61 Treatment-specific immune phenotypes in PBMCs revealed by nELISA high-throughput proteomics

Nathaniel Robichaud, Grant Ongo, Woojong Rho, Ivan Teahulos, Milad Dagher
{"title":"61 Treatment-specific immune phenotypes in PBMCs revealed by nELISA high-throughput proteomics","authors":"Nathaniel Robichaud, Grant Ongo, Woojong Rho, Ivan Teahulos, Milad Dagher","doi":"10.1136/jitc-2023-sitc2023.0061","DOIUrl":null,"url":null,"abstract":"<h3>Background</h3> High-throughput screening (HTS) programs are increasingly adopting high-content technologies that can better inform the selection of drug candidates early on in the pipelines. For cancer immunotherapy, proteomics tools to investigate interactions between cancer and immune cells compromise either content or cost, limiting access to phenotypic data. The affordable gold-standard in proteomics, the ELISA, has proven difficult to scale. At fault has been the cross-reactivity between ELISA reagents when multiplexing beyond a few dozen antibody pairs. Here, we describe the nELISA: a massively-parallelized high-throughput miniaturized ELISA with a content, cost and throughput amenable to HTS, and demonstrate its applicability to characterize immune phenotypes in co-culture systems. <h3>Methods</h3> To overcome the long-standing cross-reactivity issue, the nELISA uses DNA oligos to pre-assemble each pair of antibodies onto a spectrally barcoded microparticle set. The resulting reagents are fully-integrated nELISA sensors that can be read-out on commercial cytometers, enabling highly-multiplexed and high-throughput analysis. Using this approach, we developed a comprehensive inflammatory panel containing 191 cytokines, chemokines, proteases, growth factors, and soluble receptors. Our results show that the nELISA can maintain single-plex specificity, sensitivity, and quantification as content is scaled to 191-plex. Furthermore, the nELISA performs at a throughput of 1536 samples/cytometer/day, yielding &gt;300,000 data points in a single day, at a cost amenable to high-throughput screening. <h3>Results</h3> To demonstrate the nELISA’s utility in HTS, we ran the largest PBMC secretome screen to date, in which &gt;7000 PBMC samples were treated with various inflammatory stimuli, and further perturbed with a selected library of 80 recombinant protein ‘perturbagens’. 191 secreted proteins were profiled in all samples, resulting in ~1.4M datapoints (figure 1A). The nELISA profiles were able to capture phenotypes associated with specific stimulation conditions, individual donors, and potent cytokine perturbagens. By compensating for stimulation and donor differences, we clustered perturbagens according to their effects on PBMC secretomes, identifying well-established cell responses such as Th1 or Th2. Novel phenotypic effects were also identified, such as distinct responses to the near identical CXCL12 alpha and beta isoforms (figure 1B). Interestingly, we observed important similarities between PBMC responses to the cytokine drugs IFN beta and IL-1 Receptor antagonist, supporting the use of anakinra as a replacement for IFN beta in certain indications. <h3>Conclusions</h3> The nELISA captures broad secretome ranges and subtle differences in immune phenotypes, revealing critical insights in cell-based screens. Thus, the nELISA is a powerful new tool for cancer immunotherapy assays, including phenotypic screening, target identification/deconvolution, and discovery of markers of target engagement.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Regular and Young Investigator Award Abstracts","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1136/jitc-2023-sitc2023.0061","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background

High-throughput screening (HTS) programs are increasingly adopting high-content technologies that can better inform the selection of drug candidates early on in the pipelines. For cancer immunotherapy, proteomics tools to investigate interactions between cancer and immune cells compromise either content or cost, limiting access to phenotypic data. The affordable gold-standard in proteomics, the ELISA, has proven difficult to scale. At fault has been the cross-reactivity between ELISA reagents when multiplexing beyond a few dozen antibody pairs. Here, we describe the nELISA: a massively-parallelized high-throughput miniaturized ELISA with a content, cost and throughput amenable to HTS, and demonstrate its applicability to characterize immune phenotypes in co-culture systems.

Methods

To overcome the long-standing cross-reactivity issue, the nELISA uses DNA oligos to pre-assemble each pair of antibodies onto a spectrally barcoded microparticle set. The resulting reagents are fully-integrated nELISA sensors that can be read-out on commercial cytometers, enabling highly-multiplexed and high-throughput analysis. Using this approach, we developed a comprehensive inflammatory panel containing 191 cytokines, chemokines, proteases, growth factors, and soluble receptors. Our results show that the nELISA can maintain single-plex specificity, sensitivity, and quantification as content is scaled to 191-plex. Furthermore, the nELISA performs at a throughput of 1536 samples/cytometer/day, yielding >300,000 data points in a single day, at a cost amenable to high-throughput screening.

Results

To demonstrate the nELISA’s utility in HTS, we ran the largest PBMC secretome screen to date, in which >7000 PBMC samples were treated with various inflammatory stimuli, and further perturbed with a selected library of 80 recombinant protein ‘perturbagens’. 191 secreted proteins were profiled in all samples, resulting in ~1.4M datapoints (figure 1A). The nELISA profiles were able to capture phenotypes associated with specific stimulation conditions, individual donors, and potent cytokine perturbagens. By compensating for stimulation and donor differences, we clustered perturbagens according to their effects on PBMC secretomes, identifying well-established cell responses such as Th1 or Th2. Novel phenotypic effects were also identified, such as distinct responses to the near identical CXCL12 alpha and beta isoforms (figure 1B). Interestingly, we observed important similarities between PBMC responses to the cytokine drugs IFN beta and IL-1 Receptor antagonist, supporting the use of anakinra as a replacement for IFN beta in certain indications.

Conclusions

The nELISA captures broad secretome ranges and subtle differences in immune phenotypes, revealing critical insights in cell-based screens. Thus, the nELISA is a powerful new tool for cancer immunotherapy assays, including phenotypic screening, target identification/deconvolution, and discovery of markers of target engagement.
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
61 .利用nELISA高通量蛋白质组学技术揭示PBMCs治疗特异性免疫表型
高通量筛选(HTS)项目越来越多地采用高含量技术,可以更好地在管道早期选择候选药物。对于癌症免疫治疗,用于研究癌症和免疫细胞之间相互作用的蛋白质组学工具要么降低了内容,要么降低了成本,限制了对表型数据的获取。人们负担得起的蛋白质组学的黄金标准ELISA已被证明难以规模化。在多路复用超过几十对抗体时,ELISA试剂之间的交叉反应性是错误的。在这里,我们描述了nELISA:一种大规模并行化的高通量小型化ELISA,其含量、成本和通量适合HTS,并证明了其在共培养系统中表征免疫表型的适用性。方法为了克服长期存在的交叉反应性问题,nELISA使用DNA寡核苷酸将每对抗体预先组装到一个光谱条形码微粒集上。所得试剂是完全集成的nELISA传感器,可以在商用细胞仪上读出,实现高复用和高通量分析。使用这种方法,我们开发了一个包含191个细胞因子、趋化因子、蛋白酶、生长因子和可溶性受体的综合炎症面板。我们的研究结果表明,当含量缩放到191 plex时,nELISA可以保持单plex的特异性、灵敏度和定量。此外,nELISA的通量为1536个样品/细胞仪/天,在一天内产生300,000个数据点,成本适合高通量筛选。为了证明nELISA在HTS中的应用,我们进行了迄今为止最大的PBMC分泌组筛选,其中7000个PBMC样本接受了各种炎症刺激,并进一步用80个重组蛋白“扰动原”库进行扰动。在所有样品中分析了191种分泌蛋白,得到了约1.4万个数据点(图1A)。nELISA图谱能够捕获与特定刺激条件、个体供体和强效细胞因子扰动原相关的表型。通过补偿刺激和供体差异,我们根据它们对PBMC分泌组的影响对扰动原进行了聚类,确定了已建立的细胞反应,如Th1或Th2。新的表型效应也被发现,例如对几乎相同的CXCL12 α和β亚型的不同反应(图1B)。有趣的是,我们观察到PBMC对细胞因子药物IFN β和IL-1受体拮抗剂的反应有重要的相似性,这支持了在某些适应症中使用阿那金那作为IFN β的替代品。nELISA捕获了广泛的分泌组范围和免疫表型的细微差异,揭示了基于细胞的筛选的关键见解。因此,nELISA是癌症免疫治疗分析的一个强大的新工具,包括表型筛选、靶标识别/反卷积和靶标结合标记的发现。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
1455 Characterization of gene expression signatures of tumor immunogenicity and cellular proliferation from murine cancer models grownin vitroandin vivo 1193 Synergistic targeting of multiple activating pathways with natural killer cell engagers 1413 Immunogenicity of SARS-CoV-2 mRNA vaccines in individuals with thymic epithelial tumors 1150 DM005, an EGFR × MET bispecific antibody-drug conjugate, showed robust anti-tumor activity in PDX models 1274 Infliximab for decompensated diabetes following immune checkpoint inhibitor therapy
×
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
现在去查看 取消
×
提示
确定
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
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