JP7437648B2 - Molecular targets and their utilization for LATS2 mutant diseases - Google Patents

Molecular targets and their utilization for LATS2 mutant diseases Download PDF

Info

Publication number
JP7437648B2
JP7437648B2 JP2019191790A JP2019191790A JP7437648B2 JP 7437648 B2 JP7437648 B2 JP 7437648B2 JP 2019191790 A JP2019191790 A JP 2019191790A JP 2019191790 A JP2019191790 A JP 2019191790A JP 7437648 B2 JP7437648 B2 JP 7437648B2
Authority
JP
Japan
Prior art keywords
cells
smg6
lats2
cancer
gene
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
JP2019191790A
Other languages
Japanese (ja)
Other versions
JP2021065121A (en
Inventor
優子 村上
浩也 鈴木
孝 三井田
好孝 関戸
智美 向井
良多 山岸
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aichi Prefecture
Juntendo University
Original Assignee
Aichi Prefecture
Juntendo University
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 Aichi Prefecture, Juntendo University filed Critical Aichi Prefecture
Priority to JP2019191790A priority Critical patent/JP7437648B2/en
Publication of JP2021065121A publication Critical patent/JP2021065121A/en
Application granted granted Critical
Publication of JP7437648B2 publication Critical patent/JP7437648B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

特許法第30条第2項適用 2018年12月17日にウェブサイト(https://kaken.nii.ac.jp/ja/report/KAKENHI-PROJECT-17K18436/17K184362017hokoku/)において公開された実施状況報告書で公表Application of Article 30, Paragraph 2 of the Patent Act Implementation status published on the website (https://kaken.nii.ac.jp/ja/report/KAKENHI-PROJECT-17K18436/17K184362017hokoku/) on December 17, 2018 Published in report

特許法第30条第2項適用 2019年6月13日に大阪国際交流センターで開催された第23回日本がん分子標的治療学会学術集会で発表Application of Article 30, Paragraph 2 of the Patent Act Presented at the 23rd Academic Meeting of the Japanese Society for Molecular Targeted Cancer Therapy held at the Osaka International Exchange Center on June 13, 2019

本発明は、LATS2変異疾患のための分子標的及びその利用に関する。 The present invention relates to molecular targets and their utilization for LATS2 mutation diseases.

LATS2遺伝子は、悪性中皮腫、前立腺がん、非小細胞肺がん等のがん原因遺伝子として知られている。悪性中皮腫ではその約25%においてLATS2遺伝子の不活性化変異がみられる(非特許文献1)。LATS2遺伝子を含めて悪性中皮腫における遺伝子変異はがん抑制遺伝子変異が多いため、原因遺伝子を直接の標的とする悪性中皮腫の分子標的治療は困難とされている。 The LATS2 gene is known as a gene that causes cancers such as malignant mesothelioma, prostate cancer, and non-small cell lung cancer. Inactivating mutations of the LATS2 gene are observed in about 25% of malignant mesotheliomas (Non-Patent Document 1). Many of the genetic mutations in malignant mesothelioma, including the LATS2 gene, are tumor suppressor gene mutations, making it difficult to conduct molecular targeted therapy for malignant mesothelioma that directly targets the causative gene.

一方、近年、がん治療の新しい方法として「合成致死」の概念に基づいた治療法が提案されている。「合成致死」とは、2つの遺伝子の発現抑制によって始めて誘導される細胞死であり、遺伝子の不活性化変異を持つがんにおいてもがん細胞を特異的に死滅させる分子標的薬の開発が可能となる。
これまでに、悪性中皮腫の原因遺伝子に対して合成致死表現型を示す遺伝子の探索が行われており、例えば、NF2(Neurofibromatosis type 2)変異とFAK(focal adhesion kinase)阻害剤、BAP1(BRCA1 associated protein 1)変異とclass1 HDAC(Histone deacetylases)阻害剤の報告がある(非特許文献2,3)。しかしながら、LATS2遺伝子に対して合成致死表現型を示す合成致死パートナー遺伝子は得られていない。
On the other hand, in recent years, a new method of cancer treatment based on the concept of "synthetic lethality" has been proposed. "Synthetic lethality" is cell death that is first induced by suppressing the expression of two genes, and the development of molecular targeted drugs that specifically kill cancer cells even in cancers with inactivating mutations of genes. It becomes possible.
To date, searches have been made for genes that exhibit synthetic lethal phenotypes in relation to genes that cause malignant mesothelioma. For example, NF2 (Neurofibromatosis type 2) mutations, FAK (focal adhesion kinase) inhibitors, There are reports of BRCA1 associated protein 1) mutations and class 1 HDAC (Histone deacetylases) inhibitors (Non-patent Documents 2 and 3). However, a synthetic lethal partner gene that exhibits a synthetic lethal phenotype for the LATS2 gene has not been obtained.

H Murakami, T Mizuno, T Taniguchi, M Fujii, F Ishiguro, T Fukui, S Akatsuka, Y Horio, T Hida, Y Kondo, S Toyokuni, H Osada, and Y Sekido. “LATS2 Is a Tumor Suppressor Gene of Malignant Mesothelioma. Cancer Research”,2011, 71(3):873-83H Murakami, T Mizuno, T Taniguchi, M Fujii, F Ishiguro, T Fukui, S Akatsuka, Y Horio, T Hida, Y Kondo, S Toyokuni, H Osada, and Y Sekido. “LATS2 Is a Tumor Suppressor Gene of Malignant Mesothelioma . Cancer Research”, 2011, 71(3):873-83 NR. Shah, I Tancioni, KK. Ward, C Lawson, XL. Chen, C Jean, FJ. Sulzmaier, S Uryu, NL.G. Miller, DC.Connolly, and DD. Schlaepfer. “Analyses of merlin/NF2 connection to FAK inhibitorresponsiveness in serous ovarian cancer. Gynecologic oncology”,2015, 134(1):104-111NR. Shah, I Tancioni, KK. Ward, C Lawson, XL. Chen, C Jean, FJ. Sulzmaier, S Uryu, NL.G. Miller, DC.Connolly, and DD. Schlaepfer. “Analyses of merlin/NF2 connection to FAK inhibitor responsiveness in serous ovarian cancer. Gynecologic oncology”, 2015, 134(1):104-111 JJ. Sacco, J Kenyani, Z Butt, R Carter, HY Chew, LP. Cheeseman, S Darling, M Denny, S Urbe, MJ. Clague, JM. Coulson. “Loss of the deubiquitylase BAP1 alters class I histone deacetylase expression and sensitivity of mesothelioma cells to HDAC inhibitors. Oncotraget”, 2015,6(15):13757-13771JJ. Sacco, J Kenyani, Z Butt, R Carter, HY Chew, LP. Cheeseman, S Darling, M Denny, S Urbe, MJ. Clague, JM. Coulson. “Loss of the deubiquitylase BAP1 alters class I histone deacetylase expression and sensitivity of mesothelioma cells to HDAC inhibitors. Oncotraget”, 2015, 6(15):13757-13771

本発明の課題は、LATS2遺伝子と合成致死表現型を示す遺伝子を同定し、LATS2変異疾患の治療標的として利用することにある。 The objective of the present invention is to identify the LATS2 gene and a gene that exhibits a synthetic lethal phenotype, and to utilize this gene as a therapeutic target for LATS2 mutant diseases.

本発明者は、悪性中皮腫をモデルとして利用し、候補遺伝子群についてsiRNAによるノックダウン法を用いて個々に候補遺伝子の致死性を確認し、合成致死遺伝子の確定を進めた結果、LATS1/2とmRNAエンドヌクレアーゼの一種であるSMG6を共発現抑制することにより合成致死表現型が誘導されることが確認され、LATS2とSMG6が合成致死の関係にあることを見出した。 Using malignant mesothelioma as a model, the present inventor confirmed the lethality of each candidate gene using siRNA-based knockdown method and determined the synthetic lethal gene.As a result, LATS1/ It was confirmed that a synthetic lethal phenotype was induced by co-expressing SMG6, a type of mRNA endonuclease, with LATS2, and it was found that LATS2 and SMG6 are in a synthetic lethal relationship.

かかる知見に基づいて、本発明は、以下の〔1〕~〔3〕を提供するものである。
〔1〕がん患者由来の生体試料を用いてLATS2遺伝子の不活性化変異の有無を検出すること、当該変異を有するがん患者を、SMG6を阻害する化合物による治療対象者として選別することを含む、SMG6を阻害する化合物による治療対象者を選別する方法。
〔2〕SMG6の阻害を指標として被験化合物を評価することを含む、LATS2変異疾患のための薬剤のスクリーニング方法。
〔3〕SMG6を阻害する化合物を有効成分として含む、LATS2変異疾患のための薬剤。
Based on this knowledge, the present invention provides the following [1] to [3].
[1] Detecting the presence or absence of an inactivating mutation in the LATS2 gene using biological samples derived from cancer patients, and selecting cancer patients who have this mutation as candidates for treatment with compounds that inhibit SMG6. A method for selecting patients for treatment with a compound that inhibits SMG6, including:
[2] A method for screening drugs for LATS2 mutation diseases, which includes evaluating a test compound using SMG6 inhibition as an index.
[3] A drug for LATS2 mutation disease containing a compound that inhibits SMG6 as an active ingredient.

本発明により、SMG6がLATS2の合成致死標的として明らかになった。SMG6を阻害する治療は、LATS2遺伝子の不活性化変異を持つがん細胞を特異的に死滅させることができるので、LATS2変異疾患に対して、副作用が少なく、効果の高い治療が期待される。 The present invention reveals SMG6 as a synthetic lethal target of LATS2. Treatments that inhibit SMG6 can specifically kill cancer cells that have inactivating mutations in the LATS2 gene, and are therefore expected to be highly effective treatments for LATS2 mutation diseases with few side effects.

HOMC-D4由来LATS1/2ノックダウン細胞、MeT-5A由来LATS1 ノックアウト細胞及びLATS2 ノックアウト細胞にSMG6を発現抑制し、WST assayによる細胞生存率を測定した結果を示す。The results of suppressing SMG6 expression in HOMC-D4-derived LATS1/2 knockdown cells, MeT-5A-derived LATS1 knockdown cells, and LATS2 knockdown cells and measuring cell survival rate by WST assay are shown. (a)HOMC-D4由来LATS1/2ノックダウン細胞にTERT阻害剤(Trichostatin、Doxorubicin、BIBR1532)を反応させ、WST assayによる細胞生存率及びLD50の結果を示す。(a) HOMC-D4-derived LATS1/2 knockdown cells were reacted with TERT inhibitors (Trichostatin, Doxorubicin, BIBR1532), and the results of cell viability and LD50 by WST assay are shown. (b)MeT-5A由来LATS1 ノックアウト細胞及びLATS2 ノックアウト細胞にTERT阻害剤(Trichostatin、Doxorubicin)を反応させ、WST assayによる細胞生存率及びLD50の結果を示す。(b) MeT-5A-derived LATS1 knockout cells and LATS2 knockout cells were reacted with TERT inhibitors (Trichostatin, Doxorubicin), and the results of cell viability and LD50 by WST assay are shown. LATS1/2ノックダウン細胞にSMG6を発現抑制し、細胞をTUNEL染色後、FACSを用いて解析した結果を示す。The results of suppressing SMG6 expression in LATS1/2 knockdown cells, staining the cells with TUNEL, and analyzing the cells using FACS are shown. (a)HOMC-D4由来LATS1/2ノックダウン細胞、MeT-5A由来LATS1 ノックアウト細胞及びLATS2 ノックアウト細胞にSMG6を発現抑制し、day6における細胞を冷メタノールで固定後、γH2A.X抗体、核染色像を示す。(a) SMG6 expression was suppressed in HOMC-D4-derived LATS1/2 knockdown cells, MeT-5A-derived LATS1 knockdown cells, and LATS2 knockdown cells, and after fixing the cells with cold methanol on day 6, γH2A.X antibody, nuclear staining image shows. (b)視野内における全核数に対するγH2A.X陽性数の割合を比較解析した結果を示す。(b) Shows the results of a comparative analysis of the ratio of the number of positive γH2A.X to the total number of nuclei in the field of view.

本明細書において、「LATS2変異疾患」は、LATS2遺伝子の不活性化変異を有する疾患であり、例えば、悪性中皮腫、前立腺がん、非小細胞肺がん等が挙げられる。LATS2遺伝子の不活性化変異は、ナンセンス変異、遺伝子の部分的欠失等より生じるが、本明細書において、LATS2遺伝子を不活性化させる限り特に制限されない。
「LATS2」は、LATS腫瘍抑制因子ファミリーに属するセリントレオニンタンパク質キナーゼである。
As used herein, "LATS2 mutant disease" refers to a disease that has an inactivating mutation in the LATS2 gene, and includes, for example, malignant mesothelioma, prostate cancer, non-small cell lung cancer, and the like. Inactivating mutations of the LATS2 gene are caused by nonsense mutations, partial gene deletions, etc., but are not particularly limited herein as long as they inactivate the LATS2 gene.
"LATS2" is a serine threonine protein kinase that belongs to the LATS tumor suppressor family.

本明細書において、「SMG6」は、EST1A (ever-shorter telomeres 1A)をエンコードする17番染色体のSMG6遺伝子のことである。SMG6は、全身の器官に発現し、異常なmRNAを排除するmRNAエンドヌクレーゼの機能とTERT (telomere reverse transcriptase)と協働し、テロメア伸長を制御する機能を併せ持つ(AS. Venteicher, EB. Abreu, Z Meng, KE. McCann, RM. Terns, TD. Veenstra, MP. Terns, SE. Artandi. “A Human Telomerase Holoenzyme Protein Required for Cajal Body Localization and Telomere Synthesis. Science”, 2009,323(5914):644-8、T Fatscher, V Boehm, NH. Gehring. “Mechanism, factors, and physiological role of nonsense-mediated mRNA decay. Cellular and Molecular Life Sciences”, 2015,72(23):4523-44) As used herein, "SMG6" refers to the SMG6 gene on chromosome 17, which encodes EST1A (ever-shorter telomeres 1A). SMG6 is expressed in organs throughout the body and has both the function of mRNA endonuclease to eliminate abnormal mRNA and the function of controlling telomere elongation by cooperating with TERT (telomere reverse transcriptase) (AS. Venteicher, EB. Abreu , Z Meng, KE. McCann, RM. Terns, TD. Veenstra, MP. Terns, SE. Artandi. “A Human Telomerase Holoenzyme Protein Required for Cajal Body Localization and Telomere Synthesis. Science”, 2009, 323(5914):644 -8, T Fatscher, V Boehm, NH. Gehring. “Mechanism, factors, and physiological role of nonsense-mediated mRNA decay. Cellular and Molecular Life Sciences”, 2015, 72(23):4523-44)

本明細書において、「TERT」は、テロメア逆転写酵素のことであり、TERTの活性化に伴いテロメア長を伸長させ、ひいては体細胞の老化及び寿命を司る酵素のことである。TERTを人為的に阻害することでDNA損傷を伴うアポトーシスを誘導することが知られている(X Ling, W Yang, P Zou, G Zhang, Z Wang, X Zhang, H Chen, K Peng, F Han, J Liu, J Cao, L Ao. “TERT regulates telomere-related senescence and apoptosis through DNA damage response in male germ cells exposed to BPDE in vitro and to B[a]P in vivo. Environmental Pollution”, 2018,235:836-849) As used herein, "TERT" refers to telomere reverse transcriptase, which is an enzyme that increases the length of telomeres upon activation of TERT and, in turn, controls the aging and lifespan of somatic cells. Artificial inhibition of TERT is known to induce apoptosis accompanied by DNA damage (X Ling, W Yang, P Zou, G Zhang, Z Wang, X Zhang, H Chen, K Peng, F Han , J Liu, J Cao, L Ao. “TERT regulates telomere-related senescence and apoptosis through DNA damage response in male germ cells exposed to BPDE in vitro and to B[a]P in vivo. Environmental Pollution”, 2018, 235: 836-849)

本明細書において、「がん患者」は、がんに罹患している或いは罹患の疑いのあるヒトであり、「がん」は、悪性腫瘍(悪性新生物)全般を指す。 As used herein, a "cancer patient" is a person who is suffering from cancer or is suspected of having cancer, and "cancer" refers to malignant tumors (malignant neoplasms) in general.

本明細書において、「生体試料」は、ヒトから得られゲノムDNAを利用可能な試料であり、例えば、組織、全血、血漿、血清、リンパ球、リンパ液、血小板、単核球、顆粒球、唾液、尿などが挙げられる。 As used herein, a "biological sample" is a sample obtained from a human and for which genomic DNA can be used, such as tissue, whole blood, plasma, serum, lymphocytes, lymph fluid, platelets, mononuclear cells, granulocytes, Examples include saliva and urine.

(治療対象者を選別する方法)
本発明のSMG6を阻害する化合物による治療対象者を選別する方法は、がん患者由来の生体試料を用いてLATS2遺伝子の不活性化変異の有無を検出すること、当該変異を有するがん患者を、SMG6を阻害する化合物による治療対象者として選別すること、を含む。
LATS2遺伝子の不活性化変異が検出された場合、当該変異を有するがん患者は、SMG6を阻害する化合物による治療に応答性があると判断することができ、SMG6を阻害する化合物による治療対象者として選別すれば、LATS2とSMG6の合成致死の効果により治療成績の向上が期待される。
LATS2遺伝子の不活性化変異は、悪性中皮腫、前立腺がん、非小細胞肺がんにおいて認められることから、がん患者は当該悪性中皮腫、前立腺がん、非小細胞肺がんのがん患者であることが好ましい。
(Method of selecting treatment candidates)
The method for selecting patients for treatment with the compound that inhibits SMG6 of the present invention involves detecting the presence or absence of an inactivating mutation in the LATS2 gene using a biological sample derived from a cancer patient, and identifying cancer patients who have the mutation. , selecting patients as candidates for treatment with a compound that inhibits SMG6.
If an inactivating mutation in the LATS2 gene is detected, cancer patients with the mutation can be judged to be responsive to treatment with compounds that inhibit SMG6, and patients who are candidates for treatment with compounds that inhibit SMG6 can be judged to be responsive to treatment with compounds that inhibit SMG6. If selected as such, it is expected that the therapeutic results will improve due to the synthetic lethal effect of LATS2 and SMG6.
Inactivating mutations in the LATS2 gene are observed in malignant mesothelioma, prostate cancer, and non-small cell lung cancer, so cancer patients are patients with malignant mesothelioma, prostate cancer, and non-small cell lung cancer. It is preferable that

LATS2遺伝子の不活性化変異の検出は、特に限定されず、変異を検出できる公知の手法、例えば、直接シークエンス法、RT-PCR 法、PCR- RFLP法、PCR-SSCP法、インベーダー法、TaqMan Genotyping法、マイクロアレイ法、次世代シークエンス法等により行うことができる。 Detection of inactivating mutations in the LATS2 gene is not particularly limited, and known methods capable of detecting mutations may be used, such as direct sequencing, RT-PCR, PCR-RFLP, PCR-SSCP, Invader method, TaqMan Genotyping. This can be carried out by methods such as microarray method, microarray method, and next generation sequencing method.

SMG6を阻害する化合物は、天然に存在する物質であっても、化学的又は生物学的方法等で人工的に合成した物質であってもよく、また、組成物や混合物であってもよい。後述するスクリーニング法により同定される化合物であってもよい。
本明細書において、SMG6の阻害は、SMG6の活性の阻害、SMG6の発現の阻害の両方を含む。LATS2とSMG6の関係では、SMG6の活性変異体を用いた解析により、SMG6のヌクレアーゼ活性の変異がLATS2との合成致死性を示すのに必要であることが示唆されたため、SMG6の阻害は、好ましくはSMG6のヌクレアーゼ活性の阻害である。SMG6のヌクレアーゼ活性は、濃度既知のRNAをサンプルと37℃条件下で1時間反応させ、Bioanalyzerを用いてRNAのRIN値(RNA integrity number)を比較することで検討可能である。
The compound that inhibits SMG6 may be a naturally occurring substance, a substance artificially synthesized by a chemical or biological method, or a composition or a mixture. It may also be a compound identified by the screening method described below.
As used herein, inhibition of SMG6 includes both inhibition of SMG6 activity and inhibition of SMG6 expression. Regarding the relationship between LATS2 and SMG6, inhibition of SMG6 is preferable because analysis using active mutants of SMG6 suggested that mutations in the nuclease activity of SMG6 are required to exhibit synthetic lethality with LATS2. is an inhibition of the nuclease activity of SMG6. The nuclease activity of SMG6 can be examined by reacting RNA of known concentration with a sample for 1 hour at 37°C, and comparing the RNA RIN values (RNA integrity numbers) using a Bioanalyzer.

(薬剤のスクリーニング方法)
本発明のLATS2変異疾患のための薬剤のスクリーニング方法は、SMG6の阻害を指標として被験化合物を評価することを含む、方法である。
被験化合物は、特に制限されず、天然に存在する物質であっても、化学的又は生物学的方法等で人工的に合成した物質であってもよく、また、組成物や混合物であってもよい。
評価の指標となるSMG6の阻害は、上述したとおり、SMG6の活性の阻害であっても、SMG6の発現の阻害であってもよい。
(Drug screening method)
The method of screening for drugs for LATS2 mutation diseases of the present invention is a method that includes evaluating a test compound using SMG6 inhibition as an indicator.
The test compound is not particularly limited, and may be a naturally occurring substance, a substance artificially synthesized by chemical or biological methods, or a composition or mixture. good.
Inhibition of SMG6, which serves as an evaluation index, may be inhibition of SMG6 activity or expression of SMG6, as described above.

スクリーニングは、例えば、上記SMG6のヌクレアーゼ活性の検出系に被験化合物を作用させ、SMG6のヌクレアーゼ活性を検出し、SMG6のヌクレアーゼ活性を低下させる被験化合物をLATS2変異疾患のための薬剤として評価することによって行うことができる。被験化合物の評価は、例えば、被験化合物添加群と被験化合物非添加群もしくは対照物質添加群とを比較して行えばよい。 Screening can be carried out, for example, by causing a test compound to act on the SMG6 nuclease activity detection system, detecting SMG6 nuclease activity, and evaluating the test compound that reduces SMG6 nuclease activity as a drug for LATS2 mutant disease. It can be carried out. Evaluation of the test compound may be performed, for example, by comparing a group to which the test compound has been added and a group to which the test compound has not been added or a group to which a control substance has been added.

また、スクリーニングは、例えば、SMG6発現細胞に被験化合物を接触させること、当該細胞におけるSMG6の活性又は発現を検出すること、当該検出の結果に基づいて、SMG6の活性又は発現を阻害する被験化合物をLATS2変異疾患のための薬剤として評価することによって行うことができる。被験化合物の評価は、例えば、被験化合物の添加前後で、又は被験化合物添加群と被験化合物非添加群もしくは対照物質添加群とを比較して行えばよい。 In addition, screening includes, for example, contacting SMG6-expressing cells with a test compound, detecting the activity or expression of SMG6 in the cells, and based on the result of the detection, selecting a test compound that inhibits the activity or expression of SMG6. This can be done by evaluating it as a drug for LATS2 mutant diseases. Evaluation of the test compound may be performed, for example, before and after addition of the test compound, or by comparing a group to which the test compound has been added and a group to which no test compound has been added or a group to which a control substance has been added.

ここで、SMG6発現細胞としては、生来的にSMG6遺伝子を有し、これを発現する能力のある細胞や外来的にSMG6遺伝子を発現可能に導入された細胞が挙げられる。これらは、生体から採取された細胞、生体から採取された組織や器官に含まれる細胞、培養細胞であってよい。好ましくは、ヒト由来の細胞である。
外来的にSMG6遺伝子を発現可能にした細胞は、公知の手法を用いて、例えば、SMG6遺伝子を組み込んだ発現ベクターを任意の哺乳動物細胞に導入し、当該細胞を形質転換させることによって得ることができる。
SMG6発現細胞への被験化合物の接触は、被験化合物の細胞培養培地への添加や細胞への直接的な添加(例えば、滴下、塗布、散布、噴霧、パッチ等)が挙げられる。被験化合物の接触量や条件は、適宜設定すればよい。
Here, examples of SMG6-expressing cells include cells that naturally have the SMG6 gene and have the ability to express it, and cells that have been exogenously introduced to be able to express the SMG6 gene. These may be cells collected from a living body, cells contained in tissues or organs collected from a living body, or cultured cells. Preferably, cells are derived from humans.
Cells capable of exogenously expressing the SMG6 gene can be obtained using known techniques, for example, by introducing an expression vector incorporating the SMG6 gene into any mammalian cell and transforming the cell. can.
Examples of contacting the test compound to SMG6-expressing cells include addition of the test compound to the cell culture medium or direct addition to the cells (eg, dropping, coating, spraying, spraying, patching, etc.). The amount of contact with the test compound and the conditions may be set as appropriate.

SMG6の活性又は発現の検出は、mRNAレベルで検出する場合は、例えば、ノーザンブロット法、RT-PCR法、リアルタイムRT-PCR法、RNaseプロテクションアッセイ法等により行うことができる。
SMG6タンパク質発現を検出する場合は、例えば、SDS-PAGE、クロマトグラフィー法、免疫学的測定法(例えば、免疫組織化学、ELISA、ウェスタンブロット、免疫沈降等)、比色定量法、質量分析等により行うことができる。
SMG6遺伝子のプロモーターの活性を検出する場合は、レポーター遺伝子を用いたプロモーター活性や転写活性の蛍光・光学的測定(レポーターアッセイ)等により行うことができる。
SMG6タンパク質の活性を検出する場合は、SMG6タンパク質に対する結合基質の結合量を測定すること等により行うことができる。
When detecting the activity or expression of SMG6 at the mRNA level, it can be performed, for example, by Northern blotting, RT-PCR, real-time RT-PCR, RNase protection assay, or the like.
When detecting SMG6 protein expression, for example, SDS-PAGE, chromatography, immunoassay (e.g., immunohistochemistry, ELISA, Western blot, immunoprecipitation, etc.), colorimetry, mass spectrometry, etc. It can be carried out.
When detecting the activity of the promoter of the SMG6 gene, it can be carried out by fluorescence/optical measurement of promoter activity or transcriptional activity (reporter assay) using a reporter gene.
When detecting the activity of SMG6 protein, it can be carried out by measuring the amount of binding substrate bound to SMG6 protein.

かくして、SMG6を阻害する被験化合物は、LATS2変異疾患のための薬剤として評価され、LATS2変異疾患の治療のために用いることができる。SMG6を阻害する被験化合物は、LATS2とSMG6の合成致死の効果により、LATS2遺伝子の不活性化変異を持つがん細胞を特異的に死滅させる分子標的薬となり得る。 Thus, test compounds that inhibit SMG6 can be evaluated as drugs for LATS2 mutant diseases and used for the treatment of LATS2 mutant diseases. A test compound that inhibits SMG6 can serve as a molecular targeting drug that specifically kills cancer cells with inactivating mutations in the LATS2 gene, due to the synthetic lethal effect of LATS2 and SMG6.

(LATS2変異疾患のための薬剤)
LATS2変異疾患のための薬剤は、SMG6を阻害する化合物を有効成分として含み、当該化合物は、公知の化合物であっても、上記のスクリーニング法により同定された化合物であってもよい。SMG6の阻害は、上述したとおり、SMG6の活性の阻害であっても、SMG6の発現の阻害であってもよい。
LATS2変異疾患のための薬剤の形態は、任意であり、例えば、錠剤、カプセル剤、顆粒剤、散剤、トローチ剤、シロップ剤等による経口投与;注射剤、坐剤、吸入薬、経皮吸収剤、外用剤等による非経口投与が挙げられ、いずれであってもよい。
当該薬剤には、SMG6を阻害する化合物の他、賦形剤、結合剤、崩壊剤、滑沢剤、溶剤、溶解補助剤、懸濁化剤、等張化剤、pH調節剤・緩衝剤、防腐剤、抗酸化剤、着色剤、矯味・矯臭剤、安定化剤等、薬学的に許容される担体を適宜配合して用いることができる。
薬剤を生体に投与する場合、その用量や用法は、投与対象者の状態、体重、性別、年齢又はその他の要因に従って変動し得る。
(Drug for LATS2 mutation disease)
The drug for LATS2 mutation disease contains a compound that inhibits SMG6 as an active ingredient, and the compound may be a known compound or a compound identified by the above screening method. As described above, inhibition of SMG6 may be inhibition of SMG6 activity or expression of SMG6.
The form of the drug for LATS2 mutation disease is arbitrary, for example, oral administration in the form of tablets, capsules, granules, powders, troches, syrups, etc.; injections, suppositories, inhalants, transdermal absorption drugs, etc. , parenteral administration using external preparations, etc., and any of them may be used.
In addition to the compound that inhibits SMG6, the drug includes excipients, binders, disintegrants, lubricants, solvents, solubilizing agents, suspending agents, isotonic agents, pH regulators/buffers, Pharmaceutically acceptable carriers such as preservatives, antioxidants, coloring agents, flavoring/fragrant agents, and stabilizers can be appropriately blended and used.
When administering a drug to a living body, the dose and usage may vary depending on the condition, weight, sex, age, or other factors of the subject.

次に実施例を挙げて本発明を更に詳細に説明する。 Next, the present invention will be explained in more detail with reference to Examples.

実施例1 LATS2、SMG6共発現抑制における合成致死表現型の誘導
1. 細胞
LATS1及びLATS2を発現抑制するため、ヒト正常中皮由来の細胞株(HOMC-D4、T Kakiuchi, T Takahara, Y Kasugai, K Arita, N Yoshida, K Karube, M Suguro, K Matsuo, H Nakanishi, T Kiyono, S Nakamura, H Osada, Y Sekido, M Seto and S Tsuzuki. “Modeling mesothelioma utilizing human mesothelial cells reveals involvement of phospholipase-C beta 4 in YAP-active mesothelioma cell proliferation”,2016, Carcinogenesis, 37(11): 1098-1109)にレンチウィルスにより細胞内にshRNAを遺伝子導入した。使用したshRNA (shNT) はSIGMA (MISSION pLKO.1-puro Non-Mammalian shRNA(SHC002))であり、shLATS1/2はpLKO.1 puroに以下の表1に記載の配列のshRNAを入れたものである。
Example 1 Induction of synthetic lethal phenotype by suppressing co-expression of LATS2 and SMG6
1. Cell
In order to suppress the expression of LATS1 and LATS2, a cell line derived from human normal mesothelium (HOMC-D4, T Kakiuchi, T Takahara, Y Kasugai, K Arita, N Yoshida, K Karube, M Suguro, K Matsuo, H Nakanishi, T Kiyono, S Nakamura, H Osada, Y Sekido, M Seto and S Tsuzuki. “Modeling mesothelioma utilizing human mesothelial cells reveals involvement of phospholipase-C beta 4 in YAP-active mesothelioma cell proliferation”, 2016, Carcinogenesis, 37(11): 1098-1109), shRNA was introduced into cells using lentivirus. The shRNA (shNT) used was SIGMA (MISSION pLKO.1-puro Non-Mammalian shRNA (SHC002)), and shLATS1/2 was pLKO.1 puro containing shRNA with the sequence listed in Table 1 below. be.

Figure 0007437648000001
Figure 0007437648000001

2. LATS1及び2発現抑制細胞株
HOMC-D4細胞にshRNAを導入する手法として293T細胞に対するレンチウィルスベクターシステムにより、shRNAを封入したレンチウィルス(lenti-shRNA)を作製した。具体的には、293T細胞を6×104細胞/cm2になるように播種し、ダルベッコ改変イーグル培地:DMEM(Wako:カタログ番号04429-675)にFetal Bovine Serum:FBS (GIBCO:カタログ番号10270)を10%になるよう添加し(以下10%FBS-DMEM)、10%FBS-DMEM中で24時間培養した。8μg/ml psPAX2、2.8μg/ml pMD2.G、10μg/ml shRNAになるよう270μl OPTIMEMに混合した(1)。270μg/ml OPTIMEMにPEI-Maxを2mg/mlになるように混合した(2)。前記(2)に(1)を添加し、転倒混和した後室温で20分間静置し、レンチウィルス導入混合液とした。10%FBS-DMEMを完全に除去し、OPTIMEMを0.08ml/cm2になるように加え、レンチウィルス導入混合液を添加し、37℃で4時間培養した。培養後10%FBS-DMEMに培地交換し37℃で48時間培養した。48時間後、培養液を回収し0.22μmのMillEX-GV (Millipore:カタログ番号SLGV033RS)を通過させレンチウィルス (lenti-shNT、lenti-shLATS1/2)とした。HOMC-D4にlenti-shNT、lenti-shLATS1/2を感染させ、LATS1/2をノックダウン (KD:Knock down)した。その後、ピューロマイシン、ブラストサイジン16μg/mlで細胞をセレクションし、非単一安定細胞株 (NT細胞、LATS1/2 KD細胞)を樹立した。
2. LATS1 and 2 expression suppressed cell line
As a method for introducing shRNA into HOMC-D4 cells, we created lentivirus encapsulated with shRNA (lenti-shRNA) using a lentivirus vector system for 293T cells. Specifically, 293T cells were seeded at 6 × 10 4 cells/cm 2 in Dulbecco's modified Eagle's medium: DMEM (Wako: catalog number 04429-675) and Fetal Bovine Serum: FBS (GIBCO: catalog number 10270). ) was added to the cells at a concentration of 10% (hereinafter referred to as 10% FBS-DMEM), and cultured in 10% FBS-DMEM for 24 hours. 8 μg/ml psPAX2, 2.8 μg/ml pMD2.G, and 10 μg/ml shRNA were mixed in 270 μl OPTIMEM (1). PEI-Max was mixed with 270 μg/ml OPTIMEM at a concentration of 2 mg/ml (2). (1) was added to (2) above, mixed by inversion, and then allowed to stand at room temperature for 20 minutes to obtain a lentivirus introduction mixture. The 10% FBS-DMEM was completely removed, OPTIMEM was added at a concentration of 0.08 ml/cm 2 , the lentivirus introduction mixture was added, and the mixture was cultured at 37°C for 4 hours. After culturing, the medium was replaced with 10% FBS-DMEM and cultured at 37°C for 48 hours. After 48 hours, the culture solution was collected and passed through 0.22 μm MillEX-GV (Millipore: catalog number SLGV033RS) to obtain lentiviruses (lenti-shNT, lenti-shLATS1/2). HOMC-D4 was infected with lenti-shNT and lenti-shLATS1/2, and LATS1/2 was knocked down (KD: Knock down). Thereafter, cells were selected with puromycin and blasticidin at 16 μg/ml to establish non-single stable cell lines (NT cells, LATS1/2 KD cells).

3. siRNA遺伝子導入
SMG6の発現を抑制するため、リポフェクタミンを用い細胞内に以下の表2のsiRNAを遺伝子導入した。
3. siRNA gene transfer
In order to suppress the expression of SMG6, the siRNA shown in Table 2 below was introduced into the cells using Lipofectamine.

Figure 0007437648000002
Figure 0007437648000002

使用したsiRNA (siSMG6-1:GCCAGUGAUACAGCGAAUU、siSMG6-2:ACACCAACGGCUUCAUUGA、siNegative Control:CGUACGCGGAAUACUUCGA)は、ニッポンジーンのsiRNA合成サービスを利用し作製した。作製したsiRNAは、細胞導入直前に4% OPTIMEM、1.34% RNAiMax (Invitrogen:カタログ番号13778030)、2nM siRNAになるように混合し、15分間室温でインキュベートした。その後、RPMI (Wako:カタログ番号189-02025)にFBSを10%になるよう添加(以下、10%FBS-RPMIとする)したものを加え、ノックダウン用混合液とした。ノックダウン前処理として、樹立したNT細胞、LATS1/2 KD細胞、MeT-5A WT細胞、LATS1 KO細胞、LATS2 KO細胞をそれぞれ5×103細胞/cm2/ウェルになるように細胞培養プレート (96ウェル Corningカタログ番号:3598、6ウェル VioLAMO:カタログ番号2-8588-01)に播種し、10%FBS-RPMI中で24時間培養した。その後、10%FBS-RPMIを完全に取り除き、ノックダウン用混合液をウェルの底面積に対し0.13ml/cm2になるように添加し、72時間培養した。 The siRNAs used (siSMG6-1: GCCAGUGAUACAGCGAAUU, siSMG6-2: ACACCAACGGCUUCAUUGA, siNegative Control: CGUACGCGGAAUACUUCGA) were produced using Nippon Gene's siRNA synthesis service. The prepared siRNA was mixed with 4% OPTIMEM, 1.34% RNAiMax (Invitrogen: catalog number 13778030), and 2 nM siRNA immediately before cell introduction, and incubated at room temperature for 15 minutes. Thereafter, RPMI (Wako: catalog number 189-02025) with FBS added to 10% (hereinafter referred to as 10% FBS-RPMI) was added to prepare a mixture for knockdown. As a knockdown pretreatment, the established NT cells, LATS1/2 KD cells, MeT-5A WT cells, LATS1 KO cells, and LATS2 KO cells were each cultured in a cell culture plate (5 × 10 3 cells/cm 2 /well) ( The cells were seeded in 96-well Corning catalog number: 3598, 6-well VioLAMO: catalog number 2-8588-01) and cultured in 10% FBS-RPMI for 24 hours. Thereafter, 10% FBS-RPMI was completely removed, a knockdown mixture was added to the bottom area of the well at a concentration of 0.13 ml/cm 2 , and cultured for 72 hours.

ノックアウト用プラスミド(pX330, Addgene #42230)のBbsIサイトにLATS1またはLATS2の標的配列(LATS1; AGCAAGAAAAGTAGATACTA(配列番号7), LATS2; AGGAAACTGGACTAACAATG(配列番号8) )を導入した。MeT-5Aに作製したノックアウト用プラスミドを、Thermo Fisher社Lipofectamine 2000を用いてトランスフェクションし、CO2インキュベーターでインキュベーションした。48時間後、細胞を1 cell/wellとなるよう96wellプレートに播種し、順次スケールアップを行い、シングルクローンを得た。 The LATS1 or LATS2 target sequence (LATS1; AGCAAGAAAAGTAGATACTA (SEQ ID NO: 7), LATS2; AGGAAACTGGACTAACAATG (SEQ ID NO: 8)) was introduced into the BbsI site of the knockout plasmid (pX330, Addgene #42230). The knockout plasmid prepared in MeT-5A was transfected using Thermo Fisher's Lipofectamine 2000, and incubated in a CO 2 incubator. After 48 hours, the cells were seeded in a 96-well plate at 1 cell/well, and scaled up sequentially to obtain a single clone.

4.TERT阻害剤投与
TERTを阻害するため、トリコスタチン(SantaCruz:カタログ番号sc-3511)、ドキソルビシン (SantaCruz:カタログ番号sc-200923)、BIBR1532 (SantaCruz:カタログ番号sc-203843)を細胞に直接添加し、反応させた。各TERT阻害剤は、10%FBS-RPMIを溶媒とし、50μM、10μM、5μM、1μM、500nM、100nM、50nM、10nM、5nM、1nMになるように段階希釈した。樹立したNT細胞、LATS1/2 KD細胞、MeT-5A WT細胞、LATS1 KO細胞、LATS2 KO細胞をそれぞれ1×104細胞/cm2/ウェルになるように細胞培養プレート (96ウェル Corningカタログ番号:3598)に播種し、10%FBS-RPMI中で24時間培養した。培養後、10%FBS-RPMIを完全に取り除き、希釈した各TERT阻害剤100μlを各ウェルに添加し72時間培養した。
Four. TERT inhibitor administration
To inhibit TERT, trichostatin (SantaCruz: catalog number sc-3511), doxorubicin (SantaCruz: catalog number sc-200923), and BIBR1532 (SantaCruz: catalog number sc-203843) were directly added to cells and reacted. Each TERT inhibitor was serially diluted to 50 μM, 10 μM, 5 μM, 1 μM, 500 nM, 100 nM, 50 nM, 10 nM, 5 nM, and 1 nM using 10% FBS-RPMI as a solvent. Culture established NT cells, LATS1/2 KD cells, MeT-5A WT cells, LATS1 KO cells, and LATS2 KO cells at 1 × 10 4 cells/cm 2 /well each in a cell culture plate (96-well Corning catalog number: 3598) and cultured in 10% FBS-RPMI for 24 hours. After culturing, 10% FBS-RPMI was completely removed, 100 μl of each diluted TERT inhibitor was added to each well, and cultured for 72 hours.

5. 細胞生存率測定
ノックダウン処理した細胞において、ノックダウン処理から72時間後96ウェルプレート内のノックダウン用混合液を10%FBS-RPMI 100μlに交換し、さらに37℃で72時間培養した。培養後、10%FBS-RPMIを完全に除去し、Cell Counting Kit-8 (同仁化学研究所:カタログ番号 CK04)を10%になるように10%FBS-RPMIに添加し、各ウェルに100μl加え37℃で1時間培養した。培養後SPECTRA Max (Molecular Devices)を用いて450nm-ref.630nmの波長条件で測定した。TERT阻害剤処理した細胞において、阻害剤処理から72時間後同様に細胞生存率を測定した。未処理群における吸光度を基準に各ノックダウン群及び阻害剤処理群を正規化し、比較検討した。
5. Measurement of Cell Viability In the knockdown-treated cells, 72 hours after the knockdown treatment, the knockdown mixture in the 96-well plate was replaced with 100 μl of 10% FBS-RPMI, and the cells were further cultured at 37°C for 72 hours. After culturing, completely remove 10% FBS-RPMI, add Cell Counting Kit-8 (Dojindo Laboratories: catalog number CK04) to 10% FBS-RPMI to a concentration of 10%, and add 100 μl to each well. Culture was performed at 37°C for 1 hour. After culturing, measurements were performed using SPECTRA Max (Molecular Devices) at a wavelength of 450 nm-ref.630 nm. In cells treated with TERT inhibitor, cell viability was measured in the same manner 72 hours after treatment with the inhibitor. Each knockdown group and inhibitor-treated group were normalized based on the absorbance in the untreated group, and comparative studies were performed.

6.半数致死量の算出
TERT阻害剤を段階希釈し細胞と反応させた後、細胞生存率の測定を行い得られた生存率から半数致死量(LD50:lethal dose,50%)を算出した。Image Jの解析アルゴリズムを利用し、以下のシグモイド曲線の係数を算出した。得られたシグモイド関数よりLD50を算出した。
6. Calculation of half-lethal dose
After the TERT inhibitor was serially diluted and reacted with cells, the cell survival rate was measured and the lethal dose (LD50: 50%) was calculated from the obtained survival rate. Using Image J's analysis algorithm, we calculated the coefficients of the sigmoid curve below. LD50 was calculated from the obtained sigmoid function.

Figure 0007437648000003
Figure 0007437648000003

7.アポトーシス検出
NT細胞、LATS1/2 KD細胞をノックダウン処理から72時間後にトリプシン/EDTAを用いて細胞懸濁液を回収した。血球算定板を用い細胞数を測定した後、BD Cytofix/CytopermTM Fixation/Permeabilization Solution Kit (BD:カタログ番号BDB554714)を用いて細胞固定と膜透過処理を行った。その後TUNEL Assay Kit - BrdU-Red (Abcam:カタログ番号ab66110)を用いてTUNEL染色を行った。染色後LSRFORTESSA(BD)を用いて測定し、Flowjoにより解析を行った。
7. Apoptosis detection
72 hours after the knockdown treatment of NT cells and LATS1/2 KD cells, cell suspensions were collected using trypsin/EDTA. After measuring the number of cells using a hematology plate, cell fixation and membrane permeabilization were performed using BD Cytofix/Cytoperm TM Fixation/Permeabilization Solution Kit (BD: catalog number BDB554714). Thereafter, TUNEL staining was performed using TUNEL Assay Kit - BrdU-Red (Abcam: catalog number ab66110). After staining, it was measured using LSRFORTESSA (BD) and analyzed using Flowjo.

8.DNA損傷検出
NT細胞、LATS1/2 KD細胞、MeT-5A WT細胞、LATS1 KO細胞、LATS2 KO細胞をノックダウン処理から72時間後に冷メタノール中に浸漬し、-20℃で10分間固定した。PBSで1回洗浄した後、1%BSA含有PBSと室温で1時間反応させた。その後、マウス抗ヒトγH2AX抗体(Millipore:カタログ番号05-636、5μg/ml)に浸漬し4℃で18時間反応させた。PBSで3回洗浄した後、Alexa488標識抗マウス抗体(サーモフィッシャー:カタログ番号A32723、5μg/ml)に浸漬し室温で1時間反応させた。PBSで3回洗浄した後、DAPI(4',6-diamidino-2-phenylindole、BIO-RAD:カタログ番号133061-2、1μ/ml)と15分反応させ核染色した。PBSで3回洗浄後、Fluorescence Mounting Medium(Dako:カタログ番号s3023)に封入した。染色後LSM800(カールツァイス)を用いて観察した。
8. DNA damage detection
72 hours after knockdown treatment, NT cells, LATS1/2 KD cells, MeT-5A WT cells, LATS1 KO cells, and LATS2 KO cells were immersed in cold methanol and fixed at -20°C for 10 minutes. After washing once with PBS, it was reacted with PBS containing 1% BSA for 1 hour at room temperature. Thereafter, it was immersed in mouse anti-human γH2AX antibody (Millipore: catalog number 05-636, 5 μg/ml) and reacted at 4° C. for 18 hours. After washing three times with PBS, it was immersed in Alexa488-labeled anti-mouse antibody (Thermo Fisher: catalog number A32723, 5 μg/ml) and reacted at room temperature for 1 hour. After washing three times with PBS, nuclear staining was performed by reacting with DAPI (4',6-diamidino-2-phenylindole, BIO-RAD: catalog number 133061-2, 1 μ/ml) for 15 minutes. After washing three times with PBS, the cells were mounted in Fluorescence Mounting Medium (Dako: catalog number s3023). After staining, it was observed using LSM800 (Carl Zeiss).

9.統計解析
NT細胞とLATS1/2 KD細胞の二群間における生存率の比較検討は、スチューデントのt検定を行った。MeT-5A WT細胞、LATS1 KO細胞、LATS2 KO細胞の三群間における生存率は、Steel-Dwass検定を用い比較検討した。
結果を図1に示した。
9.Statistical analysis
A Student's t test was performed to compare the survival rates between the two groups of NT cells and LATS1/2 KD cells. The survival rates among the three groups of MeT-5A WT cells, LATS1 KO cells, and LATS2 KO cells were compared using the Steel-Dwass test.
The results are shown in Figure 1.

10.結果
図1に示すように、LATS1/2 KD細胞においてSMG6発現抑制により有意な生存細胞の減少が認められた。加えて、LATS2 KO細胞においてのみ有意な線損細胞の減少が認められLATS2とSMG6を共発現抑制することにより合成致死表現型が誘導されることが確認された。
Ten. Results As shown in Figure 1, a significant decrease in viable cells was observed in LATS1/2 KD cells by suppressing SMG6 expression. In addition, a significant decrease in linear loss cells was observed only in LATS2 KO cells, confirming that co-expression suppression of LATS2 and SMG6 induces a synthetic lethal phenotype.

図2で示すように、SMG6の下流候補であるTERT阻害剤を反応させ、細胞死を誘導するLD50を検討した結果、LATS1/2 KD細胞、LATS2 KO細胞において正常細胞と比較し低用量のTERT阻害剤による細胞死の誘導が確認された。 As shown in Figure 2, as a result of reacting with TERT inhibitor, which is a downstream candidate of SMG6, and examining the LD50 for inducing cell death, lower doses of TERT were detected in LATS1/2 KD cells and LATS2 KO cells compared to normal cells. Induction of cell death by the inhibitor was confirmed.

図3で示すように、HOMC-D4由来の安定細胞株において、SMG6の発現抑制に伴いTUNEL陽性細胞が検出された。また定量評価を行った結果、LATS1/2 KD細胞にSMG6を発現抑制した群は、NT細胞にSMG6を発現抑制した群及びLATS1/2 KD細胞にNegaive Control処理した群に対して有意にTUNEL陽性細胞の上昇が認められた。本結果より、LATS2、SMG6の共発現抑制による合成致死表現型はアポトーシスによって惹起されることが認められた。 As shown in FIG. 3, TUNEL-positive cells were detected in the stable cell line derived from HOMC-D4 due to suppression of SMG6 expression. In addition, as a result of quantitative evaluation, the group in which SMG6 expression was suppressed in LATS1/2 KD cells was significantly more TUNEL positive than the group in which SMG6 expression was suppressed in NT cells and the group in which LATS1/2 KD cells were treated with Negaive Control. An increase in the number of cells was observed. These results confirmed that the synthetic lethal phenotype caused by suppression of co-expression of LATS2 and SMG6 was induced by apoptosis.

図4で示すように、SMG6の発現抑制によってLATS1/2 KD細胞においてγH2A.Xの染色陽性数の増加が認められた。本結果より、LATS2、SMG6の共発現抑制による合成致死表現型はDNA損傷に伴うアポトーシスによって惹起されることが示唆された。 As shown in FIG. 4, an increase in the number of positive γH2A.X staining was observed in LATS1/2 KD cells due to suppression of SMG6 expression. These results suggest that the synthetic lethal phenotype caused by co-inhibition of LATS2 and SMG6 expression is caused by apoptosis associated with DNA damage.

Claims (4)

MG6を阻害する化合物による治療応答性を調べるために、がん患者由来のがん細胞におけるLATS2遺伝子の不活性化変異を検出する方法。 A method for detecting inactivating mutations in the LATS2 gene in cancer cells derived from cancer patients in order to examine the therapeutic responsiveness of compounds that inhibit S MG6. がん患者が悪性中皮腫、前立腺がん又は非小細胞肺がんの患者である請求項1記載の方法。 2. The method according to claim 1, wherein the cancer patient is a patient with malignant mesothelioma, prostate cancer, or non-small cell lung cancer. SMG6の阻害を指標として被験化合物を評価することを含む、悪性中皮腫、前立腺がん又は非小細胞肺がんであるLATS2変異疾患のための薬剤のスクリーニング方法。 A method for screening drugs for LATS2 mutant diseases such as malignant mesothelioma, prostate cancer, or non-small cell lung cancer, the method comprising evaluating a test compound using SMG6 inhibition as an index. SMG6を阻害する化合物を有効成分として含む、悪性中皮腫、前立腺がん又は非小細胞肺がんであるLATS2変異疾患のための薬剤。 A drug for LATS2 mutant disease, which is malignant mesothelioma, prostate cancer or non-small cell lung cancer, which contains a compound that inhibits SMG6 as an active ingredient.
JP2019191790A 2019-10-21 2019-10-21 Molecular targets and their utilization for LATS2 mutant diseases Active JP7437648B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2019191790A JP7437648B2 (en) 2019-10-21 2019-10-21 Molecular targets and their utilization for LATS2 mutant diseases

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
JP2019191790A JP7437648B2 (en) 2019-10-21 2019-10-21 Molecular targets and their utilization for LATS2 mutant diseases

Publications (2)

Publication Number Publication Date
JP2021065121A JP2021065121A (en) 2021-04-30
JP7437648B2 true JP7437648B2 (en) 2024-02-26

Family

ID=75635977

Family Applications (1)

Application Number Title Priority Date Filing Date
JP2019191790A Active JP7437648B2 (en) 2019-10-21 2019-10-21 Molecular targets and their utilization for LATS2 mutant diseases

Country Status (1)

Country Link
JP (1) JP7437648B2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018518980A (en) 2015-07-02 2018-07-19 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Biomarkers of response to selective inhibitors of Aurora A kinase
JP2019502753A (en) 2015-12-23 2019-01-31 ムーンショット ファーマ エルエルシー Induction of immune response by suppression of nonsense mutation-dependent mRNA degradation mechanism

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018518980A (en) 2015-07-02 2018-07-19 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Biomarkers of response to selective inhibitors of Aurora A kinase
JP2019502753A (en) 2015-12-23 2019-01-31 ムーンショット ファーマ エルエルシー Induction of immune response by suppression of nonsense mutation-dependent mRNA degradation mechanism

Also Published As

Publication number Publication date
JP2021065121A (en) 2021-04-30

Similar Documents

Publication Publication Date Title
Kang et al. The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development
Ayuso-Sacido et al. Activated EGFR signaling increases proliferation, survival, and migration and blocks neuronal differentiation in post-natal neural stem cells
Zhao et al. NOTCH-induced aldehyde dehydrogenase 1A1 deacetylation promotes breast cancer stem cells
Ito et al. A PML–PPAR-δ pathway for fatty acid oxidation regulates hematopoietic stem cell maintenance
Nakahara et al. Hes1 immortalizes committed progenitors and plays a role in blast crisis transition in chronic myelogenous leukemia
Facchino et al. BMI1 confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery
Hurtz et al. BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia
Abdouh et al. BMI1 sustains human glioblastoma multiforme stem cell renewal
Pan et al. Stromal derived factor-1 (SDF-1/CXCL12) and CXCR4 in renal cell carcinoma metastasis
Lwin et al. Cell adhesion induces p27 Kip1-associated cell-cycle arrest through down-regulation of the SCFSkp2 ubiquitin ligase pathway in mantle-cell and other non-Hodgkin B-cell lymphomas
Zhou et al. Jab1/Csn5–thioredoxin signaling in relapsed acute monocytic leukemia under oxidative stress
Yao et al. MiR-330-mediated regulation of SH3GL2 expression enhances malignant behaviors of glioblastoma stem cells by activating ERK and PI3K/AKT signaling pathways
Li et al. The p53–PUMA axis suppresses iPSC generation
Guo et al. PBX3 is essential for leukemia stem cell maintenance in MLL‐rearranged leukemia
Despeaux et al. Focal adhesion kinase splice variants maintain primitive acute myeloid leukemia cells through altered Wnt signaling
Tanaka et al. Dual function of histone H3 lysine 36 methyltransferase ASH1 in regulation of Hox gene expression
Silvestri et al. Persistence of drug-resistant leukemic stem cells and impaired NK cell immunity in CML patients depend on MIR300 antiproliferative and PP2A-activating functions
Meziane et al. Knockdown of Fbxo7 reveals its regulatory role in proliferation and differentiation of haematopoietic precursor cells
Hsu et al. 58-kDa microspherule protein (MSP58) is novel Brahma-related gene 1 (BRG1)-associated protein that modulates p53/p21 senescence pathway
Lopez et al. An essential pathway links FLT3-ITD, HCK and CDK6 in acute myeloid leukemia
Santarlasci et al. Musculin inhibits human T‐helper 17 cell response to interleukin 2 by controlling STAT5B activity
Xu et al. Prostate tumor cells with cancer progenitor properties have high telomerase activity and are rapidly killed by telomerase interference
Zhang et al. miR-125b-5p/STAT3 pathway regulated by mTORC1 plays a critical role in promoting cell proliferation and tumor growth
Achuthankutty et al. Regulation of ETAA1-mediated ATR activation couples DNA replication fidelity and genome stability
Rontauroli et al. miR-494-3p overexpression promotes megakaryocytopoiesis in primary myelofibrosis hematopoietic stem/progenitor cells by targeting SOCS6

Legal Events

Date Code Title Description
A80 Written request to apply exceptions to lack of novelty of invention

Free format text: JAPANESE INTERMEDIATE CODE: A80

Effective date: 20191120

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20191219

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A821

Effective date: 20191219

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20200129

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20200129

A621 Written request for application examination

Free format text: JAPANESE INTERMEDIATE CODE: A621

Effective date: 20220930

A977 Report on retrieval

Free format text: JAPANESE INTERMEDIATE CODE: A971007

Effective date: 20230831

A131 Notification of reasons for refusal

Free format text: JAPANESE INTERMEDIATE CODE: A131

Effective date: 20230905

A521 Request for written amendment filed

Free format text: JAPANESE INTERMEDIATE CODE: A523

Effective date: 20231102

TRDD Decision of grant or rejection written
A01 Written decision to grant a patent or to grant a registration (utility model)

Free format text: JAPANESE INTERMEDIATE CODE: A01

Effective date: 20240116

A61 First payment of annual fees (during grant procedure)

Free format text: JAPANESE INTERMEDIATE CODE: A61

Effective date: 20240201

R150 Certificate of patent or registration of utility model

Ref document number: 7437648

Country of ref document: JP

Free format text: JAPANESE INTERMEDIATE CODE: R150