Antibody Therapeutics最新文献

筛选
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
Antibody Therapeutics 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
{"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)
Antibody Therapeutics 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
{"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
Antibody Therapeutics 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
{"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
Antibody Therapeutics Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.022
Youwei Jiang
{"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
Antibody Therapeutics Pub Date : 2023-07-01 DOI: 10.1093/abt/tbad014.013
Xiaoling Jiang, Chongbing Wu, Zi Chen, Liusong Yin
{"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
Antibody Therapeutics 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
{"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
Antibody Therapeutics 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
{"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
Antibody Therapeutics 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
{"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。
Antibody Therapeutics 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
{"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
Antibody Therapeutics 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
{"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信