-10 Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same.
AU2017310163A AU2017310163B2 (en) 2016-08-10 2017-08-10 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
CA3033475A CA3033475A1 (en) 2016-08-10 2017-08-10 Heterodimeric fc-fused cytokine and pharmaceutical composition comprising the same
BR112019002394-1A BR112019002394B1 (en) 2016-08-10 2017-08-10 HETERODIMERIC PROTEIN FLUTED WITH CF AND ITS USE
JP2019506697A JP6993403B2 (en) 2016-08-10 2017-08-10 Heterodimer Fc fusion cytokine and pharmaceutical composition containing it
ZA2019/00772A ZA201900772B (en) 2016-08-10 2019-02-06 Heterodimeric fc-fused cytokine and pharmaceutical composition comprising the same
US16/886,177 US11078249B2 (en) 2016-08-10 2020-05-28 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US16/886,184 US11692019B2 (en) 2016-08-10 2020-05-28 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
AU2021273642A AU2021273642A1 (en) 2016-08-10 2021-11-26 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US18/323,124 US20230416325A1 (en) 2016-08-10 2023-05-24 Heterodimeric fc-fused cytokine and pharmaceutical composition comprising the same

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20160101823 2016-08-10
KR10-2016-0101823 2016-08-10
KR1020170101594A KR102050463B1 (en) 2016-08-10 2017-08-10 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
KR10-2017-0101594 2017-08-10

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US16/323,839 A-371-Of-International US10696722B2 (en) 2016-08-10 2017-08-10 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US16/886,177 Continuation US11078249B2 (en) 2016-08-10 2020-05-28 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US16/886,184 Division US11692019B2 (en) 2016-08-10 2020-05-28 Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same

Publications (1)

Publication Number Publication Date
WO2018030806A1 true WO2018030806A1 (en) 2018-02-15

Family

ID=61163082

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2017/008676 WO2018030806A1 (en) 2016-08-10 2017-08-10 Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same

Country Status (1)

Country Link
WO (1) WO2018030806A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110088124A (en) * 2016-09-02 2019-08-02 高丽大学校产学协力团 The Il-21 (heterodimer Fc merges IL-21) merged with immunoglobulin heavy chain constant region heterodimer (heterodimer Fc) and the pharmaceutical composition comprising it
CN110396133A (en) * 2018-04-25 2019-11-01 中国科学院生物物理研究所 It is a kind of using interleukin 12 as the fusion protein type prodrug of active constituent
WO2020086758A1 (en) * 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
WO2020072821A3 (en) * 2018-10-03 2020-06-18 Xencor, Inc. Il-12 heterodimeric fc-fusion proteins
US11078249B2 (en) 2016-08-10 2021-08-03 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
WO2021246720A1 (en) * 2020-06-01 2021-12-09 머스트바이오 주식회사 Bispecific antibody or antigen-binding fragment thereof, and preparation method therefor
US11471490B2 (en) 2017-07-03 2022-10-18 Torque Therapeutics, Inc. T cells surface-loaded with immunostimulatory fusion molecules and uses thereof
US11851466B2 (en) 2019-10-03 2023-12-26 Xencor, Inc. Targeted IL-12 heterodimeric Fc-fusion proteins
WO2024086739A1 (en) 2022-10-20 2024-04-25 Synthekine, Inc. Methods and compositions of il12 muteins and il2 muteins
RU2819097C2 (en) * 2018-10-03 2024-05-14 Ксенкор, Инк. Il-12 heterodimeric fc fusion proteins

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100015089A1 (en) * 1997-12-08 2010-01-21 Merck Patent Gmbh Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
US7695936B2 (en) 1995-03-01 2010-04-13 Genentech, Inc. Knobs and holes heteromeric polypeptides
WO2014084607A1 (en) * 2012-11-27 2014-06-05 아주대학교산학협력단 Ch3 domain variant pair inducing formation of heterodimer of heavy chain constant region of antibody at high efficiency, method for preparing same, and use thereof
WO2014145907A1 (en) * 2013-03-15 2014-09-18 Xencor, Inc. Targeting t cells with heterodimeric proteins
KR20150008012A (en) * 2013-07-12 2015-01-21 한미약품 주식회사 Conjugate of biologically active polypeptide monomer and immunoglobulin Fc fragment with reduced receptor-mediated clearance, and the method for preparing the same
KR20150142181A (en) 2014-06-11 2015-12-22 주식회사 엘지화학 Manufacturing method of vinyl chloride based resin

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7695936B2 (en) 1995-03-01 2010-04-13 Genentech, Inc. Knobs and holes heteromeric polypeptides
US20100015089A1 (en) * 1997-12-08 2010-01-21 Merck Patent Gmbh Heterodimeric fusion proteins useful for targeted immune therapy and general immune stimulation
WO2014084607A1 (en) * 2012-11-27 2014-06-05 아주대학교산학협력단 Ch3 domain variant pair inducing formation of heterodimer of heavy chain constant region of antibody at high efficiency, method for preparing same, and use thereof
KR101522954B1 (en) 2012-11-27 2015-05-27 아주대학교산학협력단 CH3 domain mutant pairs for the high yield formation of heterodimeric Fc of antibody, method of production and use thereof
WO2014145907A1 (en) * 2013-03-15 2014-09-18 Xencor, Inc. Targeting t cells with heterodimeric proteins
KR20150008012A (en) * 2013-07-12 2015-01-21 한미약품 주식회사 Conjugate of biologically active polypeptide monomer and immunoglobulin Fc fragment with reduced receptor-mediated clearance, and the method for preparing the same
KR20150142181A (en) 2014-06-11 2015-12-22 주식회사 엘지화학 Manufacturing method of vinyl chloride based resin

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
HA, JI-HEE ET AL.: "Immunoglobulin Fc Heterodimer Platform Technology: from Design to Applications in Therapeutic Antibodies and Proteins", FRONTIERS IN IMMUNOLOGY, vol. 7, 6 October 2016 (2016-10-06), pages 1 - 16, XP055377975, DOI: doi:10.3389/fimmu.2016.00394 *
LOW, S. C. ET AL.: "Oral and Pulmonary Delivery of FSH-Fc Fusion Proteins via Neonatal Fc Receptor-mediated Transcytosis", HUMAN REPRODUCTION, vol. 20, no. 7, 2005, pages 1805 - 1813, XP002455704, DOI: doi:10.1093/humrep/deh896 *
See also references of EP3511340A4 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11692019B2 (en) 2016-08-10 2023-07-04 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
US11078249B2 (en) 2016-08-10 2021-08-03 Ajou University Industry-Academic Cooperation Foundation Heterodimeric Fc-fused cytokine and pharmaceutical composition comprising the same
CN110088124A (en) * 2016-09-02 2019-08-02 高丽大学校产学协力团 The Il-21 (heterodimer Fc merges IL-21) merged with immunoglobulin heavy chain constant region heterodimer (heterodimer Fc) and the pharmaceutical composition comprising it
US11471490B2 (en) 2017-07-03 2022-10-18 Torque Therapeutics, Inc. T cells surface-loaded with immunostimulatory fusion molecules and uses thereof
CN112638938A (en) * 2018-04-25 2021-04-09 免疫靶向有限公司 Interleukin 12 fusion proteins and compositions and methods of treatment thereof
WO2019209965A3 (en) * 2018-04-25 2019-12-05 Immune Targeting Inc. Interleukin 12 fusion proteins, and compositions and therapeutic methods thereof
CN110396133B (en) * 2018-04-25 2021-07-23 免疫靶向有限公司 Fusion protein type prodrug with interleukin 12 as active component
JP2021521822A (en) * 2018-04-25 2021-08-30 イミューン ターゲティング インク. Interleukin 12 fusion protein and composition, and their therapeutic methods
CN110396133A (en) * 2018-04-25 2019-11-01 中国科学院生物物理研究所 It is a kind of using interleukin 12 as the fusion protein type prodrug of active constituent
RU2819097C2 (en) * 2018-10-03 2024-05-14 Ксенкор, Инк. Il-12 heterodimeric fc fusion proteins
WO2020072821A3 (en) * 2018-10-03 2020-06-18 Xencor, Inc. Il-12 heterodimeric fc-fusion proteins
US11655277B2 (en) 2018-10-03 2023-05-23 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
US11358999B2 (en) 2018-10-03 2022-06-14 Xencor, Inc. IL-12 heterodimeric Fc-fusion proteins
CN113286808A (en) * 2018-10-23 2021-08-20 蜻蜓疗法股份有限公司 Heterodimeric Fc fusion proteins
JP2022505871A (en) * 2018-10-23 2022-01-14 ドラゴンフライ セラピューティクス, インコーポレイテッド Heterodimer FC fusion protein
US11787864B2 (en) 2018-10-23 2023-10-17 Dragonfly Therapeutics, Inc. Heterodimeric Fc-fused proteins
WO2020086758A1 (en) * 2018-10-23 2020-04-30 Dragonfly Therapeutics, Inc. Heterodimeric fc-fused proteins
US11851466B2 (en) 2019-10-03 2023-12-26 Xencor, Inc. Targeted IL-12 heterodimeric Fc-fusion proteins
WO2021246720A1 (en) * 2020-06-01 2021-12-09 머스트바이오 주식회사 Bispecific antibody or antigen-binding fragment thereof, and preparation method therefor
WO2024086739A1 (en) 2022-10-20 2024-04-25 Synthekine, Inc. Methods and compositions of il12 muteins and il2 muteins

Similar Documents

Publication Publication Date Title
WO2018030806A1 (en) Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same
WO2020060122A1 (en) Fusion protein comprising il-2 protein and cd80 protein, and use thereof
WO2015133817A1 (en) Monoclonal antibody specifically recognizing b-cell lymphoma cells and use thereof
WO2017023138A1 (en) Chimeric antigen receptor, and t cells in which chimeric antigen receptor is expressed
WO2015058573A1 (en) Monoclonal antibody for antagonizing and inhibiting binding of programmed death (pd-1) to ligand thereof and coding sequence and use thereof
WO2020050667A1 (en) Chimeric antigen receptor for solid cancer and t cells expressing chimeric antigen receptor
WO2017065484A1 (en) Method for producing antibody ch3 domain heterodimeric mutant pair using yeast mating and ch3 mutant pair produced thereby
WO2014025198A2 (en) Lfa3 mutant, fusion protein in which target-specific polypeptides are connected to the mutant or lfa3 cd2 binding region, and use thereof
WO2021107689A1 (en) Pharmaceutical composition for treatment of cancer, comprising immune checkpoint inhibitor and fusion protein including il-2 protein and cd80 protein
WO2011142514A1 (en) Composition containing pias3 as an active ingredient for preventing or treating cancer or immune disease
WO2018026248A1 (en) Novel antibody against programmed cell death protein (pd-1), and use thereof
WO2021158073A1 (en) Fusion antibody for presenting antigen-derived t cell antigen epitope or peptide containing same on cell surface, and composition comprising same
WO2023277361A1 (en) Mesothelin-specific antibodies and use thereof
WO2022177394A1 (en) Bispecific single domain antibody to pd-l1 and cd47 and use thereof
WO2022025638A1 (en) Chimeric antigen receptor (car) t cell stabilizing immune synapse
WO2016199964A1 (en) Antibody specifically binding to isolated vimentin-derived peptide, or binding fragment of peptide
WO2019045477A1 (en) Composition for preventing and treating skin disease comprising substance specifically binding to vimentin-derived peptide
WO2022035200A1 (en) Fusion protein comprising il-12 and anti-cd20 antibody and use thereof
WO2022145739A1 (en) Humanized antibody specific for cd22 and chimeric antigen receptor using the same
WO2021071319A1 (en) Multispecific fusion protein and use thereof
WO2018026249A1 (en) Antibody against programmed death-ligand 1 (pd-l1), and use thereof
WO2020204305A1 (en) Fusion protein comprising anti-mesothelin antibody, anti-cd3 antibody or anti-egfr antibody, bispecific or trispecific antibody comprising same, and uses thereof
WO2020138868A1 (en) Type 1 interferon neutralizing fc-fusion protein and use thereof
WO2021162394A1 (en) Chimeric antigen receptor targeting b-cell maturation antigen and use thereof
WO2023121254A1 (en) Interleukin-2 fusion protein, method for preparing same, and pharmaceutical composition comprising same

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17839824

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019506697

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3033475

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112019002394

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2017310163

Country of ref document: AU

Date of ref document: 20170810

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017839824

Country of ref document: EP

Effective date: 20190311

ENP Entry into the national phase

Ref document number: 112019002394

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20190206