作者:于琦 薛超然美工:何国红 罗真真排版:马超01 引言抗体药物的传奇历程始于1975年单克隆抗体技术的诞生,这一里程碑彻底改变了疾病治疗的格局。随后的几十年里,嵌合、人源化及全人源抗体技术的不断突破,持续加速了这一领域的飞速发展。全球现已有超过170种抗体药物获批,广泛应用于肿瘤、自身免疫等重大疾病。抗体药物市场正经历爆发式增长,预计未来几年,抗体药物的市场份额将持续扩大,并有望在2030年突破数千亿美元,前景无限光明。在抗体药物蓬勃发展的浪潮中,三优生物致力于为客户提供从靶点发现到临床前候选药物(PCC)的一站式开发服务,仅需一个靶点,即可交付高质量的PCC分子,整个过程涵盖靶点调研、原材料制备、分子发现与优化、体内外药效评价以及成药性分析等环节。无论是传统的单克隆抗体、创新的双特异性抗体,还是前沿的抗体药物偶联物(ADC),三优生物均可提供定制化解决方案,助力加速创新生物药的研发进程。截至目前,三优生物已完成60余个PCC项目开发,其中10余个项目已进入临床阶段,最快推进至III期临床阶段。02 服务流程三优生物专注于抗体药物研发领域,提供涵盖靶点调研、原材料制备、分子产生、体外药效评价、分子优化、体内药效评价及成药性分析等关键环节的一站式服务。在研发过程中,首先会对靶点进行深入调研,明确其与疾病的相关性及研发价值,从而确定研发方向。随后将制备相关原材料,包括抗原蛋白、细胞株等,为后续研究奠定基础。基于超万亿创新分子库获得候选分子后,进行体外药效学评价,筛选出具有靶点活性的分子并进行特性优化,以进一步提高其生物活性和成药性。经优化后的分子将在动物模型中进行体内药效学验证,最终通过全面的成药性分析评估其安全性、药代动力学等关键特性。▲ Fig. 1 Target to PCC drug development flowchart01靶点调研在药物研发全流程中,靶点调研作为连接基础研究与临床转化的关键环节,具有核心战略意义。针对尚未满足的临床需求,开展创新抗体药物研发的首要步骤即是靶点调研。该环节会系统性地梳理目标靶点的生物学特征、在疾病发生发展中的分子机制,以及相关领域的研究进展与空白。唯有明确靶点的生物学特征、人群分布差异及与其他治疗手段的协同潜力,才能构建从机制创新到临床价值转化的完整逻辑链,让药物真正聚焦于“正确的靶点,正确的患者,正确的疗效”。三优生物在肿瘤、自身免疫性疾病、代谢性疾病、神经系统疾病、眼科疾病、心血管疾病、感染性疾病、衰老相关疾病以及宠物治疗等领域,均配备了经验丰富的高级科学家团队。针对各类靶点,公司已完成300余个靶点的系统性调研工作。目前,三优生物已有10余个项目通过授权转让进入临床阶段,这些成果充分体现了靶点调研的重要价值。▼ Table. 1 Reported IND project cases02原材料制备三优生物已构建了涵盖质粒、蛋白、mRNA、VLP及研究用细胞株等原材料的完整解决方案体系。该平台能够高效制备各类复杂蛋白,包括胞内蛋白、膜蛋白、胞外蛋白等不同细胞定位蛋白,以及GPCR靶点、离子通道靶点、细胞因子等特殊类型蛋白,充分满足客户对高难度蛋白制备的技术需求。三优生物的技术平台已达到行业领先水平,具备年产10万个蛋白和1000株研究用细胞株的制备能力。原材料制备效率卓越:mRNA包装仅需18天即可完成;足量且质检合格的蛋白可在2周内交付;高表达量、高稳定性的研究用细胞株仅需1个月即可获得。团队成员拥有丰富的项目经验,已成功完成500余个阶段性或整体性服务项目,为客户的抗体开发及药物筛选工作提供强有力的技术支持。▲ Fig. 2 Raw material preparation solution03分子产生三优生物建立了具有自主知识产权、各项性能指标全球领先的超万亿级创新分子库,可广泛应用于创新药物发现、诊断试剂开发、检测技术及科研用抗体定制等多个领域。分子库包含全人源抗体、单域抗体、动物免疫抗体和新型分子四大类,通过12种技术路径可产生全人源抗体、单域抗体、鼠源单抗、兔源单抗、双特异性抗体、多克隆抗体、靶向多肽及靶向小型蛋白等多种分子形式。结合一站式高通量自动化筛选平台,显著缩短了筛选周期,能够高效获得具有高亲和力、优异特异性和良好可开发性的候选分子,系统解决各类分子开发难题,有力推动药物研发进程。截止2025年3月份累计已完成900+项目的筛选,以合作开发或授权转让的模式协助客户完成10余个项目的IND申报,共递交国内外专利申请115件,获得专利授权28件。▲ Fig. 3 Diversified sources of molecules04体外药效评价体外药效筛选作为抗体功能评价的核心环节,可显著加速抗体药物的发现、筛选和优化进程。三优生物专注于创新抗体药物研发领域,构建了稳定、高效且可靠的体外药效筛选平台。该平台目前涵盖流式细胞分析、体外药效方法开发、体外药效样品检测及非特异性结合检测等模块,能够确保候选抗体以高效率、高通量和高标准完成体外药效筛选工作。平台建立了数十种多维度、立体化的体外药效评价体系,可满足抗原抗体结合、信号通路调控及细胞功能评估等多层次研究需求。平台目前拥有150余种经项目验证的靶点细胞株、20余种经项目验证的原代细胞、8大类51项子类检测方法,并配备92份标准化操作规程(SOP)支持。常规检测方法仅需1周即可完成,高难度检测方法可在4周内完成;常规方法开发周期为2周,高难度方法开发周期为8周。▲ Fig. 4 Solution for in vitropharmacological evaluation05分子优化三优生物凭借其完善的抗体工程改造技术平台,致力于为客户提供高效、专业的抗体优化服务。该平台涵盖了抗体优化的关键环节,包括人源化改造、亲和力成熟、成药性优化及半衰期延长。平台具有抗体各功能区域的精准改造能力,包括框架区(FR)、互补决定区(CDR)和恒定区(Fc)。通过对这些区域的精细操作,三优生物能够满足多样化的项目需求,并显著提升改造后抗体的性能,包括但不限于药效学与成药性显著提升、交叉反应性增强、靶点特异性改善、潜在修饰风险的消除。三优生物在效率方面处于行业领先地位,每轮抗体改造周期仅需6-8周即可高质量完成,显著加速了客户项目进入后续的功能筛选阶段。该平台由经验丰富的团队支撑,已成功完成500余例各类抗体分子的改造项目。其中,40余个设计抗体已进入临床申报阶段,10余个已进入临床试验阶段,充分验证了其技术平台的可靠性和成功率。▲ Fig. 5 Molecular optimization solutions06体内药效评价体内药效评价作为确定临床前候选分子的关键环节,可进一步验证候选分子在动物模型中的有效性和安全性。三优生物已建立高通量、高质量的候选分子体内药效评价平台,该平台涵盖体内药效方法开发系统、动物药效评价系统、药代动力学研究系统以及急性毒性检测系统。平台以国内外大小动物药效评价市场为核心,重点围绕肿瘤和自身免疫性疾病两大适应症,建立了完善的肿瘤CDx模型体系、自身免疫性疾病非肿瘤模型平台、细胞因子及血液标志物检测系统,且提供从小鼠到非人灵长类动物的完整药代动力学评价服务,全面满足临床前非GLP相关研究需求。目前平台已积累150余个经项目验证的CDx模型、数十种非肿瘤疾病模型、40余项标准操作规程(SOP),并成功支持10余个药效相关临床申报项目。▲ Fig. 6 Solution for in vivo drugefficacy evaluation07成药性分析三优生物经过多年的技术沉淀与升级,已建立起涵盖单抗、多抗和融合蛋白的多维表征分析平台。其中,抗体理化分析平台已开发出10种检测项目,具备多维表征、全面合规、高质高效的特点,可支持生物新药研发前期样品的质量控制。生化分析平台则通过优化各类生化检测方法,进一步丰富了检测内容。基于ELISA的生化分析技术具有周期短、重复性好、通量高、成本低等优势,可高效地从大量先导分子中筛选优质候选分子,显著提升“先导分子聚焦”效率。该技术已成功应用于数百个服务项目,覆盖早期药物研发、小试工艺开发等全研发阶段。同时,平台严格参照药典要求建立了完善的标准体系,凭借精准可靠的表征方法,为药物生产提供技术支持和工艺指导。▲ Fig.7 Solution for drug analysis03 服务特性01全流程覆盖三优生物靶点至PCC药物研发系统解决方案覆盖靶点调研、原材料制备、分子产生、体外药效评价、分子优化、体内药效评价及成药性分析等关键环节,实现研发流程的无缝整合。该模式有效规避了因技术标准差异、数据断层或沟通障碍导致的效率损失,确保从靶点发现到PCC研发的每个环节均能基于前期成果精准递进。通过全流程一体化设计,依托统一的质量控制体系和跨学科协作机制,可早期识别潜在风险(如抗体成药性不足或靶点选择性欠佳等),及时调整研发策略,从而显著缩短研发周期、降低重复投入,最终提高候选抗体的成药成功率,为创新药物研发提供高效可靠的技术支持。▲ Fig.8 Full process coverage02定制化适配三优生物靶点至PCC药物研发系统解决方案能够根据项目靶点特性及研发阶段需求,提供定制化的实验方案与技术路线。针对不同研发阶段和客户核心诉求,平台可动态调整实验方案,避免标准化研发模式导致的资源浪费与效率损失。这种高度灵活的模式既保证了研发策略与靶点生物学特性的精准匹配,又能聚焦项目核心目标,在提升候选抗体成药性的同时有效控制后期开发风险,为创新药物研发提供更加精准高效的解决方案。▲ Fig.9 Research and statistics on100+ targets of Sanyou Biotechnology03高效化推进三优靶点至PCC药物研发系统解决方案整合了多个技术平台与资深团队经验,能通过AI辅助设计和高通量实验平台加速先导分子的筛选效率,结合严格的质量控制体系确保数据可靠性,能在早期同步解决活性、选择性、成药性等关键问题,减少后期返工风险;此外,基于历史项目数据库建立的预测模型,可精准指导实验方向,大幅缩短从靶点到PCC的药物研发周期,降低研发风险,为客户提供从早期探索到候选分子确定的无缝衔接支持。▲ Fig.10 Project operation cycle04 案例展示01抗CTLA4单抗的开发基本信息:CTLA4(细胞毒性T淋巴细胞相关抗原4,CD152)属于免疫球蛋白超家族(IgSF),是一种跨膜蛋白,在细胞外区域含有IgV样结构域,与CD28具有高度同源性(约31%的氨基酸序列相似性)。两者都与抗原呈递细胞(APC)表面的B7分子(CD80/CD86)竞争性结合,但CTLA4具有更高的亲和力(约10-20倍),并通过抑制共刺激信号负调控T细胞活化。分布情况:CTLA4主要在活化的CD4+/CD8+T细胞和调节性T细胞(Treg)的表面表达,特别是在肿瘤微环境中,Treg高度表达CTLA4。竞争格局:根据Insight数据库的调查,全球有139种针对CTLA4的抗体药物正在开发中,其中5种已经上市,1种正在申请上市。首个获得美国食品药品监督管理局批准的CTLA4单克隆抗体(2011年),用于治疗黑色素瘤,2020年全球市场规模为17亿美元Tremelimumab:在癌症等适应症中进行III期临床试验。▲ Fig. 11 Mechanism of action of CTLA402抗CTLA4单抗的关键结果▶ 2.1. 细胞水平的结合活性分析通过三优生物分子产生平台获得的单域抗体候选分子大多具有较好的细胞水平结合活性。Fig. 12A展示了采用FACS分析候选抗体与huCTLA-4 CHO细胞的结合活性,候选抗体与对照抗体相比,具有相当的结合活性。Fig. 12B展示了采用FACS分析候选抗体与cynoCTLA-4 CHO细胞的亲和力测定结果,候选抗体的与对照抗体相比,具有相当的结合活性。▲ Fig. 12 Binding affinity of antibody▶ 2.2. 细胞水平的阻断活性分析采用FACS方法检测候选抗体是否阻断配体CD80-ECD、CD86-ECD与huCTLA-4 CHO细胞的结合。Fig. 13A展示了候选抗体阻断CD80-ECD与huCTLA-4 CHO细胞的结合,阻断效果与对照抗体相当或优于对照抗体。Fig. 13B展示了候选抗体阻断CD86-ECD与huCTLA-4 CHO细胞的结合,阻断效果与对照抗体相当或优于对照抗体。▲ Fig. 13 Blocking activity of antibody ▶ 2.3. 人源化抗体细胞水平的结合活性分析针对NB25B-17和NB25gb-1通过三优生物分子优化平台进行人源化改造。对获得的单域抗体在细胞水平进行结合活性检测。Fig. 14A展示了采用FACS分析候选抗体与huCTLA-4 CHO细胞的亲和力测定结果。Fig. 14B展示了采用FACS分析候选抗体与cynoCTLA-4 CHO细胞的亲和力测定结果。结果表明,NB25B-17-9-H1-IgG1和NB25gb-1-11-H1-IgG1显示出比其他候选抗体更好的结合能力和交叉活性。▲ Fig. 14 Binding affinity of antibody▶ 2.4. 人源化抗体细胞水平的阻断活性分析采用FACS方法检测候选抗体是否阻断配体CD80-ECD、CD86-ECD与huCTLA-4 CHO细胞的结合。Fig. 15A展示了候选抗体阻断CD80-ECD与huCTLA-4 CHO细胞的结合。Fig. 15B展示了候选抗体阻断CD86-ECD与huCTLA-4 CHO细胞的结合。结果表明,NB25B-17-9-H1-IgG1、NB25gb-1-11-H1-IgG1和NB25gb-1-11-H2-IgG1对huCTLA-4 CHO的阻断活性优于对照抗体。▲ Fig. 15 Blocking activity of antibody▶ 2.5. 体内药效评价在HuCTLA4小鼠皮下接种MC38肿瘤细胞,当平均肿瘤体积达到约100 mm3时,将小鼠随机分为4个实验组,其中1组为对照组,3组为给药组,分别给3个不同的剂量。通过腹部给药给予抗体(每周2次,给药三周),在第32天结束给药。当肿瘤细胞进行Anti-CTLA4抗体给药时,发现肿瘤体积明显缩小。▲ Fig. 16 In vivo efficacy of antibody05 总结展望在从靶点到PCC的抗体药物研发全流程中,三优生物依托其超万亿级创新分子库和一体化研发平台优势,已成功完成60余个靶点的项目开发(其中10余个已推进至III期临床阶段)。经过十年深耕,三优生物构建了涵盖靶点调研至PCC交付的一站式、全流程服务体系,为客户提供全程研发支持。选择三优生物,即意味着选择高效、优质、高成功率的抗体药物研发解决方案,助力创新药物快速转化。展望未来,抗体药物的研发前景无疑是广阔而激动人心的。随着科学技术的不断进步,我们有理由相信,更多创新、高效的抗体药物将不断涌现,为全球患者带来新的希望和更优的治疗选择。三优生物期待与您携手,共同探索抗体药物的无限可能,将更多具有里程碑意义的创新疗法从实验室带到临床,最终惠及每一位需要的患者。Sanyou 10th Anniversary: Service Section Target to PCC Drug Development System Solution01 IntroductionThe legendary journey of antibody drugs began in 1975 with the advent of monoclonal antibody technology, a milestone that revolutionized disease treatment. Over the following decades, breakthroughs in chimeric, humanized, and fully human antibody technologies have continuously accelerated the rapid advancement of this field. Today, more than 170 antibody drugs have been approved worldwide, widely used for major diseases such as cancer and autoimmune disorders. The antibody drug market is experiencing explosive growth, with its share expected to expand further in the coming years. By 2030, it is projected to surpass hundreds of billions of dollars, showcasing a future of immense promise.In the thriving wave of antibody drug development, Sanyou Bio is dedicated to providing clients with one-stop services from target discovery to preclinical candidate (PCC) delivery. Starting with just a target, we deliver high-quality PCC molecules through a comprehensive process covering target research, raw material preparation, molecular discovery & optimization, in vitro/in vivo efficacy evaluation, and druggability analysis. From monoclonal antibodies, bispecific antibodies, to antibody-drug conjugates (ADCs), Sanyou offers customized solutions to accelerate innovative biopharmaceutical R&D. As of now, Sanyou Bio has completed the development of over 60 PCC projects, with more than 10 advancing to clinical stages, including one project in Phase III trials as the fastest progressing candidate.02 Service ProcessSanyou Bio specializes in the field of antibody drug R&D, offering one-stop services covering key stages including target investigation, raw material preparation, molecule generation, in vitro efficacy evaluation, molecule optimization, in vivo efficacy assessment, and druggability analysis. During the R&D process, in-depth target investigation is first conducted to clarify its disease relevance and research value, thereby defining the R&D direction. Subsequently, relevant raw materials—such as antigen proteins and cell lines—are prepared to establish the foundation for further studies. After obtaining candidate molecules based on a trillion-size innovative molecular library, in vitro pharmacological evaluation is conducted to screen for molecules with target activity and optimize their characteristics, in order to further improve their biological activity and drug efficacy. The optimized molecule will undergo in vivo pharmacological validation in animal models, and ultimately its safety, pharmacokinetics, and other key characteristics will be evaluated through comprehensive pharmacokinetic analysis.▲ Fig. 1 Target to PCC drug development flowchart01Target researchTarget identification and validation is a strategically pivotal step bridging basic research and clinical translation throughout the drug discovery pipeline. For addressing unmet clinical needs, target research constitutes the primary phase in innovative antibody drug development. This process systematically evaluates the biological characteristics of target molecules, their molecular mechanisms in disease pathogenesis, as well as current research advances and gaps in relevant fields. By understanding the biological characteristics of targets, distribution variations, and synergistic potential with other therapies, we can establish a complete plan from mechanistic innovation to clinical value translation, ensuring drugs truly focus on 'the right target, the right patient, the right efficacy'. Sanyou Bio has established experienced senior scientist teams across diverse therapeutic areas, including oncology, autoimmune diseases, metabolic disorders, neurological diseases, ophthalmic diseases, cardiovascular diseases, infectious diseases, aging-related diseases, and veterinary therapeutics. The company has completed systematic research on over 300 targets. To date, more than 10 Sanyou-developed programs have advanced to clinical stages via out-licensing, demonstrating the critical value of target profiling.▼ Table. 1 Reported IND project cases02Raw material preparationSanyou Bio has established a comprehensive solution system for raw materials, including plasmids, proteins, mRNA, VLPs, and research cell lines. This platform enables the efficient production of various complex proteins, such as intracellular proteins, membrane proteins, extracellular proteins, as well as specialized targets like GPCRs, ion channels, and cytokines, fully meeting customers' technical demands for challenging protein preparation. Sanyou Bio’s technology platform has achieved industry-leading standards, with an annual production capacity of 100,000 batches of proteins and 1,000 research cell lines. The platform demonstrates exceptional efficiency in raw material preparation: mRNA packaging can be completed in just 18 days; high-quality, QC-approved proteins can be delivered within 2 weeks; and high-expression, high-stability research cell lines are obtainable in just 1 month. The team brings extensive project experience, having successfully completed 500+ milestone or full-service projects, providing robust technical support for antibody development and drug screening.▲ Fig. 2 Raw material preparation solution03Antibody discoverySanyou has established a proprietary, ultra-trillion-scale innovative molecular library with globally leading performance metrics. This library is widely applicable to innovative drug discovery, diagnostic reagent development, detection technologies, and customized research antibodies. The molecular library comprises four major categories: fully human antibodies, single-domain antibodies, animal-derived immune antibodies, and novel molecules. Through 12 technological approaches, it can generate diverse molecular formats, including fully human antibodies, single-domain antibodies, murine monoclonal antibodies, rabbit monoclonal antibodies, bispecific antibodies, polyclonal antibodies, targeting peptides, and small targeting proteins. Integrated with a one-stop high-throughput automated screening platform, this approach significantly shortens the screening cycle and efficiently identifies candidate molecules with high affinity, excellent specificity, and favorable developability. It systematically addresses various molecular development challenges, strongly accelerating drug discovery progress. As of March 2025, the platform has completed screening for 900+ projects, assisted clients in submitting INDs for over 10 projects through co-development or licensing agreements, and filed 115 patent applications globally, with 28 patents granted.▲ Fig. 3 Diversified sources of molecules04In vitro efficacy evaluationIn vitro efficacy screening, as a core component of antibody functional evaluation, significantly accelerates the discovery, screening, and optimization of antibody drugs. Sanyou Bio specializes in innovative antibody drug R&D and has established a stable, efficient, and reliable in vitro efficacy screening platform. The platform currently encompasses modules including flow cytometry analysis, in vitro efficacy method development, sample testing, and non-specific binding detection, ensuring high-efficiency, high-throughput, and high-standard screening of candidate antibodies. The platform has established dozens of multidimensional in vitro efficacy evaluation systems capable of meeting diverse research needs, ranging from antigen-antibody binding and signaling pathway modulation to cellular function assessment. The platform currently offers over 150 project-validated target cell lines, more than 20 project-validated primary cell types, and 51 subcategories of detection methods across 8 major categories, supported by 92 standardized operating procedures (SOPs). Routine assays can be completed within 1 week, while complex assays require up to 4 weeks. Method development takes 2 weeks for standard protocols and 8 weeks for challenging ones.▲ Fig. 4 Solution for in vitropharmacological evaluation05Molecule optimizationSanyou Bio its advanced antibody engineering platform to deliver efficient and specialized antibody optimization services. The platform covers key optimization processes, including humanization, affinity maturation, developability optimization, and half-life extension. It enables precise modifications of functional antibody regions, such as the framework region (FR), complementarity-determining region (CDR), and constant region (Fc).Through refined engineering, Sanyou meets diverse project demands while significantly enhancing antibody performance, including but not limited to:Improved pharmacodynamics & developability、Enhanced cross-reactivity、Refined target specificity、Elimination of potential modification risks. With industry-leading efficiency, Sanyou completes each round of antibody optimization in just 6–8 weeks, accelerating clients' transition to functional screening. Backed by an expert team, the platform has successfully optimized 500+ antibody molecules, with 40+ designed antibodies in regulatory filing and 10+ in clinical trials, demonstrating its reliability and success rate.▲ Fig. 5 Molecular optimization solutions06In vivo efficacy evaluationIn vivo efficacy evaluation, as a critical step in identifying preclinical candidate molecules, further validates their effectiveness and safety in animal models. Sanyou Bio has established a high-throughput, high-quality in vivo efficacy evaluation platform for candidate molecules, encompassing: In vivo efficacy method development system, Animal efficacy evaluation system, Pharmacokinetics (PK) study system, Acute toxicity testing system. This platform focuses on the small and large animal efficacy evaluation market, with a core emphasis on oncology and autoimmune diseases. We have established a comprehensive tumor CDx model system, a non-oncology model platform for autoimmune diseases, as well as cytokine and blood biomarker detection systems. Additionally, we provide complete pharmacokinetic evaluation services from mice to non-human primates, fully meeting preclinical non-GLP research needs.To date, the platform has accumulated over 150 project-validated CDx models, dozens of non-oncology disease models, and more than 40 standard operating procedures (SOPs), successfully supporting over 10 efficacy-related clinical submission projects.▲ Fig. 6 Solution for in vivo drug efficacy evaluation07Developability assessmentAfter years of technological refinement and advancement, Sanyou has established a multidimensional characterization platform covering monoclonal antibodies, polyclonal antibodies, and fusion proteins. The Physicochemical Characterization Platform for antibodies has developed 10 assay items, featuring multidimensional analysis, full compliance, and high-quality efficiency, supporting quality control of early-stage samples in novel biologics development. The Biochemical Analysis Platform has further expanded its testing capabilities through optimized biochemical assay methodologies. ELISA-based biochemical analysis offers advantages including short cycles, high reproducibility, high throughput, and low cost, enabling efficient screening of high-quality candidates from lead molecules and significantly improving "lead molecule focusing" efficiency. This technology has been successfully applied in hundreds of service projects across all R&D stages, from early drug discovery to pilot process development. The platform has established a comprehensive standard system in strict compliance with pharmacopoeia requirements, providing reliable technical support and process guidance for drug manufacturing through accurate characterization methods.▲ Fig.7 Solution for drug analysis03 Service Features01Full process coverageSanyou Bio's Target-to-PCC Drug Discovery Comprehensive Solution covers key stages including target profiling, raw material preparation, molecule discovery, in vitro efficacy evaluation, lead optimization, in vivo efficacy assessment, and developability analysis, ensuring seamless integration across the R&D workflow. This model eliminates efficiency losses caused by inconsistent technical standards, data fragmentation, or communication barriers, enabling precise progression from target identification to PCC development based on prior achievements. Through integrated end-to-end design supported by a unified quality control system and interdisciplinary collaboration, potential risks (e.g., inadequate antibody developability or suboptimal target selectivity) can be identified early, enabling timely strategy adjustment. This approach significantly shortens development timelines, reduces redundant investments, and ultimately improves the success rate of candidate antibodies, providing efficient and reliable technical support for innovative drug development.▲ Fig.8 Full process coverage02Customized adaptationSanyou Bio's Target-to-PCC Drug Discovery System Solution offers customized experimental solutions and technical pathways tailored to target characteristics and project requirements at different R&D stages. This platform dynamically adjusts experimental designs based on developmental phases and client priorities, eliminating resource waste and efficiency loss inherent in standardized workflows. This highly flexible approach ensures precise alignment between research strategies and target biology while focusing on core objectives. It enhances antibody drugability while mitigating late-stage development risks, delivering a more accurate and efficient solution for innovative drug development.▲ Fig.9 Research and statistics on100+ targets of Sanyou Biotechnology03Efficient promotionSanyou Bio's Target-to-PCC Drug Discovery System Solution integrates multiple technology platforms and expert team experience. It accelerates lead molecule screening through AI-assisted design and high-throughput experimental platforms, while ensuring data reliability via stringent quality control systems. This approach enables synchronous resolution of key challenges (activity, selectivity, and druggability) in early stages, reducing late-stage rework risks. Additionally, predictive models built on historical project databases precisely guide experimental direction, significantly shortening the target-to-PCC timeline and mitigating development risks. The solution provides seamless support from early exploration to candidate compound identification.▲ Fig.10 Project operation cycle04 Case Presentation01Development of anti-CTLA4 monoclonal antibodyBasic information:CTLA4 (cytotoxic T-lymphocyte-associated antigen 4, CD152), a member of the immunoglobulin superfamily (IgSF), is a transmembrane protein containing an IgV-like domain in its extracellular region that shares high homology with CD28 (~31% amino acid sequence similarity). Both competitively bind to B7 molecules (CD80/CD86) on antigen-presenting cells (APCs), but CTLA4 exhibits higher affinity (approximately 10-20-fold) and negatively regulates T-cell activation by inhibiting co-stimulatory signals.Distribution condition:CTLA4 is mainly expressed on the surface of activated CD4+/CD8+T cells and regulatory T cells (Tregs), especially in the tumor microenvironment where Tregs highly express CTLA4.Competitive landscape:According to Insight Database, there are currently 139 CTLA4-targeting antibody drugs under development globally, with 5 already marketed and 1 under regulatory review. The first CTLA4 monoclonal antibody approved by the FDA (2011) for melanoma treatment reached a global market size of $1.7 billion in 2020. Tremelimumab is undergoing Phase III clinical trials for cancer and other indications.▲ Fig. 11 Mechanism of action of CTLA402Key results of anti-CTLA4 monoclonal antibody▶ 2.1. Cell level binding activity analysisMost single-domain antibody candidates generated via SanyouBio's molecular discovery platform exhibit favorable cell-binding activity. Fig. 12A demonstrates FACS analysis of candidate antibodies binding to huCTLA-4 CHO cells, showing comparable binding activity to the control antibody. Fig. 12B presents affinity measurement results via FACS analysis against cynoCTLA-4 CHO cells, where the candidate antibodies display similar binding activity relative to the control.▲ Fig. 12 Binding affinity of antibody▶ 2.2. Analysis of blocking activity at the cellular levelFACS was employed to assess whether the candidate antibody blocks the binding of ligands CD80-ECD and CD86-ECD to huCTLA-4 CHO cells. Fig. 13A demonstrates that the candidate antibody effectively blocks CD80-ECD binding to huCTLA-4 CHO cells, with efficacy comparable or superior to the control antibody. Fig. 13B shows that the candidate antibody similarly blocks CD86-ECD binding to huCTLA-4 CHO cells, with blocking efficiency equivalent to or better than the control antibody.▲ Fig. 13 Blocking activity of antibody ▶ 2.3. Cell level binding activity analysis of humanized antibodiesNB25B-17 and NB25gb-1 were humanized using the Trianni Mo™ antibody optimization platform. The binding activities of the obtained sdAbs were evaluated at cellular level. Fig. 14A presents the affinity measurement results of candidate antibodies binding to huCTLA-4 CHO cells by FACS analysis. Fig. 14B shows the affinity measurement results of candidate antibodies binding to cynoCTLA-4 CHO cells by FACS analysis. The results demonstrate that NB25B-17-9-H1-IgG1 and NB25gb-1-11-H1-IgG1 exhibit superior binding capability and cross-reactivity compared to other candidate antibodies.▲ Fig. 14 Binding affinity of antibody▶ 2.4. Analysis of blocking activity at the cellular level of humanized antibodiesFACS was employed to assess whether candidate antibodies block the binding of ligands CD80-ECD and CD86-ECD to huCTLA-4 CHO cells. Fig. 15A demonstrates the blockade of CD80-ECD binding to huCTLA-4 CHO cells by candidate antibodies, while Fig. 15B shows their inhibition of CD86-ECD binding. Results indicate that NB25B-17-9-H1-IgG1, NB25gb-1-11-H1-IgG1, and NB25gb-1-11-H2-IgG1 exhibit superior blocking activity against huCTLA-4 CHO cells compared to the control antibody.▲ Fig. 15 Blocking activity of antibody▶ 2.5. In vivo efficacy evaluationMC38 tumor cells were subcutaneously inoculated into HuCTLA4 mice. When the average tumor volume reached approximately 100 mm³, the mice were randomly divided into four experimental groups: one control group and three treatment groups receiving different antibody doses. The antibody was administered intraperitoneally (twice weekly for three weeks), with treatment concluding on day 32. Anti-CTLA4 antibody administration significantly reduced tumor volume.▲ Fig. 16 In vivo efficacy of antibody05 Summary and OutlookIn the antibody drug development process from target to PCC, Sanyou Bio leverages its ultra-trillion innovative molecule library and integrated R&D platform to successfully complete over 60 target projects (10+ advanced to III clinical stages). With a decade of expertise, Sanyou has established a one-stop, end-to-end service system covering target assessment to PCC delivery, providing comprehensive R&D support. Sanyou provides efficient, high-quality antibody drug solutions with superior success rates, accelerating innovative drug translation.Looking ahead, the future of antibody drug development is undoubtedly vast and exciting. With continuous advancements in science and technology, we are confident that more innovative and highly effective antibody drugs will emerge, offering new hope and better treatment options for patients worldwide. Sanyou Bio looks forward to collaborating with clients globally to explore the boundless potential of antibody therapeutics, translating more landmark innovations from the lab to the clinic, ultimately benefiting every patient in need.推荐阅读三优十周年|靶点篇-六大恶性肿瘤靶点介绍创新前沿|2025ADC热门靶点与技术创新启示三优十周年|AI-STAL篇-千亿高等电点单域抗体库6类分子形式之环状多肽分子产生系统解决方案轻松玩转三优oneClick+线上9大云程序6类分子形式之mRNA分子产生系统解决方案三重好礼相送暨四种抗体制备解决方案全新上线三优生物单域抗体产生系统解决方案三优生物双抗参比品网站SY-BsAb正式上线三优ADC药物研发系统解决方案三优生物oneClick+平台再次上新三优磁阵列全人源小鼠抗体发现平台重磅发布三优超万亿全人单克隆抗体产生平台盘点三优磁阵列全人源小鼠抗体发现平台隆重上线共同轻链抗体产生之超万亿共轻库盘点关于三优生物三优生物是一家以“让天下没有难做的创新生物药”为使命,以超万亿分子库和人工智能技术双驱的生物医药智能高新技术企业。公司以智能超万亿分子库为核心,打造了干湿结合、国际领先的创新生物药临床前智能化及一体化研发平台,通过“新药发现、临床前研究、智能化药物研发及前沿科学研究”等四个维度加速全球新药发现及靶标深度研究。公司总部位于中国上海,在美国、欧洲等地设有子公司,现有投产及布局的研发及GMP场地20000多平方米。公司为合作伙伴提供“差异化CRO、整合型CDO、协同型CPO、特色CRS”于一体的“创新生物药4C综合业务”。公司已建立全球营销网络,已与全球1200多家药企、生技公司等建立了良好的合作关系;已完成了1200多个新药发现及开发服务项目;已完成了50多个合作研发项目,其中9个合作项目已完成临床申报。公司已获得国家高新技术、上海市专精特新、上海市小巨人和上海“张江之星”企业认定。