首页 > 最新文献

Antibody Therapeutics最新文献

英文 中文
CAMEL NANOBODY-BASED B7-H3 CAR-T CELLS WITH HIGH EFFICACY AGAINST SOLID TUMORS 基于骆驼纳米体的b7-h3 car-t细胞对实体肿瘤具有高效的治疗作用
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.005
Dan Li, Ruixue Wang, Tianyuzhou Liang, Hua Ren, Chaelee Park, Chin-Hsien Tai, Weiming Ni, Jing Zhou, S. Mackay, Elijah Edmondson, Javed Khan, B. S. Croix, Mitchell Ho
Abstract Background and significance Chimeric antigen receptor (CAR)-T cell therapy shows promising potency for treating patients with hematological malignancies. However, follow-up data indicate that only 30% to 50% of these patients experience long-term disease control. In solid tumors, the B7-H3 transmembrane protein is an emerging target that harbors in its ectodomain two distinct epitope motifs - IgC and IgV. Here, we developed nanobody-based CAR-T cell strategy targeting B7-H3 and investigated its anti-tumor efficacy in xenograft mouse models. Methods We isolated anti-B7-H3 VHHs from our large dromedary camel VHH nanobody libraries with great diversity (> 1012 total) by phage display technology. The binding of isolated VHHs was validated by ELISA, flow cytometry, and Octet. A B7-H3 peptide library was synthesized to predict the epitope of select VHHs. Anti-tumor effect of B7-H3 CAR-T cells was determined via cell luciferase-based cell killing assay as well as xenograft mouse models. Two tumor models, human neuroblastoma and pancreatic adenocarcinoma, were used in the present study. Single-cell transcriptome RNA sequencing coupled with single T-cell functional proteomics analysis was used to analyze the functionality of nanobody-based B7-H3 CAR-T cells. Results We analyzed the isoforms of B7-H3 at the RNA and protein levels and validated that only 4IgB7-H3 is a therapeutic target as the dominant isoform in tumors. Targeting 4Ig isoform, we obtained a panel of high-affinity nanobodies cross-reactive to human, mouse, rat, and monkey. Furthermore, we demonstrated that CAR-T cells based on the nanobodies had potent antitumor activity against tumors with rigorous T cell signaling and significant tumor infiltration. Mechanistically, we uncovered the top-upregulated genes that might be critical for the persistence of polyfunctional CAR-T cells in the tumor microenvironment. Conclusions Our results provide a novel nanobody-based B7-H3 CAR-T product for use in solid tumor therapy.
摘要背景和意义嵌合抗原受体(CAR)-T细胞疗法在治疗血液系统恶性肿瘤方面显示出良好的疗效。然而,随访数据表明,这些患者中只有30%至50%经历了长期的疾病控制。在实体瘤中,B7-H3跨膜蛋白是一个新出现的靶点,其胞外结构域中含有两个不同的表位基序——IgC和IgV。在这里,我们开发了靶向B7-H3的基于纳米体的CAR-T细胞策略,并研究了其在异种移植小鼠模型中的抗肿瘤功效。方法利用噬菌体展示技术从我们的大型单峰骆驼VHH纳米体文库中分离出具有高度多样性(总数>1012)的抗B7-H3 VHH。通过ELISA、流式细胞术和Octet验证分离的VHH的结合。合成了一个B7-H3肽文库来预测选定VHH的表位。通过基于细胞萤光素酶的细胞杀伤试验以及异种移植物小鼠模型测定B7-H3 CAR-T细胞的抗肿瘤作用。本研究使用了两种肿瘤模型,人类神经母细胞瘤和胰腺癌。单细胞转录组RNA测序结合单T细胞功能蛋白质组学分析用于分析基于纳米体的B7-H3 CAR-T细胞的功能。结果我们在RNA和蛋白质水平上分析了B7-H3的亚型,并证实只有4IgB7-H3作为肿瘤的主要亚型是治疗靶点。针对4Ig亚型,我们获得了一组对人、小鼠、大鼠和猴子具有交叉反应性的高亲和力纳米体。此外,我们证明了基于纳米体的CAR-T细胞对肿瘤具有强大的抗肿瘤活性,具有严格的T细胞信号传导和显著的肿瘤浸润。从机制上讲,我们发现了可能对多功能CAR-T细胞在肿瘤微环境中的持久性至关重要的最高上调基因。结论我们的研究结果为固体肿瘤治疗提供了一种新的基于纳米体的B7-H3 CAR-T产品。
{"title":"CAMEL NANOBODY-BASED B7-H3 CAR-T CELLS WITH HIGH EFFICACY AGAINST SOLID TUMORS","authors":"Dan Li, Ruixue Wang, Tianyuzhou Liang, Hua Ren, Chaelee Park, Chin-Hsien Tai, Weiming Ni, Jing Zhou, S. Mackay, Elijah Edmondson, Javed Khan, B. S. Croix, Mitchell Ho","doi":"10.1093/abt/tbad014.005","DOIUrl":"https://doi.org/10.1093/abt/tbad014.005","url":null,"abstract":"Abstract Background and significance Chimeric antigen receptor (CAR)-T cell therapy shows promising potency for treating patients with hematological malignancies. However, follow-up data indicate that only 30% to 50% of these patients experience long-term disease control. In solid tumors, the B7-H3 transmembrane protein is an emerging target that harbors in its ectodomain two distinct epitope motifs - IgC and IgV. Here, we developed nanobody-based CAR-T cell strategy targeting B7-H3 and investigated its anti-tumor efficacy in xenograft mouse models. Methods We isolated anti-B7-H3 VHHs from our large dromedary camel VHH nanobody libraries with great diversity (> 1012 total) by phage display technology. The binding of isolated VHHs was validated by ELISA, flow cytometry, and Octet. A B7-H3 peptide library was synthesized to predict the epitope of select VHHs. Anti-tumor effect of B7-H3 CAR-T cells was determined via cell luciferase-based cell killing assay as well as xenograft mouse models. Two tumor models, human neuroblastoma and pancreatic adenocarcinoma, were used in the present study. Single-cell transcriptome RNA sequencing coupled with single T-cell functional proteomics analysis was used to analyze the functionality of nanobody-based B7-H3 CAR-T cells. Results We analyzed the isoforms of B7-H3 at the RNA and protein levels and validated that only 4IgB7-H3 is a therapeutic target as the dominant isoform in tumors. Targeting 4Ig isoform, we obtained a panel of high-affinity nanobodies cross-reactive to human, mouse, rat, and monkey. Furthermore, we demonstrated that CAR-T cells based on the nanobodies had potent antitumor activity against tumors with rigorous T cell signaling and significant tumor infiltration. Mechanistically, we uncovered the top-upregulated genes that might be critical for the persistence of polyfunctional CAR-T cells in the tumor microenvironment. Conclusions Our results provide a novel nanobody-based B7-H3 CAR-T product for use in solid tumor therapy.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41630739","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ESTABLISHMENT OF AN ULTRA HIGH-THROUGHPUT PROTEIN PRODUCTION PLATFORM (ULTRA 96) FOR EARLY STAGE IN VITRO STUDIES 建立用于早期体外研究的超高通量蛋白生产平台(ultra 96)
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.017
Kai Jiang, Huishuai Li, Chuanlong Tang, Dawei Zhang, Xuejian Zhang, Ying Zhang, Zhengyang Zhao, Junqing Zhou, Mengjie Lu, Yongsheng Xiao, Zhumei Feng, Jiansheng Wu
Abstract Proteins are known as the building blocks of life which play many critical roles in the body. In early stage drug discovery, numerous μg to mg scale high quality proteins need to be produced for in vitro functional studies and therapeutic evaluation, including antigens, monoclonal antibodies (mAb), bispecific antibodies (bsAbs) as well as other Fc fusion proteins. High-throughput production (HTP) of various proteins with high quality in very short time is a big challenge in industry. Here we present how WuXi Biologics has created a powerful ultra HTP protein production platform (termed as “Ultra 96”) that can automatically produce proteins within 3 weeks with one step purification at 1~3mL expression scale by using 96 and 24 well DWP. High titer of expression, high quality of products, short timeline as well as low cost make “Ultra 96” a robust platform which can empower our clients and dramatically accelerate their early stage drug discovery projects.
摘要蛋白质被认为是生命的基石,在人体中起着许多重要的作用。在药物发现的早期阶段,需要生产许多μg到mg级的高质量蛋白用于体外功能研究和治疗评估,包括抗原、单克隆抗体(mAb)、双特异性抗体(bsAbs)以及其他Fc融合蛋白。在短时间内高通量生产各种高质量蛋白质是工业上的一大挑战。在这里,我们介绍了药明生物如何创建一个强大的超HTP蛋白生产平台(称为“ultra 96”),该平台可以在3周内自动生产蛋白质,通过96和24孔DWP在1~3mL表达规模下一步纯化。高滴度,高质量的产品,短时间和低成本使“Ultra 96”成为一个强大的平台,可以授权我们的客户,并大大加快他们的早期药物发现项目。
{"title":"ESTABLISHMENT OF AN ULTRA HIGH-THROUGHPUT PROTEIN PRODUCTION PLATFORM (ULTRA 96) FOR EARLY STAGE IN VITRO STUDIES","authors":"Kai Jiang, Huishuai Li, Chuanlong Tang, Dawei Zhang, Xuejian Zhang, Ying Zhang, Zhengyang Zhao, Junqing Zhou, Mengjie Lu, Yongsheng Xiao, Zhumei Feng, Jiansheng Wu","doi":"10.1093/abt/tbad014.017","DOIUrl":"https://doi.org/10.1093/abt/tbad014.017","url":null,"abstract":"Abstract Proteins are known as the building blocks of life which play many critical roles in the body. In early stage drug discovery, numerous μg to mg scale high quality proteins need to be produced for in vitro functional studies and therapeutic evaluation, including antigens, monoclonal antibodies (mAb), bispecific antibodies (bsAbs) as well as other Fc fusion proteins. High-throughput production (HTP) of various proteins with high quality in very short time is a big challenge in industry. Here we present how WuXi Biologics has created a powerful ultra HTP protein production platform (termed as “Ultra 96”) that can automatically produce proteins within 3 weeks with one step purification at 1~3mL expression scale by using 96 and 24 well DWP. High titer of expression, high quality of products, short timeline as well as low cost make “Ultra 96” a robust platform which can empower our clients and dramatically accelerate their early stage drug discovery projects.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43240558","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
3H-3000, AN FGFR2B-SPECIFIC MONOCLONAL ANTIBODY, SHOWS SUPERIOR TUMOR KILLING IN VIVO 3h-3000是一种fgfr2b特异性单克隆抗体,在体内表现出优越的肿瘤杀伤能力
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.016
You Li, Jianwei Zhang, Ruiqin Wang, Xueying Yin, Susan Liu, Jufang Lin, Shaojing Hu, M. Cai
Abstract Background and significance Fibroblast growth factor receptor 2b (FGFR2b), a splice isoform of FGFR2, is found to overexpress across multiple cancers types and promotes dysregulated tyrosine kinase activation, resulting in tumor cell proliferation and unchecked malignancy. Cohort studies reveal FGFR2b overexpression and/or amplification in Gastric cancer (~30.2%), squamous non-small cell lung cancer (~20.8%), and endometrial carcinoma (~40%). About 80–85% patients who have advanced HER2-negative gastro-esophageal cancer undergoes approximately 12-14 months of median overall survival (mOS). Thus, there is an urgent need for novel and effective molecular targeted agents. With the recent advancement of mechanistic studies, however, FGFR2b is becoming the optimal target of multiple modalities such as mAb, bispecifics, or antibody-drug conjugate. Bemarituzumab, a FGFR2b blocking antibody, developed by Five Prime clinically demonstrates the safety of FGFR2b target and exhibits preliminary efficacy in advanced GE/GEJ adenocarcinoma patients with overexpression of FGFR2 in at least 5% or 10% of tumor cells. 3H Pharmaceuticals developed a FGFR2b-specific mAb, 3H-3000, of high-affinity, differentiated epitope and enhanced ADCC effect for FGFR2b-overexpressing gastric cancer or other FGFR2b-associated cancer types. In the meantime, we will also explore the versatility of this mAb in other format and in the combination with small molecules that will bring about a wider therapeutic spectrum. Method and Result 3H-3000 is a humanized IgG1kappa antibody with a single digit nM affinity for FGFR2b and no binding to other FGFR2 isoforms. 3H-3000 shows potent proliferation-inhibitive effects on FGFR2b-overexpressing human tumor cells. In gastric cancer cell line SNU16, 3H-3000 can fully inhibit FGF7-induced phosphorylation of FGFR2b and SNU16 proliferation in vitro. On top of signaling blocking, we strongly believe efficacy of antibody-dependent cell cytotoxicity (ADCC) is another viable mechanism for growth factor-targeting therapy. Therefore, 3H-3000 was further engineered with a 20-fold ADCC enhancement which is validated through a CD16-F158 variant-based reporter system. In FGFR2b-overexpressing human gastric cancer SNU16 or OCUM-2M xenograft model, 3H-3000 was shown to inhibit tumor growth and even induce regression, which is rather unexpected given the relative low expression of FGFR2b on SNU16 and none of the competitors exhibit similar phenomenon. With potent efficacy of tumor inhibition and killing, and excellent biophysical characteristics and developability of 3H-3000, we expect to push the molecule to clinical development in the middle of 2024. Conclusion 3H-3000 is a potent FGFR2b blocker with well-defined ADCC efficacy enhancement. It demonstrates strong blocking activity of FGFR2 signaling, strikingly enhanced ADCC activity in vitro and potent efficacy of tumor inhibition in vivo. These data strongly support its clinical development in FGFR2b
背景和意义成纤维细胞生长因子受体2b (FGFR2b)是FGFR2的剪接异构体,在多种癌症类型中被发现过表达,并促进酪氨酸激酶激活失调,导致肿瘤细胞增殖和恶性肿瘤。队列研究显示FGFR2b在胃癌(~30.2%)、鳞状非小细胞肺癌(~20.8%)和子宫内膜癌(~40%)中过表达和/或扩增。约80-85%的晚期her2阴性胃食管癌患者的中位总生存期(mOS)约为12-14个月。因此,迫切需要一种新型有效的分子靶向药物。然而,随着最近机制研究的进展,FGFR2b正成为多种模式(如单抗、双特异性或抗体-药物偶联)的最佳靶点。Bemarituzumab是由Five Prime公司开发的一种FGFR2b阻断抗体,临床证明了FGFR2b靶点的安全性,并在至少5%或10%的肿瘤细胞中FGFR2过表达的晚期GE/GEJ腺癌患者中显示出初步疗效。3H制药公司开发了一种fgfr2b特异性单抗3H-3000,具有高亲和力,分化表位和增强ADCC作用,用于fgfr2b过表达的胃癌或其他fgfr2b相关癌症类型。与此同时,我们还将探索该单抗在其他形式的多功能性,并与小分子结合,从而带来更广泛的治疗范围。方法与结果3H-3000是一种人源IgG1kappa抗体,对FGFR2b具有个位数nM亲和力,不与其他FGFR2亚型结合。3H-3000对过表达fgfr2b的人肿瘤细胞显示出强大的增殖抑制作用。在胃癌细胞系SNU16中,3H-3000能在体外完全抑制fgf7诱导的FGFR2b磷酸化和SNU16的增殖。除了信号阻断之外,我们坚信抗体依赖性细胞毒性(ADCC)的功效是生长因子靶向治疗的另一个可行机制。因此,3H-3000进一步增强了20倍的ADCC,并通过基于CD16-F158变异的报告系统进行了验证。在过表达FGFR2b的人胃癌SNU16或OCUM-2M异种移植模型中,3H-3000显示出抑制肿瘤生长甚至诱导肿瘤消退的作用,这是相当出乎意料的,因为FGFR2b在SNU16上的表达相对较低,并且没有竞争对手出现类似现象。由于3H-3000具有强大的肿瘤抑制和杀伤功效,以及优异的生物物理特性和可开发性,我们预计在2024年中期将该分子推向临床开发。结论3H-3000是一种有效的FGFR2b阻滞剂,具有明显的ADCC增强作用。它显示出强大的FGFR2信号阻断活性,在体外显著增强ADCC活性,在体内具有强大的肿瘤抑制功效。这些数据有力地支持了其在过表达fgfr2b的癌症中的临床开发。同时,3H制药公司也在开发靶向FGFR2突变或融合的FGFR2选择性酪氨酸激酶抑制剂。我们提出小分子FGFR2抑制剂和FGFR2b特异性单抗的组合将显著拓宽异质性肿瘤的治疗谱,其机制以FGFR2b改变为中心,包括突变、融合和过表达。
{"title":"3H-3000, AN FGFR2B-SPECIFIC MONOCLONAL ANTIBODY, SHOWS SUPERIOR TUMOR KILLING IN VIVO","authors":"You Li, Jianwei Zhang, Ruiqin Wang, Xueying Yin, Susan Liu, Jufang Lin, Shaojing Hu, M. Cai","doi":"10.1093/abt/tbad014.016","DOIUrl":"https://doi.org/10.1093/abt/tbad014.016","url":null,"abstract":"Abstract Background and significance Fibroblast growth factor receptor 2b (FGFR2b), a splice isoform of FGFR2, is found to overexpress across multiple cancers types and promotes dysregulated tyrosine kinase activation, resulting in tumor cell proliferation and unchecked malignancy. Cohort studies reveal FGFR2b overexpression and/or amplification in Gastric cancer (~30.2%), squamous non-small cell lung cancer (~20.8%), and endometrial carcinoma (~40%). About 80–85% patients who have advanced HER2-negative gastro-esophageal cancer undergoes approximately 12-14 months of median overall survival (mOS). Thus, there is an urgent need for novel and effective molecular targeted agents. With the recent advancement of mechanistic studies, however, FGFR2b is becoming the optimal target of multiple modalities such as mAb, bispecifics, or antibody-drug conjugate. Bemarituzumab, a FGFR2b blocking antibody, developed by Five Prime clinically demonstrates the safety of FGFR2b target and exhibits preliminary efficacy in advanced GE/GEJ adenocarcinoma patients with overexpression of FGFR2 in at least 5% or 10% of tumor cells. 3H Pharmaceuticals developed a FGFR2b-specific mAb, 3H-3000, of high-affinity, differentiated epitope and enhanced ADCC effect for FGFR2b-overexpressing gastric cancer or other FGFR2b-associated cancer types. In the meantime, we will also explore the versatility of this mAb in other format and in the combination with small molecules that will bring about a wider therapeutic spectrum. Method and Result 3H-3000 is a humanized IgG1kappa antibody with a single digit nM affinity for FGFR2b and no binding to other FGFR2 isoforms. 3H-3000 shows potent proliferation-inhibitive effects on FGFR2b-overexpressing human tumor cells. In gastric cancer cell line SNU16, 3H-3000 can fully inhibit FGF7-induced phosphorylation of FGFR2b and SNU16 proliferation in vitro. On top of signaling blocking, we strongly believe efficacy of antibody-dependent cell cytotoxicity (ADCC) is another viable mechanism for growth factor-targeting therapy. Therefore, 3H-3000 was further engineered with a 20-fold ADCC enhancement which is validated through a CD16-F158 variant-based reporter system. In FGFR2b-overexpressing human gastric cancer SNU16 or OCUM-2M xenograft model, 3H-3000 was shown to inhibit tumor growth and even induce regression, which is rather unexpected given the relative low expression of FGFR2b on SNU16 and none of the competitors exhibit similar phenomenon. With potent efficacy of tumor inhibition and killing, and excellent biophysical characteristics and developability of 3H-3000, we expect to push the molecule to clinical development in the middle of 2024. Conclusion 3H-3000 is a potent FGFR2b blocker with well-defined ADCC efficacy enhancement. It demonstrates strong blocking activity of FGFR2 signaling, strikingly enhanced ADCC activity in vitro and potent efficacy of tumor inhibition in vivo. These data strongly support its clinical development in FGFR2b","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45865901","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
YEAST SURFACE DISPLAY OF FULL-LENGTH IGG FOR ENGINEERING MONOCLONAL AND BISPECIFIC ANTIBODIES 工程单克隆和双特异性抗体全长IGG的酵母表面展示
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.022
Youwei Jiang
Abstract The single-chain variable fragment (scFv) and antigen-binding fragment (Fab) are widely used in yeast surface display for antibody engineering. However, these non-IgG antibody fragments are not always well-expressed or functional. We developed a novel approach to display full-length IgG on yeast surfaces for engineering monoclonal and bispecific antibodies. The yeast surface display of full-length IgG was generated by transforming glycoengineered yeast P. pastoris with plasmids encoding for anchor protein, heavy chains, and light chains. IgG-displaying yeast cells were incubated with the fluorophore antigen and secondary detection antibody for FACS sorting. The displayed antibody with bound antigen and detection antibody was detected by FACS sorting based on both antibody affinity and display level. FACS sorting could eliminate the deviation caused by expression levels. The yeast surface display of IgG-like bispecific antibodies was also constructed, which exhibited specific binding of two distinct fluorophore antigens in flow-cytometry analysis. Thus, our yeast surface display of full-length IgG can be used to screen light chain libraries to isolate common light chains for bispecific antibodies. To assess antigen-binding efficiency between different displaying formats, an antibody was respectively displayed as full-length IgG or Fab on the surface of yeast cells for flow-cytometry analysis. Cells displaying full-length IgG showed a higher level of fluorescence than those displaying Fab fragments, indicating that yeast surface display of full-length IgG is superior for high-throughput screening. To test the specificity of the displayed antibody, we performed a demo experiment in which TNF-binding adalimumab-displaying cells were mixed with trastuzumab-displaying cells at a 1 to 1,000,000 ratio, mimicking the immune library. After three consecutive rounds of sorting, the yeast cells with high fluorescence were plated on a selective medium and the individual antibody clones were sequenced. All ten sequenced clones were confirmed to be adalimumab, demonstrating the maintenance of genotype-phenotype linkage for library screening in yeast surface display of full-length IgG. A mutation library was also generated from the initial hit and displayed at the surface of yeast cells for the screening of higher affinity maturation antibodies by FACS. A pool of high-affinity binders was plated on a selective medium. Individual clones were analyzed by flow cytometry and sequenced to identify unique antibodies with higher affinity. In summary, full-length IgG antibodies can be displayed on the yeast cell surface, mimicking their native forms in molecular structure and biophysical properties. The technique combines the high throughput of yeast display with mammalian-cell quality control. This novel approach can be used to engineer monoclonal and bispecific antibodies for high affinity and improved developability profiles.
单链可变片段(scFv)和抗原结合片段(Fab)在酵母表面展示抗体工程中应用广泛。然而,这些非igg抗体片段并不总是表达良好或具有功能。我们开发了一种在酵母表面显示全长IgG的新方法,用于工程单克隆和双特异性抗体。用编码锚蛋白、重链和轻链的质粒转化糖工程酵母酵母,在酵母表面显示全长IgG。将显示igg的酵母细胞与荧光团抗原和二检抗体孵育,进行FACS分选。结合抗原的显示抗体和检测抗体采用FACS分选,根据抗体亲和力和显示水平进行检测。FACS分选可消除表达水平引起的偏差。构建了igg样双特异性抗体的酵母表面显示,在流式细胞术分析中显示出两种不同的荧光团抗原的特异性结合。因此,我们的酵母表面展示的全长IgG可用于筛选轻链文库,以分离双特异性抗体的常见轻链。为了评估不同显示格式之间的抗原结合效率,将抗体分别以全长IgG或Fab的形式显示在酵母细胞表面进行流式细胞术分析。显示全长IgG的细胞比显示Fab片段的细胞荧光水平更高,表明酵母表面显示全长IgG更有利于高通量筛选。为了测试所显示抗体的特异性,我们进行了一个演示实验,将tnf结合的阿达木单抗显示细胞与曲妥珠单抗显示细胞以1比1,000,000的比例混合,模拟免疫文库。连续三轮分选后,将高荧光酵母细胞置于选择性培养基上,对单个抗体克隆进行测序。10个测序克隆均为阿达木单抗,证明在酵母表面展示全长IgG的文库筛选中维持了基因型-表型连锁。从初始命中生成突变文库,并在酵母细胞表面显示,用于FACS筛选高亲和力成熟抗体。在选择性培养基上镀上一池高亲和力的粘合剂。通过流式细胞术对单个克隆进行分析和测序,鉴定出具有较高亲和力的独特抗体。综上所述,全长IgG抗体可以在酵母细胞表面显示,在分子结构和生物物理性质上模仿其天然形式。该技术将酵母的高通量展示与哺乳动物细胞质量控制相结合。这种新方法可用于设计高亲和力和改进的可开发性的单克隆和双特异性抗体。
{"title":"YEAST SURFACE DISPLAY OF FULL-LENGTH IGG FOR ENGINEERING MONOCLONAL AND BISPECIFIC ANTIBODIES","authors":"Youwei Jiang","doi":"10.1093/abt/tbad014.022","DOIUrl":"https://doi.org/10.1093/abt/tbad014.022","url":null,"abstract":"Abstract The single-chain variable fragment (scFv) and antigen-binding fragment (Fab) are widely used in yeast surface display for antibody engineering. However, these non-IgG antibody fragments are not always well-expressed or functional. We developed a novel approach to display full-length IgG on yeast surfaces for engineering monoclonal and bispecific antibodies. The yeast surface display of full-length IgG was generated by transforming glycoengineered yeast P. pastoris with plasmids encoding for anchor protein, heavy chains, and light chains. IgG-displaying yeast cells were incubated with the fluorophore antigen and secondary detection antibody for FACS sorting. The displayed antibody with bound antigen and detection antibody was detected by FACS sorting based on both antibody affinity and display level. FACS sorting could eliminate the deviation caused by expression levels. The yeast surface display of IgG-like bispecific antibodies was also constructed, which exhibited specific binding of two distinct fluorophore antigens in flow-cytometry analysis. Thus, our yeast surface display of full-length IgG can be used to screen light chain libraries to isolate common light chains for bispecific antibodies. To assess antigen-binding efficiency between different displaying formats, an antibody was respectively displayed as full-length IgG or Fab on the surface of yeast cells for flow-cytometry analysis. Cells displaying full-length IgG showed a higher level of fluorescence than those displaying Fab fragments, indicating that yeast surface display of full-length IgG is superior for high-throughput screening. To test the specificity of the displayed antibody, we performed a demo experiment in which TNF-binding adalimumab-displaying cells were mixed with trastuzumab-displaying cells at a 1 to 1,000,000 ratio, mimicking the immune library. After three consecutive rounds of sorting, the yeast cells with high fluorescence were plated on a selective medium and the individual antibody clones were sequenced. All ten sequenced clones were confirmed to be adalimumab, demonstrating the maintenance of genotype-phenotype linkage for library screening in yeast surface display of full-length IgG. A mutation library was also generated from the initial hit and displayed at the surface of yeast cells for the screening of higher affinity maturation antibodies by FACS. A pool of high-affinity binders was plated on a selective medium. Individual clones were analyzed by flow cytometry and sequenced to identify unique antibodies with higher affinity. In summary, full-length IgG antibodies can be displayed on the yeast cell surface, mimicking their native forms in molecular structure and biophysical properties. The technique combines the high throughput of yeast display with mammalian-cell quality control. This novel approach can be used to engineer monoclonal and bispecific antibodies for high affinity and improved developability profiles.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43672685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A NOVEL B7H3/NKP30 BISPECIFIC NK CELL ENGAGER FOR CANCER IMMUNOTHERAPY 一种用于癌症免疫治疗的新型b7h3 / nkp30双特异性nk细胞接合剂
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.013
Xiaoling Jiang, Chongbing Wu, Zi Chen, Liusong Yin
Abstract Background and Significance Biological therapies harnessing the adaptive immune system have achieved a great success, given the clinical efficacy of CAR-T therapies and CD3 based T cell engagers in treating hematologic tumors, and the application of T cell immune checkpoint inhibitors in various indications. However, the challenges still exist due to the limitation of CAR-T therapies and T cell engagers in treating solid tumors. Side effects such as cytokine release storm and neurotoxicity remain a concern in the selection of CAR-T therapies and CD3-based T cell engagers as therapeutics. For PD1/L1-based immunotherapy, the relatively low objective response rate (ORR), short progression-free survival (PFS) of patients, and drug resistance and recurrence especially for solid malignancy, are the major clinical challenges. In contrast, innate effector cells such as NK cells and macrophages are naturally existing in human body as front line to defeat general pathogens and cancers,which will have a better safety profile. Also, NK cell activation is not limited by the antigen presentation of the MHC on the cell surface which makes it has broader anti-tumor effects. However, the innate effector cell based therapy is facing the limitation of low cell number, poor in vitro activation, and short effective duration in vivo. SunHo has generated a B7H3/NKp30 NK cell engager bispecific antibody which can directly activate and enrich NK cells to the TME when used alone or enhance the efficacy when used in combination NK cell therapy. NKp30 is a potent NK cell activation receptor with wide and persistent expression compared with other NK markers. B7H3 as a tumor associate antigen is also widely expressed on many kinds of tumor cells with great potential for multiple indications, especially for solid tumors. Methods Three anti-NKp30 VHHs were identified from Alpaca immunized library. The VHHs were fused to the N-terminus or C-terminus of an anti-B7H3 mAb heavy chain with different IgG isotypes (IgG1 or IgG4). The binding activity to NK cells was evaluated by FACS. IFNγ level was detected in the NK cell activation assay. The NK cell mediated target cell killing was evaluated using either NK92MI-cd16a or Primary NK cells as effector cells. To evaluate the non-specific self-killing of NK cell without target cell, the B7H3/NKp30 candidates were incubated with NK92MI-cd16a and cell lysis percentage were calculated. NSG mice bearing Hs.746T tumors were used to evaluate the in vivo efficacy of B7H3/NKp30 candidates. Results The B7H3/NKp30 candidates showed good binding activity and better activation to NK cells compared with benchmarks. Notably, the B7H3/NKp30 candidate IAN0982-VHH25 with NKp30 fused to the C-terminal with an IgG1 isotype showed lowest risk of non-specific NK cell killing. In the in vivo study, B7H3/NKp30 candidates in combination with 1x106 NK cells showed excellent anti-tumor activity with TGI over 95%. And we didn’t observe any significant ch
背景与意义鉴于CAR-T疗法和基于CD3的T细胞参与物治疗血液肿瘤的临床疗效,以及T细胞免疫检查点抑制剂在各种适应症中的应用,利用适应性免疫系统的生物疗法取得了巨大的成功。然而,由于CAR-T疗法和T细胞参与治疗实体瘤的局限性,挑战仍然存在。诸如细胞因子释放风暴和神经毒性等副作用仍然是选择CAR-T疗法和基于cd3的T细胞参与器作为治疗方法时需要关注的问题。对于基于PD1/ l1的免疫治疗,相对较低的客观反应率(ORR)、患者较短的无进展生存期(PFS)以及耐药和复发(尤其是实体恶性肿瘤)是临床面临的主要挑战。而天然效应细胞如NK细胞、巨噬细胞等天然存在于人体内,作为对抗一般病原体和癌症的第一线,具有更好的安全性。此外,NK细胞的活化不受细胞表面MHC抗原呈递的限制,这使其具有更广泛的抗肿瘤作用。然而,基于先天效应细胞的治疗面临着细胞数量少、体外激活能力差、体内有效时间短的限制。SunHo研制了一种B7H3/NKp30 NK细胞接合物双特异性抗体,单独使用时可直接激活并富集NK细胞至TME,联合NK细胞治疗时可增强疗效。与其他NK标志物相比,NKp30是一种有效的NK细胞激活受体,具有广泛和持久的表达。B7H3作为肿瘤相关抗原也广泛表达于多种肿瘤细胞上,具有很大的多适应症潜力,尤其是实体瘤。方法从羊驼免疫文库中鉴定3个抗nkp30的vhh。将这些vhh融合到具有不同IgG同型(IgG1或IgG4)的抗b7h3单抗重链的n端或c端。流式细胞仪检测其与NK细胞的结合活性。NK细胞活化法检测IFNγ水平。使用NK92MI-cd16a或原代NK细胞作为效应细胞,评估NK细胞介导的靶细胞杀伤作用。为了评估无靶细胞的NK细胞非特异性自杀伤作用,我们将B7H3/NKp30候选细胞与NK92MI-cd16a孵育,计算细胞裂解率。采用hs746t肿瘤NSG小鼠,评价B7H3/NKp30候选物的体内疗效。结果B7H3/NKp30候选物与NK细胞具有良好的结合活性和活化活性。值得注意的是,B7H3/NKp30候选基因IAN0982-VHH25将NKp30融合到IgG1同型的c端,显示出最低的非特异性NK细胞杀伤风险。在体内研究中,B7H3/NKp30候选物与1x106 NK细胞联合表现出良好的抗肿瘤活性,TGI超过95%。我们没有观察到动物的体重有任何明显的变化,这表明候选药物的安全性很好。此外,IAN0982-VHH25即使在NK细胞数量较低(5x105)的情况下,也表现出良好的抗肿瘤效果。我们还为IAN0982-VHH25生成了三个稳定转染的小细胞池,用于商业稳定细胞系的开发。结论IAN0982-VHH25具有良好的NK细胞活化作用和体内抗肿瘤作用,同时具有较低的非特异性NK细胞杀伤风险,是肿瘤免疫治疗中首个B7H3/NKp30双特异性NK细胞参与剂。它有望解除当前适应性和先天效应细胞为基础的治疗的局限性。
{"title":"A NOVEL B7H3/NKP30 BISPECIFIC NK CELL ENGAGER FOR CANCER IMMUNOTHERAPY","authors":"Xiaoling Jiang, Chongbing Wu, Zi Chen, Liusong Yin","doi":"10.1093/abt/tbad014.013","DOIUrl":"https://doi.org/10.1093/abt/tbad014.013","url":null,"abstract":"Abstract Background and Significance Biological therapies harnessing the adaptive immune system have achieved a great success, given the clinical efficacy of CAR-T therapies and CD3 based T cell engagers in treating hematologic tumors, and the application of T cell immune checkpoint inhibitors in various indications. However, the challenges still exist due to the limitation of CAR-T therapies and T cell engagers in treating solid tumors. Side effects such as cytokine release storm and neurotoxicity remain a concern in the selection of CAR-T therapies and CD3-based T cell engagers as therapeutics. For PD1/L1-based immunotherapy, the relatively low objective response rate (ORR), short progression-free survival (PFS) of patients, and drug resistance and recurrence especially for solid malignancy, are the major clinical challenges. In contrast, innate effector cells such as NK cells and macrophages are naturally existing in human body as front line to defeat general pathogens and cancers,which will have a better safety profile. Also, NK cell activation is not limited by the antigen presentation of the MHC on the cell surface which makes it has broader anti-tumor effects. However, the innate effector cell based therapy is facing the limitation of low cell number, poor in vitro activation, and short effective duration in vivo. SunHo has generated a B7H3/NKp30 NK cell engager bispecific antibody which can directly activate and enrich NK cells to the TME when used alone or enhance the efficacy when used in combination NK cell therapy. NKp30 is a potent NK cell activation receptor with wide and persistent expression compared with other NK markers. B7H3 as a tumor associate antigen is also widely expressed on many kinds of tumor cells with great potential for multiple indications, especially for solid tumors. Methods Three anti-NKp30 VHHs were identified from Alpaca immunized library. The VHHs were fused to the N-terminus or C-terminus of an anti-B7H3 mAb heavy chain with different IgG isotypes (IgG1 or IgG4). The binding activity to NK cells was evaluated by FACS. IFNγ level was detected in the NK cell activation assay. The NK cell mediated target cell killing was evaluated using either NK92MI-cd16a or Primary NK cells as effector cells. To evaluate the non-specific self-killing of NK cell without target cell, the B7H3/NKp30 candidates were incubated with NK92MI-cd16a and cell lysis percentage were calculated. NSG mice bearing Hs.746T tumors were used to evaluate the in vivo efficacy of B7H3/NKp30 candidates. Results The B7H3/NKp30 candidates showed good binding activity and better activation to NK cells compared with benchmarks. Notably, the B7H3/NKp30 candidate IAN0982-VHH25 with NKp30 fused to the C-terminal with an IgG1 isotype showed lowest risk of non-specific NK cell killing. In the in vivo study, B7H3/NKp30 candidates in combination with 1x106 NK cells showed excellent anti-tumor activity with TGI over 95%. And we didn’t observe any significant ch","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41445974","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DEVELOPMENT OF A SELECTIVE CD16A-BASED NK CELL ENGAGER UTILIZING ANTIBODIES TARGETING A SINGLE AMINO ACID VARIATION 利用针对单个氨基酸变异的抗体开发基于cd16的选择性NK细胞接合器
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.012
Wuxiang Liao, Christine Tumanut, Lin Li, A. Corper, D. Challa, Alex Chang, Hydari Begum, Elinaz Farokhi, C. Woods, Xiaomin Fan
Abstract Background and Significance Natural killer (NK) cells play a vital role in the human innate immune system and are being explored as a promising approach for cancer immunotherapy. Of particular interest are NK cell engagers that can target and activate NK cells to attack cancer cells. In this study, we developed novel NK cell engagers by targeting the NK cell activating receptor CD16a using antibodies that selectively distinguish between CD16a on NK cells and CD16b on granulocytes, which are highly homologous to each other. Methods and Results To generate antibodies with high developability, we employed a rational design approach to construct large yeast display libraries of human antibodies. This approach was based on the analysis of a deep sequencing dataset of human antibodies from over 500 individuals, which allowed us to determine the natural amino acid usage patterns of human antibody CDRs and mimic human antibody repertoires. Through screening these libraries, we discovered two classes of antibody clones that selectively recognize CD16a without cross-reactivity to CD16b. Epitope mapping revealed that a single amino acid difference confers over 10,000-fold selectivity for one class of antibody clones, while for the other class a second unique epitope on CD16a was identified. To evaluate the activity of these antibody clones, we produced bispecific antibody clones with one arm targeting CD16a and the other arm targeting a tumor-associated antigen (TAA). Our results demonstrated potent tumor cell-dependent activation of NK cells and effective killing of tumor cells. Several of these antibodies had greatly enhanced resistance to human IgG inhibition in killing target cells. Significantly, our anti-CD16a antibody clones exhibited superior performance compared to leading reference anti-CD16a clones in two distinct NK cell engager formats. This included higher affinity for CD16a, higher thermostability, and more potent killing activity both in the absence and presence of 10 mg/mL human IgGs as competitors. Conclusion Our findings indicate that anti-CD16a antibody-based NK cell engagers have significant potential for cancer immunotherapies.
摘要背景与意义自然杀伤细胞(NK)在人类先天免疫系统中发挥着至关重要的作用,是癌症免疫治疗的一种有前景的方法。特别感兴趣的是可以靶向并激活NK细胞以攻击癌症细胞的NK细胞掺合物。在这项研究中,我们通过靶向NK细胞激活受体CD16a,使用选择性区分NK细胞上CD16a和粒细胞上CD16b的抗体,开发了新的NK细胞接合剂,这两种抗体彼此高度同源。方法和结果为了产生具有高可开发性的抗体,我们采用合理的设计方法构建了大型的人类抗体酵母展示库。这种方法基于对来自500多个个体的人类抗体的深度测序数据集的分析,这使我们能够确定人类抗体CDR的天然氨基酸使用模式并模拟人类抗体库。通过筛选这些文库,我们发现了两类选择性识别CD16a而对CD16b没有交叉反应性的抗体克隆。表位定位显示,单个氨基酸差异对一类抗体克隆具有超过10000倍的选择性,而对另一类抗体则鉴定出CD16a上的第二个独特表位。为了评估这些抗体克隆的活性,我们制备了双特异性抗体克隆,其中一只臂靶向CD16a,另一只臂则靶向肿瘤相关抗原(TAA)。我们的结果证明了NK细胞的有效肿瘤细胞依赖性激活和肿瘤细胞的有效杀伤。这些抗体中的几种在杀死靶细胞时大大增强了对人IgG抑制的抵抗力。值得注意的是,在两种不同的NK细胞接合形式中,与领先的参考抗CD16a克隆相比,我们的抗CD16a抗体克隆表现出优异的性能。这包括在不存在和存在10mg/mL人IgG作为竞争对手的情况下,对CD16a具有更高的亲和力、更高的热稳定性和更有效的杀伤活性。结论我们的研究结果表明,抗CD16a抗体的NK细胞结合物在癌症免疫治疗中具有显著的潜力。
{"title":"DEVELOPMENT OF A SELECTIVE CD16A-BASED NK CELL ENGAGER UTILIZING ANTIBODIES TARGETING A SINGLE AMINO ACID VARIATION","authors":"Wuxiang Liao, Christine Tumanut, Lin Li, A. Corper, D. Challa, Alex Chang, Hydari Begum, Elinaz Farokhi, C. Woods, Xiaomin Fan","doi":"10.1093/abt/tbad014.012","DOIUrl":"https://doi.org/10.1093/abt/tbad014.012","url":null,"abstract":"Abstract Background and Significance Natural killer (NK) cells play a vital role in the human innate immune system and are being explored as a promising approach for cancer immunotherapy. Of particular interest are NK cell engagers that can target and activate NK cells to attack cancer cells. In this study, we developed novel NK cell engagers by targeting the NK cell activating receptor CD16a using antibodies that selectively distinguish between CD16a on NK cells and CD16b on granulocytes, which are highly homologous to each other. Methods and Results To generate antibodies with high developability, we employed a rational design approach to construct large yeast display libraries of human antibodies. This approach was based on the analysis of a deep sequencing dataset of human antibodies from over 500 individuals, which allowed us to determine the natural amino acid usage patterns of human antibody CDRs and mimic human antibody repertoires. Through screening these libraries, we discovered two classes of antibody clones that selectively recognize CD16a without cross-reactivity to CD16b. Epitope mapping revealed that a single amino acid difference confers over 10,000-fold selectivity for one class of antibody clones, while for the other class a second unique epitope on CD16a was identified. To evaluate the activity of these antibody clones, we produced bispecific antibody clones with one arm targeting CD16a and the other arm targeting a tumor-associated antigen (TAA). Our results demonstrated potent tumor cell-dependent activation of NK cells and effective killing of tumor cells. Several of these antibodies had greatly enhanced resistance to human IgG inhibition in killing target cells. Significantly, our anti-CD16a antibody clones exhibited superior performance compared to leading reference anti-CD16a clones in two distinct NK cell engager formats. This included higher affinity for CD16a, higher thermostability, and more potent killing activity both in the absence and presence of 10 mg/mL human IgGs as competitors. Conclusion Our findings indicate that anti-CD16a antibody-based NK cell engagers have significant potential for cancer immunotherapies.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46124421","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
REVOLUTIONIZING ANTIBODY DISCOVERY INDUSTRY WITH HIGHLY EFFICIENT AND ACCURATE AI-BASED EPITOPE-SPECIFIC ANTIBODY DE NOVO DESIGN WORKFLOW 通过高效准确的基于AI的表位特异性抗体从头设计工作流程,彻底改变抗体发现行业
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.024
Tianyuan Wang, Xiangrui Gao, Zhe Huai, Zhaohui Gong, Ting Mao, Xuezhe Fan, Xingxing Wu, Zhiyuan Duan, Xiaodong Wang, Jiewen Du, Mengcheng Yao, Xin Li, Min Wu, Zonghu Wang, Lin Zhang, Junjie Zhang, Wenbo Cao, Kai Yan, Yujie Fang, Shixiang Ma, Kun Yang, Lili Wu, F. An, Yezhou Yang, L. Lai, Xiaolu Huang
Abstract Background and significance The global antibody drug market is worth over $200 billion in 2021 and is expected to reach $380 billion by 2030. Antibody discovery is one of the most critical steps that determine the crucial properties of antibody drugs, such as efficacy, safety, and developability. Traditional methods based on mouse immunization have many drawbacks limiting drug discovery, which include long time periods, high costs, inability to target function-specific epitopes, unsuitable for low immunogenic and difficult-to-prepare antigens, the need to sacrifice mice, the need for further humanization to reduce immunogenicity, and so on. Here we report an antibody de novo design computational workflow that utilizes high-quality internally produced antibody data and advanced AI models. Using this workflow, we can de novo design antibodies that bind to user-specified functional epitopes with high affinity and specificity. Compared with classical wet-lab methods, the entire process is shortened from several months to several days and suitable for low immunogenicity and difficult-to-prepare antigens. It is particularly noteworthy that due to the use of humanized mouse-generated antibodies (Renlite bearing common light chain from Biocytogen) as training data for AI models, the designed antibodies have a high degree of humanization and good developability, effectively avoiding issues such as ADA and aggregation in subsequent processes. Methods First, with the help of Renlite, we comprehensively combined mouse immunization, B cell sorting with FACS, NGS single-cell sequencing, and bioinformatics analysis to internally generate a large amount of high-quality antibody sequence data. Second, we developed AI models for antigen-specific antibody selection and epitope prediction (bioRxiv, 2022: 2022.12. 22.521634.) to mine antigen-specific antibodies and corresponding antigen epitopes in the data. Based on the processed high-quality data, we trained an affinity prediction model that can accurately predict whether an antigen epitope and antibody sequence pair can bind to each other. Besides, using the sequence data, we trained an antibody sequence pre-training language model (bioRxiv, 2023: 2023.01. 19.524683.), which can generate high-quality antibody sequences to simulate the antibodies produced by mouse immunization. Finally, integrating the above AI models, we established an antibody de novo design computational workflow to simulate the biological process of antibody generation and affinity maturation in the mouse immune system, which can be seen as a “DigitalMouse”. Results In a test case, 1 million antibodies were designed aiming at binding to specific epitope of an antigen. 10 antibodies were selected and expressed. Binding affinity was determined using BLI. Two antibodies out of 10 had KD of 194 nM and 336 nM, respectively, with a concentration dependent signal increase on BLI. These antibodies have great potential as the starting point of
背景与意义2021年全球抗体药物市场价值超过2000亿美元,预计到2030年将达到3800亿美元。抗体发现是决定抗体药物关键特性(如有效性、安全性和可开发性)的最关键步骤之一。传统的基于小鼠免疫的方法存在许多限制药物发现的缺点,包括时间长、成本高、无法靶向功能特异性表位、不适合低免疫原性和难以制备的抗原、需要牺牲小鼠、需要进一步人源化以降低免疫原性等。在这里,我们报告了一个抗体从头设计计算工作流,利用高质量的内部产生的抗体数据和先进的人工智能模型。利用这一工作流程,我们可以重新设计与用户指定的功能表位结合的抗体,具有高亲和力和特异性。与传统的湿法相比,整个过程从几个月缩短到几天,适用于低免疫原性和难以制备的抗原。特别值得注意的是,由于采用人源化小鼠产生的抗体(Renlite带有Biocytogen的共轻链)作为AI模型的训练数据,设计的抗体人源化程度高,可开发性好,有效避免了后续流程中的ADA和聚集等问题。方法首先,借助Renlite,综合结合小鼠免疫、B细胞分选与FACS、NGS单细胞测序、生物信息学分析,内部生成大量高质量的抗体序列数据。其次,我们开发了抗原特异性抗体选择和表位预测的AI模型(bioRxiv, 2022: 2022.12)。22.521634.)从数据中挖掘抗原特异性抗体及相应抗原表位。基于处理后的高质量数据,我们训练了一个亲和力预测模型,可以准确预测抗原表位和抗体序列对是否可以相互结合。此外,利用序列数据,我们训练了一个抗体序列预训练语言模型(bioRxiv, 2023: 2023.01)。19.524683.),可以生成高质量的抗体序列,模拟小鼠免疫产生的抗体。最后,结合上述AI模型,我们建立了抗体从头设计计算工作流,模拟小鼠免疫系统中抗体产生和亲和成熟的生物学过程,可以看作是一个“DigitalMouse”。结果在一个试验案例中,设计了100万种针对抗原特异性表位结合的抗体。选择10个抗体进行表达。用BLI测定结合亲和力。10个抗体中有2个抗体的KD分别为194 nM和336 nM, BLI的浓度依赖性信号增加。这些抗体有很大的潜力作为候选分子的起点进行进一步的体外、体内实验验证和临床试验。结论基于人工智能的抗体从头设计工作流程将彻底改变抗体发现行业的模式,大大缩短抗体发现阶段,降低研发成本,并将抗体发现扩展到更多难以通过动物免疫的抗原靶点。计算工作流将对整个生物制药行业产生深远的影响。
{"title":"REVOLUTIONIZING ANTIBODY DISCOVERY INDUSTRY WITH HIGHLY EFFICIENT AND ACCURATE AI-BASED EPITOPE-SPECIFIC ANTIBODY DE NOVO DESIGN WORKFLOW","authors":"Tianyuan Wang, Xiangrui Gao, Zhe Huai, Zhaohui Gong, Ting Mao, Xuezhe Fan, Xingxing Wu, Zhiyuan Duan, Xiaodong Wang, Jiewen Du, Mengcheng Yao, Xin Li, Min Wu, Zonghu Wang, Lin Zhang, Junjie Zhang, Wenbo Cao, Kai Yan, Yujie Fang, Shixiang Ma, Kun Yang, Lili Wu, F. An, Yezhou Yang, L. Lai, Xiaolu Huang","doi":"10.1093/abt/tbad014.024","DOIUrl":"https://doi.org/10.1093/abt/tbad014.024","url":null,"abstract":"Abstract Background and significance The global antibody drug market is worth over $200 billion in 2021 and is expected to reach $380 billion by 2030. Antibody discovery is one of the most critical steps that determine the crucial properties of antibody drugs, such as efficacy, safety, and developability. Traditional methods based on mouse immunization have many drawbacks limiting drug discovery, which include long time periods, high costs, inability to target function-specific epitopes, unsuitable for low immunogenic and difficult-to-prepare antigens, the need to sacrifice mice, the need for further humanization to reduce immunogenicity, and so on. Here we report an antibody de novo design computational workflow that utilizes high-quality internally produced antibody data and advanced AI models. Using this workflow, we can de novo design antibodies that bind to user-specified functional epitopes with high affinity and specificity. Compared with classical wet-lab methods, the entire process is shortened from several months to several days and suitable for low immunogenicity and difficult-to-prepare antigens. It is particularly noteworthy that due to the use of humanized mouse-generated antibodies (Renlite bearing common light chain from Biocytogen) as training data for AI models, the designed antibodies have a high degree of humanization and good developability, effectively avoiding issues such as ADA and aggregation in subsequent processes. Methods First, with the help of Renlite, we comprehensively combined mouse immunization, B cell sorting with FACS, NGS single-cell sequencing, and bioinformatics analysis to internally generate a large amount of high-quality antibody sequence data. Second, we developed AI models for antigen-specific antibody selection and epitope prediction (bioRxiv, 2022: 2022.12. 22.521634.) to mine antigen-specific antibodies and corresponding antigen epitopes in the data. Based on the processed high-quality data, we trained an affinity prediction model that can accurately predict whether an antigen epitope and antibody sequence pair can bind to each other. Besides, using the sequence data, we trained an antibody sequence pre-training language model (bioRxiv, 2023: 2023.01. 19.524683.), which can generate high-quality antibody sequences to simulate the antibodies produced by mouse immunization. Finally, integrating the above AI models, we established an antibody de novo design computational workflow to simulate the biological process of antibody generation and affinity maturation in the mouse immune system, which can be seen as a “DigitalMouse”. Results In a test case, 1 million antibodies were designed aiming at binding to specific epitope of an antigen. 10 antibodies were selected and expressed. Binding affinity was determined using BLI. Two antibodies out of 10 had KD of 194 nM and 336 nM, respectively, with a concentration dependent signal increase on BLI. These antibodies have great potential as the starting point of ","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43115944","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CAMEL NANOBODIES NEUTRALIZE SARS-COV-2 VARIANTS 骆驼纳米体中和sars-cov-2变体
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.001
Jessica Hong, H. Kwon, R. Cachau, K. J. Butay, Zhijian Duan, Dan Li, Hua Ren, C. Hsieh, V. Dandey, M. Borgnia, Hang Xie, Mitchell Ho
Abstract Background and Significance With the emergence of SARS-CoV-2 variants during the global pandemic from 2020 to 2023, there is need for broadly neutralizing antibodies. Due to their small size and unique conformations, nanobodies can recognize protein cavities that are not accessible to conventional antibodies. Methods and Results Here, we used phage display libraries built from dromedary camels to isolate two VHH nanobodies (7A3 and 8A2), which have high affinity for the receptor-binding domain (RBD) of the SARS-CoV-2 spike. Cryo-EM complex structures revealed that 8A2 binds the RBD in its up mode and 7A3 targets a conserved and deeply buried site in the spike regardless of the conformational state of the RBD. At a dose of ≥5 mg/kg, nanobody 7A3 efficiently protected K18-hACE2 transgenic mice from the lethal challenge of SARS-CoV-2 variants B.1.351 or B.1.617. With the addition of omicron variant, a new VHH nanobody (J1B4) was isolated to target the S2 subunit of the SARS-CoV-2 spike that can bind across many variants including omicron. Trispecific nanobodies were made using 7A3, 8A2, and J1B4 which had increased binding signals compared to the nanobodies alone. Using this method, we hope to create a therapeutic that is able to broadly neutralize not only all pre-existing variants of SARS-CoV-2, but also be effective towards future SARS-CoV related variants. Conclusions and Future Directions By combining nanobodies targeting the RBD of the S1 subunit (7A3+8A2) with a nanobody targeting the S2 subunit (J1B4), we can increase the chance of protection against all SARS-CoV-2 infections. Due to the increased protein binding of the trispecific compared to individual nanobodies alone, it shows great promise that the trispecific may be able to enhance its activity across all variants. Nanobody-based therapeutics may be developed as a nasal spray which can be self-administered and inhaled directly to the lungs to treat the infection at its source.
背景与意义随着2020 - 2023年全球大流行期间SARS-CoV-2变异体的出现,需要广泛中和抗体。由于它们的小尺寸和独特的构象,纳米体可以识别常规抗体无法进入的蛋白质空洞。方法与结果本研究利用单峰骆驼噬菌体展示文库分离出两个VHH纳米体(7A3和8A2),这两个纳米体对SARS-CoV-2刺突的受体结合域(RBD)具有高亲和力。Cryo-EM复合物结构显示,无论RBD的构象状态如何,8A2以向上模式结合RBD,而7A3靶向刺突中一个保守且深埋的位点。当剂量≥5 mg/kg时,纳米体7A3可有效保护K18-hACE2转基因小鼠免受SARS-CoV-2变体B.1.351或B.1.617的致命攻击。通过添加组粒变体,分离出一种新的VHH纳米体(J1B4),其靶向SARS-CoV-2刺突的S2亚基,可以结合包括组粒在内的许多变体。用7A3、8A2和J1B4制备了三特异性纳米体,与单独使用纳米体相比,它们具有更高的结合信号。利用这种方法,我们希望创造一种治疗方法,不仅能够广泛中和所有已存在的SARS-CoV-2变体,而且对未来的SARS-CoV相关变体也有效。通过将靶向S1亚基(7A3+8A2) RBD的纳米体与靶向S2亚基(J1B4)的纳米体结合,我们可以增加对所有SARS-CoV-2感染的保护机会。由于与单独的单个纳米体相比,三特异性的蛋白质结合增加,这表明三特异性可能能够增强其在所有变体中的活性。基于纳米体的治疗方法可能被开发成一种鼻喷雾剂,可以自行给药并直接吸入肺部,从源头治疗感染。
{"title":"CAMEL NANOBODIES NEUTRALIZE SARS-COV-2 VARIANTS","authors":"Jessica Hong, H. Kwon, R. Cachau, K. J. Butay, Zhijian Duan, Dan Li, Hua Ren, C. Hsieh, V. Dandey, M. Borgnia, Hang Xie, Mitchell Ho","doi":"10.1093/abt/tbad014.001","DOIUrl":"https://doi.org/10.1093/abt/tbad014.001","url":null,"abstract":"Abstract Background and Significance With the emergence of SARS-CoV-2 variants during the global pandemic from 2020 to 2023, there is need for broadly neutralizing antibodies. Due to their small size and unique conformations, nanobodies can recognize protein cavities that are not accessible to conventional antibodies. Methods and Results Here, we used phage display libraries built from dromedary camels to isolate two VHH nanobodies (7A3 and 8A2), which have high affinity for the receptor-binding domain (RBD) of the SARS-CoV-2 spike. Cryo-EM complex structures revealed that 8A2 binds the RBD in its up mode and 7A3 targets a conserved and deeply buried site in the spike regardless of the conformational state of the RBD. At a dose of ≥5 mg/kg, nanobody 7A3 efficiently protected K18-hACE2 transgenic mice from the lethal challenge of SARS-CoV-2 variants B.1.351 or B.1.617. With the addition of omicron variant, a new VHH nanobody (J1B4) was isolated to target the S2 subunit of the SARS-CoV-2 spike that can bind across many variants including omicron. Trispecific nanobodies were made using 7A3, 8A2, and J1B4 which had increased binding signals compared to the nanobodies alone. Using this method, we hope to create a therapeutic that is able to broadly neutralize not only all pre-existing variants of SARS-CoV-2, but also be effective towards future SARS-CoV related variants. Conclusions and Future Directions By combining nanobodies targeting the RBD of the S1 subunit (7A3+8A2) with a nanobody targeting the S2 subunit (J1B4), we can increase the chance of protection against all SARS-CoV-2 infections. Due to the increased protein binding of the trispecific compared to individual nanobodies alone, it shows great promise that the trispecific may be able to enhance its activity across all variants. Nanobody-based therapeutics may be developed as a nasal spray which can be self-administered and inhaled directly to the lungs to treat the infection at its source.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46199194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AB-Amy: machine learning aided amyloidogenic risk prediction of therapeutic antibody light chains. AB-Amy:机器学习辅助治疗性抗体轻链淀粉样变性风险预测。
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad007
Yuwei Zhou, Ziru Huang, Yushu Gou, Siqi Liu, Wei Yang, Hongyu Zhang, Anthony Mackitz Dzisoo, Jian Huang

Over 120 FDA-approved antibody-based therapeutics are used to treat a variety of diseases.However, many candidates could fail because of unfavorable physicochemical properties. Light-chain amyloidosis is one form of aggregation that can lead to severe safety risks in clinical development. Therefore, screening candidates with a less amyloidosis risk at the early stage can not only save the time and cost of antibody development but also improve the safety of antibody drugs. In this study, based on the dipeptide composition of 742 amyloidogenic and 712 non-amyloidogenic antibody light chains, a support vector machine-based model, AB-Amy, was trained to predict the light-chain amyloidogenic risk. The AUC of AB-Amy reaches 0.9651. The excellent performance of AB-Amy indicates that it can be a useful tool for the in silico evaluation of the light-chain amyloidogenic risk to ensure the safety of antibody therapeutics under clinical development. A web server is freely available at http://i.uestc.edu.cn/AB-Amy/.

超过120种fda批准的基于抗体的治疗方法用于治疗各种疾病。然而,由于不利的物理化学性质,许多候选材料可能会失败。轻链淀粉样变性是一种聚集形式,可在临床开发中导致严重的安全风险。因此,在早期筛选淀粉样变风险较小的候选药物,不仅可以节省抗体开发的时间和成本,还可以提高抗体药物的安全性。本研究基于742个淀粉样蛋白抗体轻链和712个非淀粉样蛋白抗体轻链的二肽组成,训练基于支持向量机的AB-Amy模型来预测轻链淀粉样蛋白风险。AB-Amy的AUC达到0.9651。AB-Amy的优异表现表明,它可以作为一种有用的工具,用于轻链淀粉样变性风险的计算机评估,以确保临床开发中抗体治疗药物的安全性。在http://i.uestc.edu.cn/AB-Amy/上可以免费获得web服务器。
{"title":"AB-Amy: machine learning aided amyloidogenic risk prediction of therapeutic antibody light chains.","authors":"Yuwei Zhou,&nbsp;Ziru Huang,&nbsp;Yushu Gou,&nbsp;Siqi Liu,&nbsp;Wei Yang,&nbsp;Hongyu Zhang,&nbsp;Anthony Mackitz Dzisoo,&nbsp;Jian Huang","doi":"10.1093/abt/tbad007","DOIUrl":"https://doi.org/10.1093/abt/tbad007","url":null,"abstract":"<p><p>Over 120 FDA-approved antibody-based therapeutics are used to treat a variety of diseases.However, many candidates could fail because of unfavorable physicochemical properties. Light-chain amyloidosis is one form of aggregation that can lead to severe safety risks in clinical development. Therefore, screening candidates with a less amyloidosis risk at the early stage can not only save the time and cost of antibody development but also improve the safety of antibody drugs. In this study, based on the dipeptide composition of 742 amyloidogenic and 712 non-amyloidogenic antibody light chains, a support vector machine-based model, AB-Amy, was trained to predict the light-chain amyloidogenic risk. The AUC of AB-Amy reaches 0.9651. The excellent performance of AB-Amy indicates that it can be a useful tool for the <i>in silico</i> evaluation of the light-chain amyloidogenic risk to ensure the safety of antibody therapeutics under clinical development. A web server is freely available at http://i.uestc.edu.cn/AB-Amy/.</p>","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/07/8d/tbad007.PMC10365155.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9875993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ENGINEERING T CELLS TARGETING GPC2 FOR TREATING NEUROBLASTOMA 靶向GPC2的工程T细胞治疗神经母细胞瘤
Q2 Medicine Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.023
Alex Quan, Nan Li, Dan Li, Madeline R. Spetz, Hongbing Zhang, Cheng Liu, Mitchell Ho
Abstract Background and Significance Neuroblastoma is a rare pediatric cancer that forms in immature nerve tissue of infants and accounts for 10 to 15 percent of cancer-related deaths in children. The five-year survival for high-risk neuroblastoma is 50% with current treatment practices being a combination of surgery, chemotherapy, and radiation. A more effective therapy is therefore needed to improve overall patient outcomes. Methods The CT3 mouse antibody that targets GPC2 was previously identified in the lab and has shown activity in the chimeric antigen receptor (CAR) T cell format against neuroblastoma. Humanization of the CT3 antibody was also done through CDR grafting in human germline sequences to prevent potential adverse immunogenic effects when treating patients. In the present study, the CT3 antibody and humanized CT3 (hCT3) antibody were engineered into T cells based on the engineered gamma/delta TCR scaffold (called AbTCR). The activities of the CT3 and hCT3 AbTCRs were tested in luciferase-based cell killing assays and xenograft mouse models. Results Humanized CT3 retains a comparable binding affinity for GPC2. The hCT3 CAR T cell showed its ability to regress tumor expression in mice. Furthermore, the mice treated with the CT3 AbTCR showed tumor regression while the mice treated with the hCT3 AbTCR became tumor free three weeks after treatment. Conclusions Overall, the hCT3 AbTCR T cells are very active when combating neuroblastoma tumors in mice. The efficacy at a low treatment dosage indicates that the GPC2 targeted hCT3 AbTCRs are a promising therapeutic for the treatment of neuroblastoma and other GPC2 positive cancers in patients.
摘要背景和意义神经母细胞瘤是一种罕见的儿科癌症,在婴儿未成熟的神经组织中形成,占儿童癌症相关死亡的10-15%。高危神经母细胞瘤的五年生存率为50%,目前的治疗方法是手术、化疗和放疗相结合。因此,需要一种更有效的治疗方法来改善患者的整体预后。方法先前在实验室中鉴定了靶向GPC2的CT3小鼠抗体,该抗体在嵌合抗原受体(CAR)T细胞形式中显示出抗神经母细胞瘤的活性。CT3抗体的人源化也通过在人类种系序列中进行CDR移植来防止治疗患者时潜在的不良免疫原性影响。在本研究中,CT3抗体和人源化CT3(hCT3)抗体基于工程化γ/ΔTCR支架(称为AbTCR)被工程化为T细胞。在基于荧光素酶的细胞杀伤测定和异种移植物小鼠模型中测试CT3和hCT3-AbTCRs的活性。结果人源化CT3对GPC2具有相当的结合亲和力。hCT3 CAR T细胞在小鼠中显示出其抑制肿瘤表达的能力。此外,用CT3-AbTCR处理的小鼠显示出肿瘤消退,而用hCT3-AbTCR处理的鼠在处理后三周变得无肿瘤。结论总的来说,hCT3-AbTCR T细胞在对抗小鼠神经母细胞瘤肿瘤时非常活跃。低治疗剂量下的疗效表明,GPC2靶向的hCT3-AbTCRs是治疗患者神经母细胞瘤和其他GPC2阳性癌症的有前途的治疗方法。
{"title":"ENGINEERING T CELLS TARGETING GPC2 FOR TREATING NEUROBLASTOMA","authors":"Alex Quan, Nan Li, Dan Li, Madeline R. Spetz, Hongbing Zhang, Cheng Liu, Mitchell Ho","doi":"10.1093/abt/tbad014.023","DOIUrl":"https://doi.org/10.1093/abt/tbad014.023","url":null,"abstract":"Abstract Background and Significance Neuroblastoma is a rare pediatric cancer that forms in immature nerve tissue of infants and accounts for 10 to 15 percent of cancer-related deaths in children. The five-year survival for high-risk neuroblastoma is 50% with current treatment practices being a combination of surgery, chemotherapy, and radiation. A more effective therapy is therefore needed to improve overall patient outcomes. Methods The CT3 mouse antibody that targets GPC2 was previously identified in the lab and has shown activity in the chimeric antigen receptor (CAR) T cell format against neuroblastoma. Humanization of the CT3 antibody was also done through CDR grafting in human germline sequences to prevent potential adverse immunogenic effects when treating patients. In the present study, the CT3 antibody and humanized CT3 (hCT3) antibody were engineered into T cells based on the engineered gamma/delta TCR scaffold (called AbTCR). The activities of the CT3 and hCT3 AbTCRs were tested in luciferase-based cell killing assays and xenograft mouse models. Results Humanized CT3 retains a comparable binding affinity for GPC2. The hCT3 CAR T cell showed its ability to regress tumor expression in mice. Furthermore, the mice treated with the CT3 AbTCR showed tumor regression while the mice treated with the hCT3 AbTCR became tumor free three weeks after treatment. Conclusions Overall, the hCT3 AbTCR T cells are very active when combating neuroblastoma tumors in mice. The efficacy at a low treatment dosage indicates that the GPC2 targeted hCT3 AbTCRs are a promising therapeutic for the treatment of neuroblastoma and other GPC2 positive cancers in patients.","PeriodicalId":36655,"journal":{"name":"Antibody Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47789345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Antibody Therapeutics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1