KR20230105583A - Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein - Google Patents

Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein Download PDF

Info

Publication number
KR20230105583A
KR20230105583A KR1020220001164A KR20220001164A KR20230105583A KR 20230105583 A KR20230105583 A KR 20230105583A KR 1020220001164 A KR1020220001164 A KR 1020220001164A KR 20220001164 A KR20220001164 A KR 20220001164A KR 20230105583 A KR20230105583 A KR 20230105583A
Authority
KR
South Korea
Prior art keywords
polynucleotide
cancer
seq
cells
mrna
Prior art date
Application number
KR1020220001164A
Other languages
Korean (ko)
Inventor
이현철
구봉성
김진숙
Original Assignee
비피진 주식회사
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 비피진 주식회사 filed Critical 비피진 주식회사
Priority to KR1020220001164A priority Critical patent/KR20230105583A/en
Priority to PCT/KR2022/000131 priority patent/WO2023132380A1/en
Publication of KR20230105583A publication Critical patent/KR20230105583A/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6905Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion
    • A61K47/6911Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a colloid or an emulsion the form being a liposome
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/52Genes encoding for enzymes or proenzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/07Fusion polypeptide containing a localisation/targetting motif containing a mitochondrial localisation signal
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/09Fusion polypeptide containing a localisation/targetting motif containing a nuclear localisation signal

Abstract

신규 암 치료를 위한 폴리뉴클레오티드로서, CD47에 결합하는 바인더와 복합체를 형성하는 리포좀형 나노입자에 포집된 형태로 사용되어 암세포의 대사취약성(metabolic vulnerability)을 극대화시켜 암세포를 사멸시키는 기작을 가지는 암 치료를 위한 폴리뉴클레오티드가 개시된다. 본 발명은 서열번호 3으로 표시되는 아미노산 서열을 암호화하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.As a polynucleotide for novel cancer treatment, it is used in the form of being captured in liposomal nanoparticles that form a complex with a binder that binds to CD47, thereby maximizing the metabolic vulnerability of cancer cells and killing cancer cells. A polynucleotide for is disclosed. The present invention provides a polynucleotide for cancer treatment encoding the amino acid sequence represented by SEQ ID NO: 3.

Description

5'-뉴클레오티다아제 변형 단백질을 암호화하는 암 치료를 위한 폴리뉴클레오티드{Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein}Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein {Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein}

본 발명은 암 치료를 위한 폴리뉴클레오티드에 관한 것으로, 보다 상세하게는 약물 전달 물질에 포집된 형태로 사용되는 암 치료를 위한 폴리뉴클레오티드에 관한 것이다.The present invention relates to a polynucleotide for cancer treatment, and more particularly, to a polynucleotide for cancer treatment used in a form entrapped in a drug delivery material.

암세포는 면역세포가 CD47과 같은 다른 세포를 파괴하는 것을 막기 위해 일반 포유류 생화학에서 사용되는 신호를 남용한다. 이러한 "나를 먹지 말라(don’t eat me)"는 신호를 방해하는 것은 여러 종류의 암을 표적으로 삼을 수 있는 효과적인 암 치료법의 개발에 상당한 이득을 가져왔다.Cancer cells abuse signals used in normal mammalian biochemistry to prevent immune cells from destroying other cells, such as CD47. Interfering with these “don’t eat me” signals has had significant advantages in the development of effective cancer therapies that can target many types of cancer.

일반적으로 대식세포라 불리는 면역세포들은 암세포를 감지한 다음 이들을 삼켜 먹어 치운다. 연구자들은 최근 몇 년 사이에 세포 표면의 단백질이 대식세포에게 자신을 먹거나 파괴하지 말라는 신호를 보낸다는 사실을 발견했다. 이는 면역 시스템이 정상적인 세포들을 공격하지 않도록 보호하는 데 유용하나, 암세포가 이를 이용해 면역 시스템을 회피하기 위해 이러한 "나를 먹지 말라"는 신호를 사용한다. 연구자들은 이전에 단백질 PD-L1과 주요 조직적합성 1급 복합체의 베타-2-마이크로글로불린 서브유닛이 암세포가 면역세포로부터 자신을 보호하기 위해 활용하고 있다는 사실을 밝혀낸 바 있다. CD47을 차단하는 항체는 현재 임상시험 중이며, PD-L1 또는 PDL1 수용체를 타겟으로 하는 암 치료법이 환자 치료에 활용되고 있다.Immune cells, commonly called macrophages, detect cancer cells and then engulf and eat them. Researchers have discovered in recent years that proteins on the surface of cells send signals to macrophages not to eat or destroy them. This is useful for protecting the immune system from attacking normal cells, but cancer cells use these "don't eat me" signals to evade the immune system. Researchers have previously shown that the protein PD-L1 and the beta-2-microglobulin subunit of the major histocompatibility class 1 complex are utilized by cancer cells to protect themselves from immune cells. Antibodies that block CD47 are currently in clinical trials, and cancer therapies targeting the PD-L1 or PDL1 receptor are being used to treat patients.

이 연구는 많은 암들이 정상 세포와 주변 조직에 비해 CD47을 풍부하게 생산한다는 것을 보여주었다. 최근 연구에서는 연구자들은 종양에 침투하는 대식세포가 Sirpα라는 수용체를 통해 CD47 신호를 감지할 수 있다는 사실을 밝혀냈다. 이들은 또한 접시에 환자의 암세포와 대식세포를 섞고 CD47과 Sirpα의 상호작용을 차단하면, 대식세포가 암세포를 먹어치우기 시작한다는 사실도 보여주었다. 마지막으로, 이들은 마우스에게 인간 유방암 세포를 이식하였다. CD47 신호가 차단되자 마우스 면역계의 대식세포가 암을 공격하였다. 특히 주목할 것은 CD47 신호를 차단해 혈액암과 삼중음성 유방암에 큰 영향을 미친다는 발견이었다.This study showed that many cancers produce CD47 in abundance compared to normal cells and surrounding tissues. In a recent study, researchers showed that tumor-infiltrating macrophages can detect CD47 signals through a receptor called Sirpα. They also showed that if you mix a patient's cancer cells with macrophages in a dish and block the interaction of CD47 and Sirpα, the macrophages start eating the cancer cells. Finally, they transplanted human breast cancer cells into the mice. When the CD47 signal was blocked, the macrophages of the mouse's immune system attacked the cancer. Of particular note was the discovery that blocking the CD47 signal had a significant effect on blood cancer and triple-negative breast cancer.

[선행 특허문헌][Prior patent literature]

- 한국 공개특허 제2006-0121150호(2006.11.28.) - Korean Patent Publication No. 2006-0121150 (2006.11.28.)

본 발명은 암 치료를 위한 신규 폴리뉴클레오티드로서, CD47에 결합하는 바인더와 복합체를 형성하는 리포좀형 나노입자에 포집된 형태로 사용되어 암세포의 대사취약성(metabolic vulnerability)을 극대화시켜 암세포를 사멸시키는 기작을 가지는 암 치료를 위한 폴리뉴클레오티드를 제공하고자 한다.The present invention is a novel polynucleotide for cancer treatment, which is used in the form of being encapsulated in liposome-type nanoparticles that form a complex with a binder that binds to CD47, thereby maximizing the metabolic vulnerability of cancer cells to kill cancer cells. It is intended to provide polynucleotides for cancer treatment.

상기 과제를 해결하기 위하여 본 발명은, 서열번호 3으로 표시되는 아미노산 서열을 암호화하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In order to solve the above problems, the present invention provides a polynucleotide for cancer treatment encoding the amino acid sequence represented by SEQ ID NO: 3.

또한 상기 폴리뉴클레오티드는 서열번호 38 내지 서열번호 41로 이루어지는 군에서 선택되는 염기서열을 포함하는 것을 특징으로 하는 폴리뉴클레오티드를 제공한다.In addition, the polynucleotide is characterized by comprising a nucleotide sequence selected from the group consisting of SEQ ID NO: 38 to SEQ ID NO: 41.

또한 상기 폴리뉴클레오티드는 mRNA인 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, the polynucleotide provides a polynucleotide for cancer treatment, characterized in that the mRNA.

또한 상기 mRNA는 암세포에 진입하여 핵산대사를 저해하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, the mRNA provides a polynucleotide for cancer treatment characterized in that it enters cancer cells and inhibits nucleic acid metabolism.

또한 상기 핵산대사는 dTTP 생합성 대사인 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, the nucleic acid metabolism provides a polynucleotide for cancer treatment, characterized in that dTTP biosynthetic metabolism.

또한 상기 폴리뉴클레오티드는 서열번호 19로 표시되는 5'-UTR 및 서열번호 20으로 표시되는 3'-UTR을 더 포함하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, the polynucleotide provides a polynucleotide for cancer treatment, characterized in that it further comprises a 5'-UTR represented by SEQ ID NO: 19 and a 3'-UTR represented by SEQ ID NO: 20.

또한 상기 폴리뉴클레오티드는 서열번호 22로 표시되는 핵 위치 신호(NLS)를 암호화하는 핵산 서열을 더 포함하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, the polynucleotide provides a polynucleotide for cancer treatment, characterized in that it further comprises a nucleic acid sequence encoding a nuclear localization signal (NLS) represented by SEQ ID NO: 22.

또한 상기 폴리뉴클레오티드는 서열번호 23으로 표시되는 미토콘드리아 위치 신호(MLS)를 암호화하는 핵산 서열을 더 포함하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, the polynucleotide provides a polynucleotide for cancer treatment, characterized in that it further comprises a nucleic acid sequence encoding a mitochondrial localization signal (MLS) represented by SEQ ID NO: 23.

또한 상기 암은 대장암 또는 유방암인 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드를 제공한다.In addition, it provides a polynucleotide for the treatment of cancer, characterized in that the cancer is colon cancer or breast cancer.

본 발명은 CD47에 결합하는 바인더와 복합체를 형성하는 리포좀형 나노입자에 포집된 형태로 사용되어 암세포의 대사취약성(metabolic vulnerability)을 극대화시켜 암세포를 사멸시키는 기작을 가지는 암 치료를 위한 폴리뉴클레오티드로서, 인간세포 내에 전달되었을 때 비특이적 마이크로RNA(microRNA)로서 작용하는 부위를 최소화하고 발현을 최대화할 수 있도록 유전자 서열이 최적화된 암 치료를 위한 폴리뉴클레오티드를 제공할 수 있다.The present invention is a polynucleotide for cancer treatment having a mechanism of killing cancer cells by maximizing the metabolic vulnerability of cancer cells when used in the form of being captured in liposomal nanoparticles that form a complex with a binder that binds to CD47, It is possible to provide a polynucleotide for cancer treatment in which the gene sequence is optimized to minimize the site acting as a non-specific microRNA (microRNA) when delivered into human cells and to maximize expression.

도 1은 Sirpα, SV1 및 SV4의 아미노산 서열을 비교하여 나타낸 도면이다. 진한 글자는 부분적으로 보존되지 않은 잔기를 나타낸다. 시퀀스 정렬은 ClustalW로 이루어졌고, 이미지는 BioEdit 시퀀스 정렬 편집 프로그램으로 생성되었다.
도 2는 올바른 방향성을 위한 SV1의 돌연변이를 설명하는 도면이다. 도 2A에서는 SV 도메인(녹색 리본)이 CD47(빨간색 구면) 도메인과 결합되어 있고, 도 2A의 모델에서는 SV 도메인(빨간색 구면으로 표시)은 라이신 잔기가 올바른 방향성에 영향을 미치고 CD47에 대한 올바른 결합을 위해 변이됨을 보여준다. 도 2B는 DSPE-컨쥬게이트된 SV1 및 SV4의 질량분석법(Mass spectrometry analysis)을 이용한 분석 결과를 나타내고 있다.
도 3은 T001 및 인간 NT5M의 보존성 모티브를 나타낸 도면이다. T001 및 인간 NT5M의 시퀀스 정렬로서, 인간 NT5M 및 T001의 서열상 유사성을 확인하기 위해 ClustalW를 통해 배열하였다. 정렬에 사용된 시퀀스의 Swiss-Prot/TrEMBL 어세션 번호는 인간 NT5M 및 T001이다. 빨간색 필드는 완전히 보존된 아미노산 잔기를 나타내고, 흰색 필드의 빨간색 문자는 유사한 생화학적 기능을 가지는 부분적으로 보존된 아미노산 잔기를 나타낸다. 시퀀스 정렬은 ClustalW로 이루어졌고, 이미지는 ESPript 서버로 생성되었다.
도 4는 T001 및 NT5M의 구조를 비교하여 나타낸 모델이다. 세포질 T001(CT)과 dTMP-결합 인간 NT5M(노란색)을 중첩하여 나타내고 있으며, 질소는 흰색으로, 산소는 빨간색으로 나타내었다.
도 5는 T001의 최적 발현을 위한 UTR 스크리닝의 후보 구조를 나타낸 모식도이다.
도 6a 내지 6m은 T001의 최적 발현을 위한 UTR 스크리닝에서 FACS 분석 결과를 나타낸 사진 및 그래프이다. 분석 조건은 다음과 같다: GFP fluorescence, HCT-116, 6well (5x105 cell/well), 24h mRNA transfection, 10% FBS, 2mM Gln MEM media.
도 7은 mStrawberry의 N-말단 및 C-말단의 시퀀스를 나타낸 모식도이다. 추정되는 임포트 신호(import signal)는 도 7에 나타낸 바와 같이 위치한다.
도 8은 형광 현미경으로 드러난 mStrawberry-NLS 및 mStrawberry-MLS의 세포내 작용 위치를 나타낸 사진이다. 흰색 화살표는 핵의 위치를 나타내고, 검은색 화살표는 미토콘드리아의 위치를 나타낸다.
도 9는 암세포에서 타겟 대사의 작용모드(MOA) 및 경로를 나타낸 도면이다.
도 10은 MCF7 세포주에서 mRNA 트랜스펙션 후 생존검사(live and dead assay) 결과를 나타낸 사진이다. 형광 현미경을 사용하여 5μg/well의 mRNA 처리 후 MCF7 세포의 세포 생존성을 관찰하였다. 트랜스펙션 후 24시간에서 세포 생존성은 효과적으로 감소되었고, 이 효과는 시간 의존적이거나 용량 의존적이었다.
도 11은 MCF7 세포주에서 mRNA 트랜스펙션 후 MTT 분석 결과를 나타낸 그래프이다.
도 12a 내지 12d는 MCF7 세포주에서 mRNA 트랜스펙션 후 Annexin V 염색에 따른 세포사멸 분석 결과를 나타낸 그래프이다. 초기 사멸 부분(오른쪽 아래 사분면)은 mRNA 트랜스펙션 후 지속적으로 증가하였고, 후기 사멸 부분(오른쪽 위 사분면)도 증가하였다.
도 13은 T001과 NT5M 트랜스펙션에 따른 세포독성 및 세포 성장 억제 비교 결과를 나타낸 그래프이다.
도 14a 내지 14c는 CT 및 NT5M 트랜스펙션으로 유도된 세포사멸 결과를 나타낸 그래프이다.
도 15a 내지 15c는 T001 트랜스펙션에 의한 세포주기 정지를 나타낸 그래프이다.
도 16은 대장암 세포주 HCT-116에서 농도에 따른 세포사멸 유발 세포의 비율을 나타낸 그래프이다.
도 17a 내지 17c는 T001의 siRNA 처리에 의한 T001 상쇄 효과를 나타낸 그래프이다.
도 18은 삼중음성 유방암(TNBC)에서 농도에 따른 세포사멸 유발 세포의 비율을 나타낸 그래프이다.
도 19는 삼중음성 유방암에 대하여 CT 처리 후 DNA 손상 마커의 웨스턴 블롯 분석 결과를 나타낸 사진이다.
도 20은 SV4 바인더 및 T001 약물을 이용한 항암제를 나타낸 모식도이다.
도 21은 T001 mRNA의 구성을 개략적으로 나타낸 모식도이다.
도 22는 NLS-mStrawberry mRNA를 포함하는 면역-리포좀(iLP)으로 복합된 카복시 플루오르세인-DSPE(carboxy fluorescein-DSPE)에 대한 체외(in vitro) 분석 결과를 설명하는 모식도 및 사진이다. MCF7 세포주에서 형광 감지를 통해 핵 트랜스펙션에 의해 각 번역된 mStrawberry 단백질 위치와 mRNA 위치를 확인하였다. A는 2.5 ㎍의 NLS-mStrawberry mRNA이고, B는 5 ㎍의 NLS-mStrawberry mRNA이다.
도 23은 체외(in vitro) 및 체내(in vivo)에서 CD47 마스킹 분석(masking assay) 결과를 나타낸 사진이다.
도 24는 SV4-컨쥬게이트 iLP-NIR RFP mRNA의 정맥주사(IV) 후 MCF7 이종이식 마우스 분포를 나타낸 사진이다. A는 이종이식 마우스, B는 주사 후 1시간, C는 주사 후 3시간, D는 주사 후 6시간, E는 절단된 암조직을 나타낸다.
도 25는 체내(in vivo)에서 iLPD 정맥주사 후 기간별 종양 부피를 나타낸 그래프 및 종양 사진이다.
도 26은 iLPD 처리에 의한 마우스 장기의 독성검사 결과를 나타낸 그래프이다.
도 27은 본 발명에 따른 SV4 바인더를 이용한 항암제의 기전을 나타낸 모식도이다.
도 28은 SIRPα 및 SV1의 DNA 서열을 비교하여 나타낸 도면이다.
도 29는 DSPE-PEG2000-NHS와 SV4 단백질의 컨쥬게이션 과정을 설명하는 도면이다.
1 is a diagram showing a comparison of the amino acid sequences of Sirpα, SV1 and SV4. Bold letters indicate residues that are not partially conserved. Sequence alignment was done with ClustalW, and images were created with the BioEdit sequence alignment editing program.
Figure 2 is a diagram explaining mutation of SV1 for correct directionality. In FIG. 2A, the SV domain (green ribbon) is bound to the CD47 (red sphere) domain, and in the model of FIG. show that it is mutated for 2B shows the analysis results using mass spectrometry analysis of DSPE-conjugated SV1 and SV4.
Figure 3 is a diagram showing the conserved motif of T001 and human NT5M. As a sequence alignment of T001 and human NT5M, it was aligned through ClustalW to confirm the sequence similarity of human NT5M and T001. The Swiss-Prot/TrEMBL accession numbers of the sequences used for alignment are human NT5M and T001. Red fields represent fully conserved amino acid residues, and red letters in white fields represent partially conserved amino acid residues with similar biochemical functions. Sequence alignment was done with ClustalW, and images were created with the ESPript server.
4 is a model showing a comparison of structures of T001 and NT5M. Cytoplasmic T001 (CT) and dTMP-bound human NT5M (yellow) are shown superimposed, nitrogen is shown in white and oxygen is shown in red.
5 is a schematic diagram showing candidate structures for UTR screening for optimal expression of T001.
6a to 6m are photographs and graphs showing FACS analysis results in UTR screening for optimal expression of T001. The assay conditions were as follows: GFP fluorescence, HCT-116, 6well (5x10 5 cells/well), 24h mRNA transfection, 10% FBS, 2mM Gln MEM media.
7 is a schematic diagram showing the N-terminal and C-terminal sequences of mStrawberry. The estimated import signal is located as shown in FIG. 7 .
8 is a photograph showing the intracellular action site of mStrawberry-NLS and mStrawberry-MLS revealed by fluorescence microscopy. White arrows indicate the location of the nucleus, and black arrows indicate the location of the mitochondria.
9 is a diagram showing the mode of action (MOA) and pathway of target metabolism in cancer cells.
10 is a photograph showing the results of a live and dead assay after mRNA transfection in MCF7 cell line. Cell viability of MCF7 cells was observed after treatment with 5 μg/well of mRNA using a fluorescence microscope. Cell viability was effectively reduced at 24 hours post-transfection, and this effect was either time dependent or dose dependent.
11 is a graph showing the results of MTT analysis after mRNA transfection in MCF7 cell line.
12a to 12d are graphs showing apoptosis analysis results according to Annexin V staining after mRNA transfection in MCF7 cell line. The early apoptotic portion (lower right quadrant) increased continuously after mRNA transfection, and the late apoptotic portion (upper right quadrant) also increased.
13 is a graph showing comparison results of cytotoxicity and cell growth inhibition according to T001 and NT5M transfection.
14a to 14c are graphs showing apoptosis results induced by CT and NT5M transfection.
15a to 15c are graphs showing cell cycle arrest by T001 transfection.
16 is a graph showing the ratio of apoptosis-inducing cells according to the concentration in the colorectal cancer cell line HCT-116.
17a to 17c are graphs showing the effect of T001 offset by siRNA treatment of T001.
18 is a graph showing the ratio of apoptosis-inducing cells according to the concentration in triple-negative breast cancer (TNBC).
19 is a photograph showing the results of Western blot analysis of DNA damage markers after CT treatment for triple-negative breast cancer.
20 is a schematic diagram showing an anticancer agent using a SV4 binder and a drug T001.
21 is a schematic diagram schematically showing the composition of T001 mRNA.
22 is a schematic diagram and photographs explaining the results of in vitro analysis of carboxy fluorescein-DSPE complexed with an immuno-liposome (iLP) containing NLS-mStrawberry mRNA. The location of each translated mStrawberry protein and mRNA was confirmed by nuclear transfection through fluorescence detection in the MCF7 cell line. A is 2.5 μg of NLS-mStrawberry mRNA and B is 5 μg of NLS-mStrawberry mRNA.
23 is a photograph showing the results of CD47 masking assay in vitro and in vivo .
24 is a photograph showing the distribution of MCF7 xenograft mice after intravenous (IV) injection of SV4-conjugated iLP-NIR RFP mRNA. A is a xenograft mouse, B is 1 hour after injection, C is 3 hours after injection, D is 6 hours after injection, and E is the cut cancer tissue.
25 is a graph and a tumor photograph showing tumor volume by period after iLPD intravenous injection in vivo .
26 is a graph showing the results of toxicity test of mouse organs by iLPD treatment.
27 is a schematic diagram showing the mechanism of anticancer agents using the SV4 binder according to the present invention.
28 is a diagram showing a comparison of DNA sequences of SIRPα and SV1.
29 is a diagram explaining the conjugation process between DSPE-PEG 2000 -NHS and SV4 protein.

이하, 실시예를 통하여 본 발명을 상세히 설명하기로 한다. 이에 앞서, 본 명세서 및 청구범위에 사용된 용어나 단어는 통상적이거나 사전적인 의미로 한정해서 해석되어서는 아니 되며, 발명자는 그 자신의 발명을 가장 최선의 방법으로 설명하기 위해 용어의 개념을 적절하게 정의할 수 있다는 원칙에 입각하여, 본 발명의 기술적 사상에 부합하는 의미와 개념으로 해석되어야만 한다. 따라서, 본 명세서에 기재된 실시예의 구성은 본 발명의 가장 바람직한 일실시예에 불과할 뿐이고 본 발명의 기술적 사상을 모두 대변하는 것은 아니므로, 본 출원 시점에 있어서 이들을 대체할 수 있는 다양한 균등물과 변형예들이 있을 수 있음을 이해하여야 한다.Hereinafter, the present invention will be described in detail through examples. Prior to this, the terms or words used in this specification and claims should not be construed as being limited to the usual or dictionary meaning, and the inventor appropriately uses the concept of the term in order to explain his/her invention in the best way. Based on the principle that it can be defined, it should be interpreted as meaning and concept consistent with the technical spirit of the present invention. Therefore, since the configurations of the embodiments described in this specification are merely the most preferred embodiments of the present invention and do not represent all of the technical spirit of the present invention, various equivalents and modifications that can replace them at the time of this application It should be understood that there may be

본원에 사용된 용어는 다음과 같이 이해될 수 있다.The terms used herein can be understood as follows.

본원에서 용어 “CD47”은 특별히 제한되지 않고, 임의의 동물, 바람직하게는 포유류에서 유래한 것일 수 있으며, 더욱 바람직하게는 인간 CD47 일 수 있다. 인간 CD47의 아미노산 서열 및 뉴클레오티드 서열은 이미 공지되어 있다(J. Cell. Biol., 123, 485-496, (1993), Journal of Cell Science, 108, 3419-3425, (1995), GenBank: Z25521).The term “CD47” herein is not particularly limited, and may be derived from any animal, preferably a mammal, and more preferably human CD47. The amino acid sequence and nucleotide sequence of human CD47 are already known (J. Cell. Biol., 123, 485-496, (1993), Journal of Cell Science, 108, 3419-3425, (1995), GenBank: Z25521). .

본원에서 용어 “바인더”는 수용체, 특히, CD47에 결합하는 단백질로서, 바인더는 특히 암세포에서 CD47과 결합하여, 세포-전달물질의 인식 및/또는 상호작용을 가능하게 한다.As used herein, the term “binder” refers to a protein that binds to a receptor, in particular CD47, wherein the binder binds to CD47, particularly in cancer cells, enabling recognition and/or interaction with a cell-transmitter.

본원에서 용어 "컨쥬게이트된" 또는 "컨쥬게이트"는 둘 또는 그 이상의 화합물이 하나 또는 그 이상의 화학 결합 또는 링커와 결합하여 형성되는 화학적 화합물을 의미한다. 본 발명의 일 구체예에서, 바인더와 리포좀은 컨쥬게이트를 형성한다.As used herein, the term "conjugated" or "conjugate" refers to a chemical compound formed by combining two or more compounds with one or more chemical bonds or linkers. In one embodiment of the invention, the binder and the liposome form a conjugate.

본원에서 용어 “페길화(PEGylation)”는 대상물질에 폴리에틸렌글리콜(PEG)을 접합시켜 안정성을 높이기 위한 기술로서, 본 발명에서 페길화된 인지질이 사용될 수 있으며, 예컨대, DSPE-PEG2000 등이 사용될 수 있다. DSPE-PEG2000은 약 2000의 수평균분자량을 가지는 PEG가 부착된 DSPE를 의미한다.As used herein, the term “PEGylation” refers to a technique for conjugating polyethylene glycol (PEG) to a target material to increase stability, and in the present invention, a pegylated phospholipid may be used, for example, DSPE-PEG 2000 or the like may be used. can DSPE-PEG 2000 means DSPE attached with PEG having a number average molecular weight of about 2000.

본원에서 용어 “폴리뉴클레오티드(polynucleotide)”는 일반적으로 RNA 또는 DNA, 또는 변형된 RNA 또는 DNA일 수 있는, 단일가닥 또는 이중가닥 형태로 존재하는 디옥시리보뉴클레오티드 또는 리보뉴클레오티드의 중합체를 의미한다. 본 발명의 일 구체예에서, 폴리뉴클레오티드는 합성된 단쇄 mRNA이다.As used herein, the term “polynucleotide” generally refers to deoxyribonucleotides or polymers of ribonucleotides, which may be RNA or DNA, or modified RNA or DNA, in single-stranded or double-stranded form. In one embodiment of the present invention, the polynucleotide is a synthesized single-chain "mRNA".

본원에서 용어 “5'-UTR(5'-Untransrated Region)”은 통상적으로 mRNA의 특정 부분으로 이해되고, mRNA의 단백질 암호화 영역(즉, 개방 해독 틀(ORF))의 5'에 위치한다. 통상적으로, 5'-UTR은 전사 시작 영역에서 시작하여 개방 해독 틀의 개시 코돈에서 한 뉴클레오티드 전에 종결한다.As used herein, the term "5'-Untranslated Region (5'-UTR)" is commonly understood as a specific portion of mRNA, and is located 5' of the protein coding region (ie, open reading frame (ORF)) of mRNA. Typically, the 5'-UTR begins at the transcription start region and ends one nucleotide before the initiation codon of the open reading frame.

본원에서 용어 “3'-UTR(3'-Untransrated Region)”은 통상 mRNA의 개방 해독 틀(ORF)과 폴리A 서열 사이에 위치하는 mRNA의 일부이다. mRNA의 3'-UTR은 아미노산 서열로 번역되지 않는다. 3'-UTR 서열은 보통 유전자 발현 과정에서 각각의 mRNA로 전사되는 유전자에 의해 암호화된다.As used herein, the term "3'-Untranslated Region (3'-UTR)" is a part of mRNA that is usually located between the open reading frame (ORF) of mRNA and the polyA sequence. The 3'-UTR of mRNA is not translated into an amino acid sequence. The 3'-UTR sequence is usually encoded by a gene that is transcribed into each mRNA during gene expression.

본원에서 용어 “Kozak 서열”은 유전자 또는 개방 해독 틀(ORF)의 발현을 개선하는 번역 개시 인핸서 요소를 지칭하며, 진핵 생물에서 이는 5'-UTR에 위치하되 개시 코돈의 -3번 위치에 A 또는 G를 포함하는 서열로 위치하고, 개시 코돈의 +1번 위치에 G를 포함하는 서열(예컨대, -GTG-)을 더 포함할 수 있다.The term “Kozak sequence” herein refers to a translational initiation enhancer element that improves expression of a gene or open reading frame (ORF), which in eukaryotes is located in the 5'-UTR but at position -3 of the initiation codon is an A or It is located in a sequence containing G and may further include a sequence (eg, -GTG-) containing G at position +1 of the initiation codon.

본원에서 용어 “NLS(nuclear localization signal, 핵 위치 신호)” 및 “MLS(mitochondrial localization signal, 미토콘드리아 위치 신호)”는 각각 단백질이나 핵산과 같은 특정 물질을 세포 핵 및 미토콘드리아 내로 운반하는 역할을 하는 아미노산 서열을 의미한다.As used herein, the terms “nuclear localization signal (NLS)” and “mitochondrial localization signal (MLS)” are amino acid sequences that serve to transport specific substances such as proteins or nucleic acids into the cell nucleus and mitochondria, respectively. means

본원에서 용어 “트랜스펙션”은 세포 외부의 폴리뉴클레오티드가 수반 물질이 있는 또는 없는 상태로 숙주 세포, 특히, 암세포로 들어가는 과정을 의미한다. “트랜스펙션된 세포"는 예컨대, 세포 외부의 mRNA가 세포 내로 도입되어 세포 외부 mRNA를 가지고 있는 세포를 의미할 수 있다.As used herein, the term “transfection” refers to a process in which an extracellular polynucleotide enters a host cell, particularly a cancer cell, with or without an accompanying substance. “Transfected cell” may mean, for example, a cell having an extracellular mRNA by introducing an extracellular mRNA into the cell.

본 발명은 서열번호 3으로 표시되는 아미노산 서열을 암호화하는 암 치료를 위한 폴리뉴클레오티드를 개시한다.The present invention discloses a polynucleotide for cancer treatment encoding the amino acid sequence represented by SEQ ID NO: 3.

본 발명에 따른 암 치료를 위한 폴리뉴클레오티드는 일 구현예에 따르면, CD47에 결합하는 바인더와 복합체를 형성하는 리포좀형 나노입자에 포집된 형태로 사용되어 암세포의 대사취약성(metabolic vulnerability)을 극대화시켜 암세포를 사멸시키는 기작을 가지는 암 치료를 위한 폴리뉴클레오티드로서, 바람직하게는 인간세포 내에 전달되었을 때 비특이적 마이크로RNA(microRNA)로서 작용하는 부위를 최소화하고 발현을 최대화할 수 있도록 유전자 서열이 최적화되어 있다. 이러한 유전자 서열이 최적화된 폴리뉴클레오티드로서는 서열번호 39 내지 서열번호 41로 표시되는 염기서열 중에서 선택될 수 있고, 바람직하게는 서열번호 39로 표시되는 염기서열이 선택될 수 있다. 서열번호 38은 야생형(wild type) 서열을 나타내고 있다.According to one embodiment, the polynucleotide for cancer treatment according to the present invention is used in the form of being encapsulated in liposomal nanoparticles that form a complex with a binder that binds to CD47, thereby maximizing the metabolic vulnerability of cancer cells. As a polynucleotide for cancer treatment having a mechanism of killing, preferably, the gene sequence is optimized to minimize the site acting as a non-specific microRNA (microRNA) and maximize expression when delivered into human cells. As the polynucleotide whose gene sequence is optimized, it can be selected from the nucleotide sequences represented by SEQ ID NO: 39 to SEQ ID NO: 41, and preferably, the nucleotide sequence represented by SEQ ID NO: 39 can be selected. SEQ ID NO: 38 represents a wild type sequence.

본 발명의 일 구현예에서 상기 바인더는 암세포에서 과발현하는 CD47에 결합하는 바인더로서, 서열번호 1 또는 서열번호 2로 표시되는 아미노산 서열을 포함한다.In one embodiment of the present invention, the binder binds to CD47 overexpressed in cancer cells, and includes the amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 2.

본 발명에서 서열번호 1로 표시되는 아미노산 서열을 포함하는 바인더는 'SV1'로 명명하였으며, 서열번호 2로 표시되는 아미노산 서열을 포함하는 바인더는 'SV4'로 명명하였다.In the present invention, the binder comprising the amino acid sequence represented by SEQ ID NO: 1 was named 'SV1', and the binder comprising the amino acid sequence represented by SEQ ID NO: 2 was named 'SV4'.

먼저, SV1(Sirpα-variant-version1)은 본래(original) CD47 리간드의 수용성 도메인인 순수 Sirpα(아미노산 서열(서열번호 24) 참조)에서 유래된 돌연변이 라이브러리에서 선별되었다. SV1 돌연변이의 선별과정은 다음과 같다.First, Sirpα-variant-version1 (SV1) was selected from a mutant library derived from pure Sirpα (see amino acid sequence (SEQ ID NO: 24)), which is the soluble domain of the original CD47 ligand. The selection process for the SV1 mutation is as follows.

CD47과 결합하는 SIRPα의 결합력이 개선된 단백질을 Weiskopf K et al.(Weiskopf, K., et al. (2013). "Engineered SIRPalpha variants as immunotherapeutic adjuvants to anticancer antibodies." Science (New York, N.Y.) 341(6141): 88-91.)을 참조하여 인간 SIRPa의 단일 14-kD 결합 도메인을 합성하여 유전자를 확보하였다. Wild-type SIRPα로부터 변경된 아미노산은 각각 다음과 같다. 6번째에 위치하는 발린(valine)을 이소류신(isoleucine)으로, 27번째에 위치하는 발린(valine)을 이소류신(isoleucine)으로, 31번째에 위치하는 이소류신(isoleucine)을 페닐알라닌(phenylalanine)으로, 47번째 위치하는 글루탐산(glutamate)을 발린(valine)으로, 53번째에 위치하는 라이신(lysine)을 아르기닌(arginine)으로, 54번째에 위치하는 글루탐산(glutamate)을 글루타민(glutamine)으로, 56번째에 위치하는 히스타민(histidine)을 프롤린(proline)으로, 66번째에 위치하는 세린(serine)을 트레오닌(Threonine)으로, 92번째에 위치하는 발린(valine)을 이소류신(isoleucine)으로 치환하였다. 단백질의 대장균에서의 발현을 위해 기존 핵산 서열을 코돈 최적화를 통해 변형하여 SV1을 제조하였으며 도 28에 SIRPα 서열(서열번호 25) 및 SV1의 DNA 서열(서열번호 26)을 비교하여 나타내었다.Weiskopf K et al. (Weiskopf, K., et al. (2013). "Engineered SIRPalpha variants as immunotherapeutic adjuvants to anticancer antibodies." Science (New York, N.Y.) 341 (6141): 88-91.), the gene was obtained by synthesizing a single 14-kD binding domain of human SIRPa. Amino acids changed from wild-type SIRPα are as follows. Valine at the 6th position is converted to isoleucine, valine located at the 27th position to isoleucine, isoleucine located at the 31st position to phenylalanine, Glutamate at position valine, lysine at position 53 to arginine, glutamate at position 54 to glutamine, position at position 56 Histidine was replaced with proline, serine at position 66 with threonine, and valine at position 92 was replaced with isoleucine. For protein expression in E. coli, SV1 was prepared by modifying the existing nucleic acid sequence through codon optimization, and FIG. 28 shows a comparison between the SIRPα sequence (SEQ ID NO: 25) and the DNA sequence (SEQ ID NO: 26) of SV1.

한편, SV1의 C-말단에 CRM197 단백질을 첨가한 단백질인 SV1-(C)CRM197, N-말단에 CRM197 단백질을 첨가한 CRM197(N)-SV1, 그리고 SV1 단백질을 제작하여 SV1 N-말단과 C-말단의 단백질의 첨가의 영향을 SPR(Surface Plasmon Resonance) 분석을 통해 확인하였다. 각 시료는 Biacore X100 기기를 이용하여 SPR의 binding kinetics를 통해 분석하였다. 이때 분석을 위한 Chip으로서 Recombinant CD47 단백질을 500 RU이 되도록 Protein G 표면에 접합한 Chip을 사용하였고, HBS-P는 5 ㎕/min의 유속으로 분석하였다. 각 센소그램(sensogram) 결과는 1 : 1 바인딩 모델 및 글로벌 피팅을 사용하여 BiaEvaluation 소프트웨어로 분석하였고, 그 결과를 하기 표 1에 나타내었다.On the other hand, SV1-(C)CRM197, which is a protein in which CRM197 protein is added to the C-terminus of SV1, CRM197(N)-SV1 in which CRM197 protein is added to the N-terminus, and SV1 protein are prepared to make SV1 N-terminus and CRM197 protein. - The effect of adding the protein at the end was confirmed through SPR (Surface Plasmon Resonance) analysis. Each sample was analyzed through binding kinetics of SPR using a Biacore X100 instrument. At this time, as a chip for analysis, a chip in which recombinant CD47 protein was conjugated to the surface of Protein G to 500 RU was used, and HBS-P was analyzed at a flow rate of 5 μl/min. Each sensogram result was analyzed with BiaEvaluation software using a 1:1 binding model and global fitting, and the results are shown in Table 1 below.

AnalyteAnalyte KD (nM)K D (nM) SV1 proteinSV1 protein 0.8770.877 SV1-CRM197 proteinSV1-CRM197 protein 0.8940.894 CRM197-SV1 proteinCRM197-SV1 protein 19.919.9

표 1을 참조하면, SPR 분석 결과에서 C-말단에 CRM197이 삽입된 SV1-CRM197은 SV1에 비해 KD값이 크게 달라지지 않았으나, N-말단에 CRM197이 삽입된 CRM197-SV1 같은 경우에 affinity가 KD값이 19.9 nM까지 약 2배 가량 상승하여 친화력이 감소한 것을 확인하였다. 따라서 SV1의 N-말단에서 단백질을 첨가하는 경우, SV가 CD47에 올바르게 결합하는 것을 방해하여 발생되는 입체장애(steric hindrance)로 인해 CD47에 대한 친화도가 SV1에 비해 감소하는 것으로 확인되었다.Referring to Table 1, in the results of SPR analysis, the KD value of SV1-CRM197 with CRM197 inserted at the C-terminus did not change significantly compared to SV1, but in the case of CRM197-SV1 with CRM197 inserted at the N-terminus, affinity was It was confirmed that the KD value increased about 2 times to 19.9 nM, and the affinity decreased. Therefore, it was confirmed that when a protein was added at the N-terminus of SV1, the affinity for CD47 was reduced compared to SV1 due to steric hindrance caused by preventing SV from correctly binding to CD47.

이하, 본 발명에 관련된 SV4 바인더, 상기 바인더와 컨쥬게이트되는 리포좀에 관한 구체적인 실시예를 상세히 설명한 후, 본 발명에 따른 바인더-컨쥬케이트 리포좀에 포집되는 T001 약물과, SV4 바인더 및 T001 약물을 이용한 항암제에 관한 구체적인 실시예를 들어 본 발명을 상세히 설명한다.Hereinafter, specific examples of the SV4 binder related to the present invention and the liposome conjugated with the binder will be described in detail, and then the T001 drug encapsulated in the binder-conjugated liposome according to the present invention, and the anticancer agent using the SV4 binder and the T001 drug The present invention will be described in detail with reference to specific examples.

SV4-컨쥬케이트 리포좀SV4-conjugated liposomes

지질 나노입자(lipid nanoparticle)를 기반으로 한 약물전달 시스템은 상당한 장점을 가져 보편적으로 사용되어 왔다. 지질 나노입자는 높은 약 용량(drug capacity), 높은 안정성(stability), 높은 특이성(specificity)을 지니고 있으며, 방출 시점(release point)을 제어할 수 있다. 약물로 사용되는 유전 물질 때문에, 양이온성 리포좀은 표적 암에 약물을 전달하는 데 사용되었다. 양전하로 하전된 리포좀은 유전적 약물(genetic drug)을 끌어당겨 약물이 표적을 만날 때까지 보호를 위해 약물을 덮는 구형 콤플렉스(spherical complex)를 형성한다.Drug delivery systems based on lipid nanoparticles have been widely used with significant advantages. Lipid nanoparticles have high drug capacity, high stability, and high specificity, and the release point can be controlled. Because of the genetic material used as a drug, cationic liposomes have been used to deliver drugs to target cancers. The positively charged liposomes attract the genetic drug and form a spherical complex that covers the drug for protection until it meets its target.

본 발명에서 양이온성 리포좀을 형성하기 위해 프로-리포좀(Pro-liposome)은 양이온성 인지질, DOTAP 및 콜레스테롤 중 하나를 사용하여 준비되었다. 또한 혈관을 통해 타겟에 약물을 전달하는 동안 혈청 안정성을 높이고자 페길화 리포좀을 준비하기 위해 프로-리포좀에 PEG1000-DSPE를 추가로 첨가하였다. 페길화 리포좀 외에도, NHS 활성화 DSPE-PEG2000과 SV4 단백질을 컨쥬게이트하여 DSPE-PEG2000-SV4를 준비하였다. SV4 컨쥬게이트 리포좀을 준비하기 위한 마지막 단계로, mRNA 캡슐화 프로-리포좀과 DSPE-PEG2000-SV4를 혼합하였다. 구체적인 SV4-컨쥬케이트 리포좀 제조과정은 다음과 같다.In the present invention, to form cationic liposomes, pro-liposomes were prepared using one of cationic phospholipids, DOTAP and cholesterol. In addition, PEG 1000 -DSPE was additionally added to pro-liposomes to prepare pegylated liposomes to increase serum stability during drug delivery to the target through blood vessels. In addition to pegylated liposomes, DSPE-PEG 2000 -SV4 was prepared by conjugating NHS activated DSPE-PEG 2000 with SV4 protein. As a final step to prepare SV4 conjugated liposomes, mRNA encapsulated pro-liposomes were mixed with DSPE-PEG 2000 -SV4. The specific SV4-conjugated liposome manufacturing process is as follows.

리포좀은 드라이-필름 방식으로 준비되었다. DOTAP(Avanti Polar Lipids)과 콜레스테롤(Sigma)(1:1 몰비, 10 mM)로 구성된 양이온성 리포좀과 PEG-DSPE1000(1 mM; Avanti Polar Lipids)이 첨가되었다. 양이온성 리포좀은 둥근바닥 유리 플라스크에서 클로로포름과 메탄올(2:1 (v/v))에 용해되었다. 지질 건조는 50℃에서 회전 증발기로 진공 상태에서 수행되었다. 클로로포름과 메탄올을 완전히 제거하기 위해 지질막을 하룻밤 냉동 건조시켰다. 증발 후 지질막은 50℃에서 최대 1시간까지 뉴클레아제 프리 워터(nuclease free water)로 재수화(rehydrated)되었다. 수화된 지질막을 음파 처리하여 단일층 소포(unilamellar vesicle)를 형성하였다. 마지막으로 100 nm 공극을 가진 막을 이용하여 미니 압출기(Avanti Polar Lipids)로 지질을 압출하였다.Liposomes were prepared by dry-film method. Cationic liposomes composed of DOTAP (Avanti Polar Lipids) and cholesterol (Sigma) (1:1 molar ratio, 10 mM) and PEG-DSPE1000 (1 mM; Avanti Polar Lipids) were added. Cationic liposomes were dissolved in chloroform and methanol (2:1 (v/v)) in a round bottom glass flask. Lipid drying was performed under vacuum on a rotary evaporator at 50 °C. The lipid film was freeze-dried overnight to completely remove chloroform and methanol. After evaporation, the lipid film was rehydrated with nuclease free water at 50 °C for up to 1 hour. The hydrated lipid membrane was sonicated to form unilamellar vesicles. Finally, lipids were extruded with a mini extruder (Avanti Polar Lipids) using a membrane with a 100 nm pore.

DSPE-PEG2000-NHS와 SV4 단백질의 컨쥬게이션은 다음과 같이 준비되었다(도 29 참조). DSPE-PEG2000-NHS는 드라이-필름 방식으로 준비하였다. 클로로포름에 용해된 DSPE-PEG2000-NHS는 둥근 바닥 유리 플라스크에서 증발되었다. 30℃에서 진공 상태로 1시간 동안 회전 증발기에 의해 지질 건조를 수행하였다. 증발 후 지질막은 30℃에서 1시간까지 뉴클레아제 프리 워터에 용해된 SV4 단백질로 재수화되었다. 컨쥬게이트되지 않은 잔여 DSPE-PEG2000-NHS를 제거하기 위해 pH 6.8의 PBS에서 투석 카세트 10,000 MWCO(Thermo Scientific)를 하룻밤 수행하였다.Conjugation of DSPE-PEG 2000 -NHS with SV4 protein was prepared as follows (see Figure 29). DSPE-PEG 2000 -NHS was prepared by dry-film method. DSPE-PEG 2000 -NHS dissolved in chloroform was evaporated in a round bottom glass flask. Lipid drying was performed by rotary evaporator at 30° C. under vacuum for 1 hour. After evaporation, the lipid film was rehydrated with SV4 protein dissolved in nuclease-free water for 1 hour at 30 °C. A 10,000 MWCO (Thermo Scientific) dialysis cassette was run overnight in PBS pH 6.8 to remove residual unconjugated DSPE-PEG 2000 -NHS.

캡슐화된 약물과 결합 리간드를 함유한 리포좀을 전술한 바와 같이 준비하였다. 양이온성 리포좀 3 mg, 프로타민(protamine) 25 ㎍ 및 디에틸피로카보네이트수(diethylpyrocarbonate water)를 혼합하여 용액 A로, mRNA 50 ㎍ 및 디에틸피로카보네이트수를 혼합하여 용액 B로 하였다. 용액 A와 B는 디에틸피로카보네이트수로 부피를 같게 하여 30분간 배양되었다. 이후, 30분 동안 혼합 및 배양하여 캡슐화된 약물로 리포좀을 형성하였다. DSPE-PEG2000-NHS 컨쥬게이트 SV4 리간드의 결합을 위해 캡슐화된 약물을 함유한 리포좀(1:100 몰비)을 50℃에서 15분간 혼합하였다.Liposomes containing encapsulated drug and binding ligand were prepared as described above. 3 mg of cationic liposome, 25 μg of protamine, and diethylpyrocarbonate water were mixed to obtain solution A, and 50 μg of mRNA and diethylpyrocarbonate water were mixed to obtain solution B. Solutions A and B were equalized in volume with diethyl pyrocarbonate water and incubated for 30 minutes. Thereafter, mixing and incubation for 30 minutes formed liposomes with the encapsulated drug. For the binding of the DSPE-PEG 2000 -NHS conjugate SV4 ligand, liposomes (1:100 molar ratio) containing the encapsulated drug were mixed at 50°C for 15 minutes.

컨쥬게이션 타겟으로서 SV1SV1 as a conjugation target

자연계에 존재하는 Sirpα의 서열을 변형하여 CD47과의 결합력이 향상된 SV1을 돌연변이를 통해 선정하고, 이에 대해 리포좀 제형을 제작할 때 올바른 방향으로 삽입되어 반응할 수 있는 SV4를 돌연변이를 통해 확보하였다(도 1 참조). SV4 돌연변이 제작은 다음의 방법으로 수행되었다. 상기 확보된 SV1 서열에서 CD47과의 올바른 방향 결합을 위해 11번째와 104번째 라이신(Lysine)을 류신(leucine)으로 치환하였다. 치환을 위해 SV1 유전자를 pET28a 벡터에 삽입한 플라스미드를 이용하여 11번째와 104번째에 해당되는 서열로 점돌연변이(point-mutation)될 수 있도록 프라이머(하기 표 2 참조)를 제작하여 Quickchange Ⅱ site-directed mutagenesis kit(Agilent)의 방법대로 각각 혹은 동시에 치환시킨 돌연변이 유전자를 제작하였다.By modifying the sequence of Sirpα present in nature, SV1 with improved binding ability to CD47 was selected through mutation, and SV4, which can be inserted in the correct direction and reacted when preparing a liposome formulation, was secured through mutation (FIG. 1). reference). SV4 mutant construction was performed in the following way. In the SV1 sequence obtained above, 11th and 104th lysines were substituted with leucine for binding in the correct direction with CD47. For substitution, using a plasmid in which the SV1 gene was inserted into the pET28a vector, primers (see Table 2 below) were prepared for point-mutation to sequences corresponding to the 11th and 104th positions, and Quickchange II site-directed According to the method of the mutagenesis kit (Agilent), mutant genes each or simultaneously substituted were prepared.

구분division 서열order 서열번호sequence number Leu 11 mutagenesisLeu 11 mutagenesis (KKtoLL11_F1) Forward(KKtoLL11_F1) Forward GAT TAT TCA GCC GGA CCT GTC CGT AAG CGT TGCGAT TAT TCA GCC GGA CCT GTC CGT AAG CGT TGC 2727 (KKtoLL11_R1) Reverse(KKtoLL11_R1) Reverse GCA ACG CTT ACG GAC AGG TCC GGC TGA ATA ATCGCA ACG CTT ACG GAC AGG TCC GGC TGA ATA ATC 2828 Leu 104 mutagenesisLeu 104 mutagenesis (KKtoLL104_F2) Forward(KKtoLL104_F2) Forward CTG ACA CGG AGT TTC TGT CTG GCG CAGCTG ACA CGG AGT TTC TGT CTG GCG CAG 2929 (KKtoLL104_R2) Reverse(KKtoLL104_R2) Reverse CTG CGC CAG ACA GAA ACT CCG TGT CAGCTG CGC CAG ACA GAA ACT CCG TGT CAG 3030

SV1 단백질은 N-말단 아미노기 외에 6개의 라이신(Lysine) 잔기를 가지고 있다(도 2A 참조). SV1을 리포좀과 컨쥬게이션을 통해 연결하여 CD47과의 결합을 통해 약물을 전달하기 위해서는 결합력의 향상이 필요하다. NHS 컨쥬게이션을 통해 DSPE를 연결하는 화학반응에서 작용할 수 있는 잔기 중 CD47과의 결합을 방해하지 않으면서 올바른 방향(correct orientation)으로 작용할 수 있는 잔기를 선정하고 이들 잔기 이외의 잔기를 류신(Leucine)으로 치환함으로써 결합 활성의 손실 없이 변형할 수 있다. 도 2에 나타낸 바와 같이, 치환을 통해 DSPE에 결합하는 잔기의 경우의 수를 줄여 단순한 결합을 통해 DSPE-컨쥬게이트된 SV4를 제조하였다.SV1 protein has six lysine residues in addition to the N-terminal amino group (see FIG. 2A). In order to deliver a drug through binding to CD47 by linking SV1 with liposome through conjugation, it is necessary to improve the binding force. Among the residues that can act in the chemical reaction linking DSPE through NHS conjugation, residues that can act in the correct orientation without interfering with CD47 binding are selected, and residues other than these residues are selected as Leucine. can be modified without loss of binding activity. As shown in FIG. 2, DSPE-conjugated SV4 was prepared through simple coupling by reducing the number of residues binding to DSPE through substitution.

한편, SV4의 올바른 방향(correct orientation)을 위한 라이신(Lysine) 치환 효과를 확인하기 위하여 SV1과의 비교를 위해 DSPE-컨쥬게이션 형태 및 리포좀에 삽입된 형태를 각각 제조하여 CD47과의 친화력을 SPR(Surface Plasmon Resonance)을 통해 분석하였다. 분석을 위하여 CM5 Chip을 이용하여 CD47을 접합한 후 SV1, DSPE-SV1, LPD-SV1, SV4, DSPE-SV4 및 LPD-SV4의 회합(association)과 해리(dissociation)를 측정하여 KD 값을 비교하였다. 구체적으로 재조합(recombinant) CD47 단백질을 EDC-NHS 반응을 통해 250 RU의 표적 SPR 반응으로 CM5 칩의 표면에 접합시켜 사용하였으며 HBS-P는 30 ㎕/min의 유속으로 3분 회합 및 10분 해리 과정을 통해 확보한 센소그램(sensogram)을 1:1 바인딩 모델 및 글로벌 피팅을 사용하여 BiaEvaluation 소프트웨어로 분석하였다. 분석 시료의 특성에 따라 해리 버퍼(dissociation buffer)로, SV1 및 SV4 단백질에 대하여 10 mM 글리신(glycine) pH 2.0을, 지방산이 부착된 시료에 대해서는 2.0 M MgCl2을 사용하였다.On the other hand, in order to confirm the Lysine substitution effect for the correct orientation of SV4, a DSPE-conjugated form and a form inserted into liposomes were prepared for comparison with SV1, respectively, and affinity with CD47 was measured by SPR ( Surface Plasmon Resonance) was analyzed. For analysis, after splicing CD47 using CM5 Chip, compare KD values by measuring association and dissociation of SV1, DSPE-SV1, LPD-SV1, SV4, DSPE-SV4 and LPD-SV4 did Specifically, the recombinant CD47 protein was conjugated to the surface of the CM5 chip with a target SPR reaction of 250 RU through the EDC-NHS reaction, and HBS-P was used at a flow rate of 30 μl/min for 3 minutes of association and 10 minutes of dissociation The sensogram obtained through was analyzed with BiaEvaluation software using a 1:1 binding model and global fitting. 10 mM glycine pH 2.0 was used for the SV1 and SV4 proteins, and 2.0 M MgCl 2 was used for the sample with attached fatty acids as a dissociation buffer according to the characteristics of the sample to be analyzed.

SPR 분석 결과에서 SV1은 Sirpα(wt)에 비해 KD값이 280 nM에서 0.87 nM으로 크게 향상되었다(표 1 참조). SV1의 친화력(affinity)은 크게 향상되었지만 NHS 컨쥬게이션 반응을 통해 SV1-DSPE를 제조하였을 경우에 0.87 nM에서 2.67 nM로 KD값 상승으로 인해 친화력은 감소하였다. 또한 리포좀에 삽입한 SV1-iLP의 경우에는 더욱 친화력이 감소하여 KD값이 10.9 nM까지 상승하였다. 그에 비해 NHS 반응 잔기인 2군데의 라이신을 류신으로 치환한 SV4의 경우 단백질 자체의 친화력은 다소 감소하였으나, 올바른 방향성으로 인해 DSPE와 컨쥬게이션했을 경우와 리포좀에 삽입되었을 때조차도 큰 감소 없이 오차 범위에서 결합 활성이 유지되는 것을 확인하였다(표 3 참조).In the results of SPR analysis, the KD value of SV1 was significantly improved from 280 nM to 0.87 nM compared to Sirpα (wt) (see Table 1). The affinity of SV1 was greatly improved, but when SV1-DSPE was prepared through the NHS conjugation reaction, the affinity decreased due to the increase in KD value from 0.87 nM to 2.67 nM. Also, in the case of SV1-iLP inserted into liposomes, the affinity was further decreased and the KD value increased to 10.9 nM. In contrast, in the case of SV4, in which two lysines, which are NHS reactive residues, were substituted with leucine, the affinity of the protein itself was slightly reduced, but due to the correct orientation, it was conjugated with DSPE and even when inserted into liposomes, it was within the error range without a significant decrease. It was confirmed that the binding activity was maintained (see Table 3).

AnalyteAnalyte k a (M-1s-1) k a (M -1 s -1 ) k d (s-1) k d ( s -1 ) KD (nM)K D (nM) SirpαSirpα 6.10×105 6.10×10 5 3.70×10-1 3.70×10 -1 290.0290.0 SV1SV1 3.53×105 3.53×10 5 3.08×10-4 3.08×10 -4 0.870.87 SV1-DSPESV1-DSPE 7.34×104 7.34×10 4 1.96×10-4 1.96×10 -4 2.672.67 SV1-iLPSV1-iLP 1.73×104 1.73×10 4 1.89×10-4 1.89×10 -4 10.910.9 SV4SV4 1.64×105 1.64×10 5 2.75×10-4 2.75×10 -4 1.671.67 SV4-DSPESV4-DSPE 2.27E×105 2.27E×10 5 2.56×10-4 2.56×10 -4 1.131.13 SV4-iLPSV4-iLP 3.12E×105 3.12E×10 5 3.95×10-4 3.95×10 -4 1.271.27

바인더-컨쥬케이트 리포좀 포집 약물Binder-conjugated liposomal entrapment drugs

이하, 전술한 바인더(SV4)-컨쥬케이트 리포좀에 포집되는 약물의 구현예에 관하여 설명한다.Hereinafter, embodiments of the drug encapsulated in the binder (SV4)-conjugated liposome will be described.

인간 5'-뉴클레오티다아제(Human 5'-nucleotidase)와 유사한 활성을 가지나 이례적으로 다른 핵산에 대해서는 특이성이 없으면서 dTMP, dUMP에 대해서만 높은 특이성을 가지는 PBS2 파지(Bacteriophage PBS2) 유래 티미딜레이트 5'-포스포하이드롤라아제(Thymidylate 5'-phosphohydrolase)를 암세포의 대사취약성(metabolic vulnerability)을 극대화시켜 암세포를 사멸시키는 기작을 가지는 약물후보물질로 선정하고, 인간세포 내에 전달되었을 때 비특이적 마이크로RNA(microRNA)로서 작용하는 부위를 최소화하고 발현을 최대화할 수 있도록 유전자 서열을 최적화하여, 서열번호 3으로 표시되는 아미노산 서열과, 이를 암호화하는 폴리뉴클레오티드(서열번호 39)를 'T001' 로 명명하였고, 변이 폴리뉴클레오티드(서열번호 40 및 서열번호 41)로서 각각 'T002' 및 'T007'로 명명하였다. T001의 서열 최적화 과정은 다음과 같다.Thymidylate 5' derived from Bacteriophage PBS2, which has activity similar to that of human 5'-nucleotidase, but has exceptionally high specificity only for dTMP and dUMP without specificity for other nucleic acids. -Phosphohydrolase (Thymidylate 5'-phosphohydrolase) was selected as a drug candidate with a mechanism to kill cancer cells by maximizing the metabolic vulnerability of cancer cells, and when delivered into human cells, non-specific microRNA (microRNA) ), the amino acid sequence represented by SEQ ID NO: 3 and the polynucleotide encoding it (SEQ ID NO: 39) were named 'T001' by optimizing the gene sequence to maximize expression and minimize the site acting as Nucleotides (SEQ ID NO: 40 and SEQ ID NO: 41) were named 'T002' and 'T007', respectively. The sequence optimization process of T001 is as follows.

DNA 서열을 포유류 세포 발현(mammalian cell expression)을 위한 코돈 최적화하고자 자연 코돈(natural codon)을 다음의 최적 코돈으로 대체하였다: alanine (GCC), arginine (CGC), asparagine (AAC), aspartic acid (GAC), cysteine (TGC), glutamic acid (GAG), glutamine (CAG), glycine (GGC), histidine (CAC), isoleucine (ATC), leucine (CTG), lysine (AAG), methionine (ATG), phenylalanine (TTC), proline (CCC), serine (TCC), threonine (ACC), tryptophan (TGG), tyrosine (TAC), and valine (GTG). Runs of Cs and Gs를 회피하여 PCR 조건뿐 아니라 올리고뉴클레오티드 합성을 간략화하였다.To optimize codons for mammalian cell expression in the DNA sequence, natural codons were replaced with the following optimal codons: alanine (GCC), arginine (CGC), asparagine (AAC), aspartic acid (GAC). ), cysteine (TGC), glutamic acid (GAG), glutamine (CAG), glycine (GGC), histidine (CAC), isoleucine (ATC), leucine (CTG), lysine (AAG), methionine (ATG), phenylalanine ( TTC), proline (CCC), serine (TCC), threonine (ACC), tryptophan (TGG), tyrosine (TAC), and valine (GTG). Runs of Cs and Gs were avoided to simplify PCR conditions as well as oligonucleotide synthesis.

T001과 유사한 인간 세포 내 핵산 대사에 관여하는 효소는 5'-뉴클레오티다아제로, 작용 위치에 따라 3가지, 즉, NT5C(cytoplasm), NT5M(mitochondria), NT5E(extracellular membrane)가 알려져 있다. 이 3가지 효소는 작용 위치 뿐 아니라 NMP 또는 dNMP의 인산기를 가수분해시키는 활성 및 구조적 차이로 인해 선호하는 기질 핵산종이 다르며 대부분 NMP 또는 dNMP에 대한 광범위한 특이성을 가지고 있는 것으로 알려져 있다. 이 가운데 NT5M은 T001과 아미노산 서열의 낮은 유사성에 비해 단백질의 구조에 있어 대체적으로 유사한 것으로 추정되었다(도 3 참조). 특히 서열의 차이에도 불구하고 작용 부위에 대한 서열은 보존되어 있었고 HAD 수퍼패밀리(haloalkanoic acid dehalogenase superfamily)에 속하는 것으로 확인되었다.Similar to T001, enzymes involved in nucleic acid metabolism in human cells are 5'-nucleotidases, and three types of enzymes are known according to the location of action: NT5C (cytoplasm), NT5M (mitochondria), and NT5E (extracellular membrane). These three enzymes are known to have different preferred substrate nucleic acid species due to structural differences in activity and structural differences in hydrolyzing the phosphate group of NMP or dNMP as well as the site of action, and most of them have broad specificity for NMP or dNMP. Among them, NT5M was presumed to be generally similar in structure to T001 compared to T001 in terms of amino acid sequence (see FIG. 3). In particular, despite the difference in sequence, the sequence of the action site was conserved and it was identified as belonging to the HAD superfamily (haloalkanoic acid dehalogenase superfamily).

구조적 상동성에도 불구하고 NT5M과 T001의 구조적 차이는 T001은 NT5M에 존재하지 않는 특이한 바인딩 루프(binding loop) 구조를 가지고 있다는 점이고, 이 점이 가장 큰 특징적 차이이다(도 4 참조).Despite structural homology, the structural difference between NT5M and T001 is that T001 has a unique binding loop structure that does not exist in NT5M, and this is the biggest characteristic difference (see FIG. 4).

상기 바인딩 루프는 NT5M과 T001의 기질결합 부위와 밀접한 관계가 있으며, 기질의 특이성에 관련되어 있을 것으로 추정된다. 현재까지 상기 바인딩 루프의 기능에 대해서는 거의 알려져 있지 않지만 본 발명에서 상기 바인딩 루프로 인해 NT5M에 비해 dTMP에 대해 더 높은 친화도를 가지며 높은 dTMP 분해능을 가지는 것을 일부 확인할 수 있었다(표 4 참조).The binding loop is closely related to the substrate binding sites of NT5M and T001, and is presumed to be related to the specificity of the substrate. Until now, little is known about the function of the binding loop, but in the present invention, it was partially confirmed that the binding loop has higher affinity for dTMP than NT5M and high dTMP resolution (see Table 4).

5' NT5' NT AliasAlias Preferred substrate (Km)Preferred substrate (km) ReferencesReferences PBS2 TMP phosphohydrolasePBS2 TMP phosphohydrolase T001T001 dTMP (0.01 mM)dGMP (0.7 mM)
dUMP (0.8 mM)
dTMP (0.01 mM) dGMP (0.7 mM)
dUMP (0.8 mM)
Methods Enzymol. 51:285-290 (1978),
Also In house data
Methods Enzymol. 51:285-290 (1978);
Also In house data
Human mitochondrial 5'doxyribonucleotidaseHuman mitochondrial 5'doxyribonucleotidase NT5M(hdNT-2)NT5M (hdNT-2) dGMP (0.09 mM)
dUMP (0.16 mM)
dTMP (0.3 mM)
dGMP (0.09 mM)
dUMP (0.16 mM)
dTMP (0.3 mM)
Biochem. 46:13809-13818 (2007)
Biochemical Pharmacol. 66: 471-479 (2003)
Biochem. 46:13809-13818 (2007)
Biochemical Pharmacol. 66: 471-479 (2003)
Human cytosolic 5'deoxyribonucleotidaseHuman cytosolic 5'deoxyribonucleotidase hdNT-1hdNT-1 dUMP (1.5 mM)dTMP (1.5 mM)
dAMP (3.0 mM)
dGMP (3.3 mM)
dUMP (1.5 mM)dTMP (1.5 mM)
dAMP (3.0 mM)
dGMP (3.3 mM)
J. Biol. Chem. 265:6589-6595 (1990)
Biochemical Pharmacol. 66: 471-479 (2003)
J. Biol. Chem. 265:6589-6595 (1990)
Biochemical Pharmacol. 66: 471-479 (2003)
Murine cytosolic 5'deoxyribnucleotidaseMurine cytosolic 5'deoxyribnucleotidase mdNT-1mdNT-1 dUMP (0.8 mM)dAMP (1.0mM)
dGMP (1.2mM)
dTMP (1.4 mM)
dUMP (0.8 mM) dAMP (1.0 mM)
dGMP (1.2 mM)
dTMP (1.4 mM)
J. Biol. Chem. 275:5409-5415 (1990)
Biochemical Pharmacol. 66: 471-479 (2003)
J. Biol. Chem. 275:5409-5415 (1990)
Biochemical Pharmacol. 66: 471-479 (2003)

표 4에 나타낸 바와 같이, 현재까지 알려진 다른 5'-deoxyribonucleotidase 가운데 가장 dTMP에 대한 친화도가 높은 것으로 알려져 있다. 현재까지 보고된 바에 따르면, 다른 효소의 경우 본 발명에서 제시한 T001에 비해 dTMP에 대한 친화도가 낮을 뿐 아니라 상대적으로 광범위한 기질 특이성을 보이는 것을 알 수 있다. 특히, 구조적으로 유사한 human NT5M과 비교할 경우에도 기질에 대한 스펙트럼이 다르며 dTMP에 대한 친화도 역시 표 4에 따르면 30배 정도 낮은 것을 볼 수 있다. 이와 같은 차이는 NT5M에는 존재하지 않는 결합 루프가 T001에 존재하기 때문인 것으로 추정된다.As shown in Table 4, it is known to have the highest affinity for dTMP among other 5'-deoxyribonucleotidase known to date. According to what has been reported so far, it can be seen that other enzymes show a relatively broad substrate specificity as well as low affinity for dTMP compared to T001 presented in the present invention. In particular, even when compared with the structurally similar human NT5M, the spectrum for the substrate is different, and the affinity for dTMP is also about 30 times lower according to Table 4. This difference is presumed to be due to the presence of a binding loop in T001 that does not exist in NT5M.

T001 mRNA 약물 - 로칼리제이션(localization) 및 mRNA 구조T001 mRNA drug - localization and mRNA structure

T001의 최종 약물의 형태는 mRNA 형태로서 발현에 필요한 각 구성요소를 최적화하여 UTR(untranslated region) 및 Kozak 서열을 최적화하였다. 이를 위하여 리포터(reporter) 유전자로 GFP(Green Fluorescent Protein)를 사용하여 발현 효율을 도 5와 같은 후보 구조를 선정하여 결정하였다. 각 사용한 UTR의 서열 정보는 하기 표 5와 같다.The form of the final drug of T001 is in the form of mRNA, and UTR (untranslated region) and Kozak sequence were optimized by optimizing each component required for expression. To this end, expression efficiency was determined by selecting a candidate structure as shown in FIG. 5 using GFP (Green Fluorescent Protein) as a reporter gene. Sequence information of each used UTR is shown in Table 5 below.

NoNo 5'-UTR5′-UTR 5'-UTR source5'-UTR source 3'-UTR3′-UTR 3'-UTR source3'-UTR source RefRef 1One TTGGTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTATTGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAATTA
(서열번호 4)
TTGGTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTATTGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAATTA
(SEQ ID NO: 4)
Chimeric intronChimeric introns ACGCGTCGAGCATGCATCTAGGGCGGCCAATTCCGCCCCTCTCCCCCCCCCCCCTCTCCCTCCCCCCCCCCTAACGTTACTGGCCGAAGCCGCTTGGAATAAGGCCGGTGTGCGTTTGTCTATATGTTATTTTCCACCATATTGCCGTCTTTTGGCAATGTGAGGGCCCGGAAACCTGGCCCTGTCTTCTTGACGAGCATTCCTAGGGGTCTTTCCCCTCTCGCCAAAGGAATGCAAGGTCTGTTGA
(서열번호 5)
ACGCGTCGAGCATGCATCTAGGGCGGCCAATTCCGCCCCTCTCCCCCCCCCCTCTCCCTCCCCCCCCCTAACGTTACTGGCCGAAGCCGCTTGGAATAAGGCCGGTGTGCGTTTGTCTATATGTTATTTTCCACCATATTGCCGTCTTTTGGCAATGTGAGGGCCCGGAAACCTGGCCCTGTCTTCTTGACGAGCATTCCTAGGGGTCTTTCCCCTCTCGCAAAGGAATGCAA GGTCTGTTGA
(SEQ ID NO: 5)
IRESIRES --
22 TTGGTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTATTGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAATTA (서열번호 6)(SEQ ID NO: 6) Chimeric intronChimeric introns -- IRESIRES -- 33 TTGGTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTATTGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAATTA (서열번호 7)(SEQ ID NO: 7) Chimeric intronChimeric introns GCATCACATTTAAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAGAAAATGAAGATCAATAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACATAAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACCTAGATCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAATGCATCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCTCTTTTCAGAGCCACCAGAATT (서열번호 8)GCATCACATTTAAAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAAAATGAAGATCAATAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACATAAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACCTAGATCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAATGCATCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCTCTTTTCAGAGCCACCAGAATT (SEQ ID NO: 8) AlbuminAlbumin Andreas Thess(2015)Andreas Thess (2015) 44 TTGGTCGTGAGGCACTGGGCAGGTAAGTATCAAGGTTACAAGACAGGTTTAAGGAGACCAATAGAAACTGGGCTTGTCGAGACAGAGAAGACTCTTGCGTTTCTGATAGGCACCTATTGGTCTTACTGACATCCACTTTGCCTTTCTCTCCACAGGTGTCCACTCCCAGTTCAATTA (서열번호 9)(SEQ ID NO: 9) Chimeric intronChimeric introns GCATCACATTTAAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAGAAAATGAAGATCAATAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACATAAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACCTAGATCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAATGCATCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCTCTTTTCAGAGCCACCAGAATT (서열번호 10)GCATCACATTTAAAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAAAATGAAGATCAATAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACATAAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACCTAGATCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAATGCATCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCTCTTTTCAGAGCCACCAGAATT (SEQ ID NO: 10) AlbuminAlbumin Andreas Thess(2015)Andreas Thess (2015) 55 GGGTCCCGCAGTCGGCGTCCAGCGGCTCTGCTTGTTCGTGTGTGTGTCGTTGCAGGCCTTATTCAAGCGCCACC (서열번호 11)GGGTCCCGCAGTCGGCGTCCAGCGGCTCTGCTTGTTCGTGTGTGTGTCGTTGCAGGCCTTATTCAAGCGCCACC (SEQ ID NO: 11) Andreas Thess(2015)Andreas Thess (2015) GCATCACATTTAAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAGAAAATGAAGATCAATAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACATAAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACCTAGATCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAATGCATCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCTCTTTTCAGAGCCACCAGAATT(서열번호 12)GCATCACATTTAAAAAGCATCTCAGCCTACCATGAGAATAAGAGAAAAAATGAAGATCAATAGCTTATTCATCTCTTTTTCTTTTTCGTTGGTGTAAAGCCAACACCCTGTCTAAAAAACATAAATTTCTTTAATCATTTTGCCTCTTTTCTCTGTGCTTCAATTAATAAAAAATGGAAAGAACCTAGATCTAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAAA AAAAAAAAATGCATCCCCCCCCCCCCCCCCCCCCCCCCCCCCCCAAAGGCTCTTTTCAGAGCCACCAGAATT (SEQ ID NO: 12) AlbuminAlbumin Andreas Thess(2015)Andreas Thess (2015) 66 CTTCCTACTCAGGCTTTATACAAAGACCAAGAGGTACAGGTGCAAGGGAGAGAAGAAGAGTAAGAAGAAATATAAGAGCCACC (서열번호 13)CTTCCTACTCAGGCTTTATACAAAGACCAAGAGGTACAGGTGCAAGGGAGAGAAGAAGAGTAAGAAGAAATATAAGGCCACC (SEQ ID NO: 13) Mus musculus_alpha-globin (Hbat1)Mus musculus_alpha-globin (Hbat1) GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG (서열번호 14)GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG (SEQ ID NO: 14) Mus musculus hemoglobin alpha(Hba-a1)Mus musculus hemoglobin alpha (Hba-a1) UTRdbUTRdb 77 AGTAAGAAGAAATATAAGAGCCACC (서열번호 15)AGTAAGAAGAAATATAAGGCCACC (SEQ ID NO: 15) Rhinatrema bivittatum thiamin pyrophospho kinase 1Rhinatrema bivittatum thiamin pyrophosphokinase 1 GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG(서열번호 16)GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG (SEQ ID NO: 16) Mus musculus hemoglobin alpha(Hba-a1)Mus musculus hemoglobin alpha (Hba-a1) UTRdbUTRdb 88 GAGACTGCCACC (서열번호 17)GAGACTGCCACC (SEQ ID NO: 17) Zeljka Trepotec (2019)Zeljka Trepotec (2019) GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG(서열번호 18)GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG (SEQ ID NO: 18) Mus musculus hemoglobin alpha(Hba-a1)Mus musculus hemoglobin alpha (Hba-a1) UTRdbUTRdb 99 GAGACTGCCAAG (서열번호 19)GAGACTGCCAAG (SEQ ID NO: 19) Zeljka Trepotec (2019)Zeljka Trepotec (2019) GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG(서열번호 20)GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG (SEQ ID NO: 20) Mus musculus hemoglobin alpha(Hba-a1)Mus musculus hemoglobin alpha (Hba-a1) UTRdbUTRdb 1010 -- -- GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG
(서열번호 21)
GCTGCCTTCTGCGGGGCTTGCCTTCTGGCCATGCCCTTCTTCTCTCCCTTGCACCTGTACCTCTTGGTCTTTGAATAAAGCCTGAGTAGGAAG
(SEQ ID NO: 21)
Mus musculus hemoglobin alpha(Hba-a1)Mus musculus hemoglobin alpha (Hba-a1) UTRdbUTRdb

각 UTR에 대한 발현 효율을 GFP 형광 정도를 통해 FACS로 분석하였다. FACS 분석방법은 다음과 같다.Expression efficiency for each UTR was analyzed by FACS through the degree of GFP fluorescence. The FACS analysis method is as follows.

6구 세포 배양 플레이트의 각 구당 5×105 개의 세포를 넣고 37℃, 5% CO2 조건의 세포 배양기에서 24시간 배양하여 플레이트 바닥에 부착시킨 후, 각 UTR 구조를 가지는 녹색 형광을 발현하는 EGFP mRNA를 lipofectamine messengerMAX 시약을 사용하여 트랜스펙션 하였다. 먼저 미세 시험관에 125 ㎕의 OPTI-MEM 배지와 3.5 ㎕의 시약을 섞고 상온에서 10분간 배양 후, 다른 미세 시험관에서 125 ㎕ 배지와 각 UTR 구조를 가지는 mRNA 1.25 ㎍을 넣고 섞은 mRNA 희석 배지를 위 시약에 넣어 섞고 5분간 추가 배양 후, 각 구의 세포에 투여하였다. 4시간 뒤, 각 구의 세포를 인산 완충 생리식염수로 가볍게 세척하고 세포배양 배지로 교체한 뒤 20시간 추가 배양 후 형광 현미경 및 유 세포 분석기로 각 UTR 의 EGFP mRNA의 녹색 형광 단백질 발현 정도를 비교 분석하였다. 대조군으로는 트리링크 바이오 테크놀러지스에서 구매한 EGFP mRNA를 사용하였다. 먼저 배양이 끝난 세포의 녹색 형광의 강도를 형광 현미경을 통해 이미지로 비교 분석한 다음 대조군 포함 각각의 UTR을 가진 EGFP mRNA를 처리한 세포를 트립신 효소로 플레이트 바닥에서 떼어 내고 미세원침관에 모은 뒤 인산 완충 생리식염수에 희석하였다. 이렇게 회수한 세포 유세포 분석기를 통해 대조군 대비 녹색 형광을 발현하는 세포군의 분포를 강도에 따라 강, 중, 약 세 단계로 비교하였다.5 × 10 5 cells per each cell of a 6-well cell culture plate were cultured for 24 hours in a cell incubator at 37°C and 5% CO 2 , and then adhered to the bottom of the plate. EGFP expressing green fluorescence having each UTR structure mRNA was transfected using lipofectamine messengerMAX reagent. First, mix 125 μl of OPTI-MEM medium and 3.5 μl of reagent in a microtube and incubate for 10 minutes at room temperature. In another microtube, add 125 μl of medium and 1.25 μg of mRNA having each UTR structure, and mix the mRNA diluted medium with the above reagent. After mixing and further incubating for 5 minutes, it was administered to the cells of each sphere. After 4 hours, the cells in each sphere were lightly washed with phosphate buffered saline, replaced with cell culture medium, and then further cultured for 20 hours. The green fluorescent protein expression level of EGFP mRNA of each UTR was compared and analyzed by fluorescence microscopy and flow cytometry. . As a control, EGFP mRNA purchased from Trilink Biotechnology was used. First, the intensity of green fluorescence of the cultured cells was compared and analyzed as an image through a fluorescence microscope, and then the cells treated with EGFP mRNA with each UTR, including the control group, were detached from the bottom of the plate with trypsin enzyme, collected in a microcentrifuge tube, and phosphorus acid It was diluted in buffered physiological saline. Through the cell flow cytometer recovered in this way, the distribution of the cell population expressing green fluorescence compared to the control group was compared into three levels, strong, medium, and weak, according to intensity.

FACS 분석 결과, UTR의 길이를 줄인 형태로 최종 발현형태를 UTR 9번 형태로 확정하였다(도 6a 내지 6m 참조). As a result of FACS analysis, the final expression form in the form of a shortened UTR was confirmed as UTR No. 9 (see FIGS. 6a to 6m).

UTR 9번을 토대로 세포내 작용 위치를 다양화하기 위해 핵산합성경로가 존재하는 핵, 세포질 및 미토콘드리아에 위치시키기 위하여 도 7과 같은 위치 신호 서열(NLS: 서열번호 22, MLS: 서열번호 23)을 첨가하여 리포터 유전자 mStrawberry를 위치 신호 서열에 따라 발현시킨 후 형광으로 단백질의 위치를 확인하였다(도 8 참조). 위치 신호서열에 따른 형광 위치 확인 시험(mRNA transfection : lipofectamine)의 구체적인 과정은 다음과 같다.In order to diversify the intracellular action site based on UTR No. 9, the location signal sequence (NLS: SEQ ID NO: 22, MLS: SEQ ID NO: 23) as shown in FIG. After addition, the reporter gene mStrawberry was expressed according to the positioning signal sequence, and the position of the protein was confirmed by fluorescence (see FIG. 8). The specific process of the fluorescence localization test (mRNA transfection: lipofectamine) according to the localization signal sequence is as follows.

6구 세포 배양 플레이트의 각 구에 공초점 현미경 관찰용 커버글라스를 하나 넣고 5×105 개의 세포를 37℃, 5% CO2 조건의 세포 배양기에서 24시간 배양하여 커버글라스 위에 부착시킨다. 각각의 위치 신호 서열과 빨간 형광 단백질을 발현하는 mStrawberry 리포터 유전자 서열이 연결된 mRNA를 lipofectamine messengerMAX 시약을 사용하여 트랜스펙션 한다. 먼저 미세 시험관에 125 ㎕의 OPTI-MEM 배지와 3.5 ㎕의 시약을 섞고 상온에서 10분간 배양 후, 다른 미세 시험관에서 125 ㎕ 배지와 각 위치신호 서열을 가진 리포터 mRNA 1.25 ㎍을 넣고 섞은 mRNA 희석 배지를 위 시약에 넣어 섞고 5분간 추가 배양 후, 각 구의 세포에 투여하였다. 4시간 뒤, 각 구의 세포를 인산 완충 생리식염수로 가볍게 세척하고 세포배양 배지로 교체 한 뒤 20시간 추가 배양 후 공초점 현미경 관찰을 위한 시료를 제작하였다. 시료제작을 위해 4 % paraformaldehyde 시약을 첨가하고 10분간 배양하여 세를 고정한 뒤 인산 완충 생리식염수로 세척한 다음 슬라이드글라스 위에 보존 액 20 ㎕를 넣고 그 위에 고정 및 세척이 끝난 커버글라스를 올리고 마르지 않게 주변을 마감 후 공초점 현미경의 빨간 형광으로 위치 신호 서열이 잘 작동하여 mStrawberry 단백질이 세포 핵 및 미토콘드리아에 위치하였는지 확인하였다.One cover glass for observation under a confocal microscope is placed in each sphere of the 6-hole cell culture plate, and 5×10 5 cells are cultured for 24 hours in a cell incubator at 37° C. and 5% CO 2 , and adhered to the cover glass. mRNAs linked with each localization signal sequence and the mStrawberry reporter gene sequence expressing red fluorescent protein were transfected using lipofectamine messengerMAX reagent. First, mix 125 μl of OPTI-MEM medium and 3.5 μl of reagent in a microtube, incubate at room temperature for 10 minutes, and in another microtube, add 125 μl of medium and 1.25 μg of reporter mRNA having each positional signal sequence, and mix the mRNA diluted medium. It was added to the above reagent, mixed, and after additional incubation for 5 minutes, it was administered to the cells of each sphere. After 4 hours, the cells in each sphere were lightly washed with phosphate-buffered saline, replaced with cell culture medium, and then cultured for 20 hours, and samples for confocal microscopy were prepared. For sample preparation, 4% paraformaldehyde reagent was added, incubated for 10 minutes to fix the cells, washed with phosphate-buffered saline, and then put 20 μl of preservation solution on a slide glass and put a fixed and washed cover glass on top of it. After completion, it was confirmed that the localization signal sequence worked well with red fluorescence of a confocal microscope, and that the mStrawberry protein was located in the cell nucleus and mitochondria.

T001 mRNA 합성은 다음과 같이 수행되었다.T001 mRNA synthesis was performed as follows.

주형 DNA 준비 : IVT(In Vitro Transcribed) mRNA 합성을 위한 벡터는 pIRES 벡터에서 수정되었다. 간략히, 5'UTR-T001-3'UTR cassette는 pIRES 벡터의 MCS에 클로닝하였다. IVT 템플릿(template)을 생성하기 위해 플라스미드를 SacI/HpaI 효소로 처리하여 선형 가닥을 형성하고 T7 프로모터와 T001 카세트(cassette)가 포함된 1.5kb의 선형 가닥을 순수하게 컬럼 정제하여 PCR을 위한 템플릿으로 사용하였다. 정방향 프라이머(gtgcttctgacacaacagtctcgaacttaagc; 서열번호 36)와 역방향 프라이머(gaaGCGGCCGCCTTCCTACTCAGGCTTTATTC; 서열번호 37)를 PCR 반응에 사용하였으며, 모든 PCR 반응은 Pfu 중합 효소를 사용하여 1분 동안 95℃에서, 1분 동안 61℃; 72℃에서 3분, 총 30주기 동안 PCR 하였다. PCR 산물은 아가로스 겔에서 실행되었고, 추가 처리 전에 Qiagen 클린업 키트를 사용하여 추출되었다.Template DNA preparation: The vector for IVT (In Vitro Transcribed) mRNA synthesis was modified from the pIRES vector. Briefly, the 5'UTR-T001-3'UTR cassette was cloned into the MCS of pIRES vector. To generate the IVT template, the plasmid was treated with SacI/HpaI enzymes to form a linear strand, and the 1.5 kb linear strand containing the T7 promoter and the T001 cassette was column-purified as a template for PCR. used A forward primer (gtgcttctgacacaacagtctcgaacttaagc; SEQ ID NO: 36) and a reverse primer (gaaGCGGCCGCCTTCCTACTCAGGCTTTATTC; SEQ ID NO: 37) were used in the PCR reaction, and all PCR reactions were performed using Pfu polymerase at 95° C. for 1 min, 61° C. for 1 min; PCR was performed at 72° C. for 3 minutes, for a total of 30 cycles. PCR products were run on an agarose gel and extracted using the Qiagen cleanup kit before further processing.

IVT mNRA 합성 : PCR 후, 유전자 정보는 HiScribe ™ T7 ARCA mRNA 키트(New England Biolabs, Cot. #. E2065)를 사용하여 DNA에서 mRNA로 시험관 내 전사된다. 반응액은 10 ㎕ NTP / 캡 아날로그 혼합물, 1 ㎍ Template DNA, 2 ㎕ 1× T7 RNA 중합 효소 혼합물을 첨가하여 20 ㎕의 IVT 반응 혼합물을 조제하였다. IVT 반응 혼합물은 30분 동안 37℃에서 배양되었다. 주형 DNA를 제거하기 위해 1 ㎕ DNase를 IVT 반응 혼합물에 첨가하고 37℃에서 15분 동안 배양하였다. 폴리 (A) 테일링을 위해, 5 ㎕ 10× 폴리 (A) 폴리머라제 반응 완충액, 5 ㎕ 폴리 (A) 폴리머라제 및 20 ㎕ 뉴클레아제가 없는 물을 첨가하여 20 ㎕의 IVT 반응 혼합물을 제조하여 40분간 37℃에서 배양되었다. 이후, RNeasy Mini Kit(Qiagen, Hilden, Germany)를 사용하여 정제한 후 89 ㎕ 뉴클레아제가 없는 물로 스핀 컬럼 막에서 용출하여 합성된 mRNA를 정제하였다. 이어서 남극 포스파타제 1 ㎕를 30분 동안 37℃에서 처리하여 5'-phosphate를 제거한 후, 다시 한 번 RNeasy Mini Kit(Qiagen, Hilden, Germany)를 사용하여 정제한 후 50 ㎕ 뉴클레아제가 없는 물로 스핀 컬럼 막에서 용출하여 합성된 mRNA를 회수하였다. 다음 실험을 위하여 최종 농도가 500 ng/㎕가 되도록 맞춘 후 분주하여 -80℃에서 보관하였다.IVT mNRA synthesis: After PCR, genetic information is transcribed from DNA to mRNA in vitro using the HiScribe™ T7 ARCA mRNA kit (New England Biolabs, Cot. #. E2065). 20 μl of IVT reaction mixture was prepared by adding 10 μl of NTP/cap analog mixture, 1 μg of template DNA, and 2 μl of 1×T7 RNA polymerase mixture to the reaction solution. The IVT reaction mixture was incubated at 37°C for 30 minutes. To remove template DNA, 1 μl DNase was added to the IVT reaction mixture and incubated at 37° C. for 15 minutes. For poly (A) tailing, 20 μl of IVT reaction mixture was prepared by adding 5 μl 10× poly (A) polymerase reaction buffer, 5 μl poly (A) polymerase and 20 μl nuclease-free water to a 40 minutes at 37°C. Then, after purification using the RNeasy Mini Kit (Qiagen, Hilden, Germany), the synthesized mRNA was purified by elution from the spin column membrane with 89 μl of nuclease-free water. Subsequently, 1 μl of Antarctic phosphatase was treated at 37 ° C for 30 minutes to remove 5'-phosphate, and then purified again using the RNeasy Mini Kit (Qiagen, Hilden, Germany), followed by spin column with 50 μl nuclease-free water. The mRNA synthesized by elution from the membrane was recovered. For the next experiment, after adjusting the final concentration to 500 ng/μl, it was aliquoted and stored at -80°C.

암세포에서의 타겟 대사Target metabolism in cancer cells

TS(Thymidylate synthase)는 DNA 합성 및 복구를 위한 티미딜레이트(dTMP)의 유일한 드 노보(de novo) 원천(source)이다. TS 단백질을 타겟으로 하는 약물은 암 치료의 주종이지만 표적외 효과와 독성 때문에 사용이 제한된다. TK1(cytosolic thymidine kinase)과 TK2(mitochondrial thymidine kinase)는 종양 환경으로부터 티미딘을 회복시킴으로써 대체 dTMP 생성 경로에 기여하며, TS 타겟팅 약물에 대한 저항을 조절할 수 있다. siRNA를 가진 TKs의 하향조절(downregulation)로 종양세포를 감지하는 TS siRNA의 용량을 기존 TS 단백질 타겟팅 약물(5FUdR 및 pemetrexed)에 비해 증가시켰다는 보고가 있었기 때문에 본 발명에서는 dTTP 생합성 대사에 초점을 맞췄다.Thymidylate synthase (TS) is the only de novo source of thymidylate (dTMP) for DNA synthesis and repair. Drugs targeting TS protein are the main types of cancer treatment, but their use is limited due to off-target effects and toxicity. Cytosolic thymidine kinase (TK1) and mitochondrial thymidine kinase (TK2) contribute to alternative dTMP production pathways by restoring thymidine from the tumor milieu and may modulate resistance to TS-targeting drugs. Since there was a report that downregulation of TKs with siRNA increased the capacity of TS siRNA to detect tumor cells compared to existing TS protein-targeting drugs (5FUdR and pemetrexed), the present invention focused on dTTP biosynthetic metabolism.

이러한 효소의 복합 하향조절은 TS 타겟 항암 치료를 강화하기 위한 매력적인 전략이지만, 정상세포 독성과 암 약물에 대한 내성이 문제될 수 있다. 이러한 결함을 피할 수 있는 대안은 높은 dTMP 특이적 가수분해효소를 얻는 것으로, 바로 T001이다. T001은 다른 dNMP(deoxynucleotide mono phosphate)를 가수분해하지 않고 dTMP를 티미딘으로 가수분해할 수 있다. 세포의 불균형한 뉴클레오티드 풀은 dTTP 결핍에 의해 야기될 수 있으며, 세포의 성장과 증식에 심각한 손상을 초래할 수 있다. 기질 선택성과 T001의 활성을 고려하여, 다음과 같은 가설을 세웠다: (1) 불균형 뉴클레오티드 풀은 인간 종양 세포를 유도하여 손상을 누적시키고, (2) T001의 과발현은 불균형 뉴클레오티드 풀을 야기할 수 있으며, (3) 불균형 뉴클레오티드 풀은 과도한 복구 주기(repair frequency)에 의한 세포사멸을 초래할 수 있다.Complex downregulation of these enzymes is an attractive strategy to enhance TS-targeted anti-cancer therapy, but normal cytotoxicity and resistance to cancer drugs can be problematic. An alternative to avoiding these deficiencies is to obtain a high dTMP specific hydrolase, T001. T001 can hydrolyze dTMP to thymidine without hydrolyzing other dNMP (deoxynucleotide monophosphate). An imbalanced nucleotide pool in cells can be caused by dTTP deficiency, resulting in severe damage to cell growth and proliferation. Considering the substrate selectivity and activity of T001, we hypothesized that (1) an imbalanced nucleotide pool induces human tumor cells to accumulate damage, (2) overexpression of T001 may cause an imbalanced nucleotide pool, and , (3) imbalanced nucleotide pools can lead to cell death by excessive repair frequencies.

암세포에 T001 mRNAs 트랜스펙션(transfection)을 통한 생존 세포수 감소Reducing the number of viable cells through transfection of T001 mRNAs into cancer cells

먼저, 각 T001 mRNA를 트랜스펙션한 후 종양 세포 성장을 평가하기 위해 각 그룹의 세포 수를 추정하고, 생존검사(live and dead assay)로 배양 세포 성장을 분석하였다(도 10 참조). 생존검사(live and dead assay)는 다음과 같이 수행되었다.First, in order to evaluate tumor cell growth after transfection with each T001 mRNA, the number of cells in each group was estimated, and the growth of cultured cells was analyzed by a live and dead assay (see FIG. 10). A live and dead assay was performed as follows.

6구 세포 배양 플레이트의 한 구 마다 5×105 개의 세포를 넣고 37℃, 5% CO2 조건의 세포 배양기에서 24 시간 배양하여 바닥에 부착시킨다. 24시간 뒤 각각의 mRNA 버전별로 리포펙타민(lipofectamine)을 사용하여 트랜스펙션하고 추가로 24시간 배양하였다. 실험군 각각의 세포를 회수하고 생존검사분석 시약인 2 μM calcein AM, 4 μM EthD-1 을 회수한 세포에 처리하여 30분간 반응한 후 세포를 형광현미경을 통해 확인하였다. Calcein AM은 세포내로 들어가서 살아있는 세포의 효소에 의해 분해된 후 녹색 형광을 나타내고, EthD-1는 죽은 세포의 세포내로 들어간 뒤 핵을 염색하여 빨간 형광을 나타낸다.5×10 5 cells are put into each cell of a 6-hole cell culture plate and cultured for 24 hours in a cell incubator at 37° C. and 5% CO 2 , and adhered to the bottom. After 24 hours, each mRNA version was transfected using lipofectamine and cultured for an additional 24 hours. The cells of each experimental group were collected, and 2 μM calcein AM and 4 μM EthD-1, which are viability assay reagents, were treated on the recovered cells, reacted for 30 minutes, and then the cells were examined under a fluorescence microscope. Calcein AM enters cells and shows green fluorescence after being degraded by enzymes of living cells, and EthD-1 enters cells of dead cells and stains the nucleus to show red fluorescence.

도 10에 나타낸 바와 같이, 트랜스펙션 후 24시간에서의 생존 세포는 대조군과 비교하여 현저하게 감소하였고, mRNA 버전(NT, MT 및 CT) 사이에는 차이가 없었다.As shown in Figure 10, viable cells at 24 hours after transfection were significantly reduced compared to the control group, and there was no difference between the mRNA versions (NT, MT and CT).

암세포에 T001 mRNAs 트랜스펙션(transfection)을 통한 생존 세포 증식 억제Inhibition of viable cell proliferation through transfection of T001 mRNAs into cancer cells

세포 생존능을 정량화하기 위해 MTT 분석을 수행하였다. MTT 분석 방법은 다음과 같다.MTT assay was performed to quantify cell viability. MTT analysis method is as follows.

96구 세포 배양 플레이트에 한 구당 10,000개의 세포를 넣고 37℃, 5% CO2 조건의 세포 배양기에서 24시간 배양하여 바닥에 부착시킨다. 24시간 뒤 각 mRNA 버전 별, 농도 별로 lipofectamine reagent를 만들어서 트랜스펙션하고 추가로 24시간, 48시간, 72시간 배양 후 각각 시간별로 세포 활성 분석 실험을 수행하였다. 각 시간 별 샘플에 세포 활성 분석 시약인 EZ-Cytox를 세포 배양한 각 구 당 10 ㎕를 투여하고 2시간 반응 후, 플레이트 리더기를 사용하여 450 nm 파장의 흡광값을 측정한다. 대조군 대비 각 버전 별 mRNA 처리군의 값을 비교하여 처리 시간과 농도별 세포 생존성을 분석하였다.Put 10,000 cells per cell in a 96-hole cell culture plate and incubate for 24 hours in a cell incubator at 37° C., 5% CO 2 conditions to attach to the bottom. After 24 hours, lipofectamine reagents were prepared for each mRNA version and concentration for transfection, and cell activity assays were performed for each time after further culturing for 24 hours, 48 hours, and 72 hours. 10 μl of EZ-Cytox, a cell activity assay reagent, is administered to each sample for each time period, and after 2 hours of reaction, the absorbance value at a wavelength of 450 nm is measured using a plate reader. Cell viability was analyzed by treatment time and concentration by comparing the values of the mRNA treatment group for each version compared to the control group.

MTT 분석 결과, 대조군과 비교하여 T001 mRNA에 트랜스펙션한 세포의 증식은 트랜스펙션 후 24시간에서 현저히 억제되었으며, 억제 효과는 최대 72시간까지 유지되어, 각 세포의 생존성이 지속적으로 감소되는 것으로 나타났다. 세포 생존성은 용량에 따라 다르게 나타났다(도 11 참조).As a result of the MTT assay, compared to the control group, the proliferation of cells transfected with T001 mRNA was significantly inhibited at 24 hours after transfection, and the inhibitory effect was maintained up to 72 hours, resulting in a continuous decrease in the viability of each cell. appeared to be Cell viability was dose-dependent (see FIG. 11).

암세포에 T001 mRNAs 트랜스펙션(transfection)을 통한 세포사멸(apoptosis) 유도Induction of apoptosis through transfection of T001 mRNAs into cancer cells

상기 실험결과에 따라 Annexin V 염색 후 유세포 분석(Flow cytometric analysis)을 수행하여 T001이 세포사멸을 유도하여 세포수 감소 또는 세포 증식 억제를 유발하는지를 조사하였다. 구체적인 분석 방법은 다음과 같다.According to the above experimental results, flow cytometric analysis was performed after Annexin V staining to investigate whether T001 induces apoptosis to reduce cell number or inhibit cell proliferation. The specific analysis method is as follows.

6구 세포 배양 플레이트의 각 구당 5×105 개의 세포를 넣고 37℃, 5% CO2 조건의 세포 배양기에서 24시간 배양하여 플레이트 바닥에 부착시킨 후, 각 버전별 mRNA를 lipofectamine messengerMAX 시약을 사용하여 트랜스펙션 하였다. 먼저 미세 시험관에 125 ㎕의 OPTI-MEM 배지와 3.5 ㎕의 시약을 섞고 상온에서 10분간 배양 후, 다른 미세 시험관에서 125 ㎕ 배지와 각 UTR 구조를 가지는 mRNA 1.25 ㎍을 넣고 섞은 mRNA 희석 배지를 위 시약에 넣어 섞고 5분간 추가 배양 후, 각 구의 세포에 투여하였다. 4시간 뒤, 각 구의 세포를 인산 완충 생리식염수로 가볍게 세척하고 세포배양 배지로 교체 한 뒤 20시간 추가 배양 후, 트립신 효소로 플레이트 바닥에서 떼어 내고 미세원침관에 모은 뒤 인산 완충 생리식염수에 희석하였다. 각 미세원침관 마다 3 ㎕의 Annexin V 염색 시약과 Propidium Iodide 염색 시약을 넣고 15분간 상온에서 반응 후 유세포 분석기를 통해 T001의 버전별 세포사멸이 유도된 세포군의 정도를 비교하였다. 대조군으로는 mRNA가 없는 lipofectamine messengerMAX 시약을 사용하였다. Annexin V 시약은 세포사멸이 진행되는 세포의 세포막을 염색하고 Propidium Iodide는 죽은 세포의 세포내 핵을 염색한다. 따라서 유세포 분석 그래프 상에서 세포사멸이 진행되지 않는 세포는 3사분면에 분포하며, 세포 사멸 진행 정도에 따라 세포군의 위치는 4사분면에서 1사분면으로 이동한다.5 × 10 5 cells per each cell of a 6-well cell culture plate were cultured for 24 hours in a cell incubator at 37 ° C and 5% CO 2 to attach to the bottom of the plate, and mRNA for each version was prepared using lipofectamine messengerMAX reagent. transfected. First, mix 125 μl of OPTI-MEM medium and 3.5 μl of reagent in a microtube and incubate for 10 minutes at room temperature. In another microtube, add 125 μl of medium and 1.25 μg of mRNA having each UTR structure, and mix the mRNA diluted medium with the above reagent. After mixing and further incubating for 5 minutes, it was administered to the cells of each sphere. After 4 hours, the cells in each sphere were lightly washed with phosphate-buffered saline, replaced with cell culture medium, and then further cultured for 20 hours, then detached from the bottom of the plate with trypsin enzyme, collected in a microcentrifuge tube, and diluted in phosphate-buffered saline. . 3 μl of Annexin V staining reagent and Propidium Iodide staining reagent were added to each microcentrifuge tube, reacted at room temperature for 15 minutes, and the degree of apoptosis induced cell group by version of T001 was compared through flow cytometry. As a control, lipofectamine messengerMAX reagent without mRNA was used. Annexin V reagent stains the cell membrane of cells undergoing apoptosis, and Propidium Iodide stains the intracellular nucleus of dead cells. Therefore, on the flow cytometry graph, cells that do not undergo apoptosis are distributed in the third quadrant, and the position of the cell population moves from the fourth quadrant to the first quadrant according to the degree of apoptosis.

분석 결과, 모든 버전에서 세포사멸률은 서로 비슷하였다. 대조군의 세포사멸률은 3.75%이고, NT, MT 및 CT로 트랜스펙션한 세포의 조기사멸률은 각각 21.59%, 25.11% 및 24.65%로 나타났다. NT, MT 및 CT와 함께 전이된 세포의 사멸률은 각각 9.85%, 8.42%, 11.47%로 나타났다(도 12a 내지 12d 참조). 세포사멸률은 용량 의존적으로 증가하였고, 각각의 T001 버전 사이에 약간의 차이를 보였다.As a result of the analysis, the apoptosis rates in all versions were similar to each other. The apoptosis rate of the control group was 3.75%, and the premature apoptosis rates of cells transfected with NT, MT, and CT were 21.59%, 25.11%, and 24.65%, respectively. The death rates of cells transferred with NT, MT, and CT were 9.85%, 8.42%, and 11.47%, respectively (see FIGS. 12A to 12D). Apoptosis rates increased in a dose-dependent manner, with slight differences between each T001 version.

대장암 세포(HCT-116)에 대한 T001과 NT5M의 세포독성 비교Comparison of cytotoxicity of T001 and NT5M against colorectal cancer cells (HCT-116)

T001은 NT5M과는 다른 구조적 차이로 인해 핵산 T에 더욱 특이적이다. 따라서 전반적인 핵산의손실보다 dTTP의 손실로 인한 핵산의 불균형은 암세포 대사에 큰 영향을 미칠 수 있고 그로 인해 암세포의 성장을 저해할 수 있다. 이를 증명하기 위하여 인간 NT5M의 성숙형(mature form)과 인트론(intron)이 포함된 비성숙형(non-mature form)을 세포질 T001(CT)와 세포 생존능 및 세포 성장 억제 효과를 비교하여 각 효소간 기질특이성에 따라 세포에 어떤 영향을 미치는지 조사하였다. 각 mRNA를 대장암 세포주 HCT-116에 트랜스펙션하고 24시간 후에 트라이판 블루 분석(Trypan blue assay)을 진행하여 HCT-116 세포 생존능과 세포 성장 억제 결과 도 13에 나타내었다. 구체적인 실험방법은 다음과 같다.T001 is more specific for nucleic acid T due to other structural differences from NT5M. Therefore, the imbalance of nucleic acid due to the loss of dTTP rather than the overall loss of nucleic acid can have a greater effect on the metabolism of cancer cells, thereby inhibiting the growth of cancer cells. To prove this, the mature form of human NT5M and the non-mature form containing introns were compared with cytoplasmic T001 (CT) in terms of cell viability and cell growth inhibitory effect. Depending on the substrate specificity, the effect on cells was investigated. Each mRNA was transfected into the colorectal cancer cell line HCT-116, and 24 hours later, Trypan blue assay was performed, and the results of HCT-116 cell viability and cell growth inhibition are shown in FIG. 13 . The specific test method is as follows.

HCT-116 세포에 비성숙형(non-mature form) NT5M과 성숙형(mature form) NT5M, CT mRNA를 각각 1.25 ㎍씩 트랜스펙션한다. 24시간 후 트립신(trypsin)을 처리하고 원심분리기를 이용해 세포를 수집(harvest)하였다. 수집한 세포를 1× DPBS로 재부유(resuspension)한 뒤, 일정량의 세포를 트리판 블루(trypan blue) 시약과 1:1로 섞었다. 실온(RT)에서 2분 인큐베이션(incubation)한 뒤, 혈구계(hemocytometer)를 이용하여 세포의 수를 계수하였다. 트리판 블루 시약에 염색된 세포와 염색되지 않은 세포의 수를 구하여 생존 세포(viable cell)와 비생존 세포(non-viable cell)의 수를 구한 후, 총 세포 수로 나누어 생존도(viability)를 구하였다. 각 실험은 세 번 이상 반복하여 유의성을 평가하였다.HCT-116 cells were transfected with each of 1.25 μg of non-mature form NT5M, mature form NT5M, and CT mRNA. After 24 hours, trypsin was treated and the cells were harvested using a centrifuge. After resuspension of the collected cells with 1× DPBS, a certain amount of cells was mixed with trypan blue reagent at a ratio of 1:1. After 2 minutes of incubation at room temperature (RT), the number of cells was counted using a hemocytometer. After obtaining the number of cells stained and unstained with trypan blue reagent to obtain the number of viable cells and non-viable cells, divide by the total number of cells to obtain viability. did Each experiment was repeated three or more times to evaluate significance.

도 13에서와 같이, dTMP에 대한 친화도와 활성이 높은 T001의 경우 비슷한 활성을 가진 인간 NT5M에 비해 세포에 미치는 영향이 크게 나타났다.As shown in FIG. 13, in the case of T001 having high affinity and activity for dTMP, the effect on cells was greater than that of human NT5M having similar activity.

특히 세포에 나타난 독성은 주로 세포사멸(apoptosis) 유도에 의해 나타나는 것을 확인할 수 있었다(도 14a 내지 14c 참조). HCT-116 세포에 비성숙형 NT5M, 성숙형 NT5M 및 CT mRNA를 각각 1.25 ㎍씩 트랜스펙션한 세포를 24시간 배양 후 세포사멸 정도를 측정하여 분석한 결과, 두 종류의 NT5M mRNA는 대조군(control)에 비하여 Sub G1기가 약 12% 증가한 것에 비하여, CT mRNA는 대조군에 비하여 약 25%가 증가한 것을 확인할 수 있었다. T001 발현을 통해 야기되는 세포사멸은 세포내 핵산 결핍에 의한 세포주기의 변화에서 유래한 것으로 판단되며, 기질 가운데 특정 핵산 및 dTTP의 선택적 결핍이 세포주기의 정지(arrest)를 초래하여 이 과정이 심화될 경우 결국 세포사멸이 유발될 것으로 추정된다(도 15a 내지 15c 참조).In particular, it was confirmed that the toxicity shown in the cells was mainly caused by the induction of apoptosis (see FIGS. 14a to 14c). HCT-116 cells were transfected with 1.25 μg each of immature NT5M, mature NT5M, and CT mRNA, and the degree of apoptosis was measured and analyzed after culturing for 24 hours. ), it was confirmed that the CT mRNA increased by about 25% compared to the control group, whereas the Sub G1 phase increased by about 12% compared to the control group. Apoptosis caused by expression of T001 is believed to originate from changes in the cell cycle caused by depletion of intracellular nucleic acids, and selective deficiency of specific nucleic acids and dTTP among substrates leads to arrest of the cell cycle, intensifying this process. It is presumed that apoptosis will eventually be induced if this occurs (see FIGS. 15a to 15c).

상기 결과에서 나타난 세포사멸 유발 효과는 대장암 세포주에서 CT의 처리 농도에 따라 농도의존적인 경향이 나타난 것으로 미루어 CT의 활성이 직접적으로 세포사멸에 관여했다는 사실을 보여준다(도 16 참조).The apoptosis-inducing effect shown in the above results showed a concentration-dependent tendency according to the treatment concentration of CT in colon cancer cell lines, indicating that the activity of CT was directly involved in apoptosis (see FIG. 16).

이러한 암세포의 사멸이 T001의 효과인지 확인하기 위해 CT에 대한 siRNA를 사용하여 T001을 녹다운(knock down)시킨 후 세포사멸의 정도의 변화를 확인하였다. T001을 타겟으로 하는 두 가지 종류의 siRNA를 제작하여 HCT-116 세포에 트랜스펙션한 후 이어서 CT mRNA를 트랜스펙션하고 24시간 후에 Annexin V/PI 염색을 통해 세포사멸 정도를 확인하였다. 구체적인 실험방법은 다음과 같다.In order to confirm that the apoptosis of these cancer cells is the effect of T001, T001 was knocked down using siRNA against CT, and then the change in the degree of apoptosis was confirmed. Two types of siRNA targeting T001 were prepared and transfected into HCT-116 cells, and then CT mRNA was transfected. After 24 hours, the degree of apoptosis was confirmed by Annexin V/PI staining. The specific test method is as follows.

T001을 타겟으로 하는 siRNA를 2가지 종류로 제작하여 RNAiMAX reagent를 사용하여 트랜스펙션하였고, 1시간 뒤 CT mRNA를 트랜스펙션하였다. 24시간 뒤, 세포를 수집하여, FITC Annexin V Apoptosis Detection Kit I를 사용하여 염색하였다. 20분간 실온에서 인큐베이션한 후, 유세포분석기(flow cytometry)를 사용하여 형광의 정도를 분석하였다. 각 샘플당 10,000개의 세포를 분석하였다. 사용된 siRNA의 서열은 다음과 같다.Two types of siRNA targeting T001 were prepared and transfected using RNAiMAX reagent, and CT mRNA was transfected 1 hour later. After 24 hours, cells were collected and stained using FITC Annexin V Apoptosis Detection Kit I. After incubation at room temperature for 20 minutes, the degree of fluorescence was analyzed using flow cytometry. 10,000 cells were analyzed for each sample. The sequence of the siRNA used is as follows.

[siT001-1][siT001-1]

sense(서열번호 32) : CGAGAAGAAGUCAGAUUACAUCAAGsense (SEQ ID NO: 32): CGAGAAGAAGUCAGAUUACAUCAAG

antisense(서열번호 33) : CUUGAUGUAAUCUGACUUCUUCUCGantisense (SEQ ID NO: 33): CUUGAUGUAAUCUGACUUCUUCUCG

[siT001-2][siT001-2]

sense(서열번호 34) : CGCAAAUUCAUUGAAACCUUCCUGAsense (SEQ ID NO: 34): CGCAAAUUCAUUGAAACCUUCCUGA

antisense(서열번호 35) : UCAGGAAGGUUUCAAUGAAUUUGCGantisense (SEQ ID NO: 35): UCAGGAAGGUUUCAAUGAAUUUGCG

분석 결과, CT mRNA 단독으로 트랜스펙션한 그룹에 비하여 siRNA를 트랜스펙션한 후 CT mRNA를 트랜스펙션한 그룹에서 세포사멸이 감소하는 것을 확인할 수 있었다(도 17a 내지 17c 참조).As a result of the analysis, it was confirmed that apoptosis was reduced in the group transfected with CT mRNA after transfection with siRNA compared to the group transfected with CT mRNA alone (see FIGS. 17A to 17C ).

삼중음성 유발 암종에서 T001의 농도의존적 저해 효과Concentration-dependent inhibitory effect of T001 in triple-negative carcinoma

T001 작용기전상 대장암 이외의 암종에서도 암세포 사멸을 유도할 수 있을 것으로 사료되어 삼중음성 유방암(TNBC)에 대하여 동일한 시험을 수행하여 세포사멸 유발능을 확인하였다. 구체적인 시험방법은 다음과 같다.It is thought that T001 can induce cancer cell death in carcinomas other than colorectal cancer based on its mechanism of action, and the same test was performed on triple-negative breast cancer (TNBC) to confirm its ability to induce apoptosis. The specific test method is as follows.

MDA-MB-231, MDA-MB-468 cell을 각각 5×105 cells/well로 분주(seeding)한 뒤, CT mRNA를 각각 0, 0.625, 1.25, 2.5 ㎍씩 트랜스펙션하였다. 24시간 뒤, 세포를 수집하여, FITC Annexin V Apoptosis Detection Kit I를 사용하여 염색하였다. 20분간 실온에서 인큐베이션한 후, 유세포분석기를 사용하여 형광의 정도를 분석하였다. 각 샘플당 10,000개의 세포를 분석하였다. 세 번의 반복 실험을 통하여 유의성을 평가하였다.MDA-MB-231 and MDA-MB-468 cells were each seeded at 5×10 5 cells/well, and 0, 0.625, 1.25, and 2.5 μg of CT mRNA were respectively transfected. After 24 hours, cells were collected and stained using FITC Annexin V Apoptosis Detection Kit I. After incubation at room temperature for 20 minutes, the degree of fluorescence was analyzed using a flow cytometer. 10,000 cells were analyzed for each sample. Significance was evaluated through three repeated experiments.

세포질 T001(CT) mRNA를 농도별로 삼중음성 유방암종 2종에 대하여 처리한 후에 24시간 배양 세포를 Annexin V/PI 염색을 통해 FACS 분석을 수행한 결과, CT mRNA의 농도에 비례하여 시험한 암종에서도 역시 세포사멸 세포의 비율이 증가함을 확인하였다(도 18 참조).After treatment with cytoplasmic T001 (CT) mRNA for each concentration of two types of triple-negative breast cancer, FACS analysis was performed on cells cultured for 24 hours through Annexin V/PI staining. It was also confirmed that the proportion of apoptotic cells increased (see FIG. 18).

세포사멸의 원인에 대한 단백질 수준의 분석을 위하여 CT mRNA를 TNBC 세포주인 MDA-MB-231 및 MDA-MB-468에 1.25 ㎍을 트랜스펙션한 후 18시간 후 웨스턴 블롯(Western blot)을 수행하였다. 구체적인 분석방법은 다음과 같다.To analyze the protein level of the cause of apoptosis, 1.25 μg of CT mRNA was transfected into TNBC cell lines MDA-MB-231 and MDA-MB-468, and Western blotting was performed 18 hours later. . The specific analysis method is as follows.

CT mRNA를 트랜스펙션한 후, 18시간 뒤 세포를 수집하였다. RIPA buffer(+protease inhibitor, phosphatase inhibitor)를 사용하여 아이스에서 세포를 30분간 용균(lysis)한 후 4℃ 원심분리기를 15분간 이용하여 단백질을 분리하였다. 분리된 단백질을 정량하여 아크릴아마이드 겔(acrylamide gel)에 10 내지 20 ㎍의 단백질을 로딩하였다. 겔을 PVDF 막에 전이(transfer)한 후 5% 탈지분유(skim milk)로 RT에서 1시간 동안 블로킹(blocking)하였다. 각 1' 항체(antibody)를 넣고 4℃에서 하룻밤 보관하였다. 0.1% TBST로 세척(wash) 후, 2' 항체를 넣어 RT에서 1시간 동안 인큐베이션하였다. 0.1% TBST로 세척 후, ECL 용액을 이용하여 단백질을 검출하였다. ChemiDoc XRS+를 이용하여 단백질 발현을 분석하였다.After transfection with CT mRNA, cells were collected 18 hours later. Cells were lysed on ice for 30 minutes using RIPA buffer (+ protease inhibitor, phosphatase inhibitor), and proteins were separated using a 4° C. centrifuge for 15 minutes. Separated proteins were quantified and 10 to 20 μg of protein was loaded on an acrylamide gel. After transferring the gel to a PVDF membrane, it was blocked for 1 hour at RT with 5% skim milk. Each 1' antibody was added and stored overnight at 4°C. After washing with 0.1% TBST, the 2' antibody was added and incubated for 1 hour at RT. After washing with 0.1% TBST, proteins were detected using ECL solution. Protein expression was analyzed using ChemiDoc XRS+.

분석 결과, 도 19에 나타낸 바와 같이, 세포사멸 마커인 PARP의 절단(cleavage)과 DNA 손상(damage) 마커인 gamma-H2AX의 발현 증가를 확인할 수 있었다. CT mRNA 처리 시 dTTP의 결핍으로 인해 DNA 손상이 발생하는 것을 확인할 수 있었으며 이를 통해 세포사멸이 유발된다는 사실을 확인할 수 있었다.As a result of the analysis, as shown in FIG. 19 , it was confirmed that cleavage of PARP, which is an apoptosis marker, and increased expression of gamma-H2AX, which is a DNA damage marker, were confirmed. It was confirmed that DNA damage occurred due to dTTP deficiency during CT mRNA treatment, and it was confirmed that apoptosis was induced through this.

SV4 바인더 및 T001 약물을 이용한 항암제Anticancer drug using SV4 binder and T001 drug

이하, SV4 바인더 및 T001 약물을 이용한 항암제의 구현예에 관하여 설명한다.Hereinafter, an embodiment of an anticancer agent using the SV4 binder and the drug T001 will be described.

SV4 바인더 및 T001 약물을 이용한 항암제는 암세포 표면에서 과발현되는 CD47을 1차적으로 탐지하여 결합하고, 2차적으로 암세포에 진입한 mRNA형 핵산대사저해 약물이 암세포의 과도한 핵산합성대사를 감지하여 저해함으로써 암세포의 면역회피 및 대사취약성을 표적하여 정상세포의 부작용을 혁신적으로 줄인 4세대 타겟형 대사항암제이다. 일반적으로 암세포 특이적 표면 단백질의 경우 정상세포에도 분포하는 경우가 많아 특이적 표면 단백질을 인식하는 표적 단백질과 독성물질을 결합할 경우 정상세포의 피해는 불가피하다. 그러나 인식만으로는 독성을 주지 않고 과도하게 핵산합성을 시도하는 암세포의 대사를 표적할 경우 암세포에 대한 인식률이 높아 정상세포의 피해를 줄일 수 있다. 즉, CD47을 인식하여 T001 mRNA가 세포내로 유입되더라도 성장을 위해 핵산을 증폭하지 않는 대부분의 정상세포는 핵산에 대한 요구가 적기 때문에 피해가 거의 없으나 과도한 핵산을 합성해야만 성장할 수 있는 암세포의 경우 dTMP의 결핍으로 인한 핵산의 불균형으로 인해 큰 피해를 입는다. 따라서 암세포의 고발현 CD47을 인식하는 과정에서 일부 정상세포중 비교적 높은 CD47 수준을 갖는 세포를 표적하더라도 내부로 유입된 핵산 대사 표적을 통해 암세포에 더욱 큰 피해를 줌으로써 암세포의 인식률을 높여 정상세포의 피해를 줄일 수 있다.Anticancer drugs using SV4 binder and T001 drug primarily detect and bind to CD47, which is overexpressed on the surface of cancer cells, and secondarily, mRNA-type nucleic acid metabolism inhibitors that enter cancer cells detect and inhibit excessive nucleic acid synthesis metabolism in cancer cells, thereby inhibiting cancer cells It is a 4th-generation targeted metabolic cancer drug that innovatively reduces side effects of normal cells by targeting immune evasion and metabolic vulnerability. In general, cancer cell-specific surface proteins are often distributed even in normal cells, so when a target protein that recognizes a specific surface protein is combined with a toxic substance, damage to normal cells is inevitable. However, recognition alone does not cause toxicity, and when targeting the metabolism of cancer cells that attempt to synthesize nucleic acids excessively, the recognition rate for cancer cells is high, and damage to normal cells can be reduced. In other words, most normal cells that do not amplify nucleic acid for growth, even if CD47 is recognized and T001 mRNA is introduced into the cell, suffer little damage because they require little nucleic acid. Nucleic acid imbalance caused by deficiency causes great damage. Therefore, in the process of recognizing high-expressed CD47 in cancer cells, even if cells with relatively high CD47 levels are targeted among some normal cells, they cause more damage to cancer cells through the target of nucleic acid metabolism introduced into the inside, thereby increasing the recognition rate of cancer cells and damaging normal cells. can reduce

도 20에 나타낸 바와 같이, SV4 바인더를 이용한 항암제의 구성은 외부에 CD47 인식 단백질을 갖고 내부에 mRNA형 핵산대사저해 약물에 암종에 따라 위치 신호(핵내, 세포질내 또는 미토콘드리아내)를 다르게 조합한 mRNA를 포집한 리포좀형 나노입자 형태로 되어 있다. 도 27에는 본 발명에 따른 SV4 바인더를 이용한 항암제의 기전을 모식적으로 나타내었다.As shown in Figure 20, the composition of the anticancer agent using the SV4 binder is an mRNA that has a CD47 recognition protein on the outside and an mRNA-type nucleic acid metabolism inhibitor on the inside and a location signal (nuclear, cytoplasmic, or mitochondrial) in different combinations depending on the type of carcinoma. It is in the form of liposome-type nanoparticles encapsulated. 27 schematically shows the mechanism of the anticancer agent using the SV4 binder according to the present invention.

항암제 구성요소anticancer component

(1) CD47 바인더(SV4)(1) CD47 binder (SV4)

Sirpα 유래의 단백질을 변형한 고친화성 CD47 결합체와 이에 DSPE와의 컨쥬게이션 시 올바른 방향으로 리포좀 외부에 위치할 수 있도록 변형한 SV4를 CD47 바인더(binder)로 사용하였고, SV4의 추정 구조를 도 1A에 나타내었다. DSPE와 결합한 시료, 이를 리포좀에 삽입한 시료 각각의 KD값을 비교한 결과 서열변이에 의해 리포좀 형태에서 개선되었음을 알 수 있다(상기 표 1 참조).A high-affinity CD47 complex modified from a protein derived from Sirpα and SV4 modified to be positioned outside the liposome in the correct direction upon conjugation with DSPE were used as CD47 binders, and the estimated structure of SV4 is shown in FIG. 1A. was As a result of comparing the K D values of each of the samples combined with DSPE and the samples inserted into liposomes, it can be seen that the liposome form was improved by sequence mutation (see Table 1 above).

(2) 리포좀 기반 약물전달체 구성물질(2) Components of liposome-based drug delivery system

리포좀을 양전하로 하전시켜 표적까지 원활한 약물전달을 실행하기 위해 사용된 물질로서, 양이온성 인지질(DSPE-PEG1000), DOTAP 및 콜레스테롤 구조를 각각 하기 화학식 1 내지 3에 나타내었다.As a material used for smooth drug delivery to the target by positively charging the liposome, the structures of cationic phospholipid (DSPE-PEG 1000 ), DOTAP and cholesterol are shown in Chemical Formulas 1 to 3, respectively.

[화학식 1][Formula 1]

Figure pat00001
Figure pat00001

[화학식 2][Formula 2]

Figure pat00002
Figure pat00002

[화학식 3][Formula 3]

Figure pat00003
Figure pat00003

(3) 약물(3) Medication

전술한 바와 같이, 핵내, 세포질내 및 미토콘드리아내 발현이 가능한 mRNA로서 도 21에 T001 mRNA의 구성을 개략적으로 나타내었다.As described above, the structure of T001 mRNA is schematically shown in FIG. 21 as an mRNA capable of expression in the nucleus, cytoplasm, and mitochondria.

위치 신호로 융합된 mStrrawberry mRNA에 의해 번역된 단백질의 포지셔닝Positioning of translated proteins by mStrrawberry mRNA fused with positional signal

위치 신호가 잘 작동했는지 확인하기 위해 mRNA를 핵 및 미토콘드리아 위치 신호로 MCF7에 트랜스펙션하였다. mStrawberry 형광을 검출하여 각 mRNA의 성공적인 트랜스펙션과 위치를 확인하였다(도 22 참조). 구체적인 실험방법은 다음과 같다.To confirm that localization signals worked well, mRNA was transfected into MCF7 with nuclear and mitochondrial localization signals. mStrawberry fluorescence was detected to confirm successful transfection and localization of each mRNA (see FIG. 22). The specific test method is as follows.

6구 세포 배양 플레이트의 각 구당 5×105 개의 세포를 넣고 37℃, 5% CO2 조건의 세포 배양기에서 24시간 배양하여 플레이트 바닥에 부착시킨다. 카복시플루오레신 컨쥬게이트(carboxyfluorescein conjugated) DSPE를 사용하여 구축한 리포좀에 실험실에서 합성한 세포내 세포질 위치 신호가 연결된 빨간색 형광 단백질을 생성하는 mStrawberry mRNA를 포집하고 24시간 배양이 끝난 MCF-7 세포에 처리하여 트랜스펙션하였다. 24시간 추가 배양 뒤 공초점 레이저 형광 현미경 녹색 파장으로 리포좀의 카복시플루오레신이 발생하는 녹색 형광의 세포 상에서 위치를 확인하였고, 빨간색 파장으로 트랜스펙션된 mRNA가 mStawberry 단백질을 생성하고 세포질에 위치함을 확인하였다.5 × 10 5 cells per each cell of a 6-hole cell culture plate are cultured for 24 hours in a cell incubator at 37° C. and 5% CO 2 , and adhered to the bottom of the plate. In liposomes constructed using carboxyfluorescein conjugated DSPE, mStrawberry mRNA producing red fluorescent protein linked to intracellular cytoplasmic localization signals synthesized in the laboratory was captured and cultured for 24 hours in MCF-7 cells. treated and transfected. After 24 hours of additional incubation, confocal laser fluorescence microscopy confirmed the location on cells of green fluorescence where carboxyfluorescein of liposomes was generated with green wavelengths, and mRNA transfected with red wavelengths produced mStawberry protein and was located in the cytoplasm. Confirmed.

CD47 매개 약물전달 확인Confirmation of CD47-mediated drug delivery

약물의 전달이 CD47을 매개로 일어나는지 확인하기 위하여 MCF-7 세포에 에피루비신(epirubicin) 약물이 포집된 SV4가 결합된 iLP를 처리하였다. 이때 CD47 매개여부 확인을 위하여 CD47 항체(polyclonal)를 처리한 실험군과 처리하지 않은 실험군을 비교하였다. 구체적인 CD47 masking assay 방법은 다음과 같다.To confirm whether drug delivery occurs through CD47, MCF-7 cells were treated with SV4-conjugated iLP, in which epirubicin was captured. At this time, to confirm CD47 mediation, the experimental group treated with CD47 antibody (polyclonal) and the experimental group not treated were compared. The specific CD47 masking assay method is as follows.

6구 세포 배양 플레이트의 각 구 당 5×105 개의 세포를 넣고 37℃, 5% CO2 조건의 세포 배양기에서 24시간 배양하여 바닥에 부착시킨 뒤, 한 세포 샘플에만 최종 농도 5 ㎍/㎖의 CD47 항체를 넣고 4시간 반응하여 세포 표면의 CD47을 차단한다. 여기에 에피루비신을 포집하고 SV4가 결합된 리포좀(iLP)을 각 세포마다 에피루비신 최종 농도가 10 μM이 되도록 iLP 처리하고 24시간 배양하였다. 24시간 뒤 형광 현미경을 통해 세포내로 도입되어 세포 핵에서 빨간 형광을 나타내는 에피루비신의 형광 정도를 비교 분석하였다. 이때 대조군으로는 에피루비신을 직접 처리한 세포 샘플을 사용하였다(도 23A 참조). MCF7-이종 이식 마우스에 100 ㎍의 SV4 단백질을 먼저 투여하고 포화시켜서 마우스 체내 암세포의 CD47을 차단한 상태와 동일 부피의 인산 완충 생리식염수를 투여하여 암세포의 CD47을 차단하지 않은 상태로 만든다. 여기에 SV4-DSPE와 근적외선 영역의 빨간 형광을 나타내는 Cyanine 5.5-DSPE가 동시에 삽입되어 있는 T001 mRNA를 포집시킨 리포좀을 혈관 주사를 통해 마우스 체내 100 ㎕(mRNA 5 ㎍/Liposome 0.5 mg)를 투여하여 CD47을 매개로 한 타겟에 대한 접근성을 확인하였다.5 × 10 5 cells per each sphere of a 6-hole cell culture plate were cultured for 24 hours in a cell incubator at 37 ° C and 5% CO 2 conditions, and then attached to the bottom, and only one cell sample had a final concentration of 5 μg / ml. CD47 antibody is added and reacted for 4 hours to block CD47 on the cell surface. Epirubicin was collected here, and SV4-conjugated liposomes (iLP) were treated with iLP so that the final concentration of epirubicin was 10 μM for each cell, and cultured for 24 hours. After 24 hours, the degree of fluorescence of epirubicin, which was introduced into the cells and exhibited red fluorescence in the cell nucleus, was compared and analyzed using a fluorescence microscope. At this time, as a control, a cell sample directly treated with epirubicin was used (see FIG. 23A). 100 μg of SV4 protein was first administered to MCF7-xenotransplanted mice to saturate them, and the same volume of phosphate-buffered saline was administered to the mice in a state in which CD47 was blocked in cancer cells in the mouse, so that CD47 in cancer cells was not blocked. Here, 100 μl (mRNA 5 μg/Liposome 0.5 mg) of liposome containing T001 mRNA into which SV4-DSPE and Cyanine 5.5-DSPE, which exhibit red fluorescence in the near-infrared region are simultaneously inserted, is injected into the mouse body through intravascular injection to detect CD47 The accessibility to the target through the medium was confirmed.

실험 결과, 도 23A에 나타낸 바와 같이, CD47을 차단(blocking)한 처리군에서는 에피루비신의 형광이 나타나지 않았지만 차단하지 않은 실험군에서는 에피루비신의 형광이 나타났다.As a result of the experiment, as shown in FIG. 23A, fluorescence of epirubicin was not observed in the treatment group in which CD47 was blocked, but fluorescence of epirubicin was shown in the experimental group in which CD47 was not blocked.

또한 체내(in vivo)에서 CD47 매개 약물전달능을 확인하기 위한 MCF7-이종이식 마우스 모델 실험에서, 혈액세포에 존재하는 CD47로 인해 전반적인 노이즈가 관찰되었으나 SV4로 차단한 마우스에서보다 차단하지 않은 마우스의 암세포에서 더 강한 약물의 전달이 확인되었다(도 23B 참조).In addition, in the MCF7-xenotransplantation mouse model experiment to confirm the CD47-mediated drug delivery ability in vivo , overall noise was observed due to CD47 present in blood cells, but it was higher in unblocked mice than in SV4-blocked mice. Stronger drug delivery was confirmed in cancer cells (see Fig. 23B).

MCF7 이종이식(xenograft) 마우스 모델에서 근적외선 형광 단백질(Near Infrared Red Fluorescence Protein, NIR RFP)의 mRNA를 포함한 SV4-컨쥬게이트 iLP를 정맥주사를 통해 주입한 후 시간별로 형광 이미지를 분석하였고, 그 결과를 도 24에 나타내었다. 구체적인 실험방법은 다음과 같다.In the MCF7 xenograft mouse model, SV4-conjugated iLP containing mRNA of Near Infrared Red Fluorescence Protein (NIR RFP) was intravenously injected, and fluorescence images were analyzed over time, and the results were analyzed. 24. The specific test method is as follows.

인간 유방암 세포주인 MCF7을 배양하여 누드 마우스에 이종이식하였다. NIR-RFP(Near Infrared-Red Fluorescence Protein) mRNA가 포집된 SV4-iLP를 정맥주사를 통해 25 g 마우스당 5 ㎍ mRNA가 투여될 수 있도록 조절하여 투여한 후 in vivo optical imaging system을 사용하여 주사한 시료의 형광 정도를 추적하였다.MCF7, a human breast cancer cell line, was cultured and xenotransplanted into nude mice. SV4-iLP, in which NIR-RFP (Near Infrared-Red Fluorescence Protein) mRNA was captured, was injected intravenously, adjusted so that 5 μg mRNA was administered per 25 g mouse, and then injected using an in vivo optical imaging system. The degree of fluorescence of the sample was tracked.

도 24를 참조하면, mRNA의 전달이 일부 혈액세포의 CD47을 통해 일어나지만 비교적 높은 수준으로 암세포에 발현이 축적되는 현상을 관찰하였다.Referring to FIG. 24, although mRNA delivery occurs through CD47 in some blood cells, a phenomenon in which expression is accumulated in cancer cells at a relatively high level was observed.

MCF7 이종이식 마우스 IV 암성장 제어 효능 시험(28일)을 수행하였으며, 구체적으로 MCF7을 배양하여 누드 마우스에 피하주사(subcutaneous injection)하였다. 종양(tumor)이 자란 누드 마우스를 랜덤하게 분류한 뒤, 정맥(intravenous, IV)으로 각각 PBS, Liposome, Liposome-NT, Liposome-MT(Lipsome 20 mpk, 10 ㎍ mRNA/mg liposome)를 처리하였다. 각 mRNA는 5 ㎍을 처리하였으며, 3일 간격으로 28일 동안 총 5번을 처리하였다. 3일 간격으로 종양의 체적(volume)을 조사하여 그래프로 나타냈다. 28일째 누드 마우스를 희생시켜 종양을 분리하여 체적을 측정하였고, 그 결과를 도 25에 나타내었다.MCF7 xenograft mouse IV cancer growth control efficacy test (28 days) was performed, and specifically MCF7 was cultured and injected subcutaneously into nude mice. After randomly classifying nude mice with tumors, PBS, Liposome, Liposome-NT, and Liposome-MT (Lipsome 20 mpk, 10 μg mRNA/mg liposome) were treated intravenously (IV), respectively. Each mRNA was treated with 5 μg, a total of 5 times for 28 days at 3-day intervals. The volume of the tumor was examined at 3-day intervals and displayed in a graph. On day 28, nude mice were sacrificed, tumors were separated and the volume was measured, and the results are shown in FIG. 25 .

도 25를 참조하면, iLPD-NT 및 iLPD-MT 그룹에서 종양 성장률이 2개의 대조군 그룹(P = 0.03)과 비교하여 현저하게 감소하였다. 또한 PBS와 Void Liposome 주입 시료 사이에는 약간의 차이가 있었다.Referring to FIG. 25 , tumor growth rates in the iLPD-NT and iLPD-MT groups were significantly decreased compared to the two control groups (P = 0.03). There was also a slight difference between the PBS and Void Liposome injection samples.

체내(in vivo)에서 mRNA의 독성 효과를 평가하기 위해 실험 기간 동안 마우의 체중을 측정하였으며, 모든 그룹에서 마우스의 체중은 전체 실험 기간에 걸쳐 약간 증가하였고, 대조군에 비해 시료 주입 후 체중의 변화는 크지 않은 것으로 나타났다. 도 25에서와 같이, 실험 완료 후 희생된 마우스에서 채취한 종양 조직의 최종 크기를 비교하여 나타내었다.In order to evaluate the toxic effect of mRNA in the body ( in vivo ), the body weight of mice was measured during the experimental period. In all groups, the body weight of mice increased slightly throughout the entire experimental period, and the change in body weight after sample injection compared to the control group was It turned out not to be large. As shown in FIG. 25, the final size of the tumor tissue collected from the sacrificed mouse after completion of the experiment was compared and shown.

실험 완료 후 희생된 마우스에서 채취한 혈액과 조직으로 혈액학적 및 조직학적 검사를 실시하였고, 그 결과를 도 26에 나타내었다. 도 26을 참조하면, 대조군 대비 시료를 처리한 마우의 혈액과 간에서 WBC 수를 제외하면 유의미한 독성 효과를 거의 보이지 않았다.After completion of the experiment, hematological and histological examinations were performed with blood and tissues collected from the sacrificed mice, and the results are shown in FIG. 26 . Referring to FIG. 26, except for the number of WBCs in the blood and liver of mice treated with samples compared to the control group, no significant toxic effects were observed.

이상에서 설명한 본 발명의 바람직한 실시예들은 기술적 과제를 해결하기 위해 개시된 것으로, 본 발명이 속하는 기술분야에서 통상의 지식을 가진 자라면 본 발명의 사상 및 범위 안에서 다양한 수정, 변경, 부가 등이 가능할 것이며, 이러한 수정 변경 등은 이하의 특허청구범위에 속하는 것으로 보아야 할 것이다. Preferred embodiments of the present invention described above have been disclosed to solve the technical problems, and those skilled in the art will be able to make various modifications, changes, additions, etc. within the spirit and scope of the present invention. , such modifications and changes should be regarded as belonging to the scope of the following claims.

<110> BPGene Co., Ltd. <120> Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein <130> NP21-09070 <160> 41 <170> KoPatentIn 3.0 <210> 1 <211> 119 <212> PRT <213> Artificial Sequence <220> <223> CD47 binder SV1 <400> 1 Met Glu Glu Glu Leu Gln Ile Ile Gln Pro Asp Lys Ser Val Ser Val 1 5 10 15 Ala Ala Gly Glu Ser Ala Ile Leu His Cys Thr Ile Thr Ser Leu Phe 20 25 30 Pro Val Gly Pro Ile Gln Trp Phe Arg Gly Ala Gly Pro Ala Arg Val 35 40 45 Leu Ile Tyr Asn Gln Arg Gln Gly Pro Phe Pro Arg Val Thr Thr Val 50 55 60 Ser Glu Thr Thr Lys Arg Glu Asn Met Asp Phe Ser Ile Ser Ile Ser 65 70 75 80 Asn Ile Thr Pro Ala Asp Ala Gly Thr Tyr Tyr Cys Ile Lys Phe Arg 85 90 95 Lys Gly Ser Pro Asp Thr Glu Phe Lys Ser Gly Ala Gly Thr Glu Leu 100 105 110 Ser Val Arg Ala Lys Pro Ser 115 <210> 2 <211> 119 <212> PRT <213> Artificial Sequence <220> <223> CD47 binder SV4 <400> 2 Met Glu Glu Glu Leu Gln Ile Ile Gln Pro Asp Leu Ser Val Ser Val 1 5 10 15 Ala Ala Gly Glu Ser Ala Ile Leu His Cys Thr Ile Thr Ser Leu Phe 20 25 30 Pro Val Gly Pro Ile Gln Trp Phe Arg Gly Ala Gly Pro Ala Arg Val 35 40 45 Leu Ile Tyr Asn Gln Arg Gln Gly Pro Phe Pro Arg Val Thr Thr Val 50 55 60 Ser Glu Thr Thr Lys Arg Glu Asn Met Asp Phe Ser Ile Ser Ile Ser 65 70 75 80 Asn Ile Thr Pro Ala Asp Ala Gly Thr Tyr Tyr Cys Ile Lys Phe Arg 85 90 95 Lys Gly Ser Pro Asp Thr Glu Phe Leu Ser Gly Ala Gly Thr Glu Leu 100 105 110 Ser Val Arg Ala Lys Pro Ser 115 <210> 3 <211> 241 <212> PRT <213> Artificial Sequence <220> <223> T001 <400> 3 Met Phe Ser Ile Lys Glu Pro Phe Ser Ile Val Thr Asp Cys Asp Glu 1 5 10 15 Val Leu Thr Asp Ile Ser Pro Leu Trp Val His Lys Ile Gln Gln Asn 20 25 30 Ala Asp Tyr Phe Gly Lys Tyr Phe Asp Leu Ser Lys Leu Glu Gly Leu 35 40 45 Glu Phe Gly Thr Phe Glu His Tyr Gln Thr Val Leu Ser Arg Pro Glu 50 55 60 Phe His Leu Asn Lys Trp Leu Arg Lys Glu Asn Leu Val Leu Ser Asp 65 70 75 80 Glu Glu Glu Lys Glu Leu Phe Glu Arg Phe Tyr Ser Leu Tyr Asp Asn 85 90 95 Asp Glu Phe Tyr Glu Asp Cys Met Pro Thr Lys Met Cys Glu Gly Ile 100 105 110 Tyr Lys Leu Ser Leu Gln Lys Phe Val Asp Lys Ile Tyr Val Val Thr 115 120 125 Arg Thr Ser Glu Gly Thr Lys Glu Gly Lys Arg Lys Phe Ile Glu Thr 130 135 140 Phe Leu Asn Ser Asn Lys Val Glu Ile Ile Phe Val Gly Lys Asn Glu 145 150 155 160 Lys Lys Ser Asp Tyr Ile Lys Asn Leu Lys Asn Val Lys Met Ile Val 165 170 175 Glu Asp Glu Leu Ser Asn Ile Asn Asp Ile Val Glu Asn Cys Asn Asp 180 185 190 Gly Phe Glu Glu Val Asp Thr Tyr Ile Pro Ser Thr Gly Tyr Asn Asn 195 200 205 Lys Asp Ile Asp Gly Phe Asn Glu Asn Leu Met Lys Lys Gly Phe Asn 210 215 220 Ala Val Pro Tyr Val Ile Ile Glu Arg Pro Glu Thr Glu Glu Lys Ala 225 230 235 240 Val <210> 4 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 4 ttggtcgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 5 <211> 247 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 5 acgcgtcgag catgcatcta gggcggccaa ttccgcccct ctcccccccc cccctctccc 60 tccccccccc ctaacgttac tggccgaagc cgcttggaat aaggccggtg tgcgtttgtc 120 tatatgttat tttccaccat attgccgtct tttggcaatg tgagggcccg gaaacctggc 180 cctgtcttct tgacgagcat tcctaggggt ctttcccctc tcgccaaagg aatgcaaggt 240 ctgttga 247 <210> 6 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 6 ttggtcgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 7 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 7 ttggtcgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 8 <211> 321 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 8 gcatcacatt taaaagcatc tcagcctacc atgagaataa gagaaagaaa atgaagatca 60 atagcttatt catctctttt tctttttcgt tggtgtaaag ccaacaccct gtctaaaaaa 120 cataaatttc tttaatcatt ttgcctcttt tctctgtgct tcaattaata aaaaatggaa 180 agaacctaga tctaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 240 aaaaaaaaaa aaaaaaatgc atcccccccc cccccccccc cccccccccc cccaaaggct 300 cttttcagag ccaccagaat t 321 <210> 9 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 9 ttggtcgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 10 <211> 321 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 10 gcatcacatt taaaagcatc tcagcctacc atgagaataa gagaaagaaa atgaagatca 60 atagcttatt catctctttt tctttttcgt tggtgtaaag ccaacaccct gtctaaaaaa 120 cataaatttc tttaatcatt ttgcctcttt tctctgtgct tcaattaata aaaaatggaa 180 agaacctaga tctaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 240 aaaaaaaaaa aaaaaaatgc atcccccccc cccccccccc cccccccccc cccaaaggct 300 cttttcagag ccaccagaat t 321 <210> 11 <211> 74 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 11 gggtcccgca gtcggcgtcc agcggctctg cttgttcgtg tgtgtgtcgt tgcaggcctt 60 attcaagcgc cacc 74 <210> 12 <211> 321 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 12 gcatcacatt taaaagcatc tcagcctacc atgagaataa gagaaagaaa atgaagatca 60 atagcttatt catctctttt tctttttcgt tggtgtaaag ccaacaccct gtctaaaaaa 120 cataaatttc tttaatcatt ttgcctcttt tctctgtgct tcaattaata aaaaatggaa 180 agaacctaga tctaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 240 aaaaaaaaaa aaaaaaatgc atcccccccc cccccccccc cccccccccc cccaaaggct 300 cttttcagag ccaccagaat t 321 <210> 13 <211> 83 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 13 cttcctactc aggctttata caaagaccaa gaggtacagg tgcaagggag agaagaagag 60 taagaagaaa tataagagcc acc 83 <210> 14 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 14 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 15 <211> 25 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 15 agtaagaaga aatataagag ccacc 25 <210> 16 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 16 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 17 <211> 12 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 17 gagactgcca cc 12 <210> 18 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 18 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 19 <211> 12 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 19 gagactgcca ag 12 <210> 20 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 20 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 21 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 21 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 22 <211> 18 <212> PRT <213> Artificial Sequence <220> <223> Nucleoplasmin NLS sequence <400> 22 Lys Arg Pro Ala Ala Thr Lys Lys Ala Gly Gln Ala Lys Lys Lys Lys 1 5 10 15 Leu Asp <210> 23 <211> 25 <212> PRT <213> Artificial Sequence <220> <223> Poxvirus MLS sequence <400> 23 Lys Ile Ser Val Tyr Leu Thr Ala Ala Val Val Gly Phe Val Ala Tyr 1 5 10 15 Gly Ile Leu Lys Trp Tyr Arg Gly Thr 20 25 <210> 24 <211> 503 <212> PRT <213> Unknown <220> <223> Human Sirpa <400> 24 Met Glu Pro Ala Gly Pro Ala Pro Gly Arg Leu Gly Pro Leu Leu Cys 1 5 10 15 Leu Leu Leu Ala Ala Ser Cys Ala Trp Ser Gly Val Ala Gly Glu Glu 20 25 30 Glu Leu Gln Val Ile Gln Pro Asp Lys Ser Val Ser Val Ala Ala Gly 35 40 45 Glu Ser Ala Ile Leu His Cys Thr Val Thr Ser Leu Ile Pro Val Gly 50 55 60 Pro Ile Gln Trp Phe Arg Gly Ala Gly Pro Ala Arg Glu Leu Ile Tyr 65 70 75 80 Asn Gln Lys Glu Gly His Phe Pro Arg Val Thr Thr Val Ser Glu Ser 85 90 95 Thr Lys Arg Glu Asn Met Asp Phe Ser Ile Ser Ile Ser Asn Ile Thr 100 105 110 Pro Ala Asp Ala Gly Thr Tyr Tyr Cys Val Lys Phe Arg Lys Gly Ser 115 120 125 Pro Asp Thr Glu Phe Lys Ser Gly Ala Gly Thr Glu Leu Ser Val Arg 130 135 140 Ala Lys Pro Ser Ala Pro Val Val Ser Gly Pro Ala Ala Arg Ala Thr 145 150 155 160 Pro Gln His Thr Val Ser Phe Thr Cys Glu Ser His Gly Phe Ser Pro 165 170 175 Arg Asp Ile Thr Leu Lys Trp Phe Lys Asn Gly Asn Glu Leu Ser Asp 180 185 190 Phe Gln Thr Asn Val Asp Pro Val Gly Glu Ser Val Ser Tyr Ser Ile 195 200 205 His Ser Thr Ala Lys Val Val Leu Thr Arg Glu Asp Val His Ser Gln 210 215 220 Val Ile Cys Glu Val Ala His Val Thr Leu Gln Gly Asp Pro Leu Arg 225 230 235 240 Gly Thr Ala Asn Leu Ser Glu Thr Ile Arg Val Pro Pro Thr Leu Glu 245 250 255 Val Thr Gln Gln Pro Val Arg Ala Glu Asn Gln Val Asn Val Thr Cys 260 265 270 Gln Val Arg Lys Phe Tyr Pro Gln Arg Leu Gln Leu Thr Trp Leu Glu 275 280 285 Asn Gly Asn Val Ser Arg Thr Glu Thr Ala Ser Thr Val Thr Glu Asn 290 295 300 Lys Asp Gly Thr Tyr Asn Trp Met Ser Trp Leu Leu Val Asn Val Ser 305 310 315 320 Ala His Arg Asp Asp Val Lys Leu Thr Cys Gln Val Glu His Asp Gly 325 330 335 Gln Pro Ala Val Ser Lys Ser His Asp Leu Lys Val Ser Ala His Pro 340 345 350 Lys Glu Gln Gly Ser Asn Thr Ala Ala Glu Asn Thr Gly Ser Asn Glu 355 360 365 Arg Asn Ile Tyr Ile Val Val Gly Val Val Cys Thr Leu Leu Val Ala 370 375 380 Leu Leu Met Ala Ala Leu Tyr Leu Val Arg Ile Arg Gln Lys Lys Ala 385 390 395 400 Gln Gly Ser Thr Ser Ser Thr Arg Leu His Glu Pro Glu Lys Asn Ala 405 410 415 Arg Glu Ile Thr Gln Asp Thr Asn Asp Ile Thr Tyr Ala Asp Leu Asn 420 425 430 Leu Pro Lys Gly Lys Lys Pro Ala Pro Gln Ala Ala Glu Pro Asn Asn 435 440 445 His Thr Glu Tyr Ala Ser Ile Gln Thr Ser Pro Gln Pro Ala Ser Glu 450 455 460 Asp Thr Leu Thr Tyr Ala Asp Leu Asp Met Val His Leu Asn Arg Thr 465 470 475 480 Pro Lys Gln Pro Ala Pro Lys Pro Glu Pro Ser Phe Ser Glu Tyr Ala 485 490 495 Ser Val Gln Val Pro Arg Lys 500 <210> 25 <211> 357 <212> DNA <213> Unknown <220> <223> Human Sirpa DNA sequence <400> 25 ggtgaggagg agctgcaggt gattcagcct gacaagtccg tatcagttgc agctggagag 60 tcggccattc tgcactgcac tgtgacctcc ctgatccctg tggggcccat ccagtggttc 120 agaggagctg gaccagcccg ggaattaatc tacaatcaaa aagaaggcca cttcccccgg 180 gtaacaactg tttcagagtc cacaaagaga gaaaacatgg acttttccat cagcatcagt 240 aacatcaccc cagcagatgc cggcacctac tactgtgtga agttccggaa agggagccct 300 gacacggagt ttaagtctgg agcaggcact gagctgtctg tgcgtgccaa accctct 357 <210> 26 <211> 354 <212> DNA <213> Artificial Sequence <220> <223> SV1 DNA sequence <400> 26 gaagaggaac tgcagattat tcagccggac aagtccgtaa gcgttgcagc tggcgagagc 60 gccattctgc actgcactat tacctccctg tttccggtgg gcccgatcca gtggttccgt 120 ggcgctggcc cagcccgtgt gctgatctac aatcaacgtc agggcccgtt cccgcgtgta 180 accactgtta gcgagaccac caagcgtgaa aacatggact tttccatcag catcagcaac 240 atcaccccag cagatgccgg cacctactac tgtattaagt tccgtaaagg cagccctgac 300 acggagttta agtctggcgc aggcactgag ctgtctgtgc gtgccaaacc gtct 354 <210> 27 <211> 33 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL11_F1 Primer <400> 27 gattattcag ccggacctgt ccgtaagcgt tgc 33 <210> 28 <211> 33 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL11_R1 Primer <400> 28 gcaacgctta cggacaggtc cggctgaata atc 33 <210> 29 <211> 27 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL104_F2 Primer <400> 29 ctgacacgga gtttctgtct ggcgcag 27 <210> 30 <211> 27 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL104_R2 Primer <400> 30 ctgcgccaga cagaaactcc gtgtcag 27 <210> 31 <211> 198 <212> PRT <213> Unknown <220> <223> NT5M <400> 31 Met Gly Gly Arg Ala Leu Arg Val Leu Val Asp Met Asp Gly Val Leu 1 5 10 15 Ala Asp Phe Glu Gly Gly Phe Leu Arg Lys Phe Arg Ala Arg Phe Pro 20 25 30 Asp Gln Pro Phe Ile Ala Leu Glu Asp Arg Arg Gly Phe Trp Val Ser 35 40 45 Glu Gln Tyr Gly Arg Leu Arg Pro Gly Leu Ser Glu Lys Ala Ile Ser 50 55 60 Ile Trp Glu Ser Lys Asn Phe Phe Phe Glu Leu Glu Pro Leu Pro Gly 65 70 75 80 Ala Val Glu Ala Val Lys Glu Met Ala Ser Leu Gln Asn Thr Asp Val 85 90 95 Phe Ile Cys Thr Ser Pro Ile Lys Met Phe Lys Tyr Cys Pro Tyr Glu 100 105 110 Lys Tyr Ala Trp Val Glu Lys Tyr Phe Gly Pro Asp Phe Leu Glu Gln 115 120 125 Ile Val Leu Thr Arg Asp Lys Thr Val Val Ser Ala Asp Leu Leu Ile 130 135 140 Asp Asp Arg Pro Asp Ile Thr Gly Ala Glu Pro Thr Pro Ser Trp Glu 145 150 155 160 His Val Leu Phe Thr Ala Cys His Asn Gln His Leu Gln Leu Gln Pro 165 170 175 Pro Arg Arg Arg Leu His Ser Trp Ala Asp Asp Trp Lys Ala Ile Leu 180 185 190 Asp Ser Lys Arg Pro Cys 195 <210> 32 <211> 25 <212> RNA <213> Artificial Sequence <220> <223> siT001-1 sense <400> 32 cgagaagaag ucagauuaca ucaag 25 <210> 33 <211> 25 <212> RNA <213> Artificial Sequence <220> <223> siT001-1 antisense <400> 33 cuugauguaa ucugacuucu ucucg 25 <210> 34 <211> 25 <212> RNA <213> Artificial Sequence <220> <223> siT001-2 sense <400> 34 cgcaaauuca uugaaaccuu ccuga 25 <210> 35 <211> 25 <212> RNA <213> Artificial Sequence <220> <223> siT001-2 antisense <400> 35 ucaggaaggu uucaaugaau uugcg 25 <210> 36 <211> 32 <212> DNA <213> Artificial Sequence <220> <223> Forward Primer for PCR <400> 36 gtgcttctga cacaacagtc tcgaacttaa gc 32 <210> 37 <211> 32 <212> DNA <213> Artificial Sequence <220> <223> Reverse Primer for PCR <400> 37 gaagcggccg ccttcctact caggctttat tc 32 <210> 38 <211> 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant, wild type <400> 38 atgttttcta ttaaagaacc attttcaatt gttacagact gcgatgaggt attaactgac 60 attagtcctt tatgggttca taagattcag caaaatgctg attattttgg aaaatacttt 120 gatttaagta aactagaagg attggaattt ggtacatttg aacattatca aacagtacta 180 tcacgaccag aatttcattt aaataaatgg ctaagaaaag agaatcttgt attatcagat 240 gaagaagaaa aagaattatt tgaaagattt tattcgttat atgataatga tgaattttat 300 gaagattgta tgccaactaa aatgtgtgaa ggaatttata aattatcatt acaaaaattc 360 gtagataaaa tctatgttgt aacaagaaca agtgaaggaa ccaaagaagg aaaaagaaaa 420 tttattgaaa ctttcttaaa ttctaataaa gtagagatta tttttgttgg gaaaaatgaa 480 aagaaatcag attatattaa gaatctaaag aatgtaaaaa tgattgtaga agatgaatta 540 tcaaatatta atgatattgt agaaaattgt aatgatggtt ttgaagaagt agatacttat 600 attccatcaa ctggttataa caataaagat attgatggtt ttaatgaaaa tctaatgaaa 660 aaaggattta atgccgttcc atatgtaatt atagaaagac cagaaacaga agagaaagca 720 gtataa 726 <210> 39 <211> 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant, T001 <400> 39 atgttctcca tcaaagaacc attctccatc gtgaccgact gcgatgaggt gctgaccgac 60 attagccctc tgtgggtgca taagattcag cagaacgctg attacttcgg caaatacttt 120 gatctgagca aactggaagg cctggaattt ggtacgtttg aacattatca gacggtgctg 180 tcacgcccag agttccatct gaacaaatgg ctgcgcaaag agaacctggt gctgtcagat 240 gaagaagaga aagaactgtt tgaacgcttc tattcgctgt atgataatga tgagttctac 300 gaagattgta tgccaaccaa aatgtgcgaa ggcatctaca aactgtcact gcagaaattc 360 gtggataaaa tctatgtggt aacccgcacc agtgaaggca ccaaagaagg caaacgcaaa 420 ttcattgaaa ccttcctgaa ctctaacaaa gtggagatta tcttcgttgg caagaacgag 480 aagaagtcag attacatcaa gaacctgaag aatgtgaaaa tgattgtgga agatgaactg 540 tccaacatca acgatattgt ggagaactgt aacgatggct ttgaagaagt ggatacctac 600 attccgtcaa ctggttacaa caacaaagat attgatggct tcaacgaaaa cctgatgaaa 660 aaaggcttca acgccgttcc atacgtgatt atcgaacgcc cagaaaccga agagaaagca 720 gtgtaa 726 <210> 40 <211> 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant, T002 <400> 40 atgttctcca tcaaggagcc gttctccatc gtgaccgact gcgacgaggt gctgaccgac 60 atcagcccgc tgtgggtgca caagatccag cagaacgccg actacttcgg caagtacttc 120 gacctgagca agctggaggg cctggagttc ggcacgttcg agcactacca gacggtgctg 180 tcgcgcccgg agttccacct gaacaagtgg ctgcggaaag aaaacctcgt cctctccgac 240 gaggaggaga aggagctgtt cgagcgcttc tactcgctgt acgacaacga cgagttctac 300 gaggactgca tgccgaccaa gatgtgcgag ggcatctaca agctgtcgct gcagaagttc 360 gtggacaaga tctacgtggt cacccgcacg agcgagggca ccaaggaggg caagcgcaag 420 ttcatcgaga ccttcctgaa ctcgaacaag gtggagatca tcttcgtcgg caagaacgag 480 aagaagtcag actacatcaa gaacctgaag aacgtgaaga tgatcgtgga ggatgagctg 540 tccaacatca acgacatcgt ggagaactgc aacgacggct tcgaggaggt ggacacctac 600 atcccgtcaa ctggttacaa caacaaggat atcgacggct tcaacgagaa cctgatgaag 660 aagggcttca acgccgtgcc atacgtgatc atcgagcgcc cagagaccga ggagaaggca 720 gtgtaa 726 <210> 41 <211> 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant, T007 <400> 41 atgttcagca tcaaggagcc cttctccatc gtgaccgact gcgacgaagt cctcaccgac 60 atcagccccc tgtgggtgca caagatccag cagaacgccg actacttcgg gaaatacttc 120 gacctgagca agctggaggg cctggagttc gggaccttcg agcactacca gaccgtgctc 180 tcccgccccg aattccacct gaacaagtgg ctccgcaagg agaacctggt cctgtccgac 240 gaggaggaga aggagctgtt cgagcgcttc tacagcctgt acgacaacga cgagttctac 300 gaggactgca tgcccactaa gatgtgcgag ggcatctaca agctgagcct gcagaagttc 360 gtggataaga tctatgtcgt gacccgcacc agcgagggca ccaaggaggg caagcgcaag 420 ttcatcgaga ctttcctcaa cagcaacaag gtggagatca tcttcgtcgg caagaacgaa 480 aaaaagagcg actacatcaa gaacctgaag aacgtgaaga tgatcgtgga ggacgagctg 540 tccaacatca acgacatcgt ggagaactgc aacgacggct tcgaggaggt ggacacctac 600 atccccagca ctgggtacaa caacaaggac atcgacggct tcaacgagaa cctgatgaag 660 aagggcttca acgccgtgcc ctacgtgatc atcgagcgcc ccgagaccga ggagaaggcc 720 gtgtaa 726 <110> BPGene Co., Ltd. <120> Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein <130> NP21-09070 <160> 41 <170> KoPatentIn 3.0 <210> 1 <211> 119 <212> PRT <213> Artificial Sequence <220> < 223> CD47 binder SV1 <400> 1 Met Glu Glu Glu Leu Gln Ile Ile Gln Pro Asp Lys Ser Val Ser Val 1 5 10 15 Ala Ala Gly Glu Ser Ala Ile Leu His Cys Thr Ile Thr Ser Leu Phe 20 25 30 Pro Val Gly Pro Ile Gln Trp Phe Arg Gly Ala Gly Pro Ala Arg Val 35 40 45 Leu Ile Tyr Asn Gln Arg Gln Gly Pro Phe Pro Arg Val Thr Thr Val 50 55 60 Ser Glu Thr Thr Lys Arg Glu Asn Met Asp Phe Ser Ile Ser Ile Ser 65 70 75 80 Asn Ile Thr Pro Ala Asp Ala Gly Thr Tyr Tyr Cys Ile Lys Phe Arg 85 90 95 Lys Gly Ser Pro Asp Thr Glu Phe Lys Ser Gly Ala Gly Thr Glu Leu 100 105 110 Ser Val Arg Ala Lys Pro Ser 115 <210> 2 <211> 119 <212> PRT <213> Artificial Sequence <220> <223> CD47 binder SV4 <400> 2 Met Glu Glu Glu Leu Gln Ile Ile Gln Pro Asp Leu Ser Val Ser Val 1 5 10 15 Ala Ala Gly Glu Ser Ala Ile Leu His Cys Thr Ile Thr Ser Leu Phe 20 25 30 Pro Val Gly Pro Ile Gln Trp Phe Arg Gly Ala Gly Pro Ala Arg Val 35 40 45 Leu Ile Tyr Asn Gln Arg Gln Gly Pro Phe Pro Arg Val Thr Thr Val 50 55 60 Ser Glu Thr Thr Lys Arg Glu Asn Met Asp Phe Ser Ile Ser Ile Ser 65 70 75 80 Asn Ile Thr Pro Ala Asp Ala Gly Thr Tyr Tyr Cys Ile Lys Phe Arg 85 90 95 Lys Gly Ser Pro Asp Thr Glu Phe Leu Ser Gly Ala Gly Thr Glu Leu 100 105 110 Ser Val Arg Ala Lys Pro Ser 115 <210> 3 <211> 241 <212> PRT <213> Artificial Sequence <220> <223> T001 < 400>3 Met Phe Ser Ile Lys Glu Pro Phe Ser Ile Val Thr Asp Cys Asp Glu 1 5 10 15 Val Leu Thr Asp Ile Ser Pro Leu Trp Val His Lys Ile Gln Gln Asn 20 25 30 Ala Asp Tyr Phe Gly Lys Tyr Phe Asp Leu Ser Lys Leu Glu Gly Leu 35 40 45 Glu Phe Gly Thr Phe Glu His Tyr Gln Thr Val Leu Ser Arg Pro Glu 50 55 60 Phe His Leu Asn Lys Trp Leu Arg Lys Glu Asn Leu Val Leu Ser Asp 65 70 75 80 Glu Glu Glu Lys Glu Leu Phe Glu Arg Phe Tyr Ser Leu Tyr Asp Asn 85 90 95 Asp Glu Phe Tyr Glu Asp Cys Met Pro Thr Lys Met Cys Glu Gly Ile 100 105 110 Tyr Lys Leu Ser Leu Gln Lys Phe Val Asp Lys Ile Tyr Val Val Thr 115 120 125 Arg Thr Ser Glu Gly Thr Lys Glu Gly Lys Arg Lys Phe Ile Glu Thr 130 135 140 Phe Leu Asn Ser Asn Lys Val Glu Ile Ile Phe Val Gly Lys Asn Glu 145 150 155 160 Lys Lys Ser Asp Tyr Ile Lys Asn Leu Lys Asn Val Lys Met Ile Val 165 170 175 Glu Asp Glu Leu Ser Asn Ile Asn Asp Ile Val Glu Asn Cys Asn Asp 180 185 190 Gly Phe Glu Glu Val Asp Thr Tyr Ile Pro Ser Thr Gly Tyr Asn Asn 195 200 205 Lys Asp Ile Asp Gly Phe Asn Glu Asn Leu Met Lys Lys Gly Phe Asn 210 215 220 Ala Val Pro Tyr Val Ile Ile Glu Arg Pro Glu Thr Glu Glu Lys Ala 225 230 235 240 Val <210> 4 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 4 ttggtcgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 5 <211> 247 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 5 acgcgtcgag catgcatcta gggcggccaa ttccgcccct ctcccccccc cccctctccc 60 tccccccccc ctaacgttac tggccgaagc cgcttggaat aaggccgg tg tgcgtttgtc 120 tatatgttat tttccaccat attgccgtct tttggcaatg tgagggcccg gaaacctggc 180 cctgtcttct tgacgagcat tcctaggggt ctttcccctc tcgccaaagg aatgcaaggt 240 ctgttga 247 <210> 6 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 6 ttggtcgtga ggcactgggc aggta agtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 7 <211> 177 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 7 ttggt cgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccactttg cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 8 <211> 321 <212> DNA <213> Artificial Sequence <220> <223> 3 '-UTR sequence <400> 8 gcatcacatt taaaagcatc tcagcctacc atgagaataa gagaaagaaa atgaagatca 60 atagcttatt catctctttt tctttttcgt tggtgtaaag ccaacaccct gtctaaaaaa 120 cataaatttc tttaatcatt ttgcctcttt tctctgtgct tcaattaata aaaaatggaa 180 agaacctaga tctaaaaaaa a aaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 240 aaaaaaaaaa aaaaaaatgc atcccccccc cccccccccc cccccccccc cccaaaggct 300 cttttcagag ccaccagaat t 321 <210> 9 <211> 1 77 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 9 ttggtcgtga ggcactgggc aggtaagtat caaggttaca agacaggttt aaggagacca 60 atagaaactg ggcttgtcga gacagagaag actcttgcgt ttctgatagg cacctattgg 120 tcttactgac atccacttt g cctttctctc cacaggtgtc cactcccagt tcaatta 177 <210> 10 <211> 321 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 10 gcatcacatt taaaagcatc tcagcctacc atgagaataa gagaaagaaa atgaagatca 60 atagcttatt catctctttt tctttttcgt tggtgtaaag ccaacaccct gtctaaaaaaa 120 cataaattt c tttaatcatt ttgcctcttt tctctgtgct tcaattaata aaaaatggaa 180 agaacctaga tctaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 240 aaaaaaaaaa aaaaaaatgc atcccccccc cccccccccc cccccccccc cccaaaggct 300 cttttcagag ccaccagaat t 321 <210> 11 <211> 74 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 11 gggtcccgca gtcggcgtcc agcggct ctg cttgttcgtg tgtgtgtcgt tgcaggcctt 60 attcaagcgc cacc 74 <210> 12 <211> 321 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 12 gcatcacatt taaaagcatc tcagcctacc atgagaataa gagaaagaaa atgaagatca 60 atagcttatt catctctttt tctttttcgt tggtg taaag ccaacaccct gtctaaaaaa 120 cataaatttc tttaatcatt ttgcctcttt tctctgtgct tcaattaata aaaaatggaa 180 agaacctaga tctaaaaaaa aaaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaaaaa 240 aaaaaaaaaa aaaaaatgc atcccccccc cccccccccc cccccccccc cc caaaggct 300 cttttcagag ccaccagaat t 321 <210> 13 <211> 83 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 13 cttcctactc aggctttata caaagaccaa gaggtacagg tgcaagggag agaagaagag 60 taagaagaaa tataagagcc acc 83 <210> 14 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 14 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 15 <211> 25 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 1 5 agtaagaaga aatataagag ccacc 25 <210 > 16 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 16 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <21 0> 17 <211> 12 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 17 gagactgcca cc 12 <210> 18 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 18 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 93 <210> 19 <211> 12 <212> DNA <213> Artificial Sequence <220> <223 > 5'-UTR sequence < 400> 19 gagactgcca ag 12 <210> 20 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 3'-UTR sequence <400> 20 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttgg tct ttgaataaag cctgagtagg aag 93 <210> 21 <211> 93 <212> DNA <213> Artificial Sequence <220> <223> 5'-UTR sequence <400> 21 gctgccttct gcggggcttg ccttctggcc atgcccttct tctctccctt gcacctgtac 60 ctcttggtct ttgaataaag cctgagtagg aag 9 3 <210> 22 <211 > 18 <212> PRT <213> Artificial Sequence <220> <223> Nucleoplasmin NLS sequence <400> 22 Lys Arg Pro Ala Ala Thr Lys Lys Ala Gly Gln Ala Lys Lys Lys Lys 1 5 10 15 Leu Asp <210> 23 <211> 25 <212> PRT <213> Artificial Sequence <220> <223> Poxvirus MLS sequence <400> 23 Lys Ile Ser Val Tyr Leu Thr Ala Ala Val Val Gly Phe Val Ala Tyr 1 5 10 15 Gly Ile Leu Lys Trp Tyr Arg Gly Thr 20 25 <210> 24 <211> 503 <212> PRT <213> Unknown <220> <223> Human Sirpa <400> 24 Met Glu Pro Ala Gly Pro Ala Pro Gly Arg Leu Gly Pro Leu Leu Cys 1 5 10 15 Leu Leu Leu Ala Ala Ser Cys Ala Trp Ser Gly Val Ala Gly Glu Glu 20 25 30 Glu Leu Gln Val Ile Gln Pro Asp Lys Ser Val Ser Val Ala Ala Gly 35 40 45 Glu Ser Ala Ile Leu His Cys Thr Val Thr Ser Leu Ile Pro Val Gly 50 55 60 Pro Ile Gln Trp Phe Arg Gly Ala Gly Pro Ala Arg Glu Leu Ile Tyr 65 70 75 80 Asn Gln Lys Glu Gly His Phe Pro Arg Val Thr Thr Val Ser Glu Ser 85 90 95 Thr Lys Arg Glu Asn Met Asp Phe Ser Ile Ser Ile Ser Asn Ile Thr 100 105 110 Pro Ala Asp Ala Gly Thr Tyr Tyr Cys Val Lys Phe Arg Lys Gly Ser 115 120 125 Pro Asp Thr Glu Phe Lys Ser Gly Ala Gly Thr Glu Leu Ser Val Arg 130 135 140 Ala Lys Pro Ser Ala Pro Val Val Ser Gly Pro Ala Ala Arg Ala Thr 145 150 155 160 Pro Gln His Thr Val Ser Phe Thr Cys Glu Ser His Gly Phe Ser Pro 165 170 175 Arg Asp Ile Thr Leu Lys Trp Phe Lys Asn Gly Asn Glu Leu Ser Asp 180 185 190 Phe Gln Thr Asn Val Asp Pro Val Gly Glu Ser Val Ser Tyr Ser Ile 195 200 205 His Ser Thr Ala Lys Val Val Leu Thr Arg Glu Asp Val His Ser Gln 210 215 220 Val Ile Cys Glu Val Ala His Val Thr Leu Gln Gly Asp Pro Leu Arg 225 230 235 240 Gly Thr Ala Asn Leu Ser Glu Thr Ile Arg Val Pro Thr Leu Glu 245 250 255 Val Thr Gln Gln Pro Val Arg Ala Glu Asn Gln Val Asn Val Thr Cys 260 265 270 Gln Val Arg Lys Phe Tyr Pro Gln Arg Leu Gln Leu Thr Trp Leu Glu 275 280 285 Asn Gly Asn Val Ser Arg Thr Glu Thr Ala Ser Thr Val Thr Glu Asn 290 295 300 Lys Asp Gly Thr Tyr Asn Trp Met Ser Trp Leu Leu Val Asn Val Ser 305 310 315 320 Ala His Arg Asp Asp Val Lys Leu Thr Cys Gln Val Glu His Asp Gly 325 330 335 Gln Pro Ala Val Ser Lys Ser His Asp Leu Lys Val Ser Ala His Pro 340 345 350 Lys Glu Gln Gly Ser Asn Thr Ala Ala Glu Asn Thr Gly Ser Asn Glu 355 360 365 Arg Asn Ile Tyr Ile Val Val Gly Val Val Cys Thr Leu Leu Val Ala 370 375 380 Leu Leu Met Ala Ala Leu Tyr Leu Val Arg Ile Arg Gln Lys Lys Ala 385 390 395 400 Gln Gly Ser Thr Ser Ser Thr Arg Leu His Glu Pro Glu Lys Asn Ala 405 410 415 Arg Glu Ile Thr Gln Asp Thr Asn Asp Ile Thr Tyr Ala Asp Leu Asn 420 425 430 Leu Pro Lys Gly Lys Lys Pro Ala Pro Gln Ala Ala Glu Pro Asn Asn 435 440 445 His Thr Glu Tyr Ala Ser Ile Gln Thr Ser Pro Gln Pro Ala Ser Glu 450 455 460 Asp Thr Leu Thr Tyr Ala Asp Leu Asp Met Val His Leu Asn Arg Thr 465 470 475 480 Pro Lys Gln Pro Ala Pro Lys Pro Glu Pro Ser Phe Ser Glu Tyr Ala 485 490 495 Ser Val Gln Val Pro Arg Lys 500 <210> 25 <211> 357 <212> DNA <213> Unknown <220> <223> Human Sirpa DNA sequence <400> 25 ggtgaggagg agctgcaggt gattcagcct gacaagtccg tatcagttgc agctggagag 60 tcggccattc tgcactgcac tgtgacctcc ctgatccctg tggggcccat ccagtggttc 120 agagg agctg gaccagcccg ggaattaatc tacaatcaaa aagaaggcca cttcccccgg 180 gtaacaactg tttcagagtc cacaaagaga gaaaacatgg acttttccat cagcatcagt 240 aacatcaccc cagcagatgc cggcacctac tactgtgtga agttccggaa agggagccct 300 gacacggagt ttaagtctgg agcaggcact gagctgtctg tgcgtgccaa accctct 357 <210> 26 <211> 354 <212> DNA <213> Artificial Sequence <220> <223> SV1 DNA sequence <400> 26 gaagag gaac tgcagattat tcagccggac aagtccgtaa gcgttgcagc tggcgagagc 60 gccattctgc actgcactat tacctccctg tttccggtgg gcccgatcca gtggttccgt 120 ggcgctggcc cagcccgtgt gctgatctac aatcaacgtc agggcccgtt cccgcgtgta 180 accactgtta gcgagaccac caagcgtgaa aacatggact tttccatcag catcagcaac 240 atcaccccag cagat gccgg cacctactac tgtattaagt tccgtaaagg cagccctgac 300 acggagttta agtctggcgc aggcactgag ctgtctgtgc gtgccaaacc gtct 354 <210> 27 <211> 33 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL11_F1 Primer <400> 27 gattattcag ccggacctgt ccgtaagcgt tgc 33 <210> 28 <211> 33 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL11_R1 Primer <400> 28 gcaacgctta cgg acaggtc cggctgaata atc 33 <210> 29 <211> 27 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL104_F2 Primer <400> 29 ctgacacgga gtttctgtct ggcgcag 27 <210> 30 <211> 27 <212> DNA <213> Artificial Sequence <220> <223> KKtoLL104_R2 Primer <400> 30 ctgcgccaga cagaaactcc gtgtcag 27 <210> 31 <211> 198 <212> PRT <213> Unknown <220> <223> NT5M <400> 31 Met Gly Gly Arg Ala Leu Arg Val Leu Val Asp Met Asp Gly Val Leu 1 5 10 15 Ala Asp Phe Glu Gly Gly Phe Leu Arg Lys Phe Arg Ala Arg Phe Pro 20 25 30 Asp Gln Pro Phe Ile Ala Leu Glu Asp Arg Arg Gly Phe Trp Val Ser 35 40 45 Glu Gln Tyr Gly Arg Leu Arg Pro Gly Leu Ser Glu Lys Ala Ile Ser 50 55 60 Ile Trp Glu Ser Lys Asn Phe Phe Phe Glu Leu Glu Pro Leu Pro Gly 65 70 75 80 Ala Val Glu Ala Val Lys Glu Met Ala Ser Leu Gln Asn Thr Asp Val 85 90 95 Phe Ile Cys Thr Ser Pro Ile Lys Met Phe Lys Tyr Cys Pro Tyr Glu 100 105 110 Lys Tyr Ala Trp Val Glu Lys Tyr Phe Gly Pro Asp Phe Leu Glu Gln 115 120 125 Ile Val Leu Thr Arg Asp Lys Thr Val Val Ser Ala Asp Leu Leu Ile 130 135 140 Asp Asp Arg Pro Asp Ile Thr Gly Ala Glu Pro Thr Pro Ser Trp Glu 145 150 155 160 His Val Leu Phe Thr Ala Cys His Asn Gln His Leu Gln Leu Gln Pro 165 170 175 Pro Arg Arg Arg Leu His Ser Trp Ala Asp Asp Trp Lys Ala Ile Leu 180 185 190 Asp Ser Lys Arg Pro Cys 195 <210> 32 <211> 25 <212> RNA <213> Artificial Sequence < 220> <223> siT001-1 sense <400> 32 cgagaagaag ucagauuaca ucaag 25 <210> 33 <211> 25 <212> RNA <213> Artificial Sequence <220> <223> siT001-1 antisense <400> 33 cuugauguaa ucugacuucu ucucg 25 <210> 34 <211> 25 <212> RNA <213> Artificial Sequence <220> <223> siT001-2 sense <400> 34 cgcaaauuca uugaaaccuu ccuga 25 <210> 35 <211> 25 <212> RNA < 213> Artificial Sequence <220> <223> siT001-2 antisense <400> 35 ucaggaaggu uucaaugaau uugcg 25 <210> 36 <211> 32 <212> DNA <213> Artificial Sequence <220> <223> Forward Primer for PCR < 400> 36 gtgcttctga cacaacagtc tcgaacttaa gc 32 <210> 37 <211> 32 <212> DNA <213> Artificial Sequence <220> <223> Reverse Primer for PCR <400> 37 gaagcggccg ccttcctact caggctttat tc 32 <210> 38 <2 11 > 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant, wild type <400> 38 atgttttcta ttaaagaacc attttcaatt gttacagact gcgatgaggt attaactgac 60 attagtcctt tatgggttca taagattcag caaaatgctg attattttgg a aaatacttt 120 gatttaagta aactagaagg attggaattt ggtacatttg aacattatca aacagtacta 180 tcacgaccag aatttcattt aaataaatgg ctaagaaaag agaatcttgt attatcagat 240 gaagaagaaa aagaattattt tgaaagattt tattcgttat atgataatga tgaattttat 300 gaagattgta tgccaactaa aatgtgtgaa ggaatttata aattatcatt acaaaaattc 360 g tagataaaa tctatgttgt aacaagaaca agtgaaggaa ccaaagaagg aaaaagaaaa 420 tttattgaaa ctttcttaaa ttctaataaa gtagagatta tttttgttgg gaaaaatgaa 480 aagaaatcag attatattaa gaatctaaag aatgtaaaaa tgattgtaga aga tgaatta 540 tcaaatatta atgatattgt agaaaattgt aatgatggtt ttgaagaagt agatacttat 600 attccatcaa ctggttataa caataaagat attgatggtt ttaatgaaaa tctaatgaaa 660 aaaggattta atgccgttcc atatgtaatt atagaaagac cagaaacaga agagaaagca 720 gtataa 726 <210> 39 <211> 726 <212> DNA <213> Artificial Sequence <220> <2 23> 5'-nucleotidase variant, T001 <400> 39 atgttctcca tcaaagaacc attctccatc gtgaccgact gcgatgaggt gctgaccgac 60 attagccctc tgtgggtgca taagattcag cagaacgctg attacttcgg caaatacttt 120 gatctgagca aactggaagg cctggaattt ggtacgtttg aacattatca gacggtgctg 180 tcacgcccag agttccatct gaacaaatgg ctgcgcaaag agaacctggt gctgtcagat 240 gaagaagaga aagaactgtt tgaacgcttc tattcgctgt atgataatga tgagttct ac 300 gaagattgta tgccaaccaa aatgtgcgaa ggcatctaca aactgtcact gcagaaattc 360 gtggataaaa tctatgtggt aacccgcacc agtgaaggca ccaaagaagg caacgcaaa 420 ttcattgaaa ccttcctgaa ctctaacaaa gtggagatta tcttcgt tgg caagaacgag 480 aagaagtcag attacatcaa gaacctgaag aatgtgaaaa tgattgtgga agatgaactg 540 tccaaca acgatattgt ggagaactgt aacgatggct ttgaagaagt ggatacctac 600 attccgtcaa ctggttacaa caacaaagat attgatggct tcaacgaaaa cctgatgaaa 660 aaaggcttca acgccgttcc atacgtgatt atcgaacgcc cagaaaccga agagaaagca 720 gtgtaa 726 <210> 40 <211> 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant , T002 <400> 40 atgttctcca tcaaggagcc gttctccatc gtgaccgact gcgacgaggt gctgaccgac 60 atcagcccgc tgtgggtgca caagatccag cagaacgccg actacttcgg caagtacttc 120 gacctgagca agctggaggg cctggagttc ggcacgt tcg agcactacca gacggtgctg 180 tcgcgcccgg agttccacct gaacaagtgg ctgcggaaag aaaacctcgt cctctccgac 240 gaggaggaga aggagctgtt cgagcgcttc tactcgctgt acgacaacga cgagttctac 300 gaggactgca tgccgaccaa gat gtgcgag ggcatctaca agctgtcgct gcagaagttc 360 gtggacaaga tctacgtggt cacccgcacg agcgagggca ccaaggaggg caagcgcaag 420 ttcatcgaga ccttcctgaa ctcgaacaag gtggagatca tcttcgtcgg caagaacgag 480 aagaagtcag actacatcaa gaacctgaag aacgtgaaga tgatcgtgga ggatgagctg 540 tccaacat ca acgacatcgt ggagaactgc aacgacggct tcgaggaggt ggacacctac 600 atcccgtcaa ctggttacaa caacaaggat atcgacggct tcaacgagaa cctgatgaag 660 aagggcttca acgccgtgcc atacgtgatc atcgagcgcc cagagaccga ggagaaggca 720 gtgtaa 7 26 <210> 41 <211> 726 <212> DNA <213> Artificial Sequence <220> <223> 5'-nucleotidase variant, T007 <400> 41 atgttcagca tcaaggagcc cttctccatc gtgaccgact gcgacgaagt cctcaccgac 60 atcagccccc tgtgggtgca caagatccag cagaacgccg actacttcgg ga aatacttc 120 gacctgagca agctggaggg cctggagttc gggaccttcg agcactacca gaccgtgctc 180 tcccgccccg aattccacct gaacaagtgg ctccgcaagg agaacctggt cctgtccgac 240 gaggaggaga aggagctgtt cgagcgcttc tacagcctgt acgacaacga cgagttctac 300 gaggactgca tgcccactaa gatgtgcgag ggcatctaca agctgagcct gcagaagttc 360 gtggataaga tctatgtcgt gaccc gcacc agcgagggca ccaaggaggg caagcgcaag 420 ttcatcgaga ctttcctcaa cagcaacaag gtggagatca tcttcgtcgg caagaacgaa 480 aaaaagagcg actacatcaa gaacctgaag aacgtgaaga tgatcgtgga ggacgagctg 540 tccaaca acga catcgt ggagaactgc aacgacggct tcgaggaggt ggacacctac 600 atccccagca ctgggtacaa caacaaggac atcgacggct tcaacgagaa cctgatgaag 660 aagggcttca acgccgtgcc ctacgtgatc atcgagcgcc ccgagaccga ggagaaggcc 720gtgtaa 726

Claims (13)

서열번호 3으로 표시되는 아미노산 서열을 암호화하는 암 치료를 위한 폴리뉴클레오티드.A polynucleotide for cancer treatment encoding the amino acid sequence represented by SEQ ID NO: 3. 제1항에 있어서,
상기 폴리뉴클레오티드는 서열번호 38 내지 서열번호 41로 이루어지는 군에서 선택되는 염기서열을 포함하는 것을 특징으로 하는 폴리뉴클레오티드.
According to claim 1,
The polynucleotide characterized in that it comprises a base sequence selected from the group consisting of SEQ ID NO: 38 to SEQ ID NO: 41.
제1항에 있어서,
상기 폴리뉴클레오티드는 mRNA인 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 1,
The polynucleotide for the treatment of cancer, characterized in that the polynucleotide is mRNA.
제3항에 있어서,
상기 mRNA는 암세포에 진입하여 핵산대사를 저해하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 3,
The polynucleotide for cancer treatment, characterized in that the mRNA enters cancer cells and inhibits nucleic acid metabolism.
제4항에 있어서,
상기 핵산대사는 dTTP 생합성 대사인 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 4,
The nucleic acid metabolism is a polynucleotide for cancer treatment, characterized in that dTTP biosynthetic metabolism.
제1항에 있어서,
상기 폴리뉴클레오티드는 서열번호 19로 표시되는 5'-UTR 및 서열번호 20으로 표시되는 3'-UTR을 더 포함하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 1,
The polynucleotide for the treatment of cancer, characterized in that the polynucleotide further comprises a 5'-UTR represented by SEQ ID NO: 19 and a 3'-UTR represented by SEQ ID NO: 20.
제1항에 있어서,
상기 폴리뉴클레오티드는 서열번호 22로 표시되는 핵 위치 신호(NLS)를 암호화하는 핵산 서열을 더 포함하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 1,
The polynucleotide for the treatment of cancer, characterized in that the polynucleotide further comprises a nucleic acid sequence encoding a nuclear localization signal (NLS) represented by SEQ ID NO: 22.
제1항에 있어서,
상기 폴리뉴클레오티드는 서열번호 23로 표시되는 미토콘드리아 위치 신호(MLS)를 암호화하는 핵산 서열을 더 포함하는 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 1,
The polynucleotide for the treatment of cancer, characterized in that the polynucleotide further comprises a nucleic acid sequence encoding the mitochondrial localization signal (MLS) represented by SEQ ID NO: 23.
제1항에 있어서,
상기 암은 대장암 또는 유방암인 것을 특징으로 하는 암 치료를 위한 폴리뉴클레오티드.
According to claim 1,
The polynucleotide for cancer treatment, characterized in that the cancer is colon cancer or breast cancer.
서열번호 3으로 표시되는 아미노산 서열을 암호화하는 폴리뉴클레오티드;
상기 폴리뉴클레오티드를 포집한 리포좀; 및
상기 리포좀에 컨쥬게이트되고, 암세포에서 과발현하는 CD47에 결합하는 바인더로서, 서열번호 1 또는 서열번호 2로 표시되는 아미노산 서열을 포함하는 바인더;
를 포함하는 암 치료를 위한 복합체.
A polynucleotide encoding the amino acid sequence represented by SEQ ID NO: 3;
liposomes encapsulating the polynucleotide; and
a binder conjugated to the liposome and binding to CD47 overexpressed in cancer cells, comprising an amino acid sequence represented by SEQ ID NO: 1 or SEQ ID NO: 2;
Complex for cancer treatment comprising a.
제10항에 있어서,
상기 바인더는 DSPE(1,2-Distearoyl-sn-glycero-3-phosphorylethanolamine)-컨쥬게이트된 것을 특징으로 하는 암 치료를 위한 복합체.
According to claim 10,
The binder is DSPE (1,2-Distearoyl-sn-glycero-3-phosphorylethanolamine)-conjugated complex for cancer treatment, characterized in that.
제10항에 있어서,
상기 리포좀은 양이온성 인지질, DOTAP 및 콜레스테롤로 이루어진 군에서 선택되는 1종 이상의 물질을 이용하여 양전하로 하전된 것을 특징으로 하는 암 치료를 위한 복합체.
According to claim 10,
The liposome is a complex for cancer treatment, characterized in that positively charged using at least one material selected from the group consisting of cationic phospholipids, DOTAP and cholesterol.
제1항에 있어서,
상기 리포좀은 페길화된 것을 특징으로 하는 암 치료를 위한 복합체.
According to claim 1,
The liposome is a complex for cancer treatment, characterized in that pegylated.
KR1020220001164A 2022-01-04 2022-01-04 Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein KR20230105583A (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
KR1020220001164A KR20230105583A (en) 2022-01-04 2022-01-04 Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein
PCT/KR2022/000131 WO2023132380A1 (en) 2022-01-04 2022-01-05 Polynucleotide for cancer treatment encoding 5'-nucleotidase modified protein

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020220001164A KR20230105583A (en) 2022-01-04 2022-01-04 Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein

Publications (1)

Publication Number Publication Date
KR20230105583A true KR20230105583A (en) 2023-07-11

Family

ID=87073761

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020220001164A KR20230105583A (en) 2022-01-04 2022-01-04 Polynucleotide for treatment cancer encoding 5'-nucleotidase modified protein

Country Status (2)

Country Link
KR (1) KR20230105583A (en)
WO (1) WO2023132380A1 (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT3331902T (en) * 2015-08-07 2021-07-26 Alx Oncology Inc Constructs having a sirp-alpha domain or variant thereof
CN110386984B (en) * 2018-04-17 2022-04-22 杭州尚健生物技术有限公司 Fusion protein combined with CD47 protein and application thereof

Also Published As

Publication number Publication date
WO2023132380A1 (en) 2023-07-13

Similar Documents

Publication Publication Date Title
JP6307163B2 (en) Administration of kynurenine-depleting enzyme for tumor therapy
Zhu et al. Aptamers entirely built from therapeutic nucleoside analogues for targeted cancer therapy
US9078927B2 (en) Self-assembling complex for targeting chemical agents to cells
Wang et al. Incorporation of histone derived recombinant protein for enhanced disassembly of core-membrane structured liposomal nanoparticles for efficient siRNA delivery
Hirase et al. Dodecaborate-encapsulated extracellular vesicles with modification of cell-penetrating peptides for enhancing macropinocytotic cellular uptake and biological activity in boron neutron capture therapy
EP4124621A1 (en) Carrier peptide fragment for nucleolar localization and use thereof
CN112442129B (en) Tumor enzyme response type recombinant pyroprotein drug delivery system and anti-tumor application thereof
Zhao et al. pH low insertion peptide mediated cell division cycle-associated protein 1-siRNA transportation for prostatic cancer therapy targeted to the tumor microenvironment
US20030216345A1 (en) Histone deacetylase inhibitor and use thereof
US20090011984A1 (en) Biotin-binding receptor molecules
CN102159709B (en) New therapeutic agent for malignant mesothelioma and immunostimulant
KR102557016B1 (en) CD47 binder and liposome complex for treatment cancer
KR102579284B1 (en) Liposome complex for treatment cancer comprising new CD47 binder and polynucleotide
KR102398032B1 (en) 5&#39;-nucleotidas variants for treatment cancer
KR20190040824A (en) Fusion protein for CRISP/Cas system and complex comprising the same and uses thereof
KR20230105583A (en) Polynucleotide for treatment cancer encoding 5&#39;-nucleotidase modified protein
CN114796525B (en) Use of inhibitors of cyclin-mediated proteins in tumor therapy
Cao et al. Intracellular localization and sustained prodrug cell killing activity of TAT-HSVTK fusion protein in hepatocelullar carcinoma cells
EP4332112A1 (en) Peptide fragment and use thereof
US20040067880A1 (en) Prodrug therapy of liver diseases using receptor-mediated delivery of malarial circumsporozoite protein as a carrier
JP2000210079A (en) Nucleic acid transporter and drug for gene therapy
KR20220120910A (en) EGFR-GST-Ulixertinib complex, and pharmaceutical composition for preventing or treating cancer comprising the same
Wang et al. A Tumor‐Specific Cascade‐Activating Smart Prodrug System for Enhanced Targeted Therapy
CN116555185A (en) CAR-macrophage targeting GPC3 to block immunosuppression signals and preparation method and application thereof
KR20230161894A (en) HER2-GST-SN-38 complex and pharmaceutical composition for preventing or treating proliferative disease comprising the same