請輸入關鍵字
請輸入關鍵字
訂購
*國家
中國
美國
中國香港
中國澳門
中國台灣
阿爾巴尼亞
阿爾及利亞
阿根廷
阿拉伯聯合酋長國
阿魯巴
阿曼
阿塞拜疆
阿森鬆島
埃及
埃塞俄比亞
愛爾蘭
愛沙尼亞
安道爾
安哥拉
安圭拉
安提瓜和巴布達
奧地利
奧蘭群島
澳大利亞
巴巴多斯
巴布亞新幾內亞
巴哈馬
巴基斯坦
巴拉圭
巴勒斯坦領土
巴林
巴拿馬
巴西
白俄羅斯
百慕大
保加利亞
北馬裏亞納群島
貝寧
比利時
冰島
波多黎各
波蘭
波斯尼亞和黑塞哥維那
玻利維亞
伯利茲
博茨瓦納
不丹
布基納法索
布隆迪
朝鮮
赤道幾內亞
丹麥
德國
迪戈加西亞島
東帝汶
多哥
多米尼加共和國
多米尼克
俄羅斯
厄瓜多爾
厄立特裏亞
法國
法羅群島
法屬波利尼西亞
法屬圭亞那
法屬南部領地
梵蒂岡
菲律賓
斐濟
芬蘭
佛得角
福克蘭群島
岡比亞
剛果(布)
剛果(金)
哥倫比亞
哥斯達黎加
格恩西島
格林納達
格陵蘭
格魯吉亞
古巴
瓜德羅普
關島
圭亞那
哈薩克斯坦
海地
韓國
荷蘭
荷屬加勒比區
荷屬聖馬丁
黑山
洪都拉斯
基裏巴斯
吉布提
吉爾吉斯斯坦
幾內亞
幾內亞比紹
加拿大
加納
加納利群島
加蓬
柬埔寨
捷克
津巴布韋
喀麥隆
卡塔爾
開曼群島
科科斯(基林)群島
科摩羅
科索沃
科特迪瓦
科威特
克羅地亞
肯尼亞
庫克群島
庫拉索
拉脫維亞
萊索托
老撾
黎巴嫩
立陶宛
利比裏亞
利比亞
聯合國
列支敦士登
留尼汪
盧森堡
盧旺達
羅馬尼亞
馬達加斯加
馬恩島
馬爾代夫
馬耳他
馬拉維
馬來西亞
馬裏
馬其頓
馬紹爾群島
馬提尼克
馬約特
毛裏求斯
毛裏塔尼亞
美國本土外小島嶼
美屬薩摩亞
美屬維爾京群島
蒙古
蒙特塞拉特
孟加拉國
秘魯
密克羅尼西亞
緬甸
摩爾多瓦
摩洛哥
摩納哥
莫桑比克
墨西哥
納米比亞
南非
南極洲
南喬治亞和南桑威奇群島
南蘇丹
瑙魯
尼加拉瓜
尼泊爾
尼日爾
尼日利亞
紐埃
挪威
諾福克島
帕勞
皮特凱恩群島
葡萄牙
日本
瑞典
瑞士
薩爾瓦多
薩摩亞
塞爾維亞
塞拉利昂
塞內加爾
塞浦路斯
塞舌爾
沙特阿拉伯
聖巴泰勒米
聖誕島
聖多美和普林西比
聖赫勒拿
聖基茨和尼維斯
聖盧西亞
聖馬丁島
聖馬力諾
聖皮埃爾和密克隆群島
聖文森特和格林納丁斯
斯裏蘭卡
斯洛伐克
斯洛文尼亞
斯瓦爾巴和揚馬延
斯威士蘭
蘇丹
蘇裏南
所羅門群島
索馬裏
塔吉克斯坦
泰國
坦桑尼亞
湯加
特克斯和凱科斯群島
特裏斯坦-達庫尼亞群島
特立尼達和多巴哥
突尼斯
圖瓦盧
土耳其
土庫曼斯坦
托克勞
瓦利斯和富圖納
瓦努阿圖
危地馬拉
委內瑞拉
文萊
烏幹達
烏克蘭
烏拉圭
烏茲別克斯坦
希臘
西班牙
西撒哈拉
新加坡
新喀裏多尼亞
新西蘭
匈牙利
休達及梅利利亞
敘利亞
牙買加
亞美尼亞
也門
伊拉克
伊朗
以色列
意大利
印度
印度尼西亞
英國
英屬維爾京群島
英屬印度洋領地
約旦
越南
讚比亞
澤西島
乍得
直布羅陀
智利
中非共和國
*省份
*城市
*姓名
*電話
*單位
*職位
*郵箱
*請輸入驗證碼
*驗證碼
B-hCD3EDG mice
Strain Name
C57BL/6-Cd3etm1(CD3E)Bcgen Cd3dtm1(CD3D)Bcgen Cd3gtm1(CD3G)Bcgen/Bcgen
Common Name  B-hCD3EDG mice
Background C57BL/6 Catalog number  110039
Related Genes 
CD3E(CD3e molecule); CD3D(CD3d molecule);  CD3G(CD3g molecule)
NCBI Gene ID
12501,12500,12502

mRNA expression analysis


from clipboard


Strain specific analysis of CD3EDG gene expression in WT and homozygous B-hCD3EDG mice by RT-PCR. 
Mouse Cd3e, Cd3d and Cd3g mRNA was detectable only in thymocytes of wild type (+/+). Chimeric CD3E, CD3D and CD3G mRNA was detectable only in H/H, but not in +/+ mice. 


Protein expression analysis



from clipboard


Strain specific CD3E expression analysis in homozygous B-hCD3EDG mice by flow cytometry. 
Splenocytes were collected from wild type (WT) mice (+/+) and homozygous B-hCD3EDG mice (H/H), and analyzed by flow cytometry with species-specific anti-CD3ε antibody. Mouse CD3E was detectable in WT mice (+/+). Human CD3E was exclusively detectable in homozygous B-hCD3EDG mice (H/H) but not in WT mice (+/+).


Weight of spleen and the total cell number of splenocytes in homozygous B-hCD3EDG mice 


from clipboard

(A) Size of spleen from C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). (B) The weight of spleen in C57BL/6 and  B-hCD3EDG mice was similar. (C) The number of splenocytes in C57BL/6 and B-hCD3EDG mice was similar.


Weight of thymus and the total cell number of thymocytes in homozygous B-hCD3EDG mice 

from clipboard

(A) Size of thymus from C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). (B) The weight of thymus in C57BL/6 and  B-hCD3EDG mice was similar, but the weight of thymus in B-hCD3EDG mice was lower than that in C57BL/6 mice.(C) The number of thymocytes in C57BL/6 and B-hCD3EDG mice was similar, but the number of thymocytes in B-hCD3EDG mice was lower than that in C57BL/6.


Analysis of thymus leukocytes cell subpopulations in homozygous B-hCD3EDG mice

from clipboard


Analysis of thymus leukocyte subpopulations by FACS. Thymocytes were isolated from female C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old) . Flow cytometry analysis of the thymocytes was performed to assess leukocyte subpopulations. A. Representative FACS plots. Single live cells were gated for CD45 population and used for further analysis as indicated here. B. Results of FACS analysis. Percent of CD4/CD8 cells, B cells in homozygous B-hCD3EDG mice were similar to those in the C57BL/6 mice, demonstrating that introduction of hCD3EDG in place of its mouse counterpart does not change the overall development, differentiation or distribution of these cell types in thymus.


Analysis of thymus T cell subpopulations in homozygous B-hCD3EDG mice

from clipboard


Analysis of thymus T cell subpopulations by FACS. Thymocytes were isolated from female C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). Flow cytometry analysis of the thymocytes was performed to assess leukocyte subpopulations. A. Representative FACS plots. Single live CD45+ cells were gated for CD3 T cell population and used for further analysis as indicated here. B. Results of FACS analysis. Percent of CD8, CD4, and Treg cells in homozygous B-hCD3EDG mice were similar to those in the C57BL/6 mice, demonstrating that introduction of hCD3EDG in place of its mouse counterpart does not change the overall development, differentiation or distribution of these T cell subtypes in thymus. Values are expressed as mean ± SEM.


Analysis of spleen leukocytes cell subpopulations in homozygous B-hCD3EDG mice

from clipboard


from clipboard


Analysis of spleen leukocyte subpopulations by FACS. Splenocytes were isolated from female C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). Flow cytometry analysis of the splenocytes was performed to assess leukocyte subpopulations. A. Representative FACS plots. Single live cells were gated for CD45 population and used for further analysis as indicated here. B. Results of FACS analysis. Percent of T, B, NK, DC, Granulocyte, Monocyte, and macrophage cells in homozygous B-hCD3EDG mice were similar to those in the C57BL/6 mice, demonstrating that introduction of hCD3EDG in place of its mouse counterpart does not change the overall development, differentiation or distribution of these cell types in spleen.


Analysis of spleen T cell subpopulations in homozygous B-hCD3EDG mice

from clipboard


Analysis of spleen T cell subpopulations by FACS. Splenocytes were isolated from female C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). Flow cytometry analysis of the splenocytes was performed to assess leukocyte subpopulations. A. Representative FACS plots. Single live CD45+ cells were gated for CD3 T cell population and used for further analysis as indicated here. B. Results of FACS analysis. Percent of CD8, CD4, and Treg cells in homozygous B-hCD3EDG mice were similar to those in the C57BL/6 mice, demonstrating that introduction of hCD3EDG in place of its mouse counterpart does not change the overall development, differentiation or distribution of these T cell subtypes in spleen. Values are expressed as mean ± SEM.


Analysis of lymph node leukocytes cell subpopulations in homozygous B-hCD3EDG mice

from clipboard


Analysis of spleen leukocyte subpopulations by FACS. Lymph nodes were isolated from female C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). Flow cytometry analysis was performed to assess leukocyte subpopulations. A. Representative FACS plots. Single live cells were gated for CD45 population and used for further analysis as indicated here. B. Results of FACS analysis. Percent of T, B, NK in homozygous B-hCD3EDG mice were similar to those in the C57BL/6 mice, demonstrating that introduction of hCD3EDG in place of its mouse counterpart does not change the overall development, differentiation or distribution of these cell types in lymph nodes.


Analysis of lymph node T cell subpopulations in homozygous B-hCD3EDG mice

from clipboard


Analysis of spleen T cell subpopulations by FACS. Lymph node were isolated from female C57BL/6 and B-hCD3EDG mice (n=3, 7 week-old). Flow cytometry analysis of the splenocytes was performed to assess leukocyte subpopulations. A. Representative FACS plots. Single live CD45+ cells were gated for CD3 T cell population and used for further analysis as indicated here. B. Results of FACS analysis. Percent of CD8, CD4, and Treg cells in homozygous B-hCD3EDG mice were similar to those in the C57BL/6 mice, demonstrating that introduction of hCD3EDG in place of its mouse counterpart does not change the overall development, differentiation or distribution of these T cell subtypes in lymph node . Values are expressed as mean ± SEM.


Analysis of T cell activation stimulated with anti-CD3ε antibody in vitro



from clipboard


T cells (2×105) were isolated from splenocytes of C57BL/6 and B-hCD3EDG mice (n=3,16 week-old), and were incubated in the presence of anti-CD3ε antibody (2ug/ml) and anti-mCD28 antibody (5ug/ml) for 24h. T cell proliferation was tested by flow cytometry. T cell activation in B-hCD3EDG mice was significantly up-regulated by Anti-CD3ε antibody, similar to the activation level shown in C57BL/6 mice treated with anti-mCD3ε antibody.


Analysis of T cell activation stimulated with anti-CD3ε antibody in vitro

from clipboard


T cells (2×105) were isolated from splenocytes of C57BL/6 and B-hCD3EDG mice (n=3,16 week-old), and were incubated in the presence of anti-CD3ε antibody (2ug/ml) and anti-mCD28 antibody (5ug/ml) for 48h. T cell proliferation was tested by flow cytometry. T cell activation in B-hCD3EDG mice was significantly up-regulated by anti-hCD3ε antibody, similar to the activation level shown in C57BL/6 mice treated with anti-mCD3ε antibody.


Analysis of T cell activation stimulated with anti-CD3ε antibody in vitro



from clipboard


T cells (2×105) were isolated from splenocytes of C57BL/6 and B-hCD3EDG mice (n=3,16 week-old), and were incubated in the presence of anti-CD3ε antibody (2ug/ml) and anti-mCD28 antibody (5ug/ml) for 72h. T cell proliferation was tested by flow cytometry. T cell activation in B-hCD3EDG mice was significantly up-regulated by anti-hCD3ε antibody, similar to the activation level shown in C57BL/6 mice treated with anti-mCD3ε antibody.


Analysis of T cell activation stimulated with anti-CD3ε antibody in vitro


from clipboard


T cells (2×105) were isolated from the splenocytes of C57BL/6 and B-hCD3EDG mice (n=3, 16 week-old), and incubated in the presence of anti-CD3ε antibody (2ug/ml) and anti-mCD28 antibody (5ug/ml) for 24h, 48h and 72h. T cell proliferations were measured by flow cytometry. As a result, the T cell activation in B-hCD3EDG mice was specifically up-regulated by anti-hCD3ε antibody, similar to the level of activation in the anti-mCD3ε antibody-treated C57BL/6 mice. 


Analysis of T cell activation stimulated with anti-CD3ε antibody in vitro

from clipboard


T cells (2×105) were isolated from the splenocytes of C57BL/6 and B-hCD3EDG mice (n=3, 16 week-old), and incubated in the presence of anti-CD3ε antibody (2ug/ml) and anti-mCD28 antibody (5ug/ml) for 24h, 48h and 72h. T cell proliferations were measured by flow cytometry. As a result, the T cell activation in B-hCD3EDG mice was specifically up-regulated by anti-hCD3ε antibody, similar to the level of activation in the anti-mCD3ε antibody-treated C57BL/6 mice. 


Analysis of T cell activation stimulated with anti-CD3ε antibody in vitro

from clipboard


T cells (2×105) were isolated from the splenocytes of C57BL/6 and B-hCD3EDG mice (n=3, 16 week-old), incubated in the presence of anti-CD3ε antibody (2ug/ml) and anti-mCD28 antibody (5ug/ml) for 24h, 48h and 72h. IFN-γ and IL-2 productions were then tested using ELISA method. As a result, there is no significant difference regarding the production of IFN-γ and IL-2 between B-hCD3EDG mice and C57BL/6 mice. ND: not detectable. 

Cytotoxicity evaluation of CD3/BCMA bispecific antibody in vitro


from clipboard


Splenocytes of B-hCD3EDG mice were mixed with B-hBCMA MC38 and various concentrations of CD3/BCMA bispecific antibodies provided by the client were added. The killing activity was detected after 48 hours. When the ratio of effector cells to target cells  (E:T) were 20:1 and 40:1, the killing effect of the test sample was detected, but the killing effect was more obvious at 40:1.

Serum titers of OVA-specific antibodies 

from clipboard


B-hCD3EDG mice (n=5, 6 week-old) were immunized three times with OVA, 2 weeks apart. Blood samples were collected a week after immunization. (A) Quantification of serum subtypes of mice before immunization. (B) Serum titer test of mice after the second and third immunizations. The levels of OVA-specific antibodies titers of B-hCD3EDG mice before immunization were similar to those in C57BL/6 mice, and the specific antibody titers in the serum of each mouse were significantly increased after the third immunizations, demonstrating that introduction of hCD3EDG instead of its mouse counterpart did not affect the humoral immune response of mice. Values are expressed as mean ± SEM.

Blood routine test in B-hCD3EDG mice

from clipboard


Complete blood count (CBC). Blood from female C57BL/6 and B-hCD3EDG mice (n=8, 7-week-old) were collected and analyzed for CBC. There was no differences among any measurement between C57BL/6 and B-hCD3EDG mice, indicating that introduction of hCD3EDG in place of its mouse counterpart does not change blood cell composition and morphology. Values are expressed as mean ± SEM.


Blood chemistry of B-hCD3EDG mice

from clipboard


Blood chemistry tests of B-hCD3EDG mice. Serum from female C57BL/6 and B-hCD3EDG mice (n=8, 7-week-old) were collected and analyzed for levels of ALT (alanine aminotransferase) and AST (aspartate aminotransferase). There was no differences among any measurement between C57BL/6 and B-hCD3EDG mice, indicating that introduction of hCD3EDG in place of its mouse counterpart does not change ALT and AST levels or health of liver. Values are expressed as mean ± SEM.


In vivo efficacy of anti-hCD3ε antibody and anti-mPD-1 antibody in B-hCD3EDG mice

from clipboard


Antitumor activity of anti-hCD3ε antibody and anti-mPD-1 antibody in B-hCD3EDG mice. Murine colon cancer MC38 cells were subcutaneously implanted into B-hCD3EDG mice (female, 8 week-old, n=5). Mice were grouped when the tumor size was approximately 100±50mm3 , at which time they were treated with different antibodies with doses and schedules indicated in panel. (A) Tumor volume changes during treatment. (B) Body weight changes during treatment. As shown in panel A, anti-mPD-1 antibody significantly inhibited tumor growth in B-hCD3EDG mice, indicating their T cells function normally. However, in B-hCD3EDG mice, tumor growth was faster after anti-hCD3ε antibody treatment, which may be caused by activation induced cell death (AICD). As a result, the B-hCD3EDG mouse model is a powerful tool for in vivo CD3 antibody pharmacological efficacy studies. Values are expressed as mean ± SEM.


In vivo efficacy of anti-hCD3ε antibody and anti-mPD-1 antibody in B-hCD3EDG mice

from clipboard


T cells and B cells activation in vivo efficacy . Lymphocytes were isolated from peripheral blood at 48 hours after treatment. In the anti-hCD3ε antibody treatment group, the proportion of T cells was significantly decreased due to the activation induced cell death (AICD) effect caused by anti-hCD3ε antibody treatment. (A) The percentage of TCR-β + cells in total CD45+ cells after treatment.  (B) The percentage of CD19+ cells in total CD45+ cells after treatment. 


In vivo efficacy of anti-hCD3ε antibody and anti-mPD-1 antibody in B-hCD3EDG mice

from clipboard


T cells and B cells activation in vivo efficacy . Lymphocytes were isolated from tumor at 48 hours after treatment. In the anti-hCD3ε antibody treatment group, the proportion of T cells was significantly decreased due to the activation induced cell death (AICD) effect caused by anti-hCD3ε antibody treatment. (A) The percentage of TCR-β + cells in total CD45+ cells after treatment.  (B) The percentage of CD19+ cells in total CD45+ cells after treatment. 


In vivo efficacy of T cell bispecific antibody 


from clipboard


Antitumor activity of antibody X in B-hCD3EDG mice. B-hBCMA MC38 cells were subcutaneously implanted into B-hCD3EDG mice (female, 6 week-old, n=5). Mice were grouped when the tumor size was approximately 100mm3, at which time they were treated with antibody X provided by the client with doses and schedules indicated in panel A. (A) Tumor volume changes during treatment. (B) Body weight changes during treatment. As shown in panel A, antibodies X with different doses were efficacious in controlling tumor growth in B-hCD3EDG mice, demonstrating that the B-hCD3EDG mouse model is a powerful tool for in vivo efficacy study of T cell bispecific antibody. Values are expressed as mean ± SEM.



Efficacy evaluation of CD3 BsAbs in B-hCD3EDG mice



from clipboard

6- to 8-week-old female B-hCD3EDG mice, which are genetically modified to express human CD3E, CD3D, and CD3G, were purchased from Biocytogen Pharmaceuticals Co., Ltd (Beijing, China) and subcutaneously inoculated with MC38/BCMA cells. Tumor-bearing mice were randomized into groups and intravenously (i.v.) injected with vehicle or GR1803 when the average tumor volume reached approximately 50–100 mm3. Tumor volume was calculated as (length×width2)/2, and body weight was monitored as an indicator of general health. As shown in Fig 6A, GR1803 significantly inhibited the growth of MC38/BCMA tumors when mice were given a single intravenous injection of GR1803 at a dose of 0.1 or 0.3 mg/kg. The tumor-bearing mice tolerated these doses well, as evidenced by the absence of significant body weight loss during the course of the experiment in all groups. Consistent with this, GR1803 significantly down-regulated BCMA expression in tumor tissues, beginning at 4 h after injection and continuing for at least 10 days post-injection (Fig. 6B). The data comes from the client's literature.