WO2015182798A1 - Composition for inhibiting myeloid-derived suppressor cells (mdscs) - Google Patents

Composition for inhibiting myeloid-derived suppressor cells (mdscs) Download PDF

Info

Publication number
WO2015182798A1
WO2015182798A1 PCT/KR2014/004760 KR2014004760W WO2015182798A1 WO 2015182798 A1 WO2015182798 A1 WO 2015182798A1 KR 2014004760 W KR2014004760 W KR 2014004760W WO 2015182798 A1 WO2015182798 A1 WO 2015182798A1
Authority
WO
WIPO (PCT)
Prior art keywords
mdsc
peptide
composition
cancer
adjuvant
Prior art date
Application number
PCT/KR2014/004760
Other languages
French (fr)
Korean (ko)
Inventor
김상재
미들톤개리
Original Assignee
주식회사 카엘젬백스
김상재
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 주식회사 카엘젬백스, 김상재 filed Critical 주식회사 카엘젬백스
Priority to PCT/KR2014/004760 priority Critical patent/WO2015182798A1/en
Priority to KR1020157003185A priority patent/KR101826753B1/en
Priority to PCT/KR2014/011571 priority patent/WO2015182837A1/en
Publication of WO2015182798A1 publication Critical patent/WO2015182798A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • a composition for inhibiting myeloid-derived suppressor cells (MDSCs), a kit for inhibiting MDSC, a method for inhibiting MDSC, a composition for anticancer, a kit for anticancer, an anticancer method, or a novel use of telomerase peptide It is about.
  • MDSCs Myeloid-derived suppressor cells
  • cytotoxic T lymphocytes are a heterogeneous family of immature bone marrow cells that have had differentiation programs stagnated by various tumor-secreting factors. MDSCs inhibit the activity of cytotoxic T lymphocytes in a variety of ways.
  • the function of MDSC in vivo is, in part, to protect the body from the harmful effects of excessive immune stimulation in acute and chronic infections and to limit the development of autoimmune responses to tissue antigens released by trauma.
  • the proliferation of MDSCs is also associated with a pathological point of view. Activation is mediated through several transcription factors, resulting in upregulation and expression of immunosuppressive factors such as ARG1 and NOS2 and increased production of NO, ROS, RNS and cytokines.
  • the accumulation of MDSC allows the immunosuppressive environment to be sustained, which in turn exposes the living body to allergen and / or viral infections, which can lead to chronic inflammation. Chronic inflammation can lead to tissue damage if the body is unable to respond effectively.
  • chemotherapeutic agents known to directly reduce MDSC in preclinical models are gemcitabine and 5-fluorouracil (5-FU).
  • Gemcitabine has been reported to significantly reduce the number of MDSCs of the spleen in tumor-induced mice [Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM (2005) Gemcitabine eliminate eliminates splenic Gr-1 + / CD11b + myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 11: 6713-6721].
  • 5-FU is also known to significantly reduce the percentage of MDSC, which is reported to be greater than gemcitabine.
  • miR-142 and / or miR-223 ribonucleotides can be added to MDSC (WO 2013/082591).
  • miR-142 and / or miR-223 ribonucleotides can be reduced in number by allowing MDSCs to differentiate into macrophages, dendritic cells, and the like.
  • Bisphosphonates include clodronate, zoleronate, palmidronate, ethidronate or other bisphosphonate drugs.
  • CCR2 inhibitors include RS 1028595 or PF-04178903.
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • the object of the present invention is to inhibit MDSC.
  • the object of the present invention is to activate immunity.
  • the object of the present invention is to activate immunity inhibited by MDSC.
  • an object of the present invention is to solve the problem that the immune response of the vaccine is inhibited by MDSC.
  • an object of the present invention is to solve the problem that the immune response of a cancer vaccine is inhibited by MDSC.
  • an object of the present invention is to alleviate, treat or prevent a disease or condition associated with MDSC.
  • an object of the present invention is to provide a cancer vaccine without the side effects of lowering the immune response according to MDSC.
  • the present invention is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), the composition is a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, the peptide sequence and 80% Peptide having a sequence homology, or a fragment thereof, the peptide is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), contained in an amount effective for MDSC inhibition.
  • MDSCs myeloid-derived suppressor cells
  • the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be a composition for inhibiting MDSC containing an adjuvant.
  • the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be an anticancer composition comprising an adjuvant.
  • the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And cancer vaccine compositions comprising an adjuvant.
  • the invention may be a kit for inhibiting MDSC.
  • the kit includes a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, a peptide having 80% or more sequence homology with the peptide sequence, or a peptide thereof, wherein the peptide is effective for inhibiting MDSC.
  • MDSC inhibition composition contained in an amount; Anticancer agents; And instructions.
  • the kit may further comprise an adjuvant.
  • the kit for inhibiting MDSC may be to improve, prevent or treat a disease or symptom caused by MDSC through MDSC inhibition.
  • the kit for inhibiting MDSC may be to improve, prevent or treat cancer through MDSC inhibition.
  • the invention provides a composition comprising a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And an anticancer kit comprising instructions.
  • the kit may further comprise an adjuvant composition.
  • the present invention provides a method for inhibiting myeloid-derived suppressor cells (MDSCs), an effective amount of a peptide comprising SEQ ID NO: 1, the peptide sequence and at least 80% sequence Or a peptide that is homologous, or a fragment thereof, to a subject in need of MDSC inhibition.
  • the method may further comprise administering an anticancer agent in combination with the peptide.
  • the method may further comprise administering an adjuvant in combination with the peptide.
  • the present invention provides a method for improving, treating or preventing cancer, wherein the peptide comprises an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof.
  • the peptide comprises an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof.
  • an anticancer agent and / or adjuvant may be a method for cancer improvement, treatment or prevention comprising administering to a subject in need of improvement, treatment or prevention of cancer.
  • a peptide comprising SEQ ID NO: 1, having a sequence homology of 80% or more of the peptide sequence Peptides, or fragments thereof, may be used.
  • the present invention may inhibit MDSC.
  • the present invention may activate immune.
  • the present invention can activate immunity inhibited by MDSC.
  • the present invention can solve the problem that the immune response of the vaccine is inhibited by MDSC.
  • the present invention can solve the problem that the immune response of a cancer vaccine is inhibited by MDSC.
  • the present invention can alleviate, treat or prevent a disease or condition associated with MDSC.
  • the present invention may provide an anticancer composition, an anticancer kit, or an anticancer method that does not have the side effects of lowering the immune response according to MDSC.
  • the present invention can provide a cancer vaccine that is free of the side effects of lowering the immune response following MDSC.
  • 1 is a graph showing the relationship between MDSC and the levels of proinflammatory cytokine. Comparison of the proinflammatory cytokines of patients with high and low levels of MDSC showed no correlation between the levels of MDSC and proinflammatory cytokine.
  • Figure 2 is a log after analyzing and comparing the MDSC changes in the patient group (comparative example 2, arm2) that was administered only in combination with gemcitabine and capecitabine, and the patient group (Example 2, arm3) that was simultaneously administered GemCap and pep1 It is a graph in scale.
  • MDSC inhibition is a concept that includes not only reducing the number of MDSCs but also inhibiting the activity of MDSCs. Reducing the number includes not only inhibiting the production of cells, but also killing or differentiating cells that have already been formed. In addition, all mechanisms that are referred to as “inhibition” from a biological standpoint are included.
  • combined administration or “combined administration” refers not only to simultaneous administration in terms of physical time, but also to two or more drugs in combination, according to the unique schedule of each drug being combined as in conventional combination therapy. Administering.
  • MDSCs can inhibit the effectiveness of cancer vaccines administered for the treatment of cancer patients, and as a result can render cancer treatment impossible.
  • cancer treatment with cancer vaccines it is necessary to reduce the increased MDSC in cancer patients.
  • the present inventors used a combination of two chemotherapeutic agents, gemcitabine, and 5-fluorouracil (5-FU) or capecitabine, which are known to directly reduce MDSC. It has been found that, unlike single use, it does not reduce MDSC. Rather, a significant number of patients showed elevated MDSCs. This may act as a big limitation of the combination of gemcitabine and 5-fluorouracil (or capecitabine).
  • Using the peptides disclosed herein can significantly reduce MDSC. Reducing MDSCs can solve a series of pathological problems associated with MDSCs.
  • telomere is a genetic material repeatedly present at the end of a chromosome and is known to prevent damage to the chromosome or binding to another chromosome. Each time a cell divides, the telomeres become slightly shorter. After a certain number of cell divisions, the telomeres become very short, and the cells stop dividing and die. On the other hand, elongation of telomeres is known to prolong cell life. For example, cancer cells secrete an enzyme called telomerase, which prevents telomeres from shortening, so that cancer cells can continue to proliferate without dying.
  • the peptides disclosed herein are peptides having a sequence of SEQ ID NO: 1 or a peptide that is a fragment of sequence SEQ ID NO: 1, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, Peptides having at least 98%, at least 99% sequence homology.
  • the peptide comprises telomerase, specifically a peptide derived from human ( Homo sapiens ) telomerase.
  • the peptides disclosed herein may include peptides having sequence homology with a peptide comprising SEQ ID NO: 1 or a fragment thereof.
  • the peptide may consist of up to 30 amino acids.
  • the peptide described in SEQ ID NO: 1 is shown in Table 1 below.
  • the "name” in Table 1 below is to distinguish peptides.
  • the peptide set forth in SEQ ID NO: 1 represents the entire peptide of human telomerase.
  • a peptide having a sequence of SEQ ID NO: 1, a peptide that is a fragment of SEQ ID NO: 1, or a peptide having at least 80% sequence homology with the peptide sequence corresponds to a peptide included in telomerase.
  • synthetic peptides selected and synthesized at the positional peptides.
  • SEQ ID 2 shows the amino acid sequence of the entire telomerase.
  • amino acid changes belong to a property that allows the physicochemical properties of the peptide to be altered.
  • amino acid changes can be made, such as improving the thermal stability of the peptide, altering substrate specificity, changing the optimal pH, and the like.
  • amino acid includes not only the 22 standard amino acids that are naturally incorporated into the peptide, but also D-isomers and modified amino acids. Accordingly, in one aspect of the invention the peptide may be a peptide comprising D-amino acids. Meanwhile, in another aspect of the present invention, the peptide may include a non-standard amino acid or the like which has been post-translational modified.
  • post-translational modifications include phosphorylation, glycosylation, acylation (including, for example, acetylation, myristoylation and palmitoylation), alkylation ), Carboxylation, hydroxylation, glycation, biotinylation, ubiquitinylation, changes in chemical properties (e.g., beta-elimination deimidization) , Deamidation) and structural changes (eg, formation of disulfide bridges). It also includes changes in amino acids, such as changes in amino groups, carboxy groups or side chains, caused by chemical reactions that occur during the linkage with crosslinkers to form peptide conjugates.
  • Peptides disclosed herein can be wild-type peptides identified and isolated from a natural source.
  • a peptide disclosed herein may be an artificial variant, comprising an amino acid sequence in which one or more amino acids are substituted, deleted, and / or inserted as compared to a peptide that is SEQ ID NO: 1 or a fragment thereof.
  • Amino acid changes in the wild type polypeptide as well as in artificial variants include conservative amino acid substitutions that do not significantly affect the folding and / or activity of the protein.
  • conservative substitutions include basic amino acids (arginine, lysine and histidine), acidic amino acids (glutamic acid and aspartic acid), polar amino acids (glutamine and asparagine), hydrophobic amino acids (leucine, isoleucine, valine and methionine), aromatic amino acids (phenylalanine, Tryptophan and tyrosine), and small amino acids (glycine, alanine, serine and threonine). Amino acid substitutions that generally do not alter specific activity are known in the art.
  • the most common exchanges are Ala / Ser, Val / Ile, Asp / Glu, Thr / Ser, Ala / Gly, Ala / Thr, Ser / Asn, Ala / Val, Ser / Gly, Tyr / Phe, Ala / Pro, Lys / Arg, Asp / Asn, Leu / Ile, Leu / Val, Ala / Glu, and Asp / Gly, and vice versa.
  • Other examples of conservative substitutions are shown in the following table.
  • residue substitution Ala (A) val; leu; ile Val Arg (R) lys; gln; asn Lys Asn (N) gln; his; asp, lys; arg Gln Asp (D) glu; asn Glu Cys (C) ser; ala Ser Gln (Q) asn; glu Asn Glu (E) asp; gln Asp Gly (G) Ala Ala His (H) asn; gln; lys; arg Arg Ile (I) leu; val; met; ala; phe; norleucine Leu Leu (L) norleucine; ile; val; met; ala; phe Ile Lys (K) arg; gln; asn Arg Met (M) leu; phe; ile Leu Phe (F) leu; val; ile; ala; tyr Tyr
  • the present invention is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), the composition is a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, the peptide sequence and 80% Peptide having a sequence homology, or a fragment thereof, the peptide is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), contained in an amount effective for MDSC inhibition.
  • MDSCs myeloid-derived suppressor cells
  • the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be a composition for inhibiting MDSC containing an adjuvant.
  • the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be an anticancer composition comprising an adjuvant.
  • the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And cancer vaccine compositions comprising an adjuvant.
  • the invention may be a kit for inhibiting MDSC.
  • the kit includes the above-mentioned composition for inhibiting MDSC; Anticancer agents; And instructions.
  • the kit may further comprise an adjuvant.
  • the kit for inhibiting MDSC may be to improve, prevent or treat a disease or symptom caused by MDSC through MDSC inhibition.
  • the kit for inhibiting MDSC may be to improve, prevent or treat cancer through MDSC inhibition.
  • the invention provides a composition comprising a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And an anticancer kit comprising instructions.
  • the kit may further comprise an adjuvant composition.
  • the instructions may include a description of how the composition for inhibiting MDSC containing the peptide is used in combination with an anticancer agent. Specifically, it may include a description of the dosage, administration time, administration method and the like of each agent.
  • the instructions may include uses for inhibiting MDSC, anticancer use or cancer vaccine use. Specifically, all diseases or symptoms that need to inhibit MDSC can be included.
  • the instructions may include side effects and precautions associated with the administration.
  • the present invention provides a method for inhibiting myeloid-derived suppressor cells (MDSCs), an effective amount of a peptide comprising SEQ ID NO: 1, the peptide sequence and at least 80% sequence Or a peptide that is homologous, or a fragment thereof, to a subject in need of MDSC inhibition.
  • the method may further comprise administering an anticancer agent in combination with the peptide.
  • the method may further comprise administering an adjuvant in combination with the peptide.
  • the present invention provides a method for improving, treating or preventing cancer, wherein the peptide comprises an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof.
  • the peptide comprises an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof.
  • an anticancer agent and / or adjuvant may be a method for cancer improvement, treatment or prevention comprising administering to a subject in need of improvement, treatment or prevention of cancer.
  • the cancer is not particularly limited, but may be renal cell carcinoma (RCC), colorectal cancer (CRC), gastric cancer (GC), melanoma, lung cancer, blood cancer, prostate cancer, adenocarcinoma, prostate cancer or pancreatic cancer.
  • RCC renal cell carcinoma
  • CRC colorectal cancer
  • GC gastric cancer
  • melanoma lung cancer, blood cancer, prostate cancer, adenocarcinoma, prostate cancer or pancreatic cancer.
  • a peptide comprising SEQ ID NO: 1, having a sequence homology of 80% or more of the peptide sequence Peptides, or fragments thereof, may be used.
  • bone marrow-derived suppressor cells may be bone marrow-derived suppressor cells of an individual having a tumor.
  • the composition may be for administration to a subject in need of inhibition of MDSC.
  • MDSC functions to suppress immunity by inhibiting the activity of cytotoxic T lymphocytes.
  • cytotoxic T lymphocytes There is a net function of suppressing unnecessary and excessive immune responses, such as autoimmunity, but there is also a reverse function of inhibiting immunity in situations where an immune response is needed, causing or worsening disease, or impeding proper treatment.
  • MDSC is much increased in tumor or cancer patients, which negates the efficacy of cancer vaccines by significantly reducing the effects of cancer vaccine administration. In this situation, effectively reducing the number of MDSCs will enable smooth and effective cancer treatment.
  • Diseases or symptoms caused by MDSC are apparent in the art.
  • Diseases or symptoms caused by MDSC referred to herein include all diseases or symptoms caused by MDSC known in the art.
  • Bone diseases such as, for example, osteolytic bone disease; Multiple myeloma; Glioblastoma; Infections: bacterial & parasitic infections such as bacterial or parasitic infections; Acute or chronic inflammation; Traumatic stress; Sepsis and transplantation; Autoimmune diseases such as intraocular autoimmune diseases; Inflammatory bowel disease, Cancer cachexia; Or tuberculosis (TB), but is not limited thereto.
  • MDSC in this specification may include all MDSCs regardless of their phenotype.
  • Various phenotypes of MDSC are known in the art.
  • the phenotype is one selected from CD15, IL4Ra, CD14, CD11b, HLA-DR, CD33, Lin, FSC, DR and SSC, and optionally CD45, CD18, CD80, CD83, CD86, HLA-I and survival / killing identifiers.
  • CD15, IL4Ra, CD14, CD11b, HLA-DR, CD33, Lin, FSC, DR and SSC and optionally CD45, CD18, CD80, CD83, CD86, HLA-I and survival / killing identifiers.
  • CD14 CD11b
  • HLA-DR CD33
  • Lin Lin
  • FSC DR and SSC
  • CD45 CD18
  • CD80 CD83
  • CD86 HLA-I and survival / killing identifiers.
  • phenotypes include MDSC1 based on the labels IL4Ra + and CD14 +; MDSC2 based on the labels IL4Ra + and CD15 +; MDSC1 and MDSC2 based on the label IL4Ra +; MDSC3 based on the labels Lin-, HLA-DR- and CD33 +, and optionally CD18 + and HLA-I +; MDSC4 based on the label CD14 +, HLA-DR ( ⁇ / lo), FSChi and SSCim; MDSC5 based on the labels CD11b +, CD14- and CD15 +, and optionally FSChi, SSCim, CD80-, CD83-, CD86- and HLA-DR-; MDSC6 based on CD15 +, SClo and SSChi; And MDSC based on Lin, DR, and CD11b +, and the like.
  • the phenotype can be Lin-DR-CD11b +.
  • the concentration of peptides in the compositions disclosed herein can be routinely determined as known in the art.
  • the composition according to one aspect of the present invention comprises 10 mg / L to 1000 peptides comprising a peptide comprising the amino acid sequence of SEQ ID NO: 1, a peptide having a sequence homology of 80% or more with the amino acid sequence, or a fragment thereof. It may be included in the content of mg / L, specifically 10 mg / L to 500 mg / L, more specifically 30 mg / L to 200 mg / L, but when the difference in effect depending on the dose can be adjusted appropriately. When included in the above range or less, it is not only appropriate to exhibit the intended effect of the present invention, but also satisfies both the stability and safety of the composition, it may be appropriate to include in the above range in terms of cost-effectiveness. .
  • the dosage, administration method, administration cycle, and the like of the peptide herein are well known in the art, it may be administered according to the standards known in the art according to the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 ⁇ g / kg / day to 10 g / kg / day, more specifically 10 ⁇ g / kg / day to 100 mg / kg / day, and more specifically, 50 ⁇ g / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
  • the peptide can be administered via intradermal administration.
  • Dosage intervals may be administered once daily at two-day intervals and may be further extended over time. For example, after administration in weeks 1, 2, 3, and 4, the intervals may be spaced into weeks 6 and 10.
  • the dosage may be 0.1 to 3 mg once administered on an adult basis.
  • the dosage can be at least 0.1 mg, at least 0.2 mg, at least 0.3 mg, at least 0.4 mg at least 0.45 mg or at least 0.5 mg.
  • the dosage may be 3 mg or less, 2.5 mg or less, 2.0 mg or less, 1.5 mg or less, 1.0 mg or less, 0.9 mg or less, 0.8 mg or less, 0.7 mg or less, 0.6 mg or less.
  • the peptides described herein can be administered in combination with an anticancer agent.
  • Anticancer agents include, but are not limited to, chemotherapeutic and biotherapeutic agents. In particular, it may be a chemotherapeutic agent.
  • Chemotherapeutic agents include, but are not limited to, DNA alkylating agents, anti-metabolites, natural products, hormones, and the like.
  • the chemotherapeutic agent may be a metabolic antagonist.
  • Metabolism inhibitors refer to groups of molecules that interfere with DNA and RNA synthesis. Most metabolism inhibitors have a structure similar to the building blocks of DNA and RNA.
  • Subtypes of metabolic antagonists include, but are not limited to, folate antagonists (anti-folates), fluoropyrimidines, deoxynucleoside analogues and thiopurines.
  • Fluoropyrimidines include, but are not limited to, fluorouracil and capecitabine.
  • Capecitabine is a prodrug of 5-fluorouracil.
  • Deoxynucleoside analogues include cytarabine, gemcitabine, decitabine, vidaza, fludarabine, nelarabine, cladribine Cladribine, clofarabine and pentostatin, but are not limited thereto.
  • the chemotherapeutic agent may be one in which two or more agents are administered in combination.
  • fluoropyrimidine and deoxynucleoside analogs can be administered in combination.
  • gemcitabine and 5-fluorouracil or capecitabine, a prodrug thereof
  • gemcitabine and capecitabine may be administered in combination.
  • Appropriate dosages of the anticancer agents mentioned herein are already well known in the art and can be administered by criteria known in the art, depending on the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 ⁇ g / kg / day to 10 g / kg / day, more specifically 10 ⁇ g / kg / day to 100 mg / kg / day, and more specifically, 50 ⁇ g / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
  • gemcitabine may be administered intravenously, and the dosage and administration interval may be 1 to 6 times weekly at 2 to 8 week intervals at a dose of 100 to 10,000 mg / m 2 .
  • Dosage is at least 100 mg / m 2 , at least 200 mg / m 2, at least 300 mg / m 2, at least 400 mg / m 2, at least 500 mg / m 2, at least 600 mg / m 2 , at 700 mg / m 2 Or more, 800 mg / m 2 or more, or 900 mg / m 2 or more.
  • the dosage is 10,000 mg / m 2 or less, 9000 mg / m 2 or less, 8000 mg / m 2 or less, 7000 mg / m 2 or less, 6000 mg / m 2 or less, 5000 mg / m 2 or less, 4000 mg / m 2 or less, 3000 mg / m 2 or less, 2000 mg / m 2 or less, 1500 mg / m 2 or less, 1400 mg / m 2 or less, 1300 mg / m 2 or less, 1200 mg / m 2 or less, 1100 mg / m 2 Or less, 1050 mg / m 2 or less, 1030 mg / m 2 or less, 1020 mg / m 2 or less, or 1010 mg / m 2 or less.
  • capecitabine it may be administered orally.
  • the dosing interval may be twice daily.
  • Doses are at least 500 mg / m 2, at least 600 mg / m 2, at least 700 mg / m 2, at least 800 mg / m 2 , 900 mg / m 2 , 1,000 mg / m 2 , 1,100 mg / m 2 , 1,200 mg / m 2 , 1,300 mg / m 2 , 1,400 mg / m 2 , 1,500 mg / m 2 , 1,600 mg / m 2 or more.
  • the dosage is 15,000 mg / m 2 or less, 12,000 mg / m 2 or less, 10,000 mg / m 2 or less, 9000 mg / m 2 or less, 8000 mg / m 2 or less, 7000 mg / m 2 or less, 6000 mg / m 2 or less, 5000 mg / m 2 or less, 4000 mg / m 2 or less, 3000 mg / m 2 or less, 2000 mg / m 2 or less, 1,900 mg / m 2 or less, 1,800 mg / m 2 or less, 1,700 mg / m 2 It may be: The daily dose may be administered two or more times divided.
  • the peptides and / or anticancer agents described herein can be administered in combination with an adjuvant.
  • the adjuvant may be an adjuvant that increases MDSC. From an immunological point of view, the adjuvant is what is added to the vaccine to stimulate an immune response to the target antigen, but does not itself provide immunogenicity. All vaccines cause inflammation, resulting in the accumulation of bone marrow cells (monocytes and Neutrophils). Some of these cells are MDSCs. In addition to the purpose of stimulating the immune response, there are also adjuvants added to stabilize the formulation of the vaccine. Immunological adjuvant is well known in the art [J Biomed Biotechnol. 2012; 2012: 831486. Published online Mar 13, 2012].
  • Immunological adjuvant includes inorganic adjuvant such as aluminum salt and organic adjuvant such as oil based, virosome, squalane.
  • Organic adjuvants include, but are not limited to, emulsions, microbe-derived, synthetic adjuvants, cytokines, and the like. 9 kinds of cytokine adjuvant are known.
  • GM-CSF granulocyte-macrophage colony-stimulating factor
  • the adjuvant herein may be an adjuvant that increases MDSC.
  • the use of an adjuvant that increases MDSC inhibits the immune response and thus cannot achieve the effect of the vaccine. In this situation, a means is needed to prevent the increased action of MDSC of the adjuvant used.
  • Appropriate dosages of the adjuvant referred to herein are already well known in the art and can be administered according to criteria known in the art, depending on the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 ⁇ g / kg / day to 10 g / kg / day, more specifically 10 ⁇ g / kg / day to 100 mg / kg / day, and more specifically, 50 ⁇ g / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
  • intradermal doses of 7 to 700 mg prior to administration of the peptides disclosed herein such as from 1 minute to 150 minutes before, 5 to 80 minutes before, or 10 to 15 minutes before May be administered.
  • the administration time may be administered at least 1 minute before, at least 3 minutes, at least 5 minutes, at least 7 minutes, at least 8 minutes, at least 9 minutes or at least 10 minutes before the peptide administration.
  • the dosage can be at least 7 mg, at least 10 mg, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 60 mg or at least 70 mg.
  • the dosage may be 700 mg or less, 600 mg or less, 500 mg or less, 400 mg or less, 300 mg or less, 200 mg or less, 100 mg or less, 90 mg or less, or 80 mg or less.
  • GM-CSF which is added as an adjuvant for vaccines in preclinical trials, is known to increase MDSC in tumor micro-environments [Curran MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69: 7747-7755.
  • low doses of GM-CSF as a vaccine adjuvant are known to increase the number of MDSCs in the blood [Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani].
  • GM-CSF has a big limitation that it is difficult to use as an adjuvant for cancer vaccine.
  • the present inventors have found a breakthrough method that can solve the problem of the loss of MDSC reduction ability by the combination of gemcitabine and 5-fluorouracil (or capecitabine) and the action of increasing MDSC in the tumor micro-environment of GM-CSF. Came out.
  • administration of low dose GM-CSF as an adjuvant of PEP1 did not increase Lin-DR-CD11b + MDSC in a group of patients receiving GemCap and PEP1 at the same time.
  • the composition may be a pharmaceutical, cosmetic or food composition.
  • each active ingredient may be administered according to criteria known in the art according to the condition of each patient.
  • Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 ⁇ g / kg / day to 10 g / kg / day, more specifically 10 ⁇ g / kg / day to 100 mg / kg / day, and more specifically, 50 ⁇ g / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
  • composition according to one aspect of the present invention can be applied to all animals including humans, dogs, chickens, pigs, cattle, sheep, guinea pigs or monkeys.
  • composition according to one aspect of the present invention may be administered orally, rectal, transdermal, intravenous, intramuscular, intraperitoneal, intramedullary, intradural or subcutaneous.
  • Formulations for oral administration may be, but are not limited to, tablets, pills, soft or hard capsules, granules, powders, solutions or emulsions.
  • Formulations for parenteral administration may be, but are not limited to, injections, drops, lotions, ointments, gels, creams, suspensions, emulsions, suppositories, patches or sprays.
  • compositions according to one aspect of the invention may include additives such as diluents, excipients, lubricants, binders, disintegrants, buffers, dispersants, surfactants, colorants, flavoring or sweetening agents as needed.
  • additives such as diluents, excipients, lubricants, binders, disintegrants, buffers, dispersants, surfactants, colorants, flavoring or sweetening agents as needed.
  • Pharmaceutical compositions according to one aspect of the invention may be prepared by conventional methods in the art.
  • Preferred embodiments of the invention include the most optimal mode known to the inventors for carrying out the invention. Variations of the preferred embodiments may become apparent to those skilled in the art upon reading the foregoing description. The inventors expect those skilled in the art to make appropriate use of such variations, and the inventors expect the invention to be practiced in a manner different from that described herein. Accordingly, the invention includes all modifications and equivalents of the subject matter referred to in the appended claims, as permitted by patent law. Moreover, any combination of the abovementioned elements within all possible variations is included in the invention unless expressly stated to the contrary or apparently contradictory in context. While the invention has been particularly shown and described with reference to exemplary embodiments, those skilled in the art will understand that various changes in form and detail may be made without departing from the spirit and scope of the invention as defined by the following claims .
  • a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof, was prepared according to a conventional solid phase peptide synthesis method. Specifically, peptides were synthesized by coupling amino acids one by one from the C-terminus through Fmoc solid phase synthesis (SPPS) using ASP48S (Peptron, Inc., Daejeon, Korea). As follows, the first amino acid at the C-terminus of the peptides was attached to the resin. For example:
  • Coupling reagent is HBTU [2- (1H-Benzotriazole-1-yl) -1,1,3,3-tetamethylaminium hexafluorophosphate] / HOBt [N-Hydroxxybenzotriazole] / NMM [4-Methylmorpholine] It was. Fmoc removal was performed using piperidine in DMF in 20% of DMF.
  • Each peptide was synthesized by repeating a process of reacting the amino acids with each other, washing with a solvent, and then deprotecting the amino acid using the state in which the amino acid protecting group was bound to the solid support.
  • the synthesized peptide was separated from the resin and then purified by HPLC, and confirmed by MS and lyophilized.
  • Pep 1 (EARPALLTSRLRFIPK) consisting of SEQ ID NO: 1, the specific process is as follows.
  • Example 2 (arm3): 21 pancreatic cancer patients, gemcitabine-capecitabine (GemCap) + pep1-GM-CSF administration
  • Gemcitabine (1,000 mg / m 2 ) was administered intravenously three times weekly at four week intervals, while capecitabine was administered orally twice a day for three weeks and not for one week.
  • the dosage 1,660mg / m 2 / day was (830mg / m 2 1 twice daily).
  • the MDSCs trajectories of 19 patients with arm2 during and after GemCap treatment and two cycles of GemCap treatment were analyzed.
  • the first pep1 was administered intra-dermally 0.56 mg of PEP1 on the 1st, 3rd, and 5th days of week 1, and the 2nd, 3rd, 4th, 6th, and 10th weeks, and 75mg GM 10-15 minutes before pep1 administration.
  • -CSF was administered intradermally at the same site as pep1 with the adjuvant of pep1.
  • Peripheral blood samples were taken before and after GemCap administration. Arm2 patients receiving only gemcitabine and capecitabine were taken with blood samples after 7 weeks of treatment, before the sixth gemcitabine, and when capecitabine was administered. After 10 weeks of treatment, the 7th gemcitabine immediately and before the 8th gemcitabine, and before the capecitabine, will be consistent with the immunomonitoring timing of the arm3 patient group receiving gemcitabine, capecitabine, and pep1. When a blood sample was taken.
  • PBMCs were separated using a Ficoll-Hypaque gradient, the cells were counted, frozen at minus 80 ° C for later analysis and stored in liquid nitrogen.
  • Peripheral blood mononuclear blood cells were recovered using 0.15 M phosphate-buffed saline solution (Dulbecco's A) (Oxoid, UK). Subsamples of recovered cells were used for MDSC analysis and the LIVE / DEAD Cell Stain kit (Invitrogen, UK) was used to distinguish between living and dead cells.
  • Anti-human monoclonal antibodies were used for analysis using flow cytometry. Cells were immunostained, washed with binding buffer solution, and analyzed using a MACSQuant flow cytometer using MACSQuantify software (Miltenyi Biotec).
  • DTH Delayed-type hypersensitivity
  • DTH reaction was accompanied by erythema and sclerosis, and the average diameter was 5mm.
  • PBMCs PBMCs were aliquoted into 48-well plates (ThermoFisher Scientific, USA) to be 2x10 6 cells per well and then X-VIVO (Lonza) containing 10% pooled human serum (Innovative Research, USA), 20 ug / ml pep1 peptide. , UK) cultured for 3 days. After incubation, 10 units / ml IL-2 (Peprotech, UK) was added to the medium. On day 11 of cultivation, after harvesting PEP1 cells, 1 ⁇ 10 5 cells per well were dispensed into round-bottom 96 well plates.
  • Cytokine levels of patient serum collected during PBMC collection were analyzed using BioRad BioPlex 27 Assay of BioRad BioPlex Instrument.
  • Tumor burden was measured using the sum of the long axis measurement of the tumor lesion and the short axis measurement of the pathological lymph.
  • Welch's correction and unpaired t-test were used to compare median values of Lin-DR-CD11b + cells in advanced pancreatic cancer patients and controls. Spearman's rank test was used to analyze the association between the MDSC baseline and cytokine baseline values, and the nonparametric Mann-Withney test was used to analyze the differences in cytokine dichotomized from the median MDSC values. Paired Wilcoxon tests were used to compare cytokine levels post-chemotherapy post-treatment.
  • the pre-treated MDSCs were subtracted from the post-treatment MDSC values.
  • the calculated data are asymmetrically distributed and graphically logged at log scale, but all analyzes represent the original dimensions using a nonparametric approach.
  • Wilcoxon signed rantks test was used to analyze differences in each treatment group.
  • the Wilcoxon two-sample tests were used to compare pre-treatment MDSC values, post-treatment MDSC values, MDSC difference absolute values, and MDSC difference% absolute values in arm 2 and arm 3 treatment groups.
  • Wilcoxon two-sample tests were used to compare the% absolute value of the (PD) MDSC difference between patients with disease control (PR, SD) and patients with progressive disease.
  • sensitivity analyzes include reanalyzed data on patients who are suppressing the disease to eliminate the effects of tumor size changes, and on reconstructed data on arm3 after 10 weeks of follow-up.
  • PBMCS peripheral blood mononuclear cells
  • the pretreatment baseline of 21 patients was used to calculate the association between pretreatment median and inflammatory cytokine MDSC levels.
  • Tumor volume Eight out of 19 patients (comparative example 2, arm 2) who received a combination of gemcitabine and capecitabine had decreased Lin-DR-CD11b + cell numbers. Of seven patients with advanced cancer (progressive disease, PD), five Lin-DR-CD11b + cell levels were elevated and two levels were reduced (range -60 to +662%). Of 10 patients with no advanced cancer (Stable disease, SD), 6 Lin-DR-CD11b + cell levels were elevated and 4 levels were decreased (range -68- + 604%). Patients who responded partially to the Pep1 vaccine had decreased Lin-DR-CD11b +%. The tumor size was accurately measured and no significant increase or decrease was found in the sum of the longest diameters of the significant tumors in 8 patients out of 10.
  • Lin-DR-CD11b +% increased in 5 patients and Lin-DR-CD11b +% decreased in 3 patients.
  • Lin-DR-CD11b + was not decreased by gemcitabine and capecitabine itself.
  • Changes in Lin-DR-CD11b +% tended to track tumor response. This has been demonstrated in patients with Lin-DR-CD11b + reference values greater than the median, and the reduction of MDSCs will be of great help immunologically in this patient group (Table 3).
  • the positive proliferation assay (the development of a positive DTH to PEP1) was analyzed in 21 arm3 patients who received GemCap and PEP1 at the same time. Nine of the 21 patients had an immune response, and eight of the nine had decreased MDSC% during treatment. The baseline value of Lin-DR-CD11b +% in 6 of 9 patients was higher than the median of patients and MDSC levels decreased.
  • Both granular and mononuclear MDSCs were extracted from fresh mesothelioma tissue and subjected to inhibition assay in the microenvironment of mesothelioma.
  • the experiment also uses Pemetrexed, which, when treated with mesothelioma, induces an senescence-associated secretory phenotype that results in STAT3 activation in tumor cells.
  • pep1 on cytokine release from tumor cells, pep1 on myeloid cell differentiation and pep1 on the inhibitory activity of MDSC.
  • DC maturation experiments were performed using adherent monocytes obtained from PBMCs from healthy donors. 5-day culture was performed. The conditions were as follows.
  • MoDCs Monocyte-derived Dendritic Cells
  • Mesothelioma cells were plated in 25 flasks and treated with the following samples at 40-50% confluence.
  • Example 5 Mesothelioma cell line + GV1001 (MCLG)
  • Example 6 Mesothelioma cell line + GV1001 / pemetrexed (MCLGP)
  • the treatment was performed for 24 hours, after which the medium was removed and replaced with fresh medium. After 36 hours the supernatant was collected and stored. Thereafter, the culture was incubated for 5 days using a comparative example without pep1 and an example with pep1 as a medium.
  • the purified MDSC from the patient was treated with pep1 (also + pemetrexed) overnight, and the cells were washed and used for the mixed lymphocyte reaction (MLR) for inhibition assay.
  • MDSCs were plated in 96 wells and treated at each concentration for MLR the next day (CD3 / CD28 stimulation). Apart from that, the treatment was performed during MLR and the treatment was also added to T cell treatment as a control.

Abstract

Disclosed is a novel use of a peptide including SEQ ID NO: 1, a peptide having at least 80% sequence identity to the peptide sequence, or a peptide as a fragment thereof. Specifically, disclosed is a use of the peptide for inhibiting myeloid-derived suppressor cells (MDSCs). The suppression problem of the immune response by MDSC can be solved by inhibiting MDSC. The peptide is used in combination with other anticancer agents and adjuvants, thereby effectively inhibiting MDSC, and thus can relieve, treat, or prevent MDSC-related diseases or symptoms. Furthermore, provided can be a vaccine having no side effects associated with MDSC, especially, a cancer vaccine, an anticancer kit, or an anticancer method.

Description

골수-유래 억제세포(MYELOID-DERIVED SUPPRESSOR CELLS, MDSCS) 저해용 조성물Bone Marrow-Derived Suppressor Cells (MYELOID-DERIVED SUPPRESSOR CELLS, MDSCS)
본 명세서에서는 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물, MDSC 저해용 키트, MDSC 저해 방법, 항암용 조성물, 항암용 키트, 항암 방법 또는 텔로머라아제 펩티드의 신규한 용도에 관한 것이다. In the present specification, a composition for inhibiting myeloid-derived suppressor cells (MDSCs), a kit for inhibiting MDSC, a method for inhibiting MDSC, a composition for anticancer, a kit for anticancer, an anticancer method, or a novel use of telomerase peptide It is about.
골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs)는 다양한 종양-분비 인자들에 의해 분화 프로그램이 정체된 미성숙 골수 세포의 이종 패밀리이다. MDSC는 다양한 방법으로 세포독성 T 림프구 (cytotoxic T lymphocyte)의 활성을 저해한다.Myeloid-derived suppressor cells (MDSCs) are a heterogeneous family of immature bone marrow cells that have had differentiation programs stagnated by various tumor-secreting factors. MDSCs inhibit the activity of cytotoxic T lymphocytes in a variety of ways.
생체 내에서 MDSC의 기능은 부분적으로는 급성 및 만성 감염 시 과도한 면역 자극의 해로운 영향들로부터 생체를 보호하는 것과 트라우마에 의해 방출된 조직 항원들에 대한 자가면역 반응의 발생을 제한하는 것이다. 이와 같이, MDSC의 작용이 자가면역 반응을 억제하는 점은 유익하지만, MDSC의 증식은 병리학적 관점과도 관련된다. 여러 전사 인자들을 통해 활성화가 매개되고 그 결과 ARG1 및 NOS2와 같은 면역억제 인자의 업 레귤레이션 및 발현이 일어나며 NO, ROS, RNS 및 사이토킨의 생산이 증가된다. 특히 MDSC의 축적은 면역 억제 환경이 지속되도록 하여 생체가 지속적으로 알러겐 및/또는 바이러스 감염에 노출되도록 만들며, 이를 통해 만성 염증이 나타날 수 있다. 생체가 효과적인 면역 반응을 할 수 없게 되면 만성 염증 시 조직 손상이 일어나게 될 수 있다. The function of MDSC in vivo is, in part, to protect the body from the harmful effects of excessive immune stimulation in acute and chronic infections and to limit the development of autoimmune responses to tissue antigens released by trauma. As such, while the action of MDSCs inhibits the autoimmune response, the proliferation of MDSCs is also associated with a pathological point of view. Activation is mediated through several transcription factors, resulting in upregulation and expression of immunosuppressive factors such as ARG1 and NOS2 and increased production of NO, ROS, RNS and cytokines. In particular, the accumulation of MDSC allows the immunosuppressive environment to be sustained, which in turn exposes the living body to allergen and / or viral infections, which can lead to chronic inflammation. Chronic inflammation can lead to tissue damage if the body is unable to respond effectively.
전임상 모델에서 MDSC를 직접적으로 감소시키는 것으로 알려진 화학요법제는 젬시타빈(gemcitabine) 및 5-플루오로우라실(5-fluorouracil, 5-FU)뿐이다. 젬시타빈은 종양-유발 마우스에서 지라의 MDSC 수를 현저히 감소시키는 것으로 보고되고 있다[Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM (2005) Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 11:6713-6721]. 5-FU 또한 MDSC의 퍼센트를 현저히 감소시키는 것으로 알려져 있는데, 그 감소 정도가 젬시타빈보다 더 크다고 보고되고 있다.The only chemotherapeutic agents known to directly reduce MDSC in preclinical models are gemcitabine and 5-fluorouracil (5-FU). Gemcitabine has been reported to significantly reduce the number of MDSCs of the spleen in tumor-induced mice [Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM (2005) Gemcitabine eliminate eliminates splenic Gr-1 + / CD11b + myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 11: 6713-6721]. 5-FU is also known to significantly reduce the percentage of MDSC, which is reported to be greater than gemcitabine.
MDSC를 억제하기 위한 다른 방법으로 miR-142 및/또는 miR-223 리보뉴클레오티드를 MDSC에 가하는 방법이 있다(WO 2013/082591). miR-142 및/또는 miR-223 리보뉴클레오티드는 MDSC가 마크로파지, 수지상 세포 등으로 분화되도록 하여 그 수를 줄일 수 있다. Another method for inhibiting MDSC is to add miR-142 and / or miR-223 ribonucleotides to MDSC (WO 2013/082591). miR-142 and / or miR-223 ribonucleotides can be reduced in number by allowing MDSCs to differentiate into macrophages, dendritic cells, and the like.
MDSC를 억제하기 위한 또 다른 방법으로서, 어쥬번트로서 비스포스포네이트 또는 CCR2 저해제를 사용하는 방법이 알려져 있다(WO 2011/116299). 비스포스포네이트로는 클로드로네이트, 졸레드로네이트, 팔미드로네이트, 에티드로네이트 또는 다른 비스포스포네이트 드럭을 들 수 있다. CCR2 저해제로는 RS 1028595 또는 PF-04178903를 들 수 있다. As another method for inhibiting MDSC, a method using bisphosphonates or CCR2 inhibitors as adjuvant is known (WO 2011/116299). Bisphosphonates include clodronate, zoleronate, palmidronate, ethidronate or other bisphosphonate drugs. CCR2 inhibitors include RS 1028595 or PF-04178903.
한편, 전 임상에서 백신에 대한 어쥬번트로 첨가되는 그래뉼로사이트-마크로파지 콜로니-자극 인자(granulocyte-macrophage colony-stimulating factor, GM-CSF)가 종양 마이크로-환경에서는 MDSC를 증가시키는 것으로 알려져 있다[Curran MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69:7747-7755]. 또한, 임상에서도 백신 어쥬번트로서 GM-CSF를 저용량으로 사용한 결과 혈중 MDSC의 수를 증가시키는 것으로 알려져 있다[Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani G, Rivoltini L et al (2007) Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 25:2546-2553].Meanwhile, granulocyte-macrophage colony-stimulating factor (GM-CSF), which is added as an adjuvant for vaccines in all clinical trials, is known to increase MDSC in the tumor micro-environment [Curran]. MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69: 7747-7755. In addition, low doses of GM-CSF as a vaccine adjuvant are known to increase the number of MDSCs in the blood [Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani G]. , Rivoltini L et al (2007) Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 25: 2546-2553.
[선행기술문헌][Preceding technical literature]
[특허문헌][Patent Documents]
(1)WO 2013/082591(1) WO 2013/082591
(2)WO 2011/116299(2) WO 2011/116299
[비특허문헌][Non-Patent Documents]
(1)Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM (2005) Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 11:6713-6721(1) Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM (2005) Gemcitabine selectively eliminates splenic Gr-1 + / CD11b + myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res 11: 6713-6721
(2)Curran MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69:7747-7755(2) Curran MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69: 7747-7755
(3)Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani G, Rivoltini L et al (2007) Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 25:2546-2553(3) Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani G, Rivoltini L et al (2007) Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 25: 2546-2553
일 측면에서, 본 발명의 목적은, MDSC를 저해하는 것이다. In one aspect, the object of the present invention is to inhibit MDSC.
다른 측면에서, 본 발명의 목적은, 면역을 활성화시키는 것이다.In another aspect, the object of the present invention is to activate immunity.
다른 측면에서, 본 발명의 목적은, MDSC에 의해 저해되는 면역을 활성화시키는 것이다.In another aspect, the object of the present invention is to activate immunity inhibited by MDSC.
다른 측면에서, 본 발명의 목적은, MDSC에 의해 백신의 면역 반응이 저해되는 문제를 해결하기 위한 것이다. In another aspect, an object of the present invention is to solve the problem that the immune response of the vaccine is inhibited by MDSC.
다른 측면에서, 본 발명의 목적은, MDSC에 의해 암 백신의 면역 반응이 저해되는 문제를 해결하기 위한 것이다. In another aspect, an object of the present invention is to solve the problem that the immune response of a cancer vaccine is inhibited by MDSC.
다른 측면에서, 본 발명의 목적은, MDSC와 관련된 질병 또는 증상들을 완화, 치료 또는 예방하는 것이다.In another aspect, an object of the present invention is to alleviate, treat or prevent a disease or condition associated with MDSC.
다른 측면에서, 본 발명의 목적은, MDSC에 따른 면역 반응 저하의 부작용이 없는 항암 조성물, 항암 키트 또는 항암 방법을 제공하는 것이다. In another aspect, it is an object of the present invention to provide an anticancer composition, anticancer kit or anticancer method that is free of the side effects of lowering the immune response according to MDSC.
다른 측면에서, 본 발명의 목적은, MDSC에 따른 면역 반응 저하의 부작용이 없는 암 백신을 제공하는 것이다. In another aspect, an object of the present invention is to provide a cancer vaccine without the side effects of lowering the immune response according to MDSC.
일 측면에서, 본 발명은 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물로서, 상기 조성물은 MDSC 저해를 위한 유효성분으로서 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하며, 상기 펩티드는 MDSC 저해에 효과적인 양으로 포함된, 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물이다. In one aspect, the present invention is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), the composition is a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, the peptide sequence and 80% Peptide having a sequence homology, or a fragment thereof, the peptide is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), contained in an amount effective for MDSC inhibition.
다른 측면에서 본 발명은, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; 항암제; 및 어쥬번트를 포함하는 MDSC 저해용 조성물일 수 있다. In another aspect, the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be a composition for inhibiting MDSC containing an adjuvant.
또 다른 측면에서 본 발명은, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; 항암제; 및 어쥬번트를 포함하는 항암 조성물일 수 있다. In another aspect, the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be an anticancer composition comprising an adjuvant.
또 다른 측면에서 본 발명은, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; 항암제; 및 어쥬번트를 포함하는 암 백신 조성물일 수 있다. In another aspect, the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And cancer vaccine compositions comprising an adjuvant.
일 측면에서, 본 발명은 MDSC 저해용 키트일 수 있다. 상기 키트는, MDSC 저해를 위한 유효성분으로서 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하며, 상기 펩티드는 MDSC 저해에 효과적인 양으로 포함된 MDSC 저해용 조성물; 항암제; 및 설명서를 포함하는 것일 수 있다. 일 측면에서, 상기 키트는 어쥬번트를 더 포함할 수 있다.In one aspect, the invention may be a kit for inhibiting MDSC. The kit includes a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, a peptide having 80% or more sequence homology with the peptide sequence, or a peptide thereof, wherein the peptide is effective for inhibiting MDSC. MDSC inhibition composition contained in an amount; Anticancer agents; And instructions. In one aspect, the kit may further comprise an adjuvant.
일 측면에서, 상기 MDSC 저해용 키트는, MDSC 저해를 통해 MDSC에 의한 질병 또는 증상을 개선, 예방 또는 치료하는 것일 수 있다. In one aspect, the kit for inhibiting MDSC may be to improve, prevent or treat a disease or symptom caused by MDSC through MDSC inhibition.
다른 측면에서, 상기 MDSC 저해용 키트는, MDSC 저해를 통해 암을 개선, 예방 또는 치료하는 것일 수 있다.In another aspect, the kit for inhibiting MDSC may be to improve, prevent or treat cancer through MDSC inhibition.
일 측면에서, 본 발명은 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하는 조성물; 항암제; 및 설명서를 포함하는 항암 키트일 수 있다. 상기 키트는 어쥬번트 조성물을 더 포함할 수 있다. In one aspect, the invention provides a composition comprising a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And an anticancer kit comprising instructions. The kit may further comprise an adjuvant composition.
일 측면에서, 본 발명은 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs)를 저해하는 방법으로서, MDSC 저해에 효과적인 양의, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를, MDSC 저해가 필요한 대상에 투여하는 것을 포함하는 방법일 수 있다. 일 측면에서, 상기 방법은, 항암제를 상기 펩티드와 조합하여 투여하는 것을 더 포함할 수 있다. 다른 측면에서 상기 방법은, 어쥬번트를 상기 펩티드와 조합하여 투여하는 것을 더 포함할 수 있다. In one aspect, the present invention provides a method for inhibiting myeloid-derived suppressor cells (MDSCs), an effective amount of a peptide comprising SEQ ID NO: 1, the peptide sequence and at least 80% sequence Or a peptide that is homologous, or a fragment thereof, to a subject in need of MDSC inhibition. In one aspect, the method may further comprise administering an anticancer agent in combination with the peptide. In another aspect, the method may further comprise administering an adjuvant in combination with the peptide.
다른 측면에서 본 발명은, 암을 개선, 치료 또는 예방하는 방법으로서, 효과적인 양의, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를, 항암제 및/또는 어쥬번트와 조합하여, 암의 개선, 치료 또는 예방이 필요한 대상에 투여하는 것을 포함하는 암 개선, 치료 또는 예방 방법일 수 있다. In another aspect, the present invention provides a method for improving, treating or preventing cancer, wherein the peptide comprises an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof. And in combination with an anticancer agent and / or adjuvant, may be a method for cancer improvement, treatment or prevention comprising administering to a subject in need of improvement, treatment or prevention of cancer.
일 측면에서, 본 발명은, 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물을 제조하기 위한, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드의 용도일 수 있다.In one aspect, the present invention, to prepare a composition for inhibiting myeloid-derived suppressor cells (MDSCs), a peptide comprising SEQ ID NO: 1, having a sequence homology of 80% or more of the peptide sequence Peptides, or fragments thereof, may be used.
일 측면에서, 본 발명은, MDSC를 저해할 수 있다. In one aspect, the present invention may inhibit MDSC.
다른 측면에서, 본 발명은, 면역을 활성화킬 수 있다.In another aspect, the present invention may activate immune.
다른 측면에서, 본 발명은, MDSC에 의해 저해되는 면역을 활성화시킬 수 있다. In another aspect, the present invention can activate immunity inhibited by MDSC.
다른 측면에서, 본 발명은, MDSC에 의해 백신의 면역 반응이 저해되는 문제를 해결할 수 있다. In another aspect, the present invention can solve the problem that the immune response of the vaccine is inhibited by MDSC.
다른 측면에서, 본 발명은, MDSC에 의해 암 백신의 면역 반응이 저해되는 문제를 해결할 수 있다. In another aspect, the present invention can solve the problem that the immune response of a cancer vaccine is inhibited by MDSC.
다른 측면에서, 본 발명은, MDSC와 관련된 질병 또는 증상들을 완화, 치료 또는 예방할 수 있다.In another aspect, the present invention can alleviate, treat or prevent a disease or condition associated with MDSC.
다른 측면에서, 본 발명은, MDSC에 따른 면역 반응 저하의 부작용이 없는 항암 조성물, 항암 키트 또는 항암 방법을 제공할 수 있다. In another aspect, the present invention may provide an anticancer composition, an anticancer kit, or an anticancer method that does not have the side effects of lowering the immune response according to MDSC.
다른 측면에서, 본 발명은, MDSC에 따른 면역 반응 저하의 부작용이 없는 암 백신을 제공할 수 있다. In another aspect, the present invention can provide a cancer vaccine that is free of the side effects of lowering the immune response following MDSC.
도 1은 MDSC와 전 염증성 사이토카인(proinflammatory cytokine)의 레벨간의 관계를 나타내는 그래프이다. MDSC가 높은 환자와 낮은 환자의 전 염증성 사이토카인을 비교한 결과, MDSC와 전 염증성 사이토카인(proinflammatory cytokine)의 레벨은 서로 연관성이 없는 것으로 나타났다.1 is a graph showing the relationship between MDSC and the levels of proinflammatory cytokine. Comparison of the proinflammatory cytokines of patients with high and low levels of MDSC showed no correlation between the levels of MDSC and proinflammatory cytokine.
도 2는 젬시타빈과 카페시타빈만을 병용 투여받은 환자 군 (비교예 2, arm2)와, GemCap과 pep1을 동시에 투여받은 환자 군 (실시예 2, arm3)의 MDSC 변화를 분석하고 비교한 후 로그 스케일로 나타낸 그래프이다.Figure 2 is a log after analyzing and comparing the MDSC changes in the patient group (comparative example 2, arm2) that was administered only in combination with gemcitabine and capecitabine, and the patient group (Example 2, arm3) that was simultaneously administered GemCap and pep1 It is a graph in scale.
본 명세서에서 언급된 모든 공지 문헌들은 그 전체가 본 명세서의 일부로서 통합된다. All known references mentioned herein are incorporated in their entirety as part of this specification.
본 명세서에서 "MDSC 저해"라 함은, MDSC의 수를 감소시키는 것뿐만 아니라, MDSC의 활성을 억제시키는 것까지도 포함하는 개념이다. 수를 감소시키는 것은 세포의 생성을 억제하는 것뿐만 아니라 이미 생성된 세포를 사멸시키거나 다른 세포로 분화시키는 것도 포함한다. 그 외에도 생물학적 관점에서 "저해"라고 지칭되고 있는 모든 메커니즘이 포함된다. In the present specification, "MDSC inhibition" is a concept that includes not only reducing the number of MDSCs but also inhibiting the activity of MDSCs. Reducing the number includes not only inhibiting the production of cells, but also killing or differentiating cells that have already been formed. In addition, all mechanisms that are referred to as "inhibition" from a biological standpoint are included.
본 명세서에서 "조합하여 투여" 또는 "조합되어 투여"는 물리적인 시간 개념으로 동시에 투여하는 것뿐만이 아닌, 통상의 조합 요법에서와 같이 조합되는 각 약물 고유의 투여 일정에 따라 둘 이상의 약물을 병용하여 투여하는 것을 포함한다. As used herein, “combined administration” or “combined administration” refers not only to simultaneous administration in terms of physical time, but also to two or more drugs in combination, according to the unique schedule of each drug being combined as in conventional combination therapy. Administering.
본 발명자들은 MDSC가 암 환자들에 있어 현저히 증가한 것을 발견하였다. 이는 암 환자들의 치료를 위해 투여되는 암 백신의 효과를 MDSC가 저해할 수 있으며, 그 결과 암 치료를 불가능하게 할 수 있음을 의미한다. 암 백신을 통한 암 치료를 위해서는 암 환자에서 증가되어 있는 MDSC를 감소시키는 것이 필요하다. We have found a significant increase in MDSCs in cancer patients. This means that MDSCs can inhibit the effectiveness of cancer vaccines administered for the treatment of cancer patients, and as a result can render cancer treatment impossible. For cancer treatment with cancer vaccines, it is necessary to reduce the increased MDSC in cancer patients.
또한, 본 발명자들은 MDSC를 직접적으로 감소시키는 것으로 알려져 있는 두 가지 화학요법제인 젬시타빈(gemcitabine), 및 5-플루오로우라실(5-fluorouracil, 5-FU) 또는 카페시타빈을 조합하여 사용하였을 때, 단독 사용 시와는 달리 MDSC를 감소시키지 않는다는 점을 발견하였다. 오히려 상당한 수의 환자에서 MDSC가 상승한 것으로 나타났다. 이러한 점은 젬시타빈과 5-플루오로우라실(또는 카페시타빈)의 병용의 큰 제한으로 작용될 수 있다. In addition, the present inventors used a combination of two chemotherapeutic agents, gemcitabine, and 5-fluorouracil (5-FU) or capecitabine, which are known to directly reduce MDSC. It has been found that, unlike single use, it does not reduce MDSC. Rather, a significant number of patients showed elevated MDSCs. This may act as a big limitation of the combination of gemcitabine and 5-fluorouracil (or capecitabine).
본 명세서에 개시된 펩티드를 사용하면 MDSC를 현저히 감소시킬 수 있다. MDSC를 감소시킴으로써 MDSC와 관련된 일련의 병리학적 문제들을 해결할 수 있다. Using the peptides disclosed herein can significantly reduce MDSC. Reducing MDSCs can solve a series of pathological problems associated with MDSCs.
본 명세서에 개시된 펩티드는 텔로머라제의 일부 단편들 또는 그 유사체일 수 있다. 텔로미어(telomere)는 염색체의 말단에 반복적으로 존재하는 유전 물질로서, 해당 염색체의 손상이나 다른 염색체와의 결합을 방지한다고 알려져 있다. 세포가 분열할 때마다 텔로미어의 길이는 조금씩 짧아지는데, 일정한 횟수 이상의 세포 분열이 있게 되면 텔로미어는 매우 짧아지고, 그 세포는 분열을 멈추고 죽게 된다. 반면 텔로미어를 길게 하면 세포의 수명이 연장된다고 알려져 있으며, 그 예로 암세포에서는 텔로머라제(telomerase)라는 효소가 분비되어 텔로미어가 짧아지는 것을 막기 때문에, 암세포가 죽지 않고 계속 증식할 수 있다고 알려져 있다. The peptides disclosed herein may be some fragments or analogs of telomerase. Telomere is a genetic material repeatedly present at the end of a chromosome and is known to prevent damage to the chromosome or binding to another chromosome. Each time a cell divides, the telomeres become slightly shorter. After a certain number of cell divisions, the telomeres become very short, and the cells stop dividing and die. On the other hand, elongation of telomeres is known to prolong cell life. For example, cancer cells secrete an enzyme called telomerase, which prevents telomeres from shortening, so that cancer cells can continue to proliferate without dying.
본 명세서에 개시된 펩티드는 서열 번호 1의 서열을 갖는 펩티드 또는 서열 번호 1의 서열의 단편인 펩티드와, 80% 이상, 85% 이상, 90% 이상, 95% 이상, 96% 이상, 97% 이상, 98% 이상, 99% 이상의 서열 상동성을 갖는 펩티드를 포함할 수 있다.The peptides disclosed herein are peptides having a sequence of SEQ ID NO: 1 or a peptide that is a fragment of sequence SEQ ID NO: 1, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, Peptides having at least 98%, at least 99% sequence homology.
본 발명의 일측면에서, 펩티드는 텔로머라제, 구체적으로 인간(Homo sapiens) 텔로머라제에서 유래한 펩티드를 포함한다.In one aspect of the invention, the peptide comprises telomerase, specifically a peptide derived from human ( Homo sapiens ) telomerase.
본 명세서에 개시된 펩티드는 서열번호 1을 포함하는 펩티드 또는 그 단편과 서열 상동성을 갖는 펩티드를 포함할 수 있다. 또한, 본 명세서에 개시된 펩티드는, 서열번호 1을 포함하는 펩티드 또는 그 단편들과 1개 이상의 아미노산, 2개 이상의 아미노산, 3개 이상의 아미노산, 4개 이상의 아미노산, 5개 이상의 아미노산, 6개 이상의 아미노산 또는 7개 이상의 아미노산이 변화된 펩티드를 포함할 수 있다. The peptides disclosed herein may include peptides having sequence homology with a peptide comprising SEQ ID NO: 1 or a fragment thereof. In addition, the peptides disclosed herein, peptides or fragments thereof comprising SEQ ID NO: 1 and one or more amino acids, two or more amino acids, three or more amino acids, four or more amino acids, five or more amino acids, six or more amino acids Or peptides with seven or more amino acids changed.
본 발명의 일측면에서, 펩티드는, 30개 이하의 아미노산으로 구성될 수 있다. In one aspect of the invention, the peptide may consist of up to 30 amino acids.
서열 번호 1에 기재된 펩티드는 아래 표 1과 같다. 아래 표 1의 "이름"은 펩티드를 구별하기 위해 명명한 것이다. 본 발명의 일측면에서, 서열 번호 1에 기재된 펩티드는 인간 텔로머라제의 전체 펩티드를 나타낸다. 본 발명의 다른 일측면에서, 서열 번호 1의 서열을 갖는 펩티드, 서열 번호 1의 서열의 단편인 펩티드 또는 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드는 텔로머라제에 포함된 펩티드 중 해당 위치의 펩티드를 선별해 합성한 "합성 펩티드"를 포함한다. 서열번호 2는 전체 텔로머레이즈의 아미노산 서열을 나타낸 것이다.The peptide described in SEQ ID NO: 1 is shown in Table 1 below. The "name" in Table 1 below is to distinguish peptides. In one aspect of the invention, the peptide set forth in SEQ ID NO: 1 represents the entire peptide of human telomerase. In another aspect of the present invention, a peptide having a sequence of SEQ ID NO: 1, a peptide that is a fragment of SEQ ID NO: 1, or a peptide having at least 80% sequence homology with the peptide sequence corresponds to a peptide included in telomerase. And "synthetic peptides" selected and synthesized at the positional peptides. SEQ ID 2 shows the amino acid sequence of the entire telomerase.
표 1
Figure PCTKR2014004760-appb-T000001
Table 1
Figure PCTKR2014004760-appb-T000001
본 발명의 일측면에서, 아미노산 변화는 펩티드의 물리화학적 특성이 변경되도록 하는 성질에 속한다. 예를 들어, 펩티드의 열안정성을 향상시키고, 기질 특이성을 변경시키고, 최적의 pH를 변화시키는 등의 아미노산 변화가 수행될 수 있다.In one aspect of the invention, amino acid changes belong to a property that allows the physicochemical properties of the peptide to be altered. For example, amino acid changes can be made, such as improving the thermal stability of the peptide, altering substrate specificity, changing the optimal pH, and the like.
본 명세서에서 "아미노산"이라 함은 자연적으로 펩티드로 통합되는 22개의 표준 아미노산들 뿐만 아니라 D-아이소머 및 변형된 아미노산들을 포함한다. 이에 따라, 본 발명의 일측면에서 펩티드는 D-아미노산을 포함하는 펩티드일 수 있다. 한편, 본 발명의 다른 측면에서 펩티드는 번역 후 변형(post-translational modification)된 비표준 아미노산 등을 포함할 수 있다. 번역 후 변형의 예는 인산화(phosphorylation), 당화(glycosylation), 아실화(acylation) (예컨대, 아세틸화(acetylation), 미리스토일화(myristoylation) 및 팔미토일화(palmitoylation)를 포함), 알킬화(alkylation), 카르복실화(carboxylation), 히드록실화(hydroxylation), 당화반응(glycation), 비오티닐화(biotinylation), 유비퀴티닐화(ubiquitinylation), 화학적 성질의 변화(예컨대, 베타-제거 탈이미드화, 탈아미드화) 및 구조적 변화(예컨대, 이황화물 브릿지의 형성) 를 포함한다. 또한, 펩티드 컨쥬게이트를 형성하기 위한 가교제(crosslinker)들과의 결합과정에서 일어나는 화학 반응들에 의해 생기는 아미노산의 변화, 예컨대 아미노기, 카르복시기 또는 사이드 체인에서의 변화와 같은 아미노산의 변화를 포함한다. As used herein, "amino acid" includes not only the 22 standard amino acids that are naturally incorporated into the peptide, but also D-isomers and modified amino acids. Accordingly, in one aspect of the invention the peptide may be a peptide comprising D-amino acids. Meanwhile, in another aspect of the present invention, the peptide may include a non-standard amino acid or the like which has been post-translational modified. Examples of post-translational modifications include phosphorylation, glycosylation, acylation (including, for example, acetylation, myristoylation and palmitoylation), alkylation ), Carboxylation, hydroxylation, glycation, biotinylation, ubiquitinylation, changes in chemical properties (e.g., beta-elimination deimidization) , Deamidation) and structural changes (eg, formation of disulfide bridges). It also includes changes in amino acids, such as changes in amino groups, carboxy groups or side chains, caused by chemical reactions that occur during the linkage with crosslinkers to form peptide conjugates.
본 명세서에 개시된 펩티드는 자연 그대로의 공급원으로부터 동정 및 분리된 야생형 펩티드일 수 있다. 한편, 본 명세서에 개시된 펩티드는 서열 번호 1 또는 그 단편인 펩티드와 비교하여 하나 이상의 아미노산이 치환, 결실 및/또는 삽입된 아미노산 서열을 포함하는, 인공 변이체일 수 있다. 인공 변이체에서뿐만 아니라 야생형 폴리펩티드에서의 아미노산 변화는 단백질의 폴딩(folding) 및/또는 활성에 유의한 영향을 미치지 않는 보존성 아미노산 치환을 포함한다. 보존성 치환의 예들은 염기성 아미노산(아르기닌, 리신 및 히스티딘), 산성 아미노산(글루탐산 및 아스파르트산), 극성 아미노산(글루타민 및 아스파라긴), 소수성 아미노산(루신, 이소로이신, 발린 및 메티오닌), 방향족 아미노산(페닐알라닌, 트립토판 및 티로신), 및 작은 아미노산(글리신, 알라닌, 세린 및 트레오닌)의 군의 범위 내에 있다. 일반적으로 특이적 활성을 변경시키지 않는 아미노산 치환이 본 분야에 공지되어 있다. 가장 흔하게 발생하는 교환은 Ala/Ser, Val/Ile, Asp/Glu, Thr/Ser, Ala/Gly, Ala/Thr, Ser/Asn, Ala/Val, Ser/Gly, Tyr/Phe, Ala/Pro, Lys/Arg, Asp/Asn, Leu/Ile, Leu/Val, Ala/Glu, 및 Asp/Gly, 그리고 이들과 반대인 것들이다. 보존적 치환의 다른 예는 다음 표와 같다. Peptides disclosed herein can be wild-type peptides identified and isolated from a natural source. On the other hand, a peptide disclosed herein may be an artificial variant, comprising an amino acid sequence in which one or more amino acids are substituted, deleted, and / or inserted as compared to a peptide that is SEQ ID NO: 1 or a fragment thereof. Amino acid changes in the wild type polypeptide as well as in artificial variants include conservative amino acid substitutions that do not significantly affect the folding and / or activity of the protein. Examples of conservative substitutions include basic amino acids (arginine, lysine and histidine), acidic amino acids (glutamic acid and aspartic acid), polar amino acids (glutamine and asparagine), hydrophobic amino acids (leucine, isoleucine, valine and methionine), aromatic amino acids (phenylalanine, Tryptophan and tyrosine), and small amino acids (glycine, alanine, serine and threonine). Amino acid substitutions that generally do not alter specific activity are known in the art. The most common exchanges are Ala / Ser, Val / Ile, Asp / Glu, Thr / Ser, Ala / Gly, Ala / Thr, Ser / Asn, Ala / Val, Ser / Gly, Tyr / Phe, Ala / Pro, Lys / Arg, Asp / Asn, Leu / Ile, Leu / Val, Ala / Glu, and Asp / Gly, and vice versa. Other examples of conservative substitutions are shown in the following table.
표 2
원래 아미노산 예시적인 잔기 치환 바람직한 잔기 치환
Ala (A) val; leu; ile Val
Arg (R) lys; gln; asn Lys
Asn (N) gln; his; asp, lys; arg Gln
Asp (D) glu; asn Glu
Cys (C) ser; ala Ser
Gln (Q) asn; glu Asn
Glu (E) asp; gln Asp
Gly (G) Ala Ala
His (H) asn; gln; lys; arg Arg
Ile (I) leu; val; met; ala; phe; norleucine Leu
Leu (L) norleucine; ile ; val; met; ala; phe Ile
Lys (K) arg; gln; asn Arg
Met (M) leu; phe; ile Leu
Phe (F) leu; val; ile; ala; tyr Tyr
Pro (P) Ala Ala
Ser (S) thr Thr
Thr (T) Ser Ser
Trp (W) tyr; phe Tyr
Tyr (Y) trp; phe ; thr; ser Phe
Val (V) ile; leu; met; phe; ala; norleucine Leu
TABLE 2
Original amino acid Exemplary residue substitutions Preferred residue substitution
Ala (A) val; leu; ile Val
Arg (R) lys; gln; asn Lys
Asn (N) gln; his; asp, lys; arg Gln
Asp (D) glu; asn Glu
Cys (C) ser; ala Ser
Gln (Q) asn; glu Asn
Glu (E) asp; gln Asp
Gly (G) Ala Ala
His (H) asn; gln; lys; arg Arg
Ile (I) leu; val; met; ala; phe; norleucine Leu
Leu (L) norleucine; ile; val; met; ala; phe Ile
Lys (K) arg; gln; asn Arg
Met (M) leu; phe; ile Leu
Phe (F) leu; val; ile; ala; tyr Tyr
Pro (P) Ala Ala
Ser (S) thr Thr
Thr (T) Ser Ser
Trp (W) tyr; phe Tyr
Tyr (Y) trp; phe; thr; ser Phe
Val (V) ile; leu; met; phe; ala; norleucine Leu
일 측면에서, 본 발명은 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물로서, 상기 조성물은 MDSC 저해를 위한 유효성분으로서 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하며, 상기 펩티드는 MDSC 저해에 효과적인 양으로 포함된, 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물이다. In one aspect, the present invention is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), the composition is a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, the peptide sequence and 80% Peptide having a sequence homology, or a fragment thereof, the peptide is a composition for inhibiting myeloid-derived suppressor cells (MDSCs), contained in an amount effective for MDSC inhibition.
다른 측면에서 본 발명은, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; 항암제; 및 어쥬번트를 포함하는 MDSC 저해용 조성물일 수 있다. In another aspect, the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be a composition for inhibiting MDSC containing an adjuvant.
또 다른 측면에서 본 발명은, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; 항암제; 및 어쥬번트를 포함하는 항암 조성물일 수 있다. In another aspect, the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And it may be an anticancer composition comprising an adjuvant.
또 다른 측면에서 본 발명은, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; 항암제; 및 어쥬번트를 포함하는 암 백신 조성물일 수 있다. In another aspect, the invention provides a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And cancer vaccine compositions comprising an adjuvant.
일 측면에서, 본 발명은 MDSC 저해용 키트일 수 있다. 상기 키트는 상기 언급한 MDSC 저해용 조성물; 항암제; 및 설명서를 포함하는 것일 수 있다. 일 측면에서, 상기 키트는 어쥬번트를 더 포함할 수 있다.In one aspect, the invention may be a kit for inhibiting MDSC. The kit includes the above-mentioned composition for inhibiting MDSC; Anticancer agents; And instructions. In one aspect, the kit may further comprise an adjuvant.
일 측면에서, 상기 상기 MDSC 저해용 키트는, MDSC 저해를 통해 MDSC에 의한 질병 또는 증상을 개선, 예방 또는 치료하는 것일 수 있다. In one aspect, the kit for inhibiting MDSC may be to improve, prevent or treat a disease or symptom caused by MDSC through MDSC inhibition.
다른 측면에서, 상기 MDSC 저해용 키트는, MDSC 저해를 통해 암을 개선, 예방 또는 치료하는 것일 수 있다.In another aspect, the kit for inhibiting MDSC may be to improve, prevent or treat cancer through MDSC inhibition.
일 측면에서, 본 발명은 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하는 조성물; 항암제; 및 설명서를 포함하는 항암 키트일 수 있다. 상기 키트는 어쥬번트 조성물을 더 포함할 수 있다. In one aspect, the invention provides a composition comprising a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof; Anticancer agents; And an anticancer kit comprising instructions. The kit may further comprise an adjuvant composition.
상기 언급한 키트들에서, 설명서는 펩티드를 포함하는 MDSC 저해용 조성물을 항암제와 어떻게 병용하는지에 대한 설명을 포함할 수 있다. 구체적으로, 각 약제의 투여량, 투여시간, 투여방법 등에 대한 설명을 포함할 수 있다. 그 외에도 상기 설명서는 MDSC를 저해하기 위한 용도, 항암 용도 또는 암 백신 용도가 포함될 수 있다. 구체적으로, MDSC를 저해할 필요가 있는 모든 질병 또는 증상들이 포함될 수 있다. 또한, 설명서에는 투여에 따른 부작용과 주의사항이 함께 포함될 수 있다. In the above-mentioned kits, the instructions may include a description of how the composition for inhibiting MDSC containing the peptide is used in combination with an anticancer agent. Specifically, it may include a description of the dosage, administration time, administration method and the like of each agent. In addition, the instructions may include uses for inhibiting MDSC, anticancer use or cancer vaccine use. Specifically, all diseases or symptoms that need to inhibit MDSC can be included. In addition, the instructions may include side effects and precautions associated with the administration.
일 측면에서, 본 발명은 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs)를 저해하는 방법으로서, MDSC 저해에 효과적인 양의, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를, MDSC 저해가 필요한 대상에 투여하는 것을 포함하는 방법일 수 있다. 일 측면에서, 상기 방법은, 항암제를 상기 펩티드와 조합하여 투여하는 것을 더 포함할 수 있다. 다른 측면에서 상기 방법은, 어쥬번트를 상기 펩티드와 조합하여 투여하는 것을 더 포함할 수 있다. In one aspect, the present invention provides a method for inhibiting myeloid-derived suppressor cells (MDSCs), an effective amount of a peptide comprising SEQ ID NO: 1, the peptide sequence and at least 80% sequence Or a peptide that is homologous, or a fragment thereof, to a subject in need of MDSC inhibition. In one aspect, the method may further comprise administering an anticancer agent in combination with the peptide. In another aspect, the method may further comprise administering an adjuvant in combination with the peptide.
다른 측면에서 본 발명은, 암을 개선, 치료 또는 예방하는 방법으로서, 효과적인 양의, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를, 항암제 및/또는 어쥬번트와 조합하여, 암의 개선, 치료 또는 예방이 필요한 대상에 투여하는 것을 포함하는 암 개선, 치료 또는 예방 방법일 수 있다. In another aspect, the present invention provides a method for improving, treating or preventing cancer, wherein the peptide comprises an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof. And in combination with an anticancer agent and / or adjuvant, may be a method for cancer improvement, treatment or prevention comprising administering to a subject in need of improvement, treatment or prevention of cancer.
일 측면에서 상기 암은 특별히 한정되지는 않지만 신장세포 암종(RCC), 결장직장암(CRC), 위암(GC), 흑색종, 폐암, 혈액암, 전립선암, 선암종, 전립선암 또는 췌장암일 수 있다.In one aspect, the cancer is not particularly limited, but may be renal cell carcinoma (RCC), colorectal cancer (CRC), gastric cancer (GC), melanoma, lung cancer, blood cancer, prostate cancer, adenocarcinoma, prostate cancer or pancreatic cancer.
일 측면에서, 본 발명은, 골수-유래 억제세포(myeloid-derived suppressor cells, MDSCs) 저해용 조성물을 제조하기 위한, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드의 용도일 수 있다.In one aspect, the present invention, to prepare a composition for inhibiting myeloid-derived suppressor cells (MDSCs), a peptide comprising SEQ ID NO: 1, having a sequence homology of 80% or more of the peptide sequence Peptides, or fragments thereof, may be used.
본 명세서에서, 골수-유래 억제세포(MDSC)는 종양을 갖는 개체의 골수-유래 억제세포일 수 있다. 또한, 상기 조성물은 MDSC를 저해할 필요가 있는 개체에 투여하기 위한 것일 수 있다. MDSC는 세포독성 T 림프구 (cytotoxic T lymphocyte)의 활성을 저해함으로써 면역을 억제하는 기능을 한다. 자가면역과 같이 불필요한 과도한 면역 반응을 억제하는 순 기능이 있지만, 면역 반응이 필요한 상황에서 면역을 억제하여 질병을 발생시키거나 악화시키거나 또는 적절한 치료를 방해하는 역 기능 또한 있다. 예컨대, MDSC는 종양 또는 암 환자에서 많이 증가되어 있는데, 이는 암 백신 투여의 효과를 현저히 감소시킴으로써 암 백신의 효능을 무력화시킨다. 이러한 상황에서 MDSC의 수를 효과적으로 감소시킨다면 암 치료를 원활하고 효과적으로 수행할 수 있게 될 것이다. In the present specification, bone marrow-derived suppressor cells (MDSC) may be bone marrow-derived suppressor cells of an individual having a tumor. In addition, the composition may be for administration to a subject in need of inhibition of MDSC. MDSC functions to suppress immunity by inhibiting the activity of cytotoxic T lymphocytes. There is a net function of suppressing unnecessary and excessive immune responses, such as autoimmunity, but there is also a reverse function of inhibiting immunity in situations where an immune response is needed, causing or worsening disease, or impeding proper treatment. For example, MDSC is much increased in tumor or cancer patients, which negates the efficacy of cancer vaccines by significantly reducing the effects of cancer vaccine administration. In this situation, effectively reducing the number of MDSCs will enable smooth and effective cancer treatment.
본 명세서에서 MDSC에 의한 질병 또는 증상은 당업계에 자명하다. 본 명세서에서 언급된 MDSC에 의한 질병 또는 증상은 당업계에 알려진 MDSC에 의한 질병 또는 증상 모두를 포함한다. 예컨대, 용골성 골질환(Osteolytic bone disease)과 같은 골질환; 다발성 골수종(Multiple myeloma); 교아세포종(Glioblastoma); Infections: 박테리아성 또는 기생충성 감염과 같은 감염성 질병(bacterial & parasitic infection); 급성 또는 만성 염증; 외상 스트레스(traumatic stress); 패혈증(sepsis) 및 이식(transplantation); 안구내 자가면역증(Intraocular autoimmune diseases)과 같은 자가면역증; 염증성 장 질환(Inflammatory bowel disease), 암 악액질(Cancer cachexia); 또는 폐결핵(Tuberculosis, TB)을 포함하나, 이에 제한되는 것은 아니다. Diseases or symptoms caused by MDSC herein are apparent in the art. Diseases or symptoms caused by MDSC referred to herein include all diseases or symptoms caused by MDSC known in the art. Bone diseases such as, for example, osteolytic bone disease; Multiple myeloma; Glioblastoma; Infections: bacterial & parasitic infections such as bacterial or parasitic infections; Acute or chronic inflammation; Traumatic stress; Sepsis and transplantation; Autoimmune diseases such as intraocular autoimmune diseases; Inflammatory bowel disease, Cancer cachexia; Or tuberculosis (TB), but is not limited thereto.
본 명세서에서 MDSC는 그 표현형에 무관하게 모든 MDSC를 포함할 수 있다. MDSC의 다양한 표현형은 당업계에 알려져 있다. 예컨대, 표현형은 CD15, IL4Ra, CD14, CD11b, HLA-DR, CD33, Lin, FSC, DR 및 SSC, 및 선택적으로 CD45, CD18, CD80,CD83, CD86, HLA-I 및 생존/사멸 식별자로부터 선택된 것일 수 있다. 예컨대, 표현형은 표지 IL4Ra+ 및 CD14+에 기초하는 MDSC1; 표지 IL4Ra+ 및 CD15+에 기초하는 MDSC2; 표지 IL4Ra+에 기초하는 MDSC1 및 MDSC2; 표지 Lin-, HLA-DR- 및 CD33+, 및 선택적으로 CD18+ 및 HLA-I+에 기초하는 MDSC3; 표지 CD14+, HLA-DR(-/lo), FSChi 및 SSCim에 기초하는 MDSC4; 표지 CD11b+, CD14- 및 CD15+, 및 선택적으로 FSChi, SSCim, CD80-, CD83-, CD86- 및 HLA-DR-에 기초하는 MDSC5; CD15+, SClo 및 SSChi에 기초하는 MDSC6; 및 Lin, DR 및 CD11b+에 기초하는 MDSC 등을 포함한다. 예컨대, 표현형은 Lin-DR-CD11b+일 수 있다. MDSC in this specification may include all MDSCs regardless of their phenotype. Various phenotypes of MDSC are known in the art. For example, the phenotype is one selected from CD15, IL4Ra, CD14, CD11b, HLA-DR, CD33, Lin, FSC, DR and SSC, and optionally CD45, CD18, CD80, CD83, CD86, HLA-I and survival / killing identifiers. Can be. For example, phenotypes include MDSC1 based on the labels IL4Ra + and CD14 +; MDSC2 based on the labels IL4Ra + and CD15 +; MDSC1 and MDSC2 based on the label IL4Ra +; MDSC3 based on the labels Lin-, HLA-DR- and CD33 +, and optionally CD18 + and HLA-I +; MDSC4 based on the label CD14 +, HLA-DR (− / lo), FSChi and SSCim; MDSC5 based on the labels CD11b +, CD14- and CD15 +, and optionally FSChi, SSCim, CD80-, CD83-, CD86- and HLA-DR-; MDSC6 based on CD15 +, SClo and SSChi; And MDSC based on Lin, DR, and CD11b +, and the like. For example, the phenotype can be Lin-DR-CD11b +.
본 명세서에 개시된 조성물에 있어서 펩티드의 농도는 당업계에 공지된 바에 따라 통상적으로 결정될 수 있다. 예컨대, 본 발명의 일측면에 따른 조성물은 서열번호 1 의 아미노산 서열을 포함하는(comprising) 펩티드, 상기 아미노산 서열과 80%이상의 서열 상동성을 갖는 펩티드 또는 그 단편인 펩티드를 10 ㎎/L 내지 1000㎎/L, 구체적으로 10 ㎎/L 내지 500㎎/L, 더 구체적으로 30㎎/L 내지 200㎎/L 의 함량으로 포함할 수 있으나 용량에 따른 효과의 차이를 보이는 경우 이를 적절히 조절할 수 있다. 상기 범위 또는 그 이하의 범위로 포함하는 경우 본 발명의 의도한 효과를 나타내기에 적절할 뿐만 아니라, 조성물의 안정성 및 안전성을 모두 만족할 수 있으며, 비용 대비 효과의 측면에서도 상기 범위로 포함하는 것이 적절할 수 있다.The concentration of peptides in the compositions disclosed herein can be routinely determined as known in the art. For example, the composition according to one aspect of the present invention comprises 10 mg / L to 1000 peptides comprising a peptide comprising the amino acid sequence of SEQ ID NO: 1, a peptide having a sequence homology of 80% or more with the amino acid sequence, or a fragment thereof. It may be included in the content of mg / L, specifically 10 mg / L to 500 mg / L, more specifically 30 mg / L to 200 mg / L, but when the difference in effect depending on the dose can be adjusted appropriately. When included in the above range or less, it is not only appropriate to exhibit the intended effect of the present invention, but also satisfies both the stability and safety of the composition, it may be appropriate to include in the above range in terms of cost-effectiveness. .
본 명세서에서 펩티드의 투여량, 투여방법, 투여주기 등은 이미 당업계에 널리 알려져 있으므로, 각 환자의 상태에 따라 당업계에 알려진 기준에 의해 투여할 수 있다. 구체적인 투여량 결정은 당업자의 수준 내에 있으며, 이의 1일 투여 용량은 예를 들어 구체적으로는 1 ㎍/kg/일 내지 10 g/kg/일, 더 구체적으로는 10 ㎍/kg/일 내지 100 mg/kg/일, 보다 더 구체적으로는 50 ㎍/kg/일 내지 10 mg/kg/일이 될 수 있으나, 이에 제한되지 않으며, 투여하고자 하는 대상의 연령, 건강 상태, 합병증 등 다양한 요인에 따라 달라질 수 있다. 예컨대, 펩티드는 피내 투여를 통해 투여될 수 있다. 투여간격은 2일 간격으로 하루 1회 투여될 수 있고 시간이 지나면서 날짜 간격을 더 벌릴 수도 있다. 예컨대, 1, 2, 3, 4주차에 각각 투여한 후에는 6주차, 10주차로 간격을 벌릴 수 있다. 투여량은 성인 기준으로 1회 투여시 0.1 내지 3mg일 수 있다. 투여량은 0.1 mg 이상, 0.2 mg 이상, 0.3 mg 이상, 0.4 mg 이상 0.45 mg 이상 또는 0.5mg 이상일 수 있다. 또한, 투여량은 3 mg 이하, 2.5 mg 이하, 2.0 mg 이하, 1.5 mg 이하, 1.0 mg 이하, 0.9 mg 이하, 0.8 mg 이하, 0.7 mg 이하, 0.6mg 이하일 수 있다. Since the dosage, administration method, administration cycle, and the like of the peptide herein are well known in the art, it may be administered according to the standards known in the art according to the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 μg / kg / day to 10 g / kg / day, more specifically 10 μg / kg / day to 100 mg / kg / day, and more specifically, 50 ㎍ / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be. For example, the peptide can be administered via intradermal administration. Dosage intervals may be administered once daily at two-day intervals and may be further extended over time. For example, after administration in weeks 1, 2, 3, and 4, the intervals may be spaced into weeks 6 and 10. The dosage may be 0.1 to 3 mg once administered on an adult basis. The dosage can be at least 0.1 mg, at least 0.2 mg, at least 0.3 mg, at least 0.4 mg at least 0.45 mg or at least 0.5 mg. In addition, the dosage may be 3 mg or less, 2.5 mg or less, 2.0 mg or less, 1.5 mg or less, 1.0 mg or less, 0.9 mg or less, 0.8 mg or less, 0.7 mg or less, 0.6 mg or less.
일 측면에서, 본 명세서에 기재된 펩티드는 항암제와 조합되어 투여되는 것일 수 있다. 항암제로는 화학요법제 및 생물요법제를 모두 포함하지만 이에 제한되는 것은 아니다. 특히, 화학요법제일 수 있다. 화학요법제로는 DNA 알킬화제, 대사길항제(anti-metabolites), 천연물, 호르몬 등을 모두 포함하지만 이에 제한되는 것은 아니다. 화학요법제는 대사길항제일 수 있다. 대사길항제는, DNA 및 RNA 합성을 방해하는 분자들의 그룹을 의미한다. 대부분의 대사길항제들은 DNA 및 RNA의 빌딩 블록과 유사한 구조를 갖는다. 대사길항제의 서브 타입으로는 엽산길항제(anti-folates), 플루오로피리미딘(fluoropyrimidines), 데옥시뉴클레오시드 유사체(deoxynucleoside analogues) 및 씨오퓨린(thiopurines)을 들 수 있지만 이에 제한되는 것은 아니다. 플루오로피리미딘은 플루오로우라실 및 카페시타빈을 포함하지만 이에 제한되는 것은 아니다. 카페시타빈은 5-플루오로우라실의 프로드럭이다. 데옥시뉴클레오시드 유사체(deoxynucleoside analogues)는 사이카라빈(cytarabine), 젬시타빈(gemcitabine), 데시타빈(decitabine), 비다자(Vidaza), 플루다라빈(fludarabine), 넬라빈(nelarabine), 클라드리빈(cladribine), 클로파라빈(clofarabine) 및 펜토스타틴(pentostatin)을 들 수 있지만 이에 제한되는 것은 아니다. 화학요법제는 둘 이상의 제제가 조합되어 투여되는 것일 수 있다. 예컨대, 플루오로피리미딘과 데옥시뉴클레오시드 유사체가 조합되어 투여될 수 있다. 또 다른 구체예에서, 젬시타빈과 5-플루오로우라실(또는 그 프로드럭인 카페시타빈)이 조합되어 투여될 수 있다. 또 다른 구체예에서는 젬시타빈과 카페시타빈이 조합되어 투여될 수 있다. In one aspect, the peptides described herein can be administered in combination with an anticancer agent. Anticancer agents include, but are not limited to, chemotherapeutic and biotherapeutic agents. In particular, it may be a chemotherapeutic agent. Chemotherapeutic agents include, but are not limited to, DNA alkylating agents, anti-metabolites, natural products, hormones, and the like. The chemotherapeutic agent may be a metabolic antagonist. Metabolism inhibitors refer to groups of molecules that interfere with DNA and RNA synthesis. Most metabolism inhibitors have a structure similar to the building blocks of DNA and RNA. Subtypes of metabolic antagonists include, but are not limited to, folate antagonists (anti-folates), fluoropyrimidines, deoxynucleoside analogues and thiopurines. Fluoropyrimidines include, but are not limited to, fluorouracil and capecitabine. Capecitabine is a prodrug of 5-fluorouracil. Deoxynucleoside analogues include cytarabine, gemcitabine, decitabine, vidaza, fludarabine, nelarabine, cladribine Cladribine, clofarabine and pentostatin, but are not limited thereto. The chemotherapeutic agent may be one in which two or more agents are administered in combination. For example, fluoropyrimidine and deoxynucleoside analogs can be administered in combination. In another embodiment, gemcitabine and 5-fluorouracil (or capecitabine, a prodrug thereof) may be administered in combination. In another embodiment, gemcitabine and capecitabine may be administered in combination.
본 명세서에서 언급된 항암제의 적절한 투여량은 이미 당업계에 널리 알려져 있으므로, 각 환자의 상태에 따라 당업계에 알려진 기준에 의해 투여할 수 있다. 구체적인 투여량 결정은 당업자의 수준 내에 있으며, 이의 1일 투여 용량은 예를 들어 구체적으로는 1 ㎍/kg/일 내지 10 g/kg/일, 더 구체적으로는 10 ㎍/kg/일 내지 100 mg/kg/일, 보다 더 구체적으로는 50 ㎍/kg/일 내지 10 mg/kg/일이 될 수 있으나, 이에 제한되지 않으며, 투여하고자 하는 대상의 연령, 건강 상태, 합병증 등 다양한 요인에 따라 달라질 수 있다. Appropriate dosages of the anticancer agents mentioned herein are already well known in the art and can be administered by criteria known in the art, depending on the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 μg / kg / day to 10 g / kg / day, more specifically 10 μg / kg / day to 100 mg / kg / day, and more specifically, 50 ㎍ / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
예컨대, 젬시타빈의 경우, 정맥주사로 투여될 수 있으며, 투여량 및 투여 간격은 100~10,000 mg/m2의 투여량으로 2~8주 간격으로 매주 1~6회일 수 있다. 투여량은 100 mg/m2 이상, 200 mg/m2 이상, 300 mg/m2 이상, 400 mg/m2 이상, 500 mg/m2 이상, 600 mg/m2 이상, 700 mg/m2 이상, 800 mg/m2 이상 또는 900 mg/m2 이상일 수 있다. 또는, 투여량이 10,000 mg/m2 이하, 9000 mg/m2 이하, 8000 mg/m2 이하, 7000 mg/m2 이하, 6000 mg/m2 이하, 5000 mg/m2 이하, 4000 mg/m2 이하, 3000 mg/m2 이하, 2000 mg/m2 이하, 1500 mg/m2 이하, 1400 mg/m2 이하, 1300 mg/m2 이하, 1200 mg/m2 이하, 1100 mg/m2 이하, 1050 mg/m2 이하, 1030 mg/m2 이하, 1020 mg/m2 이하 또는 1010 mg/m2 이하일 수 있다. For example, gemcitabine may be administered intravenously, and the dosage and administration interval may be 1 to 6 times weekly at 2 to 8 week intervals at a dose of 100 to 10,000 mg / m 2 . Dosage is at least 100 mg / m 2 , at least 200 mg / m 2, at least 300 mg / m 2, at least 400 mg / m 2, at least 500 mg / m 2, at least 600 mg / m 2 , at 700 mg / m 2 Or more, 800 mg / m 2 or more, or 900 mg / m 2 or more. Or, the dosage is 10,000 mg / m 2 or less, 9000 mg / m 2 or less, 8000 mg / m 2 or less, 7000 mg / m 2 or less, 6000 mg / m 2 or less, 5000 mg / m 2 or less, 4000 mg / m 2 or less, 3000 mg / m 2 or less, 2000 mg / m 2 or less, 1500 mg / m 2 or less, 1400 mg / m 2 or less, 1300 mg / m 2 or less, 1200 mg / m 2 or less, 1100 mg / m 2 Or less, 1050 mg / m 2 or less, 1030 mg / m 2 or less, 1020 mg / m 2 or less, or 1010 mg / m 2 or less.
카페시타빈의 경우, 경구 투여될 수 있다. 투여 간격은 하루 2회 투여일 수 있다. 투여량은 1일에 500 mg/m2 이상, 600 mg/m2 이상, 700 mg/m2 이상, 800 mg/m2 이상, 900 mg/m2, 1,000 mg/m2, 1,100 mg/m2, 1,200 mg/m2, 1,300 mg/m2, 1,400 mg/m2, 1,500 mg/m2, 1,600 mg/m2 이상일 수 있다. 또는, 투여량이 15,000 mg/m2 이하, 12,000 mg/m2 이하, 10,000 mg/m2 이하, 9000 mg/m2 이하, 8000 mg/m2 이하, 7000 mg/m2 이하, 6000 mg/m2 이하, 5000 mg/m2 이하, 4000 mg/m2 이하, 3000 mg/m2 이하, 2000 mg/m2 이하, 1,900 mg/m2 이하, 1,800 mg/m2 이하, 1,700 mg/m2 이하일 수 있다. 상기 1일 투여량은 2회 이상 나누어 투여될 수 있다. In the case of capecitabine, it may be administered orally. The dosing interval may be twice daily. Doses are at least 500 mg / m 2, at least 600 mg / m 2, at least 700 mg / m 2, at least 800 mg / m 2 , 900 mg / m 2 , 1,000 mg / m 2 , 1,100 mg / m 2 , 1,200 mg / m 2 , 1,300 mg / m 2 , 1,400 mg / m 2 , 1,500 mg / m 2 , 1,600 mg / m 2 or more. Or, the dosage is 15,000 mg / m 2 or less, 12,000 mg / m 2 or less, 10,000 mg / m 2 or less, 9000 mg / m 2 or less, 8000 mg / m 2 or less, 7000 mg / m 2 or less, 6000 mg / m 2 or less, 5000 mg / m 2 or less, 4000 mg / m 2 or less, 3000 mg / m 2 or less, 2000 mg / m 2 or less, 1,900 mg / m 2 or less, 1,800 mg / m 2 or less, 1,700 mg / m 2 It may be: The daily dose may be administered two or more times divided.
일 측면에서, 본 명세서에 기재된 펩티드 및/또는 항암제는 어쥬번트와 조합되어 투여되는 것일 수 있다. 상기 어쥬번트는 MDSC를 증가시키는 어쥬번트일 수 있다. 면역학적 관점에서, 어쥬번트는 타겟 항원에 대한 면역 반응을 자극하기 위해 백신에 첨가되는 것이지만, 그 자체가 면역원성을 제공하지는 않는다. 모든 백신은 염증을 유발시키므로 골수 세포들(단핵구 및 뉴트로필)을 모으는 결과를 가져온다. 이렇게 모여진 세포들 중 일부가 MDSC이다. 면역 반응 자극 목적 외에 백신의 제형 안정화를 위해 첨가되는 어쥬번트도 있다. 면역학적 어쥬번트에 대해서는 당업계에 널리 알려져 있다[J Biomed Biotechnol. 2012; 2012: 831486. Published online Mar 13, 2012]. 면역학적 어쥬번트는 알루미늄 염과 같은 무기 어쥬번트와, 오일계, 바이로좀(virosome), 스쿠알란과 같은 유기 어쥬번트를 포함한다. 유기 어쥬번트는 에멀젼, 미생물-유래, 합성 어쥬번트, 사이토킨 등이 있지만 이에 한정되는 것은 아니다. 사이토킨 어쥬번트는 9종이 알려져 있다. 예컨대, 성숙한 그래뉼로사이트 및 마크로파지를 활성화시키는 그래뉼로사이트-마크로파지 콜로니-자극 인자(granulocyte-macrophage colony-stimulating factor, GM-CSF)를 들 수 있는데, 이는 B형 간염, HIV, 암 백신에 주로 사용된다[J Biomed Biotechnol. 2012; 2012: 831486. Published online Mar 13, 2012]. 본 명세서에서 어쥬번트는 MDSC를 증가시키는 어쥬번트일 수 있다. MDSC를 증가시키는 어쥬번트를 사용하게 되면 면역 반응이 억제되고 따라서 백신의 효과를 달성할 수 없게 된다. 이러한 상황에서는 사용된 어쥬번트의 MDSC 증가 작용을 막아 줄 수단이 필요하다. In one aspect, the peptides and / or anticancer agents described herein can be administered in combination with an adjuvant. The adjuvant may be an adjuvant that increases MDSC. From an immunological point of view, the adjuvant is what is added to the vaccine to stimulate an immune response to the target antigen, but does not itself provide immunogenicity. All vaccines cause inflammation, resulting in the accumulation of bone marrow cells (monocytes and Neutrophils). Some of these cells are MDSCs. In addition to the purpose of stimulating the immune response, there are also adjuvants added to stabilize the formulation of the vaccine. Immunological adjuvant is well known in the art [J Biomed Biotechnol. 2012; 2012: 831486. Published online Mar 13, 2012]. Immunological adjuvant includes inorganic adjuvant such as aluminum salt and organic adjuvant such as oil based, virosome, squalane. Organic adjuvants include, but are not limited to, emulsions, microbe-derived, synthetic adjuvants, cytokines, and the like. 9 kinds of cytokine adjuvant are known. For example, granulocyte-macrophage colony-stimulating factor (GM-CSF), which activates mature granulosite and macrophages, is mainly used for hepatitis B, HIV, and cancer vaccines. [J Biomed Biotechnol. 2012; 2012: 831486. Published online Mar 13, 2012]. The adjuvant herein may be an adjuvant that increases MDSC. The use of an adjuvant that increases MDSC inhibits the immune response and thus cannot achieve the effect of the vaccine. In this situation, a means is needed to prevent the increased action of MDSC of the adjuvant used.
본 명세서에서 언급된 어쥬번트의 적절한 투여량은 이미 당업계에 널리 알려져 있으므로, 각 환자의 상태에 따라 당업계에 알려진 기준에 의해 투여할 수 있다. 구체적인 투여량 결정은 당업자의 수준 내에 있으며, 이의 1일 투여 용량은 예를 들어 구체적으로는 1 ㎍/kg/일 내지 10 g/kg/일, 더 구체적으로는 10 ㎍/kg/일 내지 100 mg/kg/일, 보다 더 구체적으로는 50 ㎍/kg/일 내지 10 mg/kg/일이 될 수 있으나, 이에 제한되지 않으며, 투여하고자 하는 대상의 연령, 건강 상태, 합병증 등 다양한 요인에 따라 달라질 수 있다.Appropriate dosages of the adjuvant referred to herein are already well known in the art and can be administered according to criteria known in the art, depending on the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 μg / kg / day to 10 g / kg / day, more specifically 10 μg / kg / day to 100 mg / kg / day, and more specifically, 50 ㎍ / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
예컨대, GM-CSF 의 경우 본 명세서에 개시된 펩티드 투여 전, 예컨대 1분 내지 150분 전의 시점, 5 내지 80분 전의 시점, 또는 10 내지 15분 전의 시점에 성인 기준 7 내지 700 mg의 투여량으로 피내 투여될 수 있다. 투여시간은 펩티드 투여 전 1분 이상 전, 3분 이상 전, 5분 이상 전, 7분 이상 전, 8분 이상 전, 9분 이상 전 또는 10분 이상 전에 투여될 수 있다. 또한, 150분 이하 전, 130분 이하 전, 110분 이하 전, 100분 이하 전, 90분 이하 전, 80분 이하 전, 70분 이하 전, 60분 이하 전, 50분 이하 전, 40분 이하 전, 30분 이하 전, 20분 이하 전 또는 15분 이하 전의 시점에 투여될 수 있다. 투여량은 7 mg 이상, 10 mg 이상, 20mg 이상, 30mg 이상, 40mg 이상, 50mg 이상, 60mg 이상 또는 70mg 이상일 수 있다. 또한, 투여량은 700mg 이하, 600mg 이하, 500mg 이하, 400mg 이하, 300mg 이하, 200mg 이하, 100mg 이하, 90mg 이하 또는 80mg 이하일 수 있다. For example, in the case of GM-CSF, intradermal doses of 7 to 700 mg prior to administration of the peptides disclosed herein, such as from 1 minute to 150 minutes before, 5 to 80 minutes before, or 10 to 15 minutes before May be administered. The administration time may be administered at least 1 minute before, at least 3 minutes, at least 5 minutes, at least 7 minutes, at least 8 minutes, at least 9 minutes or at least 10 minutes before the peptide administration. In addition, before 150 minutes, before 130 minutes, before 110 minutes, before 100 minutes, before 90 minutes, before 80 minutes, before 70 minutes, before 60 minutes, before 50 minutes, and before 40 minutes Before, 30 minutes or less, 20 minutes or less, or 15 minutes or less. The dosage can be at least 7 mg, at least 10 mg, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 60 mg or at least 70 mg. In addition, the dosage may be 700 mg or less, 600 mg or less, 500 mg or less, 400 mg or less, 300 mg or less, 200 mg or less, 100 mg or less, 90 mg or less, or 80 mg or less.
전 임상에서 백신에 대한 어쥬번트로 첨가되는 GM-CSF가 종양 마이크로-환경에서는 MDSC를 증가시키는 것으로 알려져 있다[Curran MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69:7747-7755]. 또한, 임상에서도 백신 어쥬번트로서 GM-CSF를 저용량으로 사용한 결과 혈 중 MDSC의 수를 증가시키는 것으로 알려져 있다[Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani G, Rivoltini L et al (2007) Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 25:2546-2553]. 백신의 면역 작용을 돕기 위해 첨가되는 어쥬번트가 오히려 MDSC의 수를 증가시킴으로써 면역 반응을 방해하게 되는 것이다. 따라서, GM-CSF는 암 백신을 위한 어쥬번트로는 사용하기가 어렵다는 큰 제한이 있다고 할 수 있다. GM-CSF, which is added as an adjuvant for vaccines in preclinical trials, is known to increase MDSC in tumor micro-environments [Curran MA, Allison JP (2009) Tumor vaccines expressing flt3 ligand synergize with ctla-4 blockade to reject preimplanted tumors. Cancer Res 69: 7747-7755. In addition, low doses of GM-CSF as a vaccine adjuvant are known to increase the number of MDSCs in the blood [Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero MC, Mariani L, Parmiani]. G, Rivoltini L et al (2007) Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 25: 2546-2553. The adjuvant added to aid the vaccine's immune function would rather interfere with the immune response by increasing the number of MDSCs. Therefore, GM-CSF has a big limitation that it is difficult to use as an adjuvant for cancer vaccine.
본 발명자들은 젬시타빈과 5-플루오로우라실 (또는 카페시타빈)의 병용에 의한 MDSC 감소 능력의 상실과 GM-CSF의 종양 마이크로-환경에서의 MDSC 증가 작용의 문제를 해결할 수 있는 획기적인 방법을 밝혀내었다. 본 발명자들은 저용량의 GM-CSF를 PEP1의 어쥬번트로 투여 시 GemCap과 PEP1을 동시에 투여받은 환자군의 Lin-DR-CD11b+MDSC를 증가시키지 않음을 밝혀냈다.The present inventors have found a breakthrough method that can solve the problem of the loss of MDSC reduction ability by the combination of gemcitabine and 5-fluorouracil (or capecitabine) and the action of increasing MDSC in the tumor micro-environment of GM-CSF. Came out. We found that administration of low dose GM-CSF as an adjuvant of PEP1 did not increase Lin-DR-CD11b + MDSC in a group of patients receiving GemCap and PEP1 at the same time.
백신 투여 전의 높은 MDSC%는 종양 관련 항원에 대해 면역 반응을 일으키는 것을 불가능하게 하지 않는다. High MDSC% before vaccination does not render it impossible to elicit an immune response to tumor associated antigens.
본 발명의 일측면에서 상기 조성물은 약학, 화장품 또는 식품 조성물일 수 있다. In one aspect of the invention the composition may be a pharmaceutical, cosmetic or food composition.
각 유효성분의 투여량, 투여방법, 투여주기 등은 이미 당업계에 널리 알려져 있으므로, 각 환자의 상태에 따라 당업계에 알려진 기준에 의해 투여할 수 있다. 구체적인 투여량 결정은 당업자의 수준 내에 있으며, 이의 1일 투여 용량은 예를 들어 구체적으로는 1 ㎍/kg/일 내지 10 g/kg/일, 더 구체적으로는 10 ㎍/kg/일 내지 100 mg/kg/일, 보다 더 구체적으로는 50 ㎍/kg/일 내지 10 mg/kg/일이 될 수 있으나, 이에 제한되지 않으며, 투여하고자 하는 대상의 연령, 건강 상태, 합병증 등 다양한 요인에 따라 달라질 수 있다.Since the dosage, administration method, administration cycle, and the like of each active ingredient are already well known in the art, it may be administered according to criteria known in the art according to the condition of each patient. Specific dosage determinations are within the level of those skilled in the art, and their daily dosage may be, for example, specifically 1 μg / kg / day to 10 g / kg / day, more specifically 10 μg / kg / day to 100 mg / kg / day, and more specifically, 50 ㎍ / kg / day to 10 mg / kg / day, but is not limited to this, depending on a variety of factors, such as age, health conditions, complications of the subject to be administered Can be.
본 발명의 일측면에 따른 조성물은 인간, 개, 닭, 돼지, 소, 양, 기니아피그 또는 원숭이를 포함하는 모든 동물에 적용될 수 있다.The composition according to one aspect of the present invention can be applied to all animals including humans, dogs, chickens, pigs, cattle, sheep, guinea pigs or monkeys.
본 발명의 일측면에 따른 약학 조성물은 경구, 직장, 경피, 정맥 내, 근육 내, 복강 내, 골수 내, 경막 내 또는 피하 등으로 투여될 수 있다.The pharmaceutical composition according to one aspect of the present invention may be administered orally, rectal, transdermal, intravenous, intramuscular, intraperitoneal, intramedullary, intradural or subcutaneous.
경구 투여를 위한 제형은 정제, 환제, 연질 또는 경질 캅셀제, 과립제, 산제, 액제 또는 유탁제일 수 있으나, 이에 제한되는 것은 아니다. 비경구 투여를 위한 제형은 주사제, 점적제, 로션, 연고, 겔, 크림, 현탁제, 유제, 좌제, 패취 또는 분무제일 수 있으나, 이에 제한되는 것은 아니다.Formulations for oral administration may be, but are not limited to, tablets, pills, soft or hard capsules, granules, powders, solutions or emulsions. Formulations for parenteral administration may be, but are not limited to, injections, drops, lotions, ointments, gels, creams, suspensions, emulsions, suppositories, patches or sprays.
본 발명의 일측면에 따른 약학 조성물은 필요에 따라 희석제, 부형제, 활택제, 결합제, 붕해제, 완충제, 분산제, 계면 활성제, 착색제, 향료 또는 감미제 등의 첨가제를 포함할 수 있다. 본 발명의 일측면에 따른 약학 조성물은 당업계의 통상적인 방법에 의해 제조될 수 있다.Pharmaceutical compositions according to one aspect of the invention may include additives such as diluents, excipients, lubricants, binders, disintegrants, buffers, dispersants, surfactants, colorants, flavoring or sweetening agents as needed. Pharmaceutical compositions according to one aspect of the invention may be prepared by conventional methods in the art.
본 명세서에서 사용된 용어들은 특정 구체예들을 설명하기 위한 목적으로만 의도된 것이지 본 발명을 한정하고자 하는 의도가 아니다. 명사 앞에 개수가 생략된 용어는 수량을 제한하고자 하는 것이 아니라 언급된 명사 물품이 하나 이상 존재하는 것을 나타내는 것이다. 용어 "포함하는", "갖는", 및 "함유하는"은 열린 용어로 해석된다(즉, "포함하지만 이에 한정되지는 않는"의 의미). The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. The term without the number before the noun is not intended to limit the quantity but rather to the presence of one or more of the mentioned noun articles. The terms "comprising", "having", and "comprising" are interpreted as open terms (ie, meaning "including but not limited to").
수치의 범위를 언급하는 것은 단지 그 범위 내에 속하는 각각의 별개의 수치들을 개별적으로 언급하는 것을 대신하는 쉬운 방법이기 때문이며, 그것이 아님이 명시되어 있지 않는, 각 별개의 수치는 마치 개별적으로 명세서에 언급되어 있는 것처럼 본 명세서에 통합된다. 모든 범위의 끝 값들은 그 범위 내에 포함되며 독립적으로 조합 가능하다. Reference to a range of numbers is simply an easy way to substitute for referring to each individual number within the range individually, and unless stated otherwise, each individual number is referred to individually as if It is incorporated herein as is. The end values of all ranges fall within that range and can be combined independently.
본 명세서에 언급된 모든 방법들은 달리 명시되어 있거나 문맥에 의해 명백히 모순되지 않는 한 적절한 순서로 수행될 수 있다. 어느 한 실시예 및 모든 실시예 또는 예시적 언어 (예컨대, "~과 같은")를 사용하는 것은, 청구범위에 포함되어 있지 않는 한, 단지 본 발명을 더 잘 기술하기 위함이지 본 발명의 범위를 제한하고자 함이 아니다. 명세서의 어떤 언어도 어떤 비청구된 구성요소를 본 발명의 실시에 필수적인 것으로 해석되어서는 아니된다. 다른 정의가 없는 한, 본 명세서에 사용되는 기술적 및 과학적 용어들은 본 발명이 속하는 기술 분야에서 통상의 지식을 갖는 사람에 의해 통상 이해되는 것과 같은 의미를 갖는다. All methods mentioned herein may be performed in the proper order unless otherwise specified or clearly contradicted by context. The use of one embodiment and all embodiments or example language (eg, “such as”) is merely intended to better describe the invention, unless it is within the scope of the claims, and covers the scope of the invention. It is not intended to be limiting. No language in the specification should be construed as essential to the practice of the invention as to any unclaimed elements. Unless defined otherwise, 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.
본 발명의 바람직한 구체예들은 본 발명을 수행하기 위해 발명자에게 알려진 가장 최적의 모드를 포함한다. 바람직한 구체예들의 변이들이 앞선 기재를 읽으면 당업자에게 명백하게 될 수 있다. 본 발명자들은 당업자들이 그러한 변이를 적절히 이용하길 기대하고, 발명자들은 본 명세서에 기재된 것과 다른 방식으로 본 발명이 실시되기를 기대한다. 따라서, 본 발명은, 특허법에 의해 허용되는 것과 같이, 첨부된 특허청구범위에서 언급된 발명의 요지의 균등물 및 모든 변형들을 포함한다. 더욱이, 모든 가능한 변이들 내에서 상기 언급된 구성요소들의 어떤 조합이라도 여기서 반대로 명시하거나 문맥상 명백히 모순되지 않는 한 본 발명에 포함된다. 본 발명은 예시적인 구체예들을 참조하여 구체적으로 나타내어지고 기술되었지만, 당업자들은 하기 청구범위에 의해 정의되는 발명의 정신 및 범위를 벗어나지 않고서도 형태 및 디테일에서 다양한 변화가 행해질 수 있음을 잘 이해할 것이다 . Preferred embodiments of the invention include the most optimal mode known to the inventors for carrying out the invention. Variations of the preferred embodiments may become apparent to those skilled in the art upon reading the foregoing description. The inventors expect those skilled in the art to make appropriate use of such variations, and the inventors expect the invention to be practiced in a manner different from that described herein. Accordingly, the invention includes all modifications and equivalents of the subject matter referred to in the appended claims, as permitted by patent law. Moreover, any combination of the abovementioned elements within all possible variations is included in the invention unless expressly stated to the contrary or apparently contradictory in context. While the invention has been particularly shown and described with reference to exemplary embodiments, those skilled in the art will understand that various changes in form and detail may be made without departing from the spirit and scope of the invention as defined by the following claims .
실시예 1: 펩티드의 합성Example 1 Synthesis of Peptides
서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 종래에 알려진 고상 펩티드 합성법에 따라 제조하였다. 구체적으로, 펩티드들은 ASP48S(Peptron, Inc., 대한민국 대전)를 이용하여 Fmoc 고상 합성법(solid phase peptide synthesis, SPPS)을 통해 C-말단부터 아미노산 하나씩 커플링함으로써 합성하였다. 다음과 같이, 펩티드들의 C-말단의 첫번째 아미노산이 수지에 부착된 것을 사용하였다. 예컨대 다음과 같다:A peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof, was prepared according to a conventional solid phase peptide synthesis method. Specifically, peptides were synthesized by coupling amino acids one by one from the C-terminus through Fmoc solid phase synthesis (SPPS) using ASP48S (Peptron, Inc., Daejeon, Korea). As follows, the first amino acid at the C-terminus of the peptides was attached to the resin. For example:
NH2-Lys(Boc)-2-chloro-Trityl ResinNH 2 -Lys (Boc) -2-chloro-Trityl Resin
NH2-Ala-2-chloro-Trityl ResinNH 2 -Ala-2-chloro-Trityl Resin
NH2-Arg(Pbf)-2-chloro-Trityl ResinNH 2 -Arg (Pbf) -2-chloro-Trityl Resin
펩타이드 합성에 사용한 모든 아미노산 원료는 N-term이 Fmoc으로 보호(protection)되고, 잔기는 모두 산에서 제거되는 Trt, Boc, t-Bu (t-butylester), Pbf (2,2,4,6,7-pentamethyl dihydro-benzofuran-5-sulfonyl) 등으로 보호된 것을 사용하였다. 예컨대 다음과 같다: All amino acid sources used for peptide synthesis were protected by Fmoc with N-term and all residues removed from acid with Trt, Boc, t-Bu (t-butylester), Pbf (2,2,4,6, 7-pentamethyl dihydro-benzofuran-5-sulfonyl) and the like were used. For example:
Fmoc-Ala-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Glu(OtBu)-OH, Fmoc-Pro-OH, Fmoc-Leu-OH, Fmoc-Ile-OH, Fmoc-Phe-OH, Fmoc-Ser(tBu)-OH, Fmoc-Thr(tBu)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Gln(Trt)-OH, Fmoc-Trp(Boc)-OH, Fmoc-Met-OH, Fmoc-Asn(Trt)-OH, Fmoc-Tyr(tBu)-OH, Fmoc-Ahx-OH, Trt-Mercaptoacetic acid.Fmoc-Ala-OH, Fmoc-Arg (Pbf) -OH, Fmoc-Glu (OtBu) -OH, Fmoc-Pro-OH, Fmoc-Leu-OH, Fmoc-Ile-OH, Fmoc-Phe-OH, Fmoc- Ser (tBu) -OH, Fmoc-Thr (tBu) -OH, Fmoc-Lys (Boc) -OH, Fmoc-Gln (Trt) -OH, Fmoc-Trp (Boc) -OH, Fmoc-Met-OH, Fmoc -Asn (Trt) -OH, Fmoc-Tyr (tBu) -OH, Fmoc-Ahx-OH, Trt-Mercaptoacetic acid.
커플링 시약(Coupling reagent)으로는 HBTU[2-(1H-Benzotriazole-1-yl)-1,1,3,3-tetamethylaminium hexafluorophosphate] / HOBt [N-Hydroxxybenzotriazole] /NMM [4-Methylmorpholine] 를 사용하였다. Fmoc 제거는 20%의 DMF 중 피페리딘(piperidine in DMF)을 이용하였다. 합성된 펩타이드를 Resin에서 분리 및 잔기의 보호기 제거에는 절단 칵테일(Cleavage Cocktail) [TFA (trifluoroacetic acid) /TIS (triisopropylsilane) / EDT (ethanedithiol) / H2O=92.5/2.5/2.5/2.5] 를 사용하였다.Coupling reagent is HBTU [2- (1H-Benzotriazole-1-yl) -1,1,3,3-tetamethylaminium hexafluorophosphate] / HOBt [N-Hydroxxybenzotriazole] / NMM [4-Methylmorpholine] It was. Fmoc removal was performed using piperidine in DMF in 20% of DMF. The cleavage cocktail (TFA (trifluoroacetic acid) / TIS (triisopropylsilane) / EDT (ethanedithiol) / H 2 O = 92.5 / 2.5 / 2.5 / 2.5] was used to separate the synthesized peptide from Resin and remove the protecting group of the residue. It was.
아미노산 보호기가 결합된 출발 아미노산이 고상 지지체에 결합되어 있는 상태를 이용하여 여기에 해당 아미노산들을 각각 반응시키고 용매로 세척한 후 탈보호하는 과정을 반복함으로써 각 펩티드를 합성하였다. 합성된 펩티드를 수지로부터 끊어낸 후 HPLC로 정제하고, 합성 여부를 MS로 확인하고 동결 건조하였다. Each peptide was synthesized by repeating a process of reacting the amino acids with each other, washing with a solvent, and then deprotecting the amino acid using the state in which the amino acid protecting group was bound to the solid support. The synthesized peptide was separated from the resin and then purified by HPLC, and confirmed by MS and lyophilized.
서열번호 1로 구성된 Pep 1 (EARPALLTSRLRFIPK) 을 예로 들어 구체적인 과정을 설명하면 다음과 같다. Referring to Pep 1 (EARPALLTSRLRFIPK) consisting of SEQ ID NO: 1, the specific process is as follows.
1) 커플링 1) coupling
NH2-Lys(Boc)-2-chloro-Trityl Resin 에 보호된 아미노산(8당량)와 커플링 시약 HBTU(8당량)/HOBt(8당량)/NMM(16당량) 을 DMF에 녹여서 첨가한 후, 상온에서 2시간 동안 반응하고 DMF, MeOH, DMF순으로 세척하였다.Amino acid (8 equivalents) protected by NH 2 -Lys (Boc) -2-chloro-Trityl Resin and coupling reagent HBTU (8 equivalents) / HOBt (8 equivalents) / NMM (16 equivalents) were dissolved in DMF and added. Reaction was performed at room temperature for 2 hours, and washed with DMF, MeOH, and DMF in that order.
2) Fmoc 탈보호 2) Fmoc deprotection
20%의 DMF 중의 피페리딘(piperidine in DMF) 을 가하고 상온에서 5분 간 2회 반응하고 DMF, MeOH, DMF순으로 세척하였다.Piperidine in DMF at 20% was added thereto, reacted twice at room temperature for 5 minutes, and washed in the order of DMF, MeOH, and DMF.
3) 1과 2의 반응을 반복적으로 하여 펩타이드 기본 골격 NH2-E(OtBu)-A-R(Pbf)-P-A-L-L-T(tBu)-S(tBu)-R(Pbf)L-R(Pbf)-F-I-P-K(Boc)-2-chloro-Trityl Resin)을 만들었다. 3) Peptide base skeleton NH 2 -E (OtBu) -AR (Pbf) -PALLT (tBu) -S (tBu) -R (Pbf) LR (Pbf) -FIPK (Boc) -2-chloro-Trityl Resin).
4) 절단(Cleavagge): 합성이 완료된 펩타이드 Resin에 절단 칵테일(Cleavage Cocktail) 을 가하여 펩타이드를 Resin에서 분리하였다.4) Cleavage: A cleavage cocktail was added to the synthesized peptide Resin to separate the peptide from Resin.
5) 얻어진 mixture에 Cooling diethyl ether를 가한 후, 원심 분리하여 얻어진 펩타이드를 침전시켰다.5) Cooling diethyl ether was added to the mixture, followed by centrifugation to precipitate the peptide.
6) Prep-HPLC로 정제 후, LC/MS로 분자량을 확인하고 동결하여 powder로 제조하였다.6) After purification by Prep-HPLC, the molecular weight was confirmed by LC / MS and frozen to prepare a powder.
비교예 1, 2 및 실시예 2: 병용 투여Comparative Examples 1, 2 and Example 2: Concomitant Administration
40명의 췌장암 환자들 (비교예 2, 실시예 2), 및 환자들과 나이 및 성별이 일치하는 건강한 24명의 대조군(비교예 1)으로부터 각각 20~30ml의 정맥혈을 채취하였다. 대조군은 영국의 Royal Surrey Country Hospital의 surgical minor operation 클리닉에서 모집하였다. 40명의 환자들 모두 자가면역질환 또는 최근에 스테로이드 치료를 받은 적이 없으며, 대조군 또한 암에 걸린 적이 없다. 임상실험에 참가한 모든 대상은 서면 동의서를 제출 하였고 현지 휴먼 인베스티게이터스 (human investigators) 위원회에서 승인 받았다. 모든 환자들은 젬시타빈과 카페시타빈을 병용으로 투여(GemCap) 받았다. 40명의 환자를 두 그룹인 arm2 (비교예 2)와 arm3 (실시예 2)로 나누었다. Arm2 (비교예 2)는 19명이었고, arm3 (실시예 2)는 21명이었다. Arm2 (비교예 2)는 젬시타빈과 카페시타빈을 병용투여하였고, arm3 (실시예 2)는 젬시타빈과 카페시타빈에 더해 pep1 펩티드와 그 어쥬번트인 GM-CSF를 동시에 투여하였다. 20-30 ml of venous blood were collected from 40 pancreatic cancer patients (Comparative Example 2, Example 2) and 24 healthy controls (Comparative Example 1) of age and gender matched with the patients. Controls were recruited from a surgical minor operation clinic at Royal Surrey Country Hospital in the United Kingdom. All 40 patients have never had autoimmune diseases or recent steroid treatments, and controls have never had cancer. All subjects who participated in the study submitted written informed consent and were approved by the local human investigators committee. All patients received gemcitabine and capecitabine in combination (GemCap). Forty patients were divided into two groups, arm2 (Comparative Example 2) and arm3 (Example 2). Arm2 (Comparative Example 2) was 19, and arm3 (Example 2) was 21. Arm2 (Comparative Example 2) was treated with gemcitabine and capecitabine in combination, and arm3 (Example 2) was simultaneously administered pep1 peptide and its adjuvant GM-CSF in addition to gemcitabine and capecitabine.
- 비교예 1 (대조군): 24명의 건강한 대조군, 젬시타빈-카페시타빈(GemCap) 투여Comparative Example 1 (Control): 24 healthy controls, gemcitabine-capecitabine (GemCap) administration
- 비교예 2 (arm2): 19명의 췌장암 환자들, 젬시타빈-카페시타빈(GemCap) 투여Comparative Example 2 (arm2): 19 pancreatic cancer patients, gemcitabine-capecitabine (GemCap) administration
- 실시예 2 (arm3): 21명의 췌장암 환자들, 젬시타빈-카페시타빈(GemCap) + pep1- GM-CSF 투여Example 2 (arm3): 21 pancreatic cancer patients, gemcitabine-capecitabine (GemCap) + pep1-GM-CSF administration
젬시타빈 (1,000 mg/m2)은 정맥주사로 4주 간격으로 매주 3번 투여하였으며 이와 동시에 카페시타빈은 3주 동안은 하루에 2회 경구 투여하고 1주 간은 투여하지 않았다. 투여량은 1,660mg/m2/day (830mg/m2 1일 2회)이었다. arm2의 19명 환자들의 GemCap 치료 중 및 GemCap 치료 두 사이클 후의 MDSCs 궤도를 분석하였다. GemCap 과 pep1을 동시에 투여한 arm3의 21명 환자들의 MDSCs 궤도 또한 분석하였다. 1차 pep1은 1주차에 1,3,5일째 0.56mg의 PEP1을 피내(intra-dermally) 투여하고, 2, 3, 4, 6 및 10주차에 투여하였으며, pep1 투여 10~15분 전에 75mg GM-CSF를 pep1의 어쥬번트로 pep1과 같은 부위에 피내 투여하였다. Gemcitabine (1,000 mg / m 2 ) was administered intravenously three times weekly at four week intervals, while capecitabine was administered orally twice a day for three weeks and not for one week. The dosage 1,660mg / m 2 / day was (830mg / m 2 1 twice daily). The MDSCs trajectories of 19 patients with arm2 during and after GemCap treatment and two cycles of GemCap treatment were analyzed. We also analyzed the MDSCs trajectories of 21 patients in arm3 who received both GemCap and pep1. The first pep1 was administered intra-dermally 0.56 mg of PEP1 on the 1st, 3rd, and 5th days of week 1, and the 2nd, 3rd, 4th, 6th, and 10th weeks, and 75mg GM 10-15 minutes before pep1 administration. -CSF was administered intradermally at the same site as pep1 with the adjuvant of pep1.
말초혈액 샘플 (Peripheral blood sample)을 GemCap 투여 전 및 투여 후에 채취하였다. 젬시타빈과 카페시타빈만 병용 투여 받고 있는 arm2 환자들은 7주간의 치료 후, 6번째 젬시타빈 투여 전, 및 카페시타빈을 투여하고 있을 때 혈액 샘플을 채취하였다. 젬시타빈, 카페시타빈 및 pep1을 함께 투여 받는 arm3 환자군의 면역 모니터링(immunomonitoring) 시기와 일치하도록 10주간의 치료 후, 7번째 젬시타빈 투여 즉시 및 8번째 젬시타빈 전 및 카페시타빈을 투여하고 있을 때 혈액 샘플을 채취하였다. Peripheral blood samples were taken before and after GemCap administration. Arm2 patients receiving only gemcitabine and capecitabine were taken with blood samples after 7 weeks of treatment, before the sixth gemcitabine, and when capecitabine was administered. After 10 weeks of treatment, the 7th gemcitabine immediately and before the 8th gemcitabine, and before the capecitabine, will be consistent with the immunomonitoring timing of the arm3 patient group receiving gemcitabine, capecitabine, and pep1. When a blood sample was taken.
Arm 3군의 환자 3명은 14주째에 혈액을 채취하였고, 환자 2명은 18주에 채취하였는데, 이는 pep1의 투여시기와 일치하였다. 채취된 혈액 샘플은 li-heparin 튜브 (BD Biosciencs, Europe) 또는 CPT 튜브에 옮겨 영국의 리버풀 캔서 트라이얼 유닛 (Liverpool Cancer Trial Unit, UK)에 위치한 바이오마커 저장소 (Biomarker repository)로 옮겨졌다. PBMC는 피콜-하이파크 그레디언트(Ficoll-Hypaque gradient)를 사용하여 분리하고, 세포 수를 센 후, 차 후 분석을 위하여 마이너스 80℃에서 동결한 후 액체질소에 보관하였다. Three patients from Arm 3 group were taken at week 14 and two patients were taken at week 18, which coincided with the timing of pep1 administration. The collected blood samples were transferred to li-heparin tubes (BD Biosciencs, Europe) or CPT tubes to a Biomarker repository located in Liverpool Cancer Trial Unit (UK). PBMCs were separated using a Ficoll-Hypaque gradient, the cells were counted, frozen at minus 80 ° C for later analysis and stored in liquid nitrogen.
세포의 면역표현형 분석 (Immunophenotypic analysis of cells)Immunophenotypic analysis of cells
0.15M 포스페이트 버퍼 살린(phosphate-buffed saline) 용액 (Dulbecco's A) (Oxoid, UK)을 사용하여 말초혈액 단핵세포 (Peripherl blood mononuclear blood cells) 를 회수하였다. 회수한 세포의 부분 표본을 MDSC 분석에 사용하였으며, LIVE/DEAD Cell Stain 키트 (Invitrogen, UK)를 사용하여, 생존 한 세포와 죽은 세포를 구별하였다. Binding buffer 용액 (BD Biosciences, Europe)으로 세포 세척 후 anti-HLA-DR-APC-Cy7, anti-Lin1 (CD3,14,16,19,20,56)-FITC 및 anti-CD1b-PECy7 와 같은 항 인간 모노클로널 항체 (anti-human monoclonal antibody)는 유세포 분석기 (flow cytometry)를 이용한 분석에 사용하였다. 세포는 면역염색 (immunostaining) 후, 바인딩 버퍼(binding buffer) 용액으로 세척하고, MACSQuantify 소프트웨어 (Miltenyi Biotec)을 이용한 MACSQuant flow cytometer를 사용하여 분석하였다. Peripheral blood mononuclear blood cells were recovered using 0.15 M phosphate-buffed saline solution (Dulbecco's A) (Oxoid, UK). Subsamples of recovered cells were used for MDSC analysis and the LIVE / DEAD Cell Stain kit (Invitrogen, UK) was used to distinguish between living and dead cells. Anti-HLA-DR-APC-Cy7, anti-Lin1 (CD3,14,16,19,20,56) -FITC and anti-CD1b-PECy7 after cell washing with Binding buffer solution (BD Biosciences, Europe) Anti-human monoclonal antibodies were used for analysis using flow cytometry. Cells were immunostained, washed with binding buffer solution, and analyzed using a MACSQuant flow cytometer using MACSQuantify software (Miltenyi Biotec).
지연형 과민증 반응 검사 (Delayed-type hypersensitivity (DTH) skin tests)Delayed-type hypersensitivity (DTH) skin tests
100 μg의 pep1을 아랫배 백신 사이트 반대쪽에 피내 주사하였다. 환자들은 피내 주사 48시간 후 DTH 반응의 사이즈를 측정하고 의사에게 보고하도록 하였다. DTH 반응은 홍반 및 경화작용을 동반하였고 평균 지름 5mm 크기로 나타내었다. 100 μg of pep1 was injected intradermal opposite the belly vaccine site. Patients were asked to measure and report the size of the DTH response 48 hours after intradermal injection. DTH reaction was accompanied by erythema and sclerosis, and the average diameter was 5mm.
생체 외 세포 증식 분석 (In vitro proliferation assays)In vitro proliferation assays
해동한 PBMC는 웰 당 2x106 세포가 되도록 48-웰 플레이트 (ThermoFisher Scientific, USA)에 분주한 후 10% pooled 인간 혈청 (Innovative Research, USA), 20 ug/ml pep1 펩타이드를 포함한 X-VIVO (Lonza, UK) 배지에 3일 간 배양하였다. 배양 후, 10 units/ml IL-2 (Peprotech, UK)를 배지에 첨가하였다. 배양일 11 째, PEP1 뷰유된 세포들을 수확한 후, round-bottom 96웰 플레이트에 웰 당 1x105 세포를 분주하였다. 사전에 자극시킨 세포에 웰당 50μl 의 1x105 가 되도록irradiated (45Gy) 자가유래된 PBMC를 더해, 항원표출세포 (antigen-presenting cell) 로써 역할을 하도록 하였다, pep1에 특정한 세포증식은 100μl 대조군 배지 (pep1 (20μg/ml) 또는 양성 대조군 (5μg/ml)를 첨가하여 이틀간 배양 후 분석하였다. 배양 후, 세포 수를 세기 전 웰 당 1μCi의 3H-thymidine을 16시간 동안 첨가하였다. 4번의 계수(counting)를 통해 얻은 분당 세포 수 차이보다 stimulation index (SI)가 1.8이상 되는 것은 pep1에 대한 양성적인 증식 반응으로 규정하였다. Thawed PBMCs were aliquoted into 48-well plates (ThermoFisher Scientific, USA) to be 2x10 6 cells per well and then X-VIVO (Lonza) containing 10% pooled human serum (Innovative Research, USA), 20 ug / ml pep1 peptide. , UK) cultured for 3 days. After incubation, 10 units / ml IL-2 (Peprotech, UK) was added to the medium. On day 11 of cultivation, after harvesting PEP1 cells, 1 × 10 5 cells per well were dispensed into round-bottom 96 well plates. 50 μl of 1 × 10 5 per well was added to pre-stimulated cells to serve as antigen-presenting cells, with pep1 specific cell proliferation to 100 μl control medium (pep1). (20 μg / ml) or positive control (5 μg / ml) was added and analyzed after two days of culture After incubation, 1 μCi of 3 H-thymidine per well was counted for 16 hours before counting the cells. A higher stimulation index (SI) of 1.8 than the number of cells per minute was determined as a positive proliferative response to pep1.
사이토카인 분석 (Cytokine Analysis)Cytokine Analysis
PBMC 채취 시 수집한 환자 혈청의 사이토카인 레벨은 BioRad BioPlex Instrument의 BioRad BioPlex 27 Assay를 사용하여 분석하였다. Cytokine levels of patient serum collected during PBMC collection were analyzed using BioRad BioPlex 27 Assay of BioRad BioPlex Instrument.
종양 버든 평가(Tumor Burden assessment) Tumor Burden assessment
CT 촬영을 통해 병변 (lesion) 측정을 위한 PRECIST V1.1 CRITERIA 를 사용하여 화학요법 사전 및 사후의 종양 버든에 대해 개별적인 분석을 수행하였다. 종양 병변의 long axis 측정 값과 병리적 림프의 short axis 측정값의 합계를 사용하여 종양 버든(mm) 을 측정하였다. Individual analyzes of pre and post chemotherapy tumor burden were performed using PRECIST V1.1 CRITERIA for lesion measurement via CT scan. Tumor burden (mm) was measured using the sum of the long axis measurement of the tumor lesion and the short axis measurement of the pathological lymph.
통계학적 분석 (Statistical Analysis)Statistical Analysis
Welch's correction과 unpaired t-test를 사용하여 진행된 췌장암 환자 및 대조군의 Lin-DR-CD11b+ 세포의 중간 값을 비교하였다. Spearman's rank test를 사용하여 MDSC baseline 값과 사이토카인 베이스라인 값의 연관성을 분석하였으며, nonparametric Mann-Withney test를 사용하여 사이토카인이 MDSC 중간 값에서 이분화 (dichotomize) 될 때의 차이점을 분석하였다. Paired Wilcoxon 테스트를 통해 화학요법 사전 사후의 사이토카인 레벨을 비교하였다.Welch's correction and unpaired t-test were used to compare median values of Lin-DR-CD11b + cells in advanced pancreatic cancer patients and controls. Spearman's rank test was used to analyze the association between the MDSC baseline and cytokine baseline values, and the nonparametric Mann-Withney test was used to analyze the differences in cytokine dichotomized from the median MDSC values. Paired Wilcoxon tests were used to compare cytokine levels post-chemotherapy post-treatment.
MDSC 차이의 절대값을 산출하기 위해 치료 후의 MDSC 값에서 치료 이전 측정한 MDSCs 값을 제(subtract) 하였고 MDSC 변화 절대값을 치료 이전에 측정한 값으로 나누어 MDSC 차이의 절대값을 %로 계산하였다. 이 산출된 데이터는 비대칭으로 분포되어 있어서 로그 스케일(log scale)로 그래픽화하였으나 실시한 모든 분석은 비모수 접근법 (nonparametric approach)을 사용하여 원래의 치수를 나타낸다. Wilcoxon signed rantks test를 사용하여 각 치료 군 내의 차이점을 분석하였다. Wilcoxon two-sample tests를 사용하여 arm 2 와 arm 3 치료 군의 치료 이전 MDSC 값, 치료 이후 MDSC 값, MDSC 차이 절대 값, MDSC 차이 % 절대 값을 비교하였다. Wilcoxon two-sample tests를 사용하여 질병을 억제하고 있는 환자군 (patients with disease control) (PR, SD)와 진행성 질환의 환자군의 (PD) MDSC 차이 % 절대 값을 비교하였다. 동향의 일관성을 위해 민감도 분석은 종양 크기 변화로 인한 효과를 제거하기 위하여 질병을 억제하고 있는 환자군에 대해 재 분석한 데이터와 10주간 의 사후 치료를 한 arm3에 대해 재분석한 데이터가 포함되어 있다.  In order to calculate the absolute value of the MDSC difference, the pre-treated MDSCs were subtracted from the post-treatment MDSC values. The calculated data are asymmetrically distributed and graphically logged at log scale, but all analyzes represent the original dimensions using a nonparametric approach. Wilcoxon signed rantks test was used to analyze differences in each treatment group. The Wilcoxon two-sample tests were used to compare pre-treatment MDSC values, post-treatment MDSC values, MDSC difference absolute values, and MDSC difference% absolute values in arm 2 and arm 3 treatment groups. Wilcoxon two-sample tests were used to compare the% absolute value of the (PD) MDSC difference between patients with disease control (PR, SD) and patients with progressive disease. For consistency of trends, sensitivity analyzes include reanalyzed data on patients who are suppressing the disease to eliminate the effects of tumor size changes, and on reconstructed data on arm3 after 10 weeks of follow-up.
결과result
1. MDSC와 전 염증성 사이토카인 레벨과의 연관성1. Association of MDSC with Proinflammatory Cytokine Levels
도 1에 나타난 바와 같이, 췌장암 환자의 Lin-DR-CD11b+ 세포의 기준값과 전 염증성 사이토카인 (pro-inflammatory cytokine)의 레벨은 연관성이 없었다. As shown in FIG. 1, the reference value of Lin-DR-CD11b + cells and the level of pro-inflammatory cytokine in pancreatic cancer patients were not related.
젬시타빈과 카페시타빈을 투여 받은 40명의 진행된 췌장암 환자들의 동결보존한 말초혈액 단핵세포 (PBMCS)를 분석하였다. 40명 중 21명의 환자들은 텔로머라제 백신인 PEP1과 젬시타빈, 카페시타빈을 동시에 투여 받았다. 21명 환자들의 치료 전 기준값을 사용하여, 치료 전 중간 값과 염증성 사이토카인 MDSC 레벨의 연관성을 산출하였다. Lin-DR-CD11b+ 세포에 대한 젬시타빈과 카페시타빈의 효과를 분석하기 위해 젬시타빈과 카페시타빈만을 연속으로 투여한 19명의 환자들의 샘플을 분석하였다. 분석을 통해 얻은 표현형은 Kotsakis의 연구결과에 근거하여 MDSCs를 표시하기 위해 사용하였다. Cryopreserved peripheral blood mononuclear cells (PBMCS) of 40 patients with advanced pancreatic cancer who received gemcitabine and capecitabine were analyzed. Twenty-one of the 40 patients received the telomerase vaccine PEP1, gemcitabine and capecitabine at the same time. The pretreatment baseline of 21 patients was used to calculate the association between pretreatment median and inflammatory cytokine MDSC levels. To analyze the effects of gemcitabine and capecitabine on Lin-DR-CD11b + cells, a sample of 19 patients who received only gemcitabine and capecitabine consecutively was analyzed. The phenotype obtained from the analysis was used to represent MDSCs based on Kotsakis' findings.
대조군(비교예 1)과 비교 하였을 때 40명의 췌장암 환자군의 Lin-DR-CD11b+ 세포 수는 증가하였다 (p < 0.0001). 환자군의 Lin-DR-CD11b+ 세포의 중간 값은 1.85 (범위 0.62-845)이며, 이에 해당하는 24명 대조군의 세포 중간 값은 0.82 (범위 0.16-0.22) 이다. Compared with the control group (Comparative Example 1), the number of Lin-DR-CD11b + cells was increased in 40 pancreatic cancer patients ( p <0.0001). The median value of Lin-DR-CD11b + cells in the patient group is 1.85 (range 0.62-845), and the median value of the 24 control groups is 0.82 (range 0.16-0.22).
전체 사이토카인에 대한 데이터 (Speraman's coefficient: IL-6 = 0.153, IL-1ㅯ = 0.22, VEGF = -0.0389, TNFа = 0.0587, MCP-1 = -0.226)를 가지고 있는 33명의 췌장암 환자들에서는 염증성 사이토카인과 MDSCs 기준 값의 연관성은 발견하지 못하였다. MDSC 레벨이 미디언 값에서 이분화 될 때, MDSCs 레벨이 낮은 사람과 레벨이 높은 사람 사이에서 상기 사이토카인의 기준값의 유의한 차이는 확인할 수 없었다 (도 1). Inflammatory cytokines in 33 pancreatic cancer patients with data on total cytokines (Speraman's coefficient: IL-6 = 0.153, IL-1 '= 0.22, VEGF = -0.0389, TNFа = 0.0587, MCP-1 = -0.226) No association between Cain and MDSCs reference values was found. When the MDSC levels were divided into median values, no significant difference in the reference value of the cytokine was found between those with low MDSCs and those with high levels (FIG. 1).
2. GemCap만을 투여한 비교예 2(arm2)의 결과2. Results of Comparative Example 2 (arm2) administered with GemCap only
GemCap만을 투여한 비교예 2(arm2)의 결과, 일부에서는 Lin-DR-CD11b+ 세포 수가 감소하긴 하였으나, 이는 GemCap에 의한 것이 아니라, 질병 컨트롤이나 암 관련 염증의 정도에 의한 것으로 나타났다. 구체적으로 설명하면 다음과 같다. As a result of Comparative Example 2 (arm2) in which only GemCap was administered, the number of Lin-DR-CD11b + cells was decreased in some cases, but it was not caused by GemCap, but by disease control or the degree of cancer-related inflammation. Specifically, it is as follows.
종양의 부피 젬시타빈과 카페시타빈을 병용 투여받은 환자 19명 (비교예 2, arm 2) 중 8명은 Lin-DR-CD11b+ 세포 수가 감소하였다. 암이 진행중인 7명의 환자 (progressive disease, PD) 중 5명의 Lin-DR-CD11b+ 세포 레벨은 상승 하였으며 2명의 레벨은 감소 하였다 (범위 -60~ +662 %). 암이 진행 중이지 않은 (Stable disease, SD) 10명의 환자 중 6명의 Lin-DR-CD11b+ 세포 레벨은 상승하였으며, 4명의 레벨은 감소하였다 (범위 -68~+604 %). Pep1 백신에 대해 부분적으로 반응을 보인 환자들은 Lin-DR-CD11b+ % 가 감소하였다. 정확하게 종양의 크기를 측정하였으며, 암이 진행 중이지 않은 10명 중 8명의 환자에게서는 유의한 종양의 가장 긴 직경의 합에 증가 또는 감소를 확인 할 수 없었다. 종양 부피의 변화의 직접적인 결과가 Lin-DR-CD11b+ 세포에 미치는 영향은 크지 않으며, 5명의 환자에서 Lin-DR-CD11b+ %는 증가하였으며, 3명의 환자에서는 Lin-DR-CD11b+ %가 감소하였다. 이러한 데이터는 젬시타빈과 카페시타빈 자체에 의해서 Lin-DR-CD11b+가 감소한 것이 아님을 나타낸다. Lin-DR-CD11b+%의 변화는 종양반응을 추적하는 경향을 보였다. 이는 Lin-DR-CD11b+ 기준 값이 중간 값보다 큰 환자군에서 입증이 되었으며, MDSCs의 감소는 이 환자군에게 면역적으로 큰 도움이 될 것이다 (표 3). Tumor volume Eight out of 19 patients (comparative example 2, arm 2) who received a combination of gemcitabine and capecitabine had decreased Lin-DR-CD11b + cell numbers. Of seven patients with advanced cancer (progressive disease, PD), five Lin-DR-CD11b + cell levels were elevated and two levels were reduced (range -60 to +662%). Of 10 patients with no advanced cancer (Stable disease, SD), 6 Lin-DR-CD11b + cell levels were elevated and 4 levels were decreased (range -68- + 604%). Patients who responded partially to the Pep1 vaccine had decreased Lin-DR-CD11b +%. The tumor size was accurately measured and no significant increase or decrease was found in the sum of the longest diameters of the significant tumors in 8 patients out of 10. The direct effect of the change in tumor volume on the Lin-DR-CD11b + cells was not significant, Lin-DR-CD11b +% increased in 5 patients and Lin-DR-CD11b +% decreased in 3 patients. These data indicate that Lin-DR-CD11b + was not decreased by gemcitabine and capecitabine itself. Changes in Lin-DR-CD11b +% tended to track tumor response. This has been demonstrated in patients with Lin-DR-CD11b + reference values greater than the median, and the reduction of MDSCs will be of great help immunologically in this patient group (Table 3).
표 3
Number Stage Baseline Lin-DR-CD11b+ Post therapy Lin-DR-CD11b+ Change (%) Radiological response
1 III 2.58 18.16 +604 SD (0 %)
2 IV 3.65 4.12 +13 PD
3 III 2.25 3.33 +48 SD (0 %)
4 IV 2.54 1.59 -37 SD (-11 %)
5 IV 2.0 1.08 -46 SD (0 %)
6 IV 2.69 3.64 +35 PD
7 IV 2.23 4.12 +85 PD
8 IV 2.96 0.94 -68 PR
9 III 1.86 2.02 +9 SD (0 %)
TABLE 3
Number Stage Baseline Lin-DR-CD11b + Post therapy Lin-DR-CD11b + Change (%) Radiological response
One III 2.58 18.16 +604 SD (0%)
2 IV 3.65 4.12 +13 PD
3 III 2.25 3.33 +48 SD (0%)
4 IV 2.54 1.59 -37 SD (-11%)
5 IV 2.0 1.08 -46 SD (0%)
6 IV 2.69 3.64 +35 PD
7 IV 2.23 4.12 +85 PD
8 IV 2.96 0.94 -68 PR
9 III 1.86 2.02 +9 SD (0%)
Lin-DR-CD11b+ %의 기준 값이 중간 값보다 큰 환자 중 6명은 Lin-DR-CD11b+%가 증가하였으며, 3명은 종양의 부피 변화 (0%) 가 없었다. Lin-DR-CD11b+가 감소한 3명의 환자 중 1명은 부분적인 반응을 보였으며 11% 종양의 부피 변화가 있었다. 이 중 명 만이 Lin-DR-CD11b+%가 젬시타빈과 카페시타빈응 병용 투여 받은 후의 중간 값보다 작았다. Arm2와 arm3를 결합하였을 때, MDSC 변화의 절대 값은 항암제에 대한 반응 (p = 0.02)과 관련 이 있었으며 암이 진행 중인 9명의 환자들의 MDSC 레벨은 증가하였으며 (median = 0.47), 암을 억제하고 있는 환자 31명의 MDSC 레벨 (median = -0.49)은 감소하였다. Of the patients with a baseline value of Lin-DR-CD11b +% greater than the median, 6 had increased Lin-DR-CD11b +% and 3 had no change in tumor volume (0%). One of three patients with decreased Lin-DR-CD11b + had a partial response and a 11% tumor volume change. Among them, only Lin-DR-CD11b +% was lower than the median value after the combination of gemcitabine and capecitabine. When arm2 and arm3 were combined, the absolute value of MDSC change was related to the response to anticancer drugs ( p = 0.02), and the MDSC levels of nine patients with advanced cancer increased (median = 0.47). The MDSC level (median = -0.49) of 31 patients who had decreased.
치료 중 다른 염증성 사이토카인 및 IL-6를 사용하여 암 관련 염증의 정도와 MDSC 레벨이 관련성을 분석하였다. 그 결과는 표 4에 나타나 있다. Different inflammatory cytokines and IL-6 during treatment were used to analyze the association between the extent of cancer-related inflammation and MDSC levels. The results are shown in Table 4.
표 4
Figure PCTKR2014004760-appb-T000002
Table 4
Figure PCTKR2014004760-appb-T000002
종양의 부피 변화에 변화가 거의 없으나 MDSC%가 증가한 환자 군에, 지속적인 MDSC 생성을 유도하는 암 관련 염증성이 유발되고 있을 것이라 가정하였다. 젬시타빈과 카페시타빈 투여 중 Arm2의 환자 19명 중 7명의 IL-6레벨이 증가하였으며, 이 중 4명은 암이 진행 되고 있었다. 암이 진행되지 않고 있는 3명의 환자들의 MDSC%는 증가하였다. 표 4는 암이 진행중이지 않은 19명의 환자 중 10명의 환자들의 젬시타빈과 카페시타빈의 투여 중 염증성 사이토카인에 대한 MDSC%의 변화를 나타낸다. MDSC%가 감소한 4명의 환자들의 IL-5 레벨은 4명 모두 감소하였으며, 이 중 1명의 환자(환자 8) 의 IL-6 레벨은 7주간의 화학요법 동안 152.72에서 8.66 pg/ml로 감소하였으며, 이는 2.54 에서 1.59로 감소한 MDSC%와 연관성을 보였다. MDSC%가 증가한 암이 진행중이지 않은 6명의 환자들 3명의 MDSC 레벨 기본 값은 중간 값 보다 낮았으며, 젬시타빈과 카페시타빈 투여 이후에도 중간 값보다 낮았다. MDSC 기본 값이 중간 값 보다 높았던 2명의 환자는 치료 중에도 MDSC 레벨이 지속적으로 증가 하였으며, IL-6 레벨에서도 유의한 증가를 보였다 (환자 1, 3). 환자3의 혈관내피 성장인자 (Vascular Endothelial Growth Factor)또한 37.59 에서 70.69 pg/ml로 증가하였으며 이는 암이 진행되지 않고 있는 환자 중 유일하게 VEGF 레벨이 증가하였다. It was hypothesized that cancer-related inflammatory stimuli that induce sustained MDSC production were induced in a group of patients with little change in tumor volume but with increased MDSC%. During the administration of gemcitabine and capecitabine, IL-6 levels in seven of 19 patients with Arm2 increased, and four of them had advanced cancer. MDSC% in three patients with no cancer progressed. Table 4 shows the change in MDSC% for inflammatory cytokines during administration of gemcitabine and capecitabine in 10 of 19 patients with no cancer progression. IL-5 levels in four patients with reduced MDSC% decreased in all four patients, of whom one patient (Patient 8) had reduced IL-6 levels from 152.72 to 8.66 pg / ml during 7 weeks of chemotherapy. This was associated with MDSC%, which decreased from 2.54 to 1.59. The baseline MDSC levels of three patients in six non-advanced cancers with increased MDSC% were lower than the median and lower than the median after gemcitabine and capecitabine. Two patients whose MDSC baseline values were higher than the median had increased MDSC levels continuously during treatment and showed significant increases at IL-6 levels (Patients 1 and 3). Vascular Endothelial Growth Factor of Patient 3 also increased from 37.59 to 70.69 pg / ml, the only increase in VEGF levels among patients without cancer.
3. 비교예 2(arm2)와 실시예 2(arm3)의 MDSC 수 변화 비교 3. Comparison of MDSC Number Changes of Comparative Example 2 (arm2) and Example 2 (arm3)
19명의 췌장암 환자들에게 젬시타빈-카페시타빈(GemCap) 투여한 비교예 2 (arm2)와, 21명의 췌장암 환자들에게 젬시타빈-카페시타빈(GemCap) + pep1-GM-CSF를 투여한 실시예 2 (arm3)에 따른 MDSC 수의 변화를 비교한 결과는 도 2 및 표 5에 나타나 있다. Comparative Example 2 (arm2) administered gemcitabine-capecitabine (GemCap) to 19 pancreatic cancer patients, and gemcitabine-capecitabine (GemCap) + pep1-GM-CSF administered to 21 pancreatic cancer patients The result of comparing the change of MDSC number according to Example 2 (arm3) is shown in FIG.
표 5
Figure PCTKR2014004760-appb-T000003
Table 5
Figure PCTKR2014004760-appb-T000003
젬시타빈과 카페시타빈 (GemCap) 만을 병용 투여받은 환자 군 (비교예 2, arm2)와 GemCap과 PEP1을 함께 투여받은 환자 군 (실시예 2, arm3)의 MDSC 변화를 분석하고 비교하였다. Arm2와 arm3의 투여 전 투여 후의 MDSC를 비교한 요약 통계는 표 5에 나타내었으며, MDSC 변화 값은 도 1에 로그화하여 나타내었다. 치료전 MDSC 값은 arm3 (실시예 2)가 arm 2(비교예 2)보다 높았음에도 불구하고 (p = 0.08), arm 3 (실시예 2)환자군에서는 MDSC 레벨 ((p = 0.007 and p = 0.006 for absolute and percentage change) 의 유의한 감소가 있었으나, arm2 (비교예 2) 환자군 (p = 0.60 and p = 0.62 for absolute and percentage change )에서는 유의한 감소는 없었다. 그 결과, 치료 후 arm2 환자 군과 arm 3 환자군의 MDSC 값 (P > 0.99)은 차이가 거의 없었다. MDSC 레벨의 변화에 있어 arm 2와 arm3은 통계적으로 유의하다 (p = 0.04 and p = 0.06 for absolute and percentage change). 민감도 분석은 arm 2와 arm 3 환자군 간에 다른 경향을 보였으나, SD subgroup 과 치료 10주 후에는 일관성을 유지하였다. MDSC%가 감소한 환자의 수를 비교했을 때, arm3 (실시예 2)의 경우 전체 21명 환자 중 19명에서 감소를 보인 반면, arm 2(비교예 2)에서는 전체 19명 환자 중 8명만이 감소를 나타내었다. MDSC changes were analyzed and compared in the group of patients receiving only gemcitabine and capecitabine (GemCap) (Comparative Example 2, arm2) and the group of patients receiving GemCap and PEP1 (Example 2, arm3). Summary statistics comparing the MDSCs after administration of Arm2 and arm3 are shown in Table 5, and the MDSC change values are shown by logging in FIG. The pre-treatment MDSC values were higher in arm3 (Example 2) than in Arm 2 (Comparative Example 2) (p = 0.08), and in the arm 3 (Example 2) patient group, MDSC levels (( p = 0.007 and p = 0.006). for absolute and percentage change, but no significant reduction in arm2 (Comparative Example 2) patient group ( p = 0.60 and p = 0.62 for absolute and percentage change). There was little difference in MDSC values (P> 0.99) in the arm 3 patient group, and arm 2 and arm 3 were statistically significant (p = 0.04 and p = 0.06 for absolute and percentage change) in changes in MDSC levels. There was a different trend between arm 2 and arm 3 groups, but remained consistent after 10 weeks of treatment with the SD subgroup.Comparing the number of patients with decreased MDSC%, a total of 21 patients for arm3 (Example 2) were compared. Of 19 patients showed a decrease, whereas in arm 2 (Comparative Example 2) only 8 of the 19 patients Showed a decrease.
GemCap과 PEP1을 동시에 투여받은 21명의 arm3 환자군에서 PEP1 에 대한 면역반응(positive proliferation assay, the development of a positive DTH to PEP1)을 분석하였다. 21명 중 9 명의 환자들이 면역반응을 보였으며, 9명 중 8명은 치료 중 MDSC% 가 감소하였다. 9명 중 6명의 Lin-DR-CD11b+%의 기준 값은 환자들의 중간 값 보다 높았으며, MDSC 레벨은 감소하였다. The positive proliferation assay (the development of a positive DTH to PEP1) was analyzed in 21 arm3 patients who received GemCap and PEP1 at the same time. Nine of the 21 patients had an immune response, and eight of the nine had decreased MDSC% during treatment. The baseline value of Lin-DR-CD11b +% in 6 of 9 patients was higher than the median of patients and MDSC levels decreased.
면역 반응을 보인 모든 환자 군은 MDSC 분석 시기와 동일하게 증식반응 분석을 위해 혈액 채취 시 방사선 질병을 통제하였다. All groups of patients with immune responses controlled radiation sickness during blood collection for analysis of proliferative responses in the same way as for MDSC analysis.
Pep1의 단일 투여 효과에 대한 실험예Experimental Example for the Single Dose Effect of Pep1
그래뉼성 및 단핵구성 MDSC 둘다를 신선한 중피종(mesothelioma) 조직에서 추출하여 중피종의 미세환경에서 억제 어세이를 수행하였다. 또한, 본 실험은 페메트렉스드(Pemetrexed)를 사용하는데, 이를 중피종에 처리하면 종양세포에서 STAT3 활성화를 가져오는 노화-관련 분비성 표현형(senescence-associated secretory phenotype)이 유도된다. 이를 통해 본 실험은 종양 세포로부터의 사이토카인 방출에 미치는 pep1의 효과, 골수 세포 분화에 미치는 pep1의 효과 및 MDSC의 억제 활성에 미치는 pep1의 효과를 알아낼 수 있었다. Both granular and mononuclear MDSCs were extracted from fresh mesothelioma tissue and subjected to inhibition assay in the microenvironment of mesothelioma. The experiment also uses Pemetrexed, which, when treated with mesothelioma, induces an senescence-associated secretory phenotype that results in STAT3 activation in tumor cells. Through this experiment, we could find out the effect of pep1 on cytokine release from tumor cells, pep1 on myeloid cell differentiation and pep1 on the inhibitory activity of MDSC.
1. 비교예 3-4 및 실시예 3-4: pep1 단일 투여가 수지상 세포(Dendritic Cell, DC) 성숙에 미치는 영향 1. Comparative Example 3-4 and Example 3-4: Effect of single pep1 administration on dendritic cell (DC) maturation
건강한 공여자의 PBMC로부터 얻어진 부착성 단핵구(adherent monocytes)를 사용하여 DC 성숙 실험을 수행하였다. 5일 배양을 수행하였다. 조건은 하기와 같았다.DC maturation experiments were performed using adherent monocytes obtained from PBMCs from healthy donors. 5-day culture was performed. The conditions were as follows.
비교예 3: MoDCs + GM-CSF /IL4 Comparative Example 3: MoDCs + GM-CSF / IL4
실시예 3: MoDCs + GM-CSF /IL4 + pep1Example 3: MoDCs + GM-CSF / IL4 + pep1
비교예 4: MoDCs + GM-CSF /IL4 + pemetrexedComparative Example 4: MoDCs + GM-CSF / IL4 + pemetrexed
실시예 4: MoDCs + GM-CSF /IL4 + pep1/ pemetrexedExample 4: MoDCs + GM-CSF / IL4 + pep1 / pemetrexed
*MoDCs : Monocyte-derived Dendritic CellMoDCs: Monocyte-derived Dendritic Cells
모든 샘플은 LPS (Lipopolysaccharides) 를 하룻밤 처리하거나 처리하지 않았다. 상기 배양된 샘플들을 다음에 대하여 테스트하였다:All samples were treated with or without LPS (Lipopolysaccharides) overnight. The cultured samples were tested for:
DC 성숙 마커;DC maturation markers;
인비트로에서 DC의 T 세포 자극 기능; 및T cell stimulating function of DC in vitro; And
CD3/CD28에서의 T 세포 증식 억제 능력.Ability to inhibit T cell proliferation in CD3 / CD28.
아르기네이즈(Arginase), Inos , Ros, Stat 3 및 Stat 6 또한 분석되었다. Arginase, Inos, Ros, Stat 3 and Stat 6 were also analyzed.
2. 비교예 5-6 및 실시예 5-6: 종양 유래 인자들의 존부에 따른 DC 성숙과pep1의 영향 2. Comparative Examples 5-6 and 5-6: Effect of DC Maturation and pep1 on Presence of Tumor-derived Factors
중피종 세포는 25 플라스크에 플레이팅되었고, 40-50% 컨플루언스에서 하기 샘플들로 처리되었다. Mesothelioma cells were plated in 25 flasks and treated with the following samples at 40-50% confluence.
비교예 5: Mesothelioma cell line (MCL)Comparative Example 5: Mesothelioma cell line (MCL)
실시예 5: Mesothelioma cell line + GV1001 (MCLG)Example 5: Mesothelioma cell line + GV1001 (MCLG)
비교예 6: Mesothelioma cell line + pemetrexed (MCLP)Comparative Example 6: Mesothelioma cell line + pemetrexed (MCLP)
실시예 6: Mesothelioma cell line + GV1001/ pemetrexed (MCLGP)Example 6: Mesothelioma cell line + GV1001 / pemetrexed (MCLGP)
상기 처리는 24시간 동안 수행된 후 배지를 제거하고 새로운 배지로 교체해 주었다. 36시간 후에 상청액을 수거하여 저장하였다. 그 후 하기와 같이 pep1을 처리하지 않은 비교예와 pep1을 처리한 실시예를 배지로 하여 5일간 배양하였다. The treatment was performed for 24 hours, after which the medium was removed and replaced with fresh medium. After 36 hours the supernatant was collected and stored. Thereafter, the culture was incubated for 5 days using a comparative example without pep1 and an example with pep1 as a medium.
대조군: MoDCs +GMCSF /IL4 Control: MoDCs + GMCSF / IL4
비교예 7: MoDCs +GMCSF /IL4 + MLCComparative Example 7: MoDCs + GMCSF / IL4 + MLC
실시예 7: MoDCs +GMCSF /IL4 + MCLGExample 7: MoDCs + GMCSF / IL4 + MCLG
비교예 8: MoDCs +GMCSF /IL4 + MLCPComparative Example 8: MoDCs + GMCSF / IL4 + MLCP
실시예 8: MoDCs +GMCSF /IL4 + MLCGPExample 8 MoDCs + GMCSF / IL4 + MLCGP
모든 샘플은 LPS를 하룻밤 처리하거나 처리하지 않았다. 상기 배양된 샘플들을 다음에 대하여 테스트하였다:All samples were treated with or without LPS overnight. The cultured samples were tested for:
DC 성숙 마커;DC maturation markers;
인비트로에서 DC의 T 세포 자극 기능; 및T cell stimulating function of DC in vitro; And
CD3/CD28에서의 T 세포 증식 억제 능력.Ability to inhibit T cell proliferation in CD3 / CD28.
아르기네이즈(Arginase), Inos , Ros, Stat 3 및 Stat 6 또한 분석되었다. Arginase, Inos, Ros, Stat 3 and Stat 6 were also analyzed.
3. MDSC의 억제 활성에 미치는 pep1의 영향3. Effect of pep1 on Inhibitory Activity of MDSC
환자로부터 정제한 MDSC에 pep1 (또한 + pemetrexed)을 하룻밤 처리하고, 세포를 세척한 후 억제 어세이를 위한 MLR (Mixed lymphocyte reaction) 에 사용하였다. MDSC를 96웰에 플레이트하고 다음 날 MLR을 위해 각각의 농도로 처리하였다(CD3/CD28 자극). 그것과 별개로, MLR 동안 상기 처리를 하고 상기 처리를 대조군으로서 T 세포 처리에도 추가하였다. The purified MDSC from the patient was treated with pep1 (also + pemetrexed) overnight, and the cells were washed and used for the mixed lymphocyte reaction (MLR) for inhibition assay. MDSCs were plated in 96 wells and treated at each concentration for MLR the next day (CD3 / CD28 stimulation). Apart from that, the treatment was performed during MLR and the treatment was also added to T cell treatment as a control.

Claims (31)

  1. 골수-유래 억제세포(myeloid-derived suppressor cells, 이하"MDSC") 저해용 조성물로서,As a composition for inhibiting myeloid-derived suppressor cells ("MDSC"),
    상기 조성물은 MDSC 저해를 위한 유효성분으로서 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하며,The composition comprises a peptide comprising SEQ ID NO: 1 as an active ingredient for MDSC inhibition, a peptide having 80% or more sequence homology with the peptide sequence, or a fragment thereof.
    상기 펩티드는 MDSC 저해에 효과적인 양으로 포함된, MDSC 저해용 조성물. The peptide is contained in an amount effective to inhibit the MDSC, composition for inhibiting MDSC.
  2. 제1항에 있어서, 상기 조성물은 MDSC를 저해할 필요가 있는 개체에 투여하기 위한 것인, MDSC 저해용 조성물.The composition of claim 1, wherein the composition is for administration to a subject in need of inhibiting MDSC.
  3. 제1항에 있어서, 상기 골수-유래 억제세포는 종양을 갖는 개체의 골수-유래 억제세포인, MDSC 저해용 조성물.The composition of claim 1, wherein the bone marrow-derived suppressor cells are bone marrow-derived suppressor cells of a subject having a tumor.
  4. 제3항에 있어서, 상기 종양이 신장세포 암종(RCC), 결장직장암(CRC), 위암(GC), 흑색종, 폐암, 혈액암, 전립선암, 선암종, 전립선암 및 췌장암에서 선택되는 것인, MDSC 저해용 조성물.According to claim 3, wherein the tumor is selected from renal cell carcinoma (RCC), colorectal cancer (CRC), gastric cancer (GC), melanoma, lung cancer, blood cancer, prostate cancer, adenocarcinoma, prostate cancer and pancreatic cancer, MDSC inhibition composition.
  5. 제1항에 있어서, 상기 조성물은 항암제와 조합되어 투여되는 것인 MDSC 저해용 조성물.The composition of claim 1, wherein the composition is administered in combination with an anticancer agent.
  6. 제5항에 있어서, 상기 항암제는 화학요법제이고,The method of claim 5, wherein the anticancer agent is a chemotherapeutic agent,
    상기 화학요법제는 데옥시뉴클레오시드 유사체 및 플루오로피리미딘 중 어느 하나 이상인 MDSC 저해용 조성물.Wherein said chemotherapeutic agent is any one or more of deoxynucleoside analogs and fluoropyrimidines.
  7. 제6항에 있어서, The method of claim 6,
    상기 데옥시뉴클레오시드 유사체는 젬시타빈이고, 상기 플루오로피리미딘은 5-플루오로우라실 또는 카페시타빈인 MDSC 저해용 조성물.The deoxynucleoside analog is gemcitabine, and the fluoropyrimidine is 5-fluorouracil or capecitabine.
  8. 제5항에 있어서, 상기 조성물은 어쥬번트(adjuvant)와 조합되어 투여되는 것인 MDSC 저해용 조성물.The composition of claim 5, wherein the composition is administered in combination with an adjuvant.
  9. 제8항에 있어서, 상기 어쥬번트는 사이토킨 어쥬번트인 MDSC 저해용 조성물.The composition of claim 8, wherein the adjuvant is a cytokine adjuvant.
  10. 제9항에 있어서, 상기 사이토킨 어쥬번트는 그래뉼로사이트-마크로파지 콜로니-자극 인자(granulocyte-macrophage colony-stimulating factor, GM-CSF)인 MDSC 저해용 조성물.10. The composition of claim 9, wherein the cytokine adjuvant is granulocyte-macrophage colony-stimulating factor (GM-CSF).
  11. 제1항 내지 제10항 중 어느 한 항에 있어서, 상기 MDSC는 표현형이 Lin-DR-CD11b+인, MDSC 저해용 조성물.The composition for inhibiting MDSC according to any one of claims 1 to 10, wherein the MDSC has a phenotype of Lin-DR-CD11b +.
  12. 제1항 내지 제10항 중 어느 한 항에 따른 MDSC 저해용 조성물; 및Composition for inhibiting MDSC according to any one of claims 1 to 10; And
    설명서를 포함하는 MDSC 저해용 키트.Kit for inhibiting MDSC containing instructions.
  13. 제 12항에 있어서, 상기 키트는,The method of claim 12, wherein the kit,
    항암제를 더 포함하는 MDSC 저해용 키트. Kit for inhibition of MDSC further comprising an anticancer agent.
  14. 제13항에 있어서, 상기 항암제는 화학요법제이고,The method of claim 13, wherein the anticancer agent is a chemotherapeutic agent,
    상기 화학요법제는 데옥시뉴클레오시드 유사체 및 플루오로피리미딘 중 어느 하나 이상인 MDSC 저해용 키트.Wherein said chemotherapeutic agent is any one or more of deoxynucleoside analogs and fluoropyrimidines.
  15. 제14항에 있어서, The method of claim 14,
    상기 데옥시뉴클레오시드 유사체는 젬시타빈이고, 상기 플루오로피리미딘은 5-플루오로우라실 또는 카페시타빈인 MDSC 저해용 키트. The deoxynucleoside analog is gemcitabine, and the fluoropyrimidine is 5-fluorouracil or capecitabine.
  16. 제12항에 있어서, 상기 키트는 어쥬번트를 더 포함하는 것인 MDSC 저해용 키트.The kit of claim 12, wherein the kit further comprises an adjuvant.
  17. 제16항에 있어서, 상기 어쥬번트는 사이토킨 어쥬번트인 MDSC 저해용 키트.17. The kit of claim 16, wherein the adjuvant is a cytokine adjuvant.
  18. 제17항에 있어서, 상기 어쥬번트는 그래뉼로사이트-마크로파지 콜로니-자극 인자(granulocyte-macrophage colony-stimulating factor, GM-CSF)인 MDSC 저해용 키트.18. The kit of claim 17, wherein the adjuvant is granulocyte-macrophage colony-stimulating factor (GM-CSF).
  19. 제12항에 있어서, 상기 설명서는 상기 MDSC 저해용 조성물과, 상기 항암제 및/또는 상기 어쥬번트를 조합하여 투여하는 내용을 포함하는 MDSC 저해용 키트.The kit for inhibiting MDSC of claim 12, wherein the instruction manual comprises a combination of the composition for inhibiting MDSC and the anticancer agent and / or the adjuvant.
  20. 제12항에 있어서, 상기 MDSC 저해용 키트는, MDSC 저해를 통해 MDSC에 의한 질병 또는 증상을 개선, 예방 또는 치료하는 것인 MDSC 저해용 키트.The kit for inhibiting MDSC of claim 12, wherein the kit for inhibiting MDSC improves, prevents or treats a disease or symptom caused by MDSC through MDSC inhibition.
  21. 제12항에 있어서, 상기 MDSC 저해용 키트는, MDSC 저해를 통해 암을 개선, 예방 또는 치료하는 것인 MDSC 저해용 키트.The kit for inhibiting MDSC of claim 12, wherein the kit for inhibiting MDSC improves, prevents or treats cancer through MDSC inhibition.
  22. 제21항에 있어서, 상기 암이 신장세포 암종(RCC), 결장직장암(CRC), 위암(GC), 흑색종, 폐암, 혈액암, 전립선암, 선암종, 전립선암 및 췌장암에서 선택되는 것인, MDSC 저해용 키트.The method of claim 21, wherein the cancer is selected from renal cell carcinoma (RCC), colorectal cancer (CRC), gastric cancer (GC), melanoma, lung cancer, blood cancer, prostate cancer, adenocarcinoma, prostate cancer and pancreatic cancer. MDSC Inhibition Kit.
  23. 골수-유래 억제세포(myeloid-derived suppressor cells, "MDSC")를 저해하는 방법으로서,As a method of inhibiting myeloid-derived suppressor cells ("MDSC"),
    MDSC 저해에 효과적인 양의, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를, MDSC 저해가 필요한 대상에 투여하는 것을 포함하는 MDSC 저해 방법. MDSC inhibition comprising administering to a subject in need of MDSC inhibition an effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof, that is effective for MDSC inhibition. Way.
  24. 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; A peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof;
    항암제; 및Anticancer agents; And
    어쥬번트를 포함하는 MDSC 저해용 조성물. MDSC inhibition composition comprising an adjuvant.
  25. 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; A peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof;
    항암제; 및Anticancer agents; And
    어쥬번트를 포함하는 항암 조성물. An anticancer composition comprising an adjuvant.
  26. 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드; A peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof;
    항암제; 및Anticancer agents; And
    어쥬번트를 포함하는 암 백신 조성물. Cancer vaccine composition comprising an adjuvant.
  27. 제24항 내지 제26항 중 어느 한 항에 있어서,The method according to any one of claims 24 to 26,
    상기 항암제는 화학요법제이고, 상기 화학요법제는 데옥시뉴클레오시드 유사체 및 플루오로피리미딘 중 어느 하나 이상이며,The anticancer agent is a chemotherapeutic agent, the chemotherapeutic agent is any one or more of deoxynucleoside analogs and fluoropyrimidines,
    상기 어쥬번트는 사이토킨 어쥬번트인, 조성물.Wherein said adjuvant is a cytokine adjuvant.
  28. 제27항에 있어서,The method of claim 27,
    상기 펩티드는 서열번호 1로 구성된 펩티드이며,The peptide is a peptide consisting of SEQ ID NO: 1,
    상기 데옥시뉴클레오시드 유사체는 젬시타빈이고, 상기 플루오로피리미딘은 5-플루오로우라실 또는 카페시타빈이며,The deoxynucleoside analog is gemcitabine, the fluoropyrimidine is 5-fluorouracil or capecitabine,
    상기 어쥬번트는 GM-CSF인, 조성물.The adjuvant is GM-CSF.
  29. 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를 포함하는 조성물;A composition comprising a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof;
    항암제; 및Anticancer agents; And
    설명서를 포함하는 항암 키트. Anticancer kit including instructions.
  30. 암을 개선, 치료 또는 예방하는 방법으로서,As a method of improving, treating or preventing cancer,
    효과적인 양의, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드를, 항암제 및/또는 어쥬번트와 조합하여, 암의 개선, 치료 또는 예방이 필요한 대상에 투여하는 것을 포함하는 암 개선, 치료 또는 예방 방법. An effective amount of a peptide comprising SEQ ID NO: 1, a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof, in combination with an anticancer agent and / or adjuvant to improve, treat or A method for improving, treating, or preventing cancer, comprising administering to a subject in need thereof.
  31. 골수-유래 억제세포(myeloid-derived suppressor cells, "MDSC") 저해용 조성물을 제조하기 위한, 서열번호 1을 포함하는 펩티드, 상기 펩티드 서열과 80% 이상의 서열 상동성을 갖는 펩티드, 또는 그것의 단편인 펩티드의 용도.A peptide comprising SEQ ID NO: 1 for preparing a composition for inhibiting myeloid-derived suppressor cells ("MDSC"), a peptide having at least 80% sequence homology with the peptide sequence, or a fragment thereof Use of Phosphorus Peptides.
PCT/KR2014/004760 2014-05-28 2014-05-28 Composition for inhibiting myeloid-derived suppressor cells (mdscs) WO2015182798A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
PCT/KR2014/004760 WO2015182798A1 (en) 2014-05-28 2014-05-28 Composition for inhibiting myeloid-derived suppressor cells (mdscs)
KR1020157003185A KR101826753B1 (en) 2014-05-28 2014-11-28 Composition for inhibiting myeloid-derived suppressor cells
PCT/KR2014/011571 WO2015182837A1 (en) 2014-05-28 2014-11-28 Composition for inhibiting myeloid-derived suppressor cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/KR2014/004760 WO2015182798A1 (en) 2014-05-28 2014-05-28 Composition for inhibiting myeloid-derived suppressor cells (mdscs)

Publications (1)

Publication Number Publication Date
WO2015182798A1 true WO2015182798A1 (en) 2015-12-03

Family

ID=54699114

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/KR2014/004760 WO2015182798A1 (en) 2014-05-28 2014-05-28 Composition for inhibiting myeloid-derived suppressor cells (mdscs)
PCT/KR2014/011571 WO2015182837A1 (en) 2014-05-28 2014-11-28 Composition for inhibiting myeloid-derived suppressor cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/KR2014/011571 WO2015182837A1 (en) 2014-05-28 2014-11-28 Composition for inhibiting myeloid-derived suppressor cells

Country Status (2)

Country Link
KR (1) KR101826753B1 (en)
WO (2) WO2015182798A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105848667B (en) * 2013-11-22 2020-05-19 珍白斯凯尔有限公司 Peptide having angiogenesis inhibitory activity and composition comprising the same
KR20210023306A (en) 2019-08-22 2021-03-04 숙명여자대학교산학협력단 Composition for inhibiting myeloid-derived suppressor cells comprising MITF inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011116299A2 (en) * 2010-03-18 2011-09-22 Colorado State University Research Foundation Myeloid derived suppressor cell inhibiting agents
WO2013135266A1 (en) * 2012-03-12 2013-09-19 Gemvax As Treatment of non-small cell lung carcinoma by active immunotherapy

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011116299A2 (en) * 2010-03-18 2011-09-22 Colorado State University Research Foundation Myeloid derived suppressor cell inhibiting agents
WO2013135266A1 (en) * 2012-03-12 2013-09-19 Gemvax As Treatment of non-small cell lung carcinoma by active immunotherapy

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
"Findings from Royal Marsden hospital has provided new data on pancreatic cancer", WEBSITE: PHARMACY CHOICE - PHARMACY NEWS ARTICLE, 31 March 2014 (2014-03-31), Retrieved from the Internet <URL:http://www.pharmacychoice.com/news/article.cfm?Article_ID=1182638> *
ANNELS ET AL.: "The effects of gemcitabine and capecitabine combination chemotherapy and of low-dose adjuvant GM-CSF on the levels of myeloid-derived suppressor cells in patients with advanced pancreatic cancer", CANCER IMMUNOLOGY, IMMUNOTHERAPY, vol. 63, no. 2, pages 175 - 183, XP055240759, ISSN: 0340-7004 *
MAZZOLA ET AL.: "Aging, cancer, and cancer vaccines", IMMUNITY & AGEING, vol. 9, 2012, pages 1 - 11, XP055240764 *
RASMUSSEN ET AL.: "Ex vivo expansion protocol for human tumor specific T cells for adoptive T cell therapy", JOURNAL OF IMMUNOLOGICAL METHODS, vol. 355, no. 1-2, 2010, pages 52 - 60, XP055240763, ISSN: 0022-1759 *

Also Published As

Publication number Publication date
KR20160000881A (en) 2016-01-05
KR101826753B1 (en) 2018-02-08
WO2015182837A1 (en) 2015-12-03

Similar Documents

Publication Publication Date Title
WO2013169077A1 (en) Composition for preventing or treating cachexia
WO2015076621A1 (en) Peptide having angiogenesis inhibitory activity and composition containing same
WO2015156649A1 (en) Peptide having fibrosis inhibitory activity and composition containing same
WO2015093854A1 (en) Composition for treating prostate cancer
WO2015167067A1 (en) Composition for organ, tissue, or cell transplantation, kit, and transplantation method
US20070081991A1 (en) Compositions and methods for modulation of immune responses
WO2015060673A1 (en) Composition for treating and preventing benign prostatic hyperplasia
JP6166858B2 (en) Compositions and methods for the treatment of intracellular damage
WO2017023138A1 (en) Chimeric antigen receptor, and t cells in which chimeric antigen receptor is expressed
WO2011052883A2 (en) Method for activating a natural killer cell by adjusting the expression of the socs2 gene
WO2021096275A1 (en) Fusion protein including modified interleukin-7 and tgf beta receptor ii and use thereof
WO2015182798A1 (en) Composition for inhibiting myeloid-derived suppressor cells (mdscs)
WO2021158091A2 (en) Composition comprising chemokine inhibitor, colony stimulating factor inhibitor, and cancer immunotherapy agent for prevention or treatment of cancer and combination therapy
WO2017034244A1 (en) Peptide having effect of preventing or treating central nervous system diseases and pharmaceutical composition for preventing and treating central nervous system diseases, containing same as active ingredient
WO2021177518A1 (en) Pharmaceutical composition for lowering blood cholesterol, preventing or treating cardiovascular diseases and reducing inflammation
WO2019039700A1 (en) Composition including melittin for removing m2-type tumor-associated macrophage
WO2013129852A1 (en) Repebody for novel interleukin-6 and use thereof
WO2016027990A1 (en) Pharmaceutical composition containing dusp5 as active ingredient for preventing or treating bone metabolic diseases
WO2015072750A1 (en) Peptide having anti-inflammatory activity and composition comprising same
WO2023195802A1 (en) Novel-peptide-based anticancer immunotherapeutic agent
WO2023120844A1 (en) Composition for mass proliferating natural killer cells, comprising novel peptide, and method for mass proliferating natural killer cells by using same
WO2023153773A1 (en) Cancer vaccine comprising epitope of c-met and use thereof
WO2021235685A1 (en) Pharmaceutical composition for preventing or treating cancer
WO2015170790A1 (en) Composition for treating and preventing ischemic damage
WO2021107635A1 (en) Composition for anticancer treatment, comprising nk cells and fusion protein which comprises il-2 protein and cd80 protein

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: 14893414

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14893414

Country of ref document: EP

Kind code of ref document: A1