TWI777479B - A use of cytisine derivatives - Google Patents

A use of cytisine derivatives Download PDF

Info

Publication number
TWI777479B
TWI777479B TW110112331A TW110112331A TWI777479B TW I777479 B TWI777479 B TW I777479B TW 110112331 A TW110112331 A TW 110112331A TW 110112331 A TW110112331 A TW 110112331A TW I777479 B TWI777479 B TW I777479B
Authority
TW
Taiwan
Prior art keywords
formula
cells
dengue virus
present
denv
Prior art date
Application number
TW110112331A
Other languages
Chinese (zh)
Other versions
TW202239406A (en
Inventor
林振文
茵娜 彩比謝娃
恩彤 喬
阿麗娜 科沃斯卡婭
魯斯塔姆 薩菲林
Original Assignee
中國醫藥大學
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 中國醫藥大學 filed Critical 中國醫藥大學
Priority to TW110112331A priority Critical patent/TWI777479B/en
Application granted granted Critical
Publication of TWI777479B publication Critical patent/TWI777479B/en
Publication of TW202239406A publication Critical patent/TW202239406A/en

Links

Images

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

A cytisine derivative of following general formula (I), a synthesis method thereof, a pharmaceutical composition thereof and a use thereof are disclosed.
In formula (I), each symbol is as defined in the specification. The cytisine derivative can inhibit the cytopathic effect and viral infectivity of dengue virus. Therefore, it is useful as a prophylactic and therapeutic drug for treating dengue virus.

Description

金雀花鹼衍生物之用途 Uses of cystein derivatives

本發明是有關於一種金雀花鹼衍生物,特別是有關於一種可預防、治療及改善登革病毒之金雀花鹼衍生物、其合成方法、其醫藥組合物及其用途。 The present invention relates to a cystein derivative, in particular to a cystein derivative that can prevent, treat and improve dengue virus, its synthesis method, its pharmaceutical composition and its use.

登革病毒(Dengue virus,DENV)屬於黃病毒屬(Flavivirus),其是經由斑蚊屬的病媒蚊和靈長類動物形成病毒傳播的循環,並使帶有登革病毒的病媒蚊叮咬後的靈長類動物罹患登革熱、登革出血熱或登革休克症候群。 Dengue virus (DENV) belongs to the genus Flavivirus, which forms a cycle of viral transmission through vector mosquitoes and primates of the genus Aedes, and causes primates to be bitten by vector mosquitoes with dengue virus. Animals suffering from dengue fever, dengue hemorrhagic fever or dengue shock syndrome.

登革病毒於地理上的分佈範圍甚廣,在世界各國均有發現,其中以北緯35度到南緯35度且海拔1,000公尺以下地區最為常見,受感染者除了罹患登革熱、登革出血熱或登革休克症候群之外,亦可能併發其他神經系統疾病,如橫貫性脊髓炎和格林-巴利症候群(Guillain-Barre Syndrome)、飛蚊症,其他罕見的併發症有心臟感染和急性肝衰竭。 Dengue virus has a wide geographical distribution and has been found in all countries in the world. The most common areas are 35 degrees north latitude to 35 degrees south latitude and below 1,000 meters above sea level. In addition to dengue fever, dengue hemorrhagic fever or In addition to dengue shock syndrome, other neurological diseases may also be complicated, such as transverse myelitis and Guillain-Barre syndrome (Guillain-Barre Syndrome), floaters, and other rare complications include heart infection and acute liver failure.

雖登革病毒所導致之疾病具有相當之嚴重性,然時至今日僅可透過休息與症狀治療改善,尚無藥物或疫苗可 供登革病毒的預防或治療,是故開發一種新穎的藥物來對抗此類病毒有其重要性。 Although the disease caused by dengue virus is quite serious, it can only be improved by rest and symptomatic treatment. There is no drug or vaccine. For the prevention or treatment of dengue virus, it is important to develop a novel drug to fight against this virus.

本發明之一目的在於提供一種金雀花鹼衍生物、其合成方法、其醫藥組合物及其用途,並將金雀花鹼衍生物應用於抗登革病毒之用途中,藉由金雀花鹼衍生物具有抑制病毒的效果,以有效對登革病毒進行治療。 One object of the present invention is to provide a genus derivative, its synthesis method, its pharmaceutical composition and its use, and to apply the genus derivative to the purpose of resisting dengue virus. The base derivative has a virus-inhibiting effect to effectively treat dengue virus.

本發明之一實施方式提供一種金雀花鹼衍生物之用途,其係用於製備一預防、治療或改善登革病毒感染症之藥物,其中所述金雀花鹼衍生物具有如式(I)所示之一結構:

Figure 110112331-A0305-02-0004-4
其中式(I)中,R1為甲基、如式(MAI)所示之一基團或如式(MAII)所示之一基團,R2為氫原子或硫原子,R3為氫原子或硫原子,R4為氫原子、硝基、如式(MBI)所示之一基團或如式(MBII)所示之一基團,R5為氧原子或硫原子,
Figure 110112331-A0305-02-0004-5
表示單鍵或雙鍵以便滿足所有價數:
Figure 110112331-A0305-02-0005-6
One embodiment of the present invention provides the use of a cystein derivative, which is used to prepare a medicament for preventing, treating or ameliorating dengue virus infection, wherein the cyphine derivative has the formula (I ) shown in one of the structures:
Figure 110112331-A0305-02-0004-4
Wherein in formula (I), R 1 is a methyl group, a group shown in formula (MAI) or a group shown in formula (MAII), R 2 is a hydrogen atom or a sulfur atom, and R 3 is hydrogen atom or sulfur atom, R 4 is a hydrogen atom, a nitro group, a group shown in formula (MBI) or a group shown in formula (MBII), R 5 is an oxygen atom or a sulfur atom,
Figure 110112331-A0305-02-0004-5
Represent single or double bonds to satisfy all valences:
Figure 110112331-A0305-02-0005-6

藉此,本發明之金雀花鹼衍生物經由實驗數據證實具有抗登革病毒之功效,能夠在體外抑制登革病毒之細胞病變效應和病毒感染性,並能阻斷病毒之早期和晚期複製的步驟。是以本發明之金雀花鹼衍生物適於作為預防、治療及改善登革病毒感染症的藥物,或與醫藥上可接受之賦形劑或載體搭配作為可預防、治療及改善登革病毒疾病的醫藥組合物,具有運用於生醫市場之潛能。 Thereby, the cystein derivatives of the present invention have been proved to have anti-dengue virus efficacy through experimental data, can inhibit the cytopathic effect and virus infectivity of dengue virus in vitro, and can block the early and late replication of the virus A step of. Therefore, the cystein derivatives of the present invention are suitable as medicines for preventing, treating and ameliorating dengue virus infection, or can be used in combination with pharmaceutically acceptable excipients or carriers to prevent, treat and improve dengue virus. The pharmaceutical composition for diseases has the potential to be used in the biomedical market.

100:金雀花鹼衍生物之合成方法 100: Synthesis method of cystein derivatives

110:前驅物提供步驟 110: Precursor providing step

120:化學反應步驟 120: Chemical Reaction Steps

130:產物純化步驟 130: Product purification step

為讓本發明之上述和其他目的、特徵、優點與實施例能更明顯易懂,所附圖式之說明如下:第1圖係繪示本發明另一實施方式之金雀花鹼衍生物合成方法之步驟流程圖;第2A圖、第2B圖、第2C圖、第2D圖、第2E圖、第2F圖和第2G圖係繪示本發明之實施例對Vero E6細胞之細胞存活率試驗結果長條圖; 第3A圖、第3B圖、第3C圖、第3D圖、第3E圖、第3F圖和第3G圖係繪示本發明之實施例對A549細胞之細胞存活率試驗結果長條圖;第4圖為本發明之實施例1對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像;第5圖為本發明之實施例1對DENV-2誘導A549細胞病變之抑制能力的顯微影像;第6A圖、第6B圖、第6C圖、第6D圖、第6E圖、第6F圖和第6G圖係繪示本發明之實施例對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果;第7A圖、第7B圖、第7C圖、第7D圖、第7E圖、第7F圖和第7G圖係繪示本發明之實施例對受DENV-2感染之A549細胞的感染抑制力試驗結果;第8A圖、第8B圖、第8C圖、第8D圖和第8E圖係繪示本發明之實施例對受DENV-1感染之Vero E6細胞的感染抑制力試驗結果;第9圖為本發明之實施例5在病毒感染前處理試驗中(不清洗藥物下)對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像;第10A圖、第10B圖和第10C圖係繪示本發明之實施例在病毒感染前處理試驗中(不清洗藥物下)對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果;第11圖為本發明之實施例5在病毒感染前處理試驗中(以PBS清洗藥物)對DENV-2誘導Vero E6細胞病變之抑 制能力的顯微影像;第12A圖、第12B圖和第12C圖係繪示本發明之實施例在病毒感染前處理試驗中(以PBS清洗藥物)對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果;第13圖為本發明之實施例1在病毒感染時處理試驗中對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像;第14A圖、第14B圖、第14C圖、第14D圖、第14E圖、第14F圖和第14G圖係繪示本發明之實施例在病毒感染時處理試驗中對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果;第15圖為本發明之實施例1在病毒感染後處理試驗中對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像;以及第16A圖、第16B圖、第16C圖、第16D圖、第16E圖、第16F圖和第16G圖係繪示本發明之實施例在病毒感染後處理試驗中對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果。 In order to make the above-mentioned and other objects, features, advantages and embodiments of the present invention more clearly understood, the accompanying drawings are described as follows: Fig. 1 shows the synthesis of cystein derivatives according to another embodiment of the present invention. The flow chart of the steps of the method; Fig. 2A, Fig. 2B, Fig. 2C, Fig. 2D, Fig. 2E, Fig. 2F and Fig. 2G illustrate the cell viability test of Vero E6 cells according to an embodiment of the present invention result bar chart; Fig. 3A, Fig. 3B, Fig. 3C, Fig. 3D, Fig. 3E, Fig. 3F and Fig. 3G are bar graphs showing the results of the cell viability test on A549 cells according to the embodiments of the present invention; Fig. 4 Figure 1 is a microscopic image of the inhibitory ability of Example 1 of the present invention to DENV-2-induced Vero E6 cytopathies; Figure 5 is a microscopic image of the inhibitory ability of Example 1 of the present invention to DENV-2-induced A549 cytopathic changes ; Figure 6A, Figure 6B, Figure 6C, Figure 6D, Figure 6E, Figure 6F, and Figure 6G illustrate the infection inhibition of DENV-2-infected Vero E6 cells by embodiments of the present invention Test results; Fig. 7A, Fig. 7B, Fig. 7C, Fig. 7D, Fig. 7E, Fig. 7F and Fig. 7G show the infection inhibition of DENV-2 infected A549 cells by the examples of the present invention The results of the force test; Figure 8A, Figure 8B, Figure 8C, Figure 8D and Figure 8E show the results of the infection inhibition test results of the embodiments of the present invention on Vero E6 cells infected with DENV-1; Figure 9 The figure is a microscopic image of the inhibitory ability of DENV-2 to induce Vero E6 cytopathic changes in the pretreatment test of virus infection (without washing the drug) in Example 5 of the present invention; Figure 10A, Figure 10B and Figure 10C are series Figure 11 shows the results of the infection inhibition test on Vero E6 cells infected with DENV-2 in the pretreatment test of virus infection (without cleaning drugs) according to the embodiment of the present invention; In the pretreatment test (washing the drug with PBS), the inhibition of DENV-2-induced Vero E6 cytopathic changes Figure 12A, Figure 12B, and Figure 12C show the microscopic images of DENV-2-infected Vero E6 cells in a pre-treatment test for virus infection (drug washing with PBS) according to an example of the present invention. Infection inhibition test results; Figure 13 is a microscopic image of the inhibitory ability of DENV-2-induced Vero E6 cytopathies in the treatment test of Example 1 of the present invention during virus infection; Figure 14A, Figure 14B, Figure 14C Figure, Figure 14D, Figure 14E, Figure 14F and Figure 14G show the results of the infection inhibition test on DENV-2-infected Vero E6 cells in the treatment test of the embodiment of the present invention; Figure 15 is a microscopic image of the inhibitory ability of DENV-2 to induce Vero E6 cytopathic changes in the post-treatment test of virus infection in Example 1 of the present invention; and Figure 16A, Figure 16B, Figure 16C, Figure 16D, Figure 16E, Figure 16F and Figure 16G show the results of the infection inhibition test on DENV-2-infected Vero E6 cells in the virus infection post-treatment test according to the embodiment of the present invention.

以下將參照圖式說明本發明之複數個實施例。為明確說明起見,許多實務上的細節將在以下敘述中一併說明。然而,應瞭解到,這些實務上的細節不應用以限制本發明。也就是說,在本發明部分實施例中,這些實務上的細節是非必要的。此外,為簡化圖式起見,一些習知慣用 的結構與元件在圖式中將以簡單示意的方式繪示之;並且重複之元件將可能使用相同的編號表示之。 Several embodiments of the present invention will be described below with reference to the drawings. For the sake of clarity, many practical details are set forth in the following description. It should be understood, however, that these practical details should not be used to limit the invention. That is, in some embodiments of the present invention, these practical details are unnecessary. In addition, in order to simplify the diagram, some conventional The structures and elements in the drawings will be shown in a simplified schematic manner; and repeated elements will possibly be designated by the same reference numerals.

<金雀花鹼衍生物> <Cycylate Derivatives>

本發明之一實施方式提供一種金雀花鹼衍生物,其具有如式(I)所示之一結構:

Figure 110112331-A0305-02-0008-7
其中式(I)中,R1為甲基、如式(MAI)所示之一基團或如式(MAII)所示之一基團,R2為氫原子或硫原子,R3為氫原子或硫原子,R4為氫原子、硝基、如式(MBI)所示之一基團或如式(MBII)所示之一基團,R5為氧原子或硫原子:
Figure 110112331-A0305-02-0008-8
One embodiment of the present invention provides a cystein derivative, which has a structure as shown in formula (I):
Figure 110112331-A0305-02-0008-7
Wherein in formula (I), R 1 is a methyl group, a group shown in formula (MAI) or a group shown in formula (MAII), R 2 is a hydrogen atom or a sulfur atom, and R 3 is hydrogen atom or sulfur atom, R 4 is a hydrogen atom, a nitro group, a group as shown in formula (MBI) or a group as shown in formula (MBII), R 5 is an oxygen atom or a sulfur atom:
Figure 110112331-A0305-02-0008-8

<金雀花鹼衍生物之合成方法> <Synthesis method of cystein derivatives>

本發明之另一實施方式提供一種如前述之金雀花 鹼衍生物之合成方法。請參照第1圖,第1圖係繪示本發明之金雀花鹼衍生物之合成方法100之步驟流程圖。金雀花鹼衍生物之合成方法100包含步驟110、步驟120及步驟130。 Another embodiment of the present invention provides a gorse flower as described above Synthesis of base derivatives. Please refer to FIG. 1. FIG. 1 is a flow chart showing the steps of the method 100 for synthesizing the cystein derivatives of the present invention. The method 100 for synthesizing cystein derivatives includes step 110 , step 120 and step 130 .

步驟110為一前驅物提供步驟,其係提供一前驅物以進行一化學反應,所使用之前驅物為金雀花鹼((-)-Cytisine)。 Step 110 is a step of providing a precursor, which provides a precursor to perform a chemical reaction, and the used precursor is cytisine ((-)-Cytisine).

步驟120為一化學反應步驟,其係對前驅物以一反應方式進行化學反應,以獲得一產物,所述反應方式為於室溫攪拌或高溫回流。 Step 120 is a chemical reaction step, which is to chemically react the precursor in a reaction manner to obtain a product, and the reaction manner is stirring at room temperature or refluxing at high temperature.

步驟130為一產物純化步驟,其係將產物進行純化,以獲得一金雀花鹼衍生物。其中純化的方式可為薄層層析法(Thin layer chromatography,TLC),但本發明不以此為限。 Step 130 is a product purification step, which is to purify the product to obtain a cysteine derivative. The purification method may be thin layer chromatography (TLC), but the present invention is not limited thereto.

茲以下列具體試驗例進一步示範說明本發明,用以有利於本發明所屬技術領域之通常知識者,可在不需過度解讀的情形下完整利用並實踐本發明,而不應將其視為對本發明範圍的限制,但用於說明如何實施本發明的材料及方法。 The following specific test examples are hereby used to further demonstrate the present invention, so as to help those of ordinary knowledge in the technical field to which the present invention pertains to fully utilize and practice the present invention without excessive interpretation, and should not be regarded as a It is intended to limit the scope of the invention, but to illustrate how the materials and methods of the invention may be practiced.

<試驗例> <Test example>

一、本發明之金雀花鹼衍生物的合成與結構鑑定 1. Synthesis and structural identification of cystein derivatives of the present invention

請參照下表一,為本發明之金雀花鹼衍生物之實施例1至實施例10之化合物式(IA)至化合物式(IJ)的結構式。 Please refer to Table 1 below, which are the structural formulas of compound formula (IA) to compound formula (IJ) of Example 1 to Example 10 of the cystein derivatives of the present invention.

Figure 110112331-A0305-02-0010-9
Figure 110112331-A0305-02-0010-9
Figure 110112331-A0305-02-0011-10
Figure 110112331-A0305-02-0011-10
Figure 110112331-A0305-02-0012-11
Figure 110112331-A0305-02-0012-11
Figure 110112331-A0305-02-0013-12
Figure 110112331-A0305-02-0013-12

請參照上表一,本發明之金雀花鹼衍生物之各實施例,其結構式如表一所示。 Please refer to the above Table 1, the various embodiments of the cystein derivatives of the present invention, the structural formulas are shown in Table 1.

本發明之實施例1及實施例3的合成方法如下:於56℃下以碘甲烷及碳酸鉀於丙酮中與(-)-cytisine進行反應,反應完成後蒸發丙酮,得到N-methylcytisine(下稱中間物1),將中間物1於室溫下以硝酸鈉在硫酸中進行硝化,將產物倒在冰上以碳酸鈉中和,以乙酸乙酯萃取後以無水硫酸鈉乾燥並濃縮,得到實施例3之金雀花鹼衍生物,將實施例3之金雀花鹼衍生物溶於乙酸乙酯中,並於氫氣氣氛下以鈀碳催化劑將實施例3之金雀花鹼衍生物所含之硝基還原為胺,得到9-amino-N-methylcytisine(下稱中間物2),將中間物2與1,3-二苯甲基甲酰基尿嘧啶反應後,溶於甲醇中以硼氫化鈉進行羰基還原,產物以三氯甲烷萃取後以無水硫酸鈉乾燥,得到實施例1之金雀花鹼衍生物。 The synthesis methods of Example 1 and Example 3 of the present invention are as follows: react with (-)-cytisine in acetone with methyl iodide and potassium carbonate at 56° C. After the reaction is completed, acetone is evaporated to obtain N-methylcytisine (hereinafter referred to as N-methylcytisine). Intermediate 1), the intermediate 1 was nitrated with sodium nitrate in sulfuric acid at room temperature, the product was poured on ice and neutralized with sodium carbonate, extracted with ethyl acetate, dried with anhydrous sodium sulfate and concentrated to obtain the implementation The cytidine derivative of Example 3 was dissolved in ethyl acetate, and the cytidine derivative of Example 3 was dissolved in ethyl acetate with a palladium-carbon catalyst under a hydrogen atmosphere. The nitro group is reduced to amine to obtain 9-amino-N-methylcytisine (hereinafter referred to as intermediate 2). After the intermediate 2 is reacted with 1,3-benzylformyluracil, it is dissolved in methanol for hydroboration Sodium is subjected to carbonyl reduction, and the product is extracted with chloroform and dried with anhydrous sodium sulfate to obtain the cysteine derivative of Example 1.

本發明之實施例2的合成方法如下:於室溫下以硝酸鈉對溶於濃硫酸中的(-)-cytisine進行硝化後,以碳酸鈉中和,以乙酸乙酯萃取後以無水硫酸鈉乾燥並濃縮,得到9-nitrocytisine(下稱中間物3),將中間物3溶於苯中以異氰酸苯基酯於室溫下進行反應後濃縮,得到9-Nitro-8-oxo-N-phenyl-1,5,6,8-tetrahydro-2H-1,5-methanopyrido[1,2-a][1,5]diazocine-3(4H)-carboxamide(下稱中間物4),將中間物4溶於乙酸乙酯中,於氫氣氣氛下以鈀碳催化劑將中間物2-2所含 之硝基還原為胺,得到9-Amino-8-oxo-N-phenyl-1,5,6,8-tetrahydro-2H-1,5-methanopyrido[1,2-a][1,5]diazocine-3(4H)-carboxamide(下稱中間物5),將中間物5與1,3-二甲基-5-甲酰基尿嘧啶於80℃在苯中進行回流,將產物溶於甲醇中以硼氫化鈉在0℃下進行羰基還原,以三氯化碳萃取後以無水硫酸鈉乾燥,得到實施例2之金雀花鹼衍生物。 The synthesis method of Example 2 of the present invention is as follows: at room temperature, the (-)-cytisine dissolved in concentrated sulfuric acid is nitrified with sodium nitrate, neutralized with sodium carbonate, extracted with ethyl acetate, and then extracted with anhydrous sodium sulfate. Dry and concentrate to obtain 9-nitrocytisine (hereinafter referred to as intermediate 3), which is dissolved in benzene and reacted with phenyl isocyanate at room temperature and concentrated to obtain 9-Nitro-8-oxo-N -phenyl-1,5,6,8-tetrahydro-2H-1,5-methanopyrido[1,2-a][1,5]diazocine-3(4H)-carboxamide (hereinafter referred to as intermediate 4), the intermediate Compound 4 was dissolved in ethyl acetate, and the intermediate compound 2-2 was mixed with palladium-carbon catalyst under a hydrogen atmosphere. The nitro group is reduced to an amine to give 9-Amino-8-oxo-N-phenyl-1,5,6,8-tetrahydro-2H-1,5-methanopyrido[1,2-a][1,5]diazocine -3(4H)-carboxamide (hereinafter referred to as intermediate 5), the intermediate 5 and 1,3-dimethyl-5-formyluracil were refluxed in benzene at 80 °C, and the product was dissolved in methanol to The carbonyl group was reduced by sodium borohydride at 0° C., extracted with carbon trichloride, and dried with anhydrous sodium sulfate to obtain the cysteine derivative of Example 2.

本發明之實施例4的合成方法如下:將中間物2與4-羥基苯甲醛於苯中在80℃下進行反應後,將苯蒸發並將產物溶於甲醇中,於0℃下以硼氫化鈉進行羰基還原後,以三氯化碳萃取後以無水硫酸鈉乾燥,得到實施例4之金雀花鹼衍生物。 The synthesis method of Example 4 of the present invention is as follows: After the intermediate 2 is reacted with 4-hydroxybenzaldehyde in benzene at 80°C, the benzene is evaporated and the product is dissolved in methanol, and the hydroboration is carried out at 0°C. After carbonyl reduction with sodium, extraction with carbon trichloride and drying with anhydrous sodium sulfate, the cystein derivative of Example 4 was obtained.

本發明之實施例5的合成方法如下:於35℃-40℃下以異硫氰酸烯丙酯在苯中對(-)-cytisine進行反應,得到實施例5之金雀花鹼衍生物。 The synthesis method of Example 5 of the present invention is as follows: react (-)-cytisine with allyl isothiocyanate in benzene at 35°C to 40°C to obtain the cytisine derivative of Example 5.

本發明之實施例6的合成方法如下:將中間物1溶於甲苯以勞森試劑(Lawesson's Reagent)於110℃下進行回流,反應完成後蒸發溶劑,產物以碳酸鈉進行鹼化,並以三氯甲烷萃取後以無水硫酸鈉乾燥並濃縮,得到實施例6之金雀花鹼衍生物。 The synthesis method of Example 6 of the present invention is as follows: the intermediate 1 is dissolved in toluene and refluxed at 110° C. with Lawesson's Reagent, the solvent is evaporated after the reaction is completed, the product is alkalized with sodium carbonate, and the reaction is carried out with three After extraction with methyl chloride, drying over anhydrous sodium sulfate and concentration, the cystein derivative of Example 6 was obtained.

本發明之實施例7及實施例10的合成方法如下:將中間物1與過錳酸鉀於室溫下在乙腈水溶液(乙腈:水=10:1)中反應2小時後,得到 (1R,5R)-3-Methyl-3,4,5,6-tetrahydro-2H-1,5-methanopyrido[1,2-a][1,5]diazocine-2,8(1H)-dione(下稱中間物6),將中間物6溶於甲苯以勞森試劑於110℃下進行回流,反應完成後蒸發溶劑,產物以碳酸鈉進行鹼化,並以三氯甲烷萃取後以無水硫酸鈉乾燥並濃縮,得到實施例7及實施例10之金雀花鹼衍生物。 The synthesis methods of Example 7 and Example 10 of the present invention are as follows: the intermediate 1 and potassium permanganate are reacted in an aqueous acetonitrile solution (acetonitrile:water=10:1) at room temperature for 2 hours to obtain (1R,5R)-3-Methyl-3,4,5,6-tetrahydro-2H-1,5-methanopyrido[1,2-a][1,5]diazocine-2,8(1H)-dione( Hereinafter referred to as intermediate 6), the intermediate 6 was dissolved in toluene and refluxed at 110 ° C with Lawesson's reagent. After the reaction was completed, the solvent was evaporated, and the product was alkalized with sodium carbonate, extracted with chloroform and then extracted with anhydrous sodium sulfate. Drying and concentration gave the cystein derivatives of Example 7 and Example 10.

本發明之實施例8及實施例9的合成方法如下:將中間物1與過錳酸鉀於室溫下在乙腈水溶液(乙腈:水=10:1)中反應2小時後,溶於甲苯以0.5當量勞森試劑於110℃下進行回流,得到實施例8及實施例9之金雀花鹼衍生物。 The synthesis methods of Example 8 and Example 9 of the present invention are as follows: Intermediate 1 and potassium permanganate are reacted in an aqueous acetonitrile solution (acetonitrile:water=10:1) at room temperature for 2 hours, then dissolved in toluene to 0.5 equivalent of Lawson's reagent was refluxed at 110°C to obtain the cystein derivatives of Example 8 and Example 9.

本發明之各實施例以紅外光譜儀及核磁共振光譜儀,進行紅外光譜及核磁共振光譜分析數據如下表二所示。 In each embodiment of the present invention, infrared spectroscopy and nuclear magnetic resonance spectroscopy are used to perform infrared spectroscopy and nuclear magnetic resonance spectroscopy analysis data as shown in Table 2 below.

Figure 110112331-A0305-02-0016-13
Figure 110112331-A0305-02-0016-13
Figure 110112331-A0305-02-0017-14
Figure 110112331-A0305-02-0017-14
Figure 110112331-A0305-02-0018-15
Figure 110112331-A0305-02-0018-15
Figure 110112331-A0305-02-0019-16
Figure 110112331-A0305-02-0019-16
Figure 110112331-A0305-02-0020-17
Figure 110112331-A0305-02-0020-17
Figure 110112331-A0305-02-0021-18
Figure 110112331-A0305-02-0021-18
Figure 110112331-A0305-02-0022-19
Figure 110112331-A0305-02-0022-19
Figure 110112331-A0305-02-0023-20
Figure 110112331-A0305-02-0023-20
Figure 110112331-A0305-02-0024-21
Figure 110112331-A0305-02-0024-21

二、本發明之金雀花鹼衍生物之抗登革病毒試驗 2. Anti-dengue virus test of the genistein derivatives of the present invention

2-1:細胞存活率試驗 2-1: Cell Viability Test

進行細胞存活率試驗的細胞株為非洲綠猴腎上皮細胞Vero E6(ATCC® No.CRL-1586,以下簡稱Vero E6細胞)及人類肺腺癌細胞A549(ATCC® No.CLL-185TM,以下簡稱A549細胞)。首先將Vero E6細胞或A549細胞以每孔5×103的細胞量培養於96孔盤中,以含有10% FBS(Fetal bovine serum,Gibco®) 的DMEM(Dulbecco’s modified eagle medium,Gibco®)配置至總體積160μL/孔,在37℃及5% CO2的培養箱中培養過夜。隔日,在不去除培養液情況下,分別加入實施例1-實施例10之金雀花鹼衍生物,且使每一實施例的藥物濃度分別達0.1μM、1μM、10μM及50μM,而各濃度進行四重複試驗。藥物處理96小時後,於每孔細胞添加10μL的MTT溶液,避光反應4小時,然後加入100μL的Solution C(J.T.Baker®),將結晶紫打散,使用ELISA讀取器或高通亮光學測讀儀分別測量背景值波長670nm和樣品波長570nm的吸光度,並以檢測到之樣品波長570nm的吸光度減去背景值波長670nm的吸光度,對每孔處理的細胞進行評估,以沒有加入金雀花鹼衍生物處理之組別的吸光度(OD值)為100%,而推算出細胞存活率(Survival rate,%)及衍生物對Vero E6細胞或A549細胞的半毒殺劑量(CC50)。 The cell lines used for the cell viability test were African green monkey kidney epithelial cells Vero E6 (ATCC ® No.CRL-1586, hereinafter referred to as Vero E6 cells) and human lung adenocarcinoma cells A549 (ATCC ® No. CLL-185 TM , hereinafter referred to as referred to as A549 cells). First, Vero E6 cells or A549 cells were cultured in a 96-well plate at a cell volume of 5 × 10 3 per well, and were prepared in DMEM (Dulbecco's modified eagle medium, Gibco ® ) containing 10% FBS (Fetal bovine serum, Gibco ® ). To a total volume of 160 μL/well, incubate overnight in an incubator at 37°C and 5% CO 2 . On the next day, without removing the culture medium, the cystein derivatives of Example 1-Example 10 were added respectively, and the drug concentration of each example reached 0.1 μM, 1 μM, 10 μM and 50 μM, respectively. Four replicate experiments were performed. After 96 hours of drug treatment, add 10 μL of MTT solution to each well of cells, react in the dark for 4 hours, and then add 100 μL of Solution C (JT Baker ® ) to disperse the crystal violet, and use an ELISA reader or high-brightness optical reading. The instrument measures the absorbance at the background wavelength of 670nm and the sample at the wavelength of 570nm respectively, and subtracts the absorbance of the background value at 670nm from the absorbance of the detected sample at the wavelength of 570nm, and evaluates the cells treated in each well. The absorbance (OD value) of the treated group was 100%, and the cell survival rate (%) and the half-toxic dose (CC 50 ) of the derivatives to Vero E6 cells or A549 cells were calculated.

第2A圖、第2B圖、第2C圖、第2D圖、第2E圖、第2F圖和第2G圖分別繪示本發明之實施例1、實施例2、實施例3、實施例4、實施例5、實施例6和實施例7對Vero E6細胞之細胞存活率試驗結果長條圖;第3A圖、第3B圖、第3C圖、第3D圖、第3E圖、第3F圖和第3G圖分別繪示本發明之實施例1、實施例2、實施例3、實施例4、實施例5、實施例6和實施例7對A549細胞之細胞存活率試驗結果長條圖。由試驗結果可見,即使所添加之實施例1-實施例7的藥物濃度達50μM,亦未 明顯導致Vero E6細胞或A549細胞死亡,顯示本發明之金雀花鹼衍生物對Vero E6細胞及對A549細胞無毒性作用(CC50

Figure 110112331-A0305-02-0026-44
50μM)。 Fig. 2A, Fig. 2B, Fig. 2C, Fig. 2D, Fig. 2E, Fig. 2F, and Fig. 2G respectively illustrate Embodiment 1, Embodiment 2, Embodiment 3, Embodiment 4, and Embodiment of the present invention Bar graphs of cell viability assay results of Example 5, Example 6 and Example 7 on Vero E6 cells; Figure 3A, Figure 3B, Figure 3C, Figure 3D, Figure 3E, Figure 3F, and Figure 3G The figure shows a bar graph of the cell viability test results of Example 1, Example 2, Example 3, Example 4, Example 5, Example 6 and Example 7 of the present invention on A549 cells, respectively. It can be seen from the test results that even if the added drug concentration of Example 1-Example 7 reaches 50 μM, it does not obviously lead to the death of Vero E6 cells or A549 cells, which shows that the cytidine derivatives of the present invention are effective against Vero E6 cells and on Vero E6 cells. A549 cells had no toxic effects (CC 50
Figure 110112331-A0305-02-0026-44
50 μM).

2-2:細胞病變抑制試驗及免疫螢光染色分析 2-2: Cytopathic inhibition test and immunofluorescence staining analysis

進行細胞病變抑制試驗及免疫螢光染色分析的細胞株為Vero E6細胞及A549細胞,病毒株為第一型登革病毒(Dengue virus type 1,DENV-1)及第二型登革病毒(Dengue virus type 2,DENV-2)之16681病毒株(Strain 16681)。 The cell lines for cytopathic inhibition test and immunofluorescence staining analysis were Vero E6 cells and A549 cells, and the virus strains were Dengue virus type 1 (DENV-1) and Dengue virus type 2 (Dengue virus type 1, DENV-1). virus type 2, DENV-2) of the 16681 strain (Strain 16681).

首先將Vero E6細胞或A549細胞以每孔2×105的細胞量培養於6孔盤中,以含有10% FBS的DMEM配製至總體積達2mL/孔,並於37℃及5% CO2的培養箱中培養過夜。隔日,去除培養液並以磷酸鹽緩衝溶液(Phosphate buffered saline,PBS)洗滌6孔盤兩次,再使用不含FBS的DMEM清洗一次後,加入DENV-1或DENV-2,詳細來說,以病毒感染劑量(Multiplicity of infection,MOI)為0.1的DENV-1感染Vero E6細胞及A549細胞,以MOI為0.005的DENV-2感染Vero E6細胞,以MOI為0.05的DENV-2感染A549細胞,並分別加入實施例1-實施例10的金雀花鹼衍生物,使其藥物濃度達0.1μM、1μM及10μM,而針對最有效的衍生物會增加進行0.001μM和0.01μM濃度,以對細胞進行處理。培養96小時後,藉由觀察細胞病變狀態並拍照即可得到細胞病變抑制試驗之結果。 First, Vero E6 cells or A549 cells were cultured in a 6-well dish at 2×10 5 cells per well, prepared in DMEM containing 10% FBS to a total volume of 2 mL/well, and incubated at 37°C and 5% CO 2 Incubator overnight. On the next day, the culture medium was removed and the 6-well plate was washed twice with Phosphate buffered saline (PBS) and once with DMEM without FBS, and then DENV-1 or DENV-2 was added. Vero E6 cells and A549 cells were infected with DENV-1 with a multiplicity of infection (MOI) of 0.1, Vero E6 cells were infected with DENV-2 with an MOI of 0.005, and A549 cells were infected with DENV-2 with an MOI of 0.05. The genistein derivatives of Example 1-Example 10 were added to their drug concentrations of 0.1 μM, 1 μM, and 10 μM, respectively, while the most effective derivatives were increased to 0.001 μM and 0.01 μM to treat cells. deal with. After 96 hours of culture, the results of the cytopathic inhibition test can be obtained by observing the cytopathic state and taking pictures.

請參考第4圖及第5圖,第4圖為本發明之實施例1對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像,第5圖為本發明之實施例1對DENV-2誘導A549細胞病變之抑制能力的顯微影像。從圖中可見,實施例1以濃度依賴的方式抑制了DENV-2誘導的Vero E6細胞病變及A549細胞病變,且在藥物濃度為10μM時,呈現顯著抑制的效果。以第4圖所顯示之抑制效果為顯著抑制(+++),在此試驗下之各實施例衍生物對於受感染Vero E6細胞之細胞病變的抑制效果整理如下表三。圖表之結果表明,本發明之金雀花鹼衍生物對於DENV-2誘導之細胞病變具有顯著的抑制效果。 Please refer to Figure 4 and Figure 5. Figure 4 is a microscopic image of the inhibitory ability of Example 1 of the present invention to DENV-2-induced Vero E6 cell lesions, and Figure 5 is a microscopic image of Example 1 of the present invention to DENV-2 2 Microscopic images of the inhibitory ability to induce pathological changes in A549 cells. As can be seen from the figure, Example 1 inhibited DENV-2-induced Vero E6 cytopathic and A549 cytopathic in a concentration-dependent manner, and showed a significant inhibitory effect when the drug concentration was 10 μM. Taking the inhibitory effect shown in Figure 4 as a significant inhibitory effect (+++), the inhibitory effects of the derivatives of each example under this test on the cytopathic effects of infected Vero E6 cells are listed in Table 3 below. The results of the graphs show that the genistein derivatives of the present invention have a significant inhibitory effect on DENV-2-induced cytopathies.

Figure 110112331-A0305-02-0027-22
Figure 110112331-A0305-02-0027-22

承上(培養96小時後),於每孔細胞中加入1mL 4%的福馬林將細胞固定,之後於震盪器上室溫反應30分鐘,接著以PBS清洗兩次,而後於每孔細胞中加入1mL 50mM的NH4Cl,之後於震盪器上室溫反應30分鐘,以去除自體螢光,接著再以PBS清洗一次,而後加入BSA:Triton-100為1000:1比例的封閉液,然後於4℃冷房震盪反應4小時,經PBS清洗後,再以抗體進行染色。詳細而言,係以Rabbit anti-DENV2-NS4B(GeneTex,Inc.)作為一抗,然後於4℃冷房震盪過夜。隔日以PBS清洗三次後,以Goat anti-rabbit IgG H & L(Alexa Fluor® 555,ThermoFisher)抗體做為二抗進行染色。去除抗體後使用DAPI(4’6-diamidino-2-phenylindole)螢光染料,將細胞核染色,以計數細胞數,並使用Image J軟體來確定受感染細胞中NS4B陽性細胞的百分比。感染抑制力的計算方式為:(未加入衍生物處理的感染細胞中NS4B陽性百分比-加入衍生物處理的感染細胞中NS4B陽性百分比)/加入衍生物處理的感染細胞中NS4B陽性百分比。根據感染抑制力的結果,計算得到半數最大抑制濃度(IC50),即為本發明之金雀花鹼衍生物對於DENV-2之抗病毒活性。 Continue on (after 96 hours of culture), add 1 mL of 4% formalin to each well of cells to fix the cells, then react on a shaker for 30 minutes at room temperature, then wash twice with PBS, and then add to each well of cells 1mL of 50mM NH 4 Cl, then reacted on a shaker for 30 minutes at room temperature to remove autofluorescence, then washed once with PBS, and then added BSA:Triton-100 as a 1000:1 blocking solution, and then in The cells were reacted with shaking in a cold room at 4°C for 4 hours, washed with PBS, and then stained with antibodies. Specifically, Rabbit anti-DENV2-NS4B (GeneTex, Inc.) was used as the primary antibody, and then the cells were shaken in a refrigerated room at 4°C overnight. After washing three times with PBS every other day, Goat anti-rabbit IgG H & L (Alexa Fluor ® 555, ThermoFisher) antibody was used as the secondary antibody for staining. Nuclei were stained with DAPI (4'6-diamidino-2-phenylindole) fluorescent dye after antibody removal to count the number of cells, and Image J software was used to determine the percentage of NS4B positive cells in the infected cells. Infection inhibition was calculated as: (percent NS4B positive in infected cells not treated with derivative - percent NS4B positive in infected cells treated with derivative)/percent NS4B positive in infected cells treated with derivative. According to the results of the infection inhibition, the half-maximum inhibitory concentration (IC 50 ) was calculated, which is the antiviral activity of the genistein derivatives of the present invention against DENV-2.

第6A圖、第6B圖、第6C圖、第6D圖、第6E圖、第6F圖和第6G圖分別繪示本發明之實施例1、實施例2、實施例3、實施例4、實施例5、實施例6和實施例7對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果,第7A圖、第7B圖、第7C圖、第7D圖、第7E圖、第7F圖和第7G圖分別繪示本發明之實施例1、實施例2、實施例3、實施例4、實施例5、實施例6和實施例 7對受DENV-2感染之A549細胞的感染抑制力試驗結果,第8A圖、第8B圖、第8C圖、第8D圖和第8E圖分別繪示本發明之實施例1、實施例2、實施例3、實施例4及實施例7對受DENV-1感染之Vero E6細胞的感染抑制力試驗結果。同時請參照下表四。表四為本發明實施例1-實施例10對受DENV-2感染細胞的感染抑制力及抗病毒活性數據。圖表之結果表明,本發明之各實施例皆以濃度依賴的方式,顯著降低了受感染之細胞中NS4B陽性細胞的百分比,亦即本發明之各實施例能夠顯著降低DENV-2對於Vero E6細胞及A549細胞的感染性,這表明了本發明之金雀花鹼衍生物能夠顯著降低登革病毒對於細胞的感染性並對於DENV-2誘導之細胞病變具有顯著的抑制效果。 Fig. 6A, Fig. 6B, Fig. 6C, Fig. 6D, Fig. 6E, Fig. 6F, and Fig. 6G respectively illustrate Embodiment 1, Embodiment 2, Embodiment 3, Embodiment 4, and Embodiment of the present invention Infection inhibition test results of Example 5, Example 6 and Example 7 on Vero E6 cells infected with DENV-2, Figure 7A, Figure 7B, Figure 7C, Figure 7D, Figure 7E, Figure 7F and FIG. 7G respectively illustrate Embodiment 1, Embodiment 2, Embodiment 3, Embodiment 4, Embodiment 5, Embodiment 6 and Embodiment of the present invention. 7. The results of the infection inhibition test on A549 cells infected with DENV-2. Figure 8A, Figure 8B, Figure 8C, Figure 8D and Figure 8E respectively depict Example 1, Example 2, and Figure 8E of the present invention. Infection inhibition test results of Example 3, Example 4 and Example 7 on Vero E6 cells infected with DENV-1. Please also refer to Table 4 below. Table 4 shows the data on the infection inhibition and antiviral activity of DENV-2 infected cells in Examples 1 to 10 of the present invention. The results of the graph show that each embodiment of the present invention significantly reduces the percentage of NS4B positive cells in the infected cells in a concentration-dependent manner, that is, each embodiment of the present invention can significantly reduce the effect of DENV-2 on Vero E6 cells. and the infectivity of A549 cells, which indicates that the cystein derivatives of the present invention can significantly reduce the infectivity of dengue virus to cells and have a significant inhibitory effect on DENV-2-induced cytopathic effects.

Figure 110112331-A0305-02-0029-23
Figure 110112331-A0305-02-0029-23
Figure 110112331-A0305-02-0030-24
Figure 110112331-A0305-02-0030-24

2-3 病毒感染前處理試驗 2-3 Virus infection pretreatment test

進行病毒感染前處理試驗的細胞株為Vero E6細胞,第二型登革病毒(Dengue virus type 2,DENV-2)之16681病毒株(Strain 16681)。 The cell line used for virus infection pretreatment test was Vero E6 cell, the 16681 strain of Dengue virus type 2 (DENV-2) (Strain 16681).

首先,將Vero E6細胞以2×105每孔細胞數培養於6孔盤中,以含有10% FBS的DMEM配製至總體積達2mL/孔,並於37℃及5% CO2的培養箱中培養過夜。隔日,去除培養液並以磷酸鹽緩衝溶液(Phosphate buffered saline,PBS)洗滌6孔盤兩次,再使用不含FBS的DMEM清洗一次後,加入實施例1-實施例10之藥物,使藥物濃度達到0.1μM、1μM和10μM,在37℃及5% CO2的培養箱中培養24小時後,在不清洗藥物下/以PBS清洗藥物後,以MOI為0.005的DENV-2感染Vero E6細胞,最後加入不含FBS的DMEM讓總體積達2mL,再放入37℃及5% CO2的培養箱中96小時以觀察細胞病變抑制效果,並進行免疫螢光染色,免疫螢光染色之過程已於前段提及,在此不再贅述,利用螢光顯微鏡下觀察NS4B病毒蛋白和DAPI螢光數量並拍照,再利用Image J軟體計算NS4B病毒蛋白和DAPI量,計算出病毒感染的比率。 First, Vero E6 cells were cultured at 2×10 5 cells per well in a 6-well dish, prepared in DMEM containing 10% FBS to a total volume of 2 mL/well, and incubated at 37°C in an incubator with 5% CO 2 . Incubate overnight. The next day, the culture medium was removed and the 6-well plate was washed twice with Phosphate buffered saline (PBS), and then washed once with DMEM without FBS, and then the drugs of Example 1-Example 10 were added to make the drug concentration. At 0.1 μM, 1 μM and 10 μM, Vero E6 cells were infected with DENV-2 at an MOI of 0.005 after culturing for 24 hours in an incubator at 37°C and 5% CO 2 without washing the drug/after washing the drug with PBS, Finally, DMEM without FBS was added to bring the total volume to 2 mL, and then placed in an incubator at 37°C and 5% CO 2 for 96 hours to observe the inhibitory effect of cytopathic changes, and immunofluorescence staining was performed. The process of immunofluorescence staining has been completed. As mentioned in the previous paragraph, I will not repeat them here. The fluorescence quantity of NS4B viral protein and DAPI was observed under a fluorescence microscope and photographed, and then Image J software was used to calculate the quantity of NS4B viral protein and DAPI to calculate the ratio of virus infection.

第9圖為本發明之實施例5在病毒感染前處理試驗中(不清洗藥物下)對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像,第10A圖、第10B圖和第10C圖分別繪示本發明之實施例5、實施例6和實施例7在病毒感染前處理試驗中(不清洗藥物下)對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果,第11圖為本發明之實施例5在病毒感染前處理試驗中(以PBS清洗藥物)對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影 像,第12A圖、第12B圖和第12C圖係繪示本發明之實施例5、實施例6和實施例7在病毒感染前處理試驗中(以PBS清洗藥物)對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果。由第9圖、第10A圖至第10C圖、第11圖及第12A圖至第12C圖之結果顯示,本發明之實施例5顯著降低了受感染之細胞中NS4B陽性細胞的百分比,其IC50於不清洗藥物的狀態下為0.45μM,其IC50於以PBS清洗藥物的狀態下為5.42μM,亦即本發明實施例5之金雀花鹼衍生物能夠顯著預防DENV-2對於Vero E6細胞的感染性。 Fig. 9 is a microscopic image of the inhibitory ability of DENV-2 to induce Vero E6 cytopathic changes in the pretreatment test of virus infection (without cleaning drugs) according to Example 5 of the present invention, Fig. 10A, Fig. 10B and Fig. 10C The figure shows the results of the infection inhibition test of DENV-2-infected Vero E6 cells in the pretreatment test of virus infection (without washing the drug) in Example 5, Example 6 and Example 7 of the present invention, the 11th The figure is a microscopic image of the inhibitory ability of DENV-2 to induce Vero E6 cytopathies in the pretreatment test of virus infection (washing the drug with PBS) in Example 5 of the present invention. Figure 12A, Figure 12B and Figure 12C are the series The results of the infection inhibition test of DENV-2-infected Vero E6 cells in the pretreatment test of virus infection (washing the drug with PBS) of Example 5, Example 6 and Example 7 of the present invention are shown. From the results of Figure 9, Figure 10A to Figure 10C, Figure 11, and Figure 12A to Figure 12C, Example 5 of the present invention significantly reduced the percentage of NS4B positive cells in the infected cells, and its IC 50 is 0.45 μM in the state of not washing the drug, and its IC 50 is 5.42 μM in the state of washing the drug with PBS, that is, the cytidine derivative of Example 5 of the present invention can significantly prevent DENV-2 for Vero E6 Infectivity of cells.

2-4 病毒感染時及病毒感染後處理試驗 2-4 Virus infection and post-virus treatment test

首先,將兩組Vero E6細胞以前述方式於6孔盤中培養,並以DENV-2以前述方式感染之,且一組細胞在病毒感染同時添加實施例之金雀花鹼衍生物(病毒感染時處理試驗),另一組細胞在感染後1小時添加實施例之金雀花鹼衍生物(病毒感染後處理試驗)。所使用的實施例為實施例1、實施例2、實施例3、實施例4、實施例5、實施例6或實施例7,各實施例的藥物濃度分別達0.1μM、1μM及10μM。放入37℃及5% CO2的培養箱培養1小時後,以PBS清洗兩次以清除藥物,最後加入不含FBS的DMEM讓總體積達2mL,再放入37℃及5% CO2的培養箱中96小時以觀察細胞病變抑制效果,並進行免疫螢光染色,免疫螢光染色之過程已於前段提及,在此不再贅述,利用螢光顯微鏡下觀察NS4B病毒蛋白和DAPI螢 光數量並拍照,再利用Image J軟體計算NS4B病毒蛋白和DAPI量,計算出病毒感染的比率。 First, two groups of Vero E6 cells were cultured in a 6-well dish in the aforementioned manner, and infected with DENV-2 in the aforementioned manner, and one group of cells was infected with the virus while adding the cysteine derivative of the embodiment (virus infection). Time treatment test), another group of cells was added with the cytidine derivative of the example 1 hour after infection (virus infection post treatment test). The examples used are Example 1, Example 2, Example 3, Example 4, Example 5, Example 6 or Example 7, and the drug concentration of each example reached 0.1 μM, 1 μM and 10 μM, respectively. After culturing in an incubator at 37°C and 5% CO 2 for 1 hour, wash twice with PBS to remove the drug, and finally add FBS-free DMEM to make the total volume 2 mL, and then put it into an incubator at 37° C. and 5% CO 2 . Incubator for 96 hours to observe the cytopathic inhibition effect, and perform immunofluorescence staining. The process of immunofluorescence staining has been mentioned in the previous paragraph and will not be repeated here. NS4B virus protein and DAPI fluorescence were observed under a fluorescence microscope. Quantity and taking pictures, and then use Image J software to calculate the amount of NS4B virus protein and DAPI, and calculate the ratio of virus infection.

第13圖為本發明之實施例1在病毒感染時處理試驗中對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像,第14A圖、第14B圖、第14C圖、第14D圖、第14E圖、第14F圖和第14G圖分別繪示本發明之實施例1、實施例2、實施例3、實施例4、實施例5、實施例6和實施例7在病毒感染時處理試驗中對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果。同時請參照下表五,表五為本發明實施例1-實施例7對DENV-2於進入細胞階段的抗病毒活性數據。圖表之結果顯示,本發明之各實施例皆以濃度依賴的方式,顯著降低了受感染之細胞中NS4B陽性細胞的百分比,亦即本發明之各實施例能夠顯著降低DENV-2對於Vero E6細胞的感染性,這表明了本發明之金雀花鹼衍生物能夠顯著降低登革病毒對於細胞的感染性並對於DENV-2的早期複製具有顯著的抑制效果。 Figure 13 is a microscopic image of the inhibitory ability of DENV-2 to induce Vero E6 cytopathic changes in the treatment test of Example 1 of the present invention during virus infection, Figure 14A, Figure 14B, Figure 14C, Figure 14D, Fig. 14E, Fig. 14F and Fig. 14G respectively illustrate the treatment test of the present invention in Example 1, Example 2, Example 3, Example 4, Example 5, Example 6 and Example 7 of the present invention Infection inhibition test results on Vero E6 cells infected with DENV-2. Please refer to the following table 5 at the same time, table 5 is the antiviral activity data of the embodiments 1-7 of the present invention to DENV-2 in the cell entry stage. The results of the graph show that each embodiment of the present invention significantly reduces the percentage of NS4B positive cells in the infected cells in a concentration-dependent manner, that is, each embodiment of the present invention can significantly reduce the effect of DENV-2 on Vero E6 cells. The infectivity of the present invention shows that the genistein derivatives of the present invention can significantly reduce the infectivity of dengue virus to cells and have a significant inhibitory effect on the early replication of DENV-2.

Figure 110112331-A0305-02-0033-25
Figure 110112331-A0305-02-0033-25
Figure 110112331-A0305-02-0034-26
Figure 110112331-A0305-02-0034-26

第15圖為本發明之實施例1在病毒感染後處理試驗中對DENV-2誘導Vero E6細胞病變之抑制能力的顯微影像,第16A圖、第16B圖、第16C圖、第16D圖、第16E圖、第16F圖和第16G圖係繪示本發明之實施例1、實施例2、實施例3、實施例4、實施例5、實施例6和實施例7在病毒感染後處理試驗中對受DENV-2感染之Vero E6細胞的感染抑制力試驗結果。同時請參照下表六,表六為本發明實施例1-實施例7對DENV-2於感染細胞後的抗病毒活性數據。圖表之結果顯示,本發明之各實施例皆以濃度依賴的方式,顯著降低了受感染之細胞中NS4B陽性細胞的百分比,亦即本發明之各實施例能夠顯著降低DENV-2進入Vero E6細胞後的複製能力,這表明了本發明之金雀花鹼衍生物能夠顯著降低登革病毒進入細胞後複製能力並對於DENV-2的晚期複製具有顯著的抑制效果。 Figure 15 is a microscopic image of the inhibitory ability of DENV-2 to induce Vero E6 cytopathic changes in the virus infection post-treatment test of Example 1 of the present invention, Figure 16A, Figure 16B, Figure 16C, Figure 16D, Fig. 16E, Fig. 16F and Fig. 16G illustrate the post-virus treatment test of Example 1, Example 2, Example 3, Example 4, Example 5, Example 6 and Example 7 of the present invention Infection inhibition test results on Vero E6 cells infected with DENV-2. Please refer to the following table 6 at the same time. Table 6 is the antiviral activity data of DENV-2 after infecting cells in Examples 1-7 of the present invention. The results of the graphs show that each embodiment of the present invention significantly reduces the percentage of NS4B positive cells in the infected cells in a concentration-dependent manner, that is, each embodiment of the present invention can significantly reduce the entry of DENV-2 into Vero E6 cells This shows that the genistein derivatives of the present invention can significantly reduce the replication ability of dengue virus after entering cells and have a significant inhibitory effect on the late replication of DENV-2.

Figure 110112331-A0305-02-0034-27
Figure 110112331-A0305-02-0034-27
Figure 110112331-A0305-02-0035-28
Figure 110112331-A0305-02-0035-28

綜上所述,本發明結構如式(I)所示之金雀花鹼衍生物,可在體外抑制DENV-1和DENV-2的細胞病變效應和病毒感染性,亦能阻斷病毒之早期和晚期複製的步驟,且其對DENV-1和DENV-2的半數最大抑制濃度皆小於10μM,故除降低了半數最大抑制濃度外,還能提高治療指數(Therapeutic Index,TI)。附帶一提,所述治療指數為半毒殺劑量與半數最大抑制濃度之比值。因此,本發明之金雀花鹼衍生物,具有預防、治療及改善登革病毒感染症的效果,適於作為預防、治療及改善登革病毒的藥物,或與醫藥上可接受之賦形劑或載體搭配作為可預防、治療及改善登革病毒疾病的醫藥組合物,更能進一步擴大作為抗登革病毒藥物的用途,深具生醫市場之潛能。 To sum up, the genistein derivatives represented by the structure of the present invention as shown in formula (I) can inhibit the cytopathic effect and virus infectivity of DENV-1 and DENV-2 in vitro, and can also block the early stage of the virus and late replication steps, and its half-maximal inhibitory concentration on DENV-1 and DENV-2 are both less than 10 μM, so in addition to reducing the half-maximal inhibitory concentration, it can also improve Therapeutic Index (TI). Incidentally, the therapeutic index is the ratio of the half-toxic dose to the half-maximal inhibitory concentration. Therefore, the cystein derivatives of the present invention have the effect of preventing, treating and improving dengue virus infection, and are suitable as medicines for preventing, treating and improving dengue virus, or together with pharmaceutically acceptable excipients Or the carrier can be used as a pharmaceutical composition that can prevent, treat and improve dengue virus disease, and can further expand the use as an anti-dengue virus drug, which has deep potential in the biomedical market.

本發明已以實施方式揭露如上,然其並非用以限定本發明,任何熟習此技藝者,在不脫離本發明的精神和範圍內,當可作各種的更動與潤飾,因此本發明的保護範圍當視後附的申請專利範圍所界定者為準。 The present invention has been disclosed as above in embodiments, but it is not intended to limit the present invention. Anyone who is familiar with the art can make various changes and modifications without departing from the spirit and scope of the present invention. Therefore, the protection scope of the present invention is The scope of the patent application attached shall prevail.

Figure 110112331-A0305-02-0002-3
Figure 110112331-A0305-02-0002-3

Claims (8)

一種金雀花鹼衍生物之用途,其係用於製備一預防、治療或改善登革病毒感染症之藥物;其中該金雀花鹼衍生物具有如式(I)所示之一結構:
Figure 110112331-A0305-02-0037-29
其中該式(I)中,R1為甲基、如式(MAI)所示之一基團或如式(MAII)所示之一基團,R2為氫原子或硫原子,R3為氫原子或硫原子,R4為氫原子、硝基、如式(MBI)所示之一基團或如式(MBII)所示之一基團,R5為氧原子或硫原子,
Figure 110112331-A0305-02-0037-30
表示單鍵或雙鍵以便滿足所有價數:
Figure 110112331-A0305-02-0037-31
A use of a cyphine derivative, which is used to prepare a medicine for preventing, treating or improving dengue virus infection; wherein the cyphine derivative has a structure as shown in formula (I):
Figure 110112331-A0305-02-0037-29
Wherein in the formula (I), R 1 is a methyl group, a group shown in formula (MAI) or a group shown in formula (MAII), R 2 is a hydrogen atom or a sulfur atom, and R 3 is A hydrogen atom or a sulfur atom, R 4 is a hydrogen atom, a nitro group, a group shown in formula (MBI) or a group shown in formula (MBII), R 5 is an oxygen atom or a sulfur atom,
Figure 110112331-A0305-02-0037-30
Represent single or double bonds to satisfy all valences:
Figure 110112331-A0305-02-0037-31
如請求項1所述之金雀花鹼衍生物之用途,其中該金雀花鹼衍生物具有如式(IA)、式(IB)、式(IC)、式(ID)、式(IE)、式(IF)、式(IG)、式(IH)、式(II)或式(IJ)所示之一結構:
Figure 110112331-A0305-02-0038-33
Figure 110112331-A0305-02-0038-34
Figure 110112331-A0305-02-0038-35
Figure 110112331-A0305-02-0038-36
Figure 110112331-A0305-02-0039-38
Figure 110112331-A0305-02-0039-39
Figure 110112331-A0305-02-0039-40
Figure 110112331-A0305-02-0039-41
Figure 110112331-A0305-02-0039-42
Figure 110112331-A0305-02-0040-43
The use of the cystein derivative according to claim 1, wherein the cystein derivative has formula (IA), formula (IB), formula (IC), formula (ID), formula (IE) , one of the structures shown in formula (IF), formula (IG), formula (IH), formula (II) or formula (IJ):
Figure 110112331-A0305-02-0038-33
Figure 110112331-A0305-02-0038-34
Figure 110112331-A0305-02-0038-35
Figure 110112331-A0305-02-0038-36
Figure 110112331-A0305-02-0039-38
Figure 110112331-A0305-02-0039-39
Figure 110112331-A0305-02-0039-40
Figure 110112331-A0305-02-0039-41
Figure 110112331-A0305-02-0039-42
Figure 110112331-A0305-02-0040-43
如請求項1所述之金雀花鹼衍生物之用途,其中該預防、治療或改善登革病毒感染症之藥物中之一有效劑量為0.1μM至10μM。 The use of cystein derivatives according to claim 1, wherein an effective dose of the drugs for preventing, treating or ameliorating dengue virus infection is 0.1 μM to 10 μM. 如請求項3所述之金雀花鹼衍生物之用途,其中該有效劑量為10μM。 The use of cystein derivatives according to claim 3, wherein the effective dose is 10 μM. 如請求項1所述之金雀花鹼衍生物之用途,其中該預防、治療或改善登革病毒感染症之藥物更包含一醫藥上可接受之賦形劑或載體。 The use of cystein derivatives according to claim 1, wherein the medicine for preventing, treating or ameliorating dengue virus infection further comprises a pharmaceutically acceptable excipient or carrier. 如請求項1所述之金雀花鹼衍生物之用途,其中該登革病毒感染症由第一型登革病毒或第二型登革病毒引起。 The use of a cystein derivative according to claim 1, wherein the dengue virus infection is caused by type 1 dengue virus or type 2 dengue virus. 如請求項1所述之金雀花鹼衍生物之用途,其中該預防、治療或改善登革病毒感染症之藥物為降低登革病毒對於細胞感染性之藥物。 The use of the cystein derivatives according to claim 1, wherein the drug for preventing, treating or improving dengue virus infection is a drug for reducing the infectivity of dengue virus to cells. 如請求項1所述之金雀花鹼衍生物之用途,其中該預防、治療或改善登革病毒感染症之藥物為抑制登革病毒的早期複製及晚期複製之藥物。 The use of cystein derivatives as claimed in claim 1, wherein the drug for preventing, treating or improving dengue virus infection is a drug for inhibiting early and late replication of dengue virus.
TW110112331A 2021-04-06 2021-04-06 A use of cytisine derivatives TWI777479B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW110112331A TWI777479B (en) 2021-04-06 2021-04-06 A use of cytisine derivatives

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW110112331A TWI777479B (en) 2021-04-06 2021-04-06 A use of cytisine derivatives

Publications (2)

Publication Number Publication Date
TWI777479B true TWI777479B (en) 2022-09-11
TW202239406A TW202239406A (en) 2022-10-16

Family

ID=84957944

Family Applications (1)

Application Number Title Priority Date Filing Date
TW110112331A TWI777479B (en) 2021-04-06 2021-04-06 A use of cytisine derivatives

Country Status (1)

Country Link
TW (1) TWI777479B (en)

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
期刊 Koval'skaya A. V., et al.Thionation of quinolizidine alkaloids and their derivatives via Lawesson's reagent Natural Product Research online only Natural Product Research 2021 January pages 1-6. 2021/01/04 *
期刊 Kulakov, I. V., et al.Synthesis and intramolecular heterocyclization of n-allylthiocarbamide derivatives of the alkaloids cytisine and anabasine into 1,3-thiazoline derivatives and features of their molecular structures Chemistry of Natural Compounds Vol. 46(2) Chemistry of Natural Compounds 2010 May pages 257-261. 2010/05/25; *
期刊 Tsypysheva, I. P., et al.Synthesis and Cytotoxic Activity of Conjugates of (-)-Cytisine and Thermopsin Amine Derivatives with 1,3-Dimethyl-5-Formyluracil Chemistry of Natural Compounds Vol. 54(5) Chemistry of Natural Compounds 2018 September pages 938-946. 2018/09/01; *
期刊 Tsypysheva, I. P., et al.Synthesis and Nootropic Activity of new 3-Amino-12- N-Methylcytisine Derivatives Chemistry of Natural Compounds Vol. 51(5) Chemistry of Natural Compounds 2015 September pages 910-915. 2015/10/05; *
期刊 Tsypysheva, I. P., et al.Synthesis of 3- and 5-Amino Derivatives of Methylcytisine Chemistry of Natural Compounds Vol. 49(5) Chemistry of Natural Compounds 2013 November pages 902-906. 2013/11/22; *

Also Published As

Publication number Publication date
TW202239406A (en) 2022-10-16

Similar Documents

Publication Publication Date Title
CN107427521A (en) The inhibitor of cell cycle protein dependent kinase
US20240287014A1 (en) Biflavone compound against i-type herpes simplex virus, and preparation method therefor and use thereof
CN113289018A (en) Application of old medicine such as auranofin and composition thereof in resisting single positive strand RNA virus
WO2024037520A1 (en) Amide compound, and preparation method therefor, pharmaceutical composition and use thereof
CN101948467B (en) Thiazoleamide compound and use thereof for the preparation of anti-malignant tumor medicines
TWI777479B (en) A use of cytisine derivatives
WO2022213262A1 (en) Cytisine derivative, and synthesis method therefor, pharmaceutical composition thereof and use thereof
CN113354684A (en) Novel compound and application thereof
CN106349160B (en) A kind of 8-aminoquinoline derivatives and its preparation and application
Bialer et al. Structure-activity relationships of pyrroleamidine antiviral antibiotics. 1. Modifications of the alkylamidine side chain
CN109081818B (en) Novel indoleamine 2, 3-bis-oxidase inhibitors
TWI770767B (en) A use of glycyrrhetic acid derivatives
JP5711822B2 (en) Use of quinazoline compounds in the preparation of therapeutic drugs for Raviviridae viruses
CN109867636B (en) Compound for resisting CVA16 type hand-foot-and-mouth disease and synthetic method thereof
CN106399319A (en) Fejervarya multistriata gene, polypeptide encoded by same and application of polypeptide
CN106632373A (en) A D crystal form solid substance of bergenin, a preparing method thereof and uses of the substance
WO2022151002A1 (en) Glycyrrhetinic acid derivative, synthesis method therefor, pharmaceutical composition thereof and use thereof
CN106466320A (en) A kind of application of sulfur-bearing thick miscellaneous Fourth Ring Azasugar
CN111518090B (en) Flavane flavone derivative and preparation method and application thereof
CN115745830B (en) Binaphthyl dicarboxylic acid derivative, preparation method and application thereof
CN100438867C (en) Application of flavone monomer in antivirus drug preparing process
CN114409595B (en) 1-Styryl isoquinoline derivative and preparation and application thereof
CN108785296B (en) Application of diterpenoid compounds in preparation of antiviral drugs
CN115006386B (en) Application of nifurazite in preparation of medicines for resisting tick-borne encephalitis virus west nile virus yellow fever virus and chikungunya fever virus infection
CN103070876B (en) The compositions that the anti-encephalitis b virus of one class infects and application thereof

Legal Events

Date Code Title Description
GD4A Issue of patent certificate for granted invention patent