首页 > 最新文献

Regular and Young Investigator Award Abstracts最新文献

英文 中文
1220 Orthogonal CRISPR screens to identify transcriptional and epigenetic regulators of human CD8 T cell function 1220正交CRISPR筛选鉴定人类CD8 T细胞功能的转录和表观遗传调控因子
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1220
Sean McCutcheon, Adam Swartz, Michael Brown, Alejandro Barrera, Christian McRoberts Amador, Keith Siklenka, Lucas Humayun, James Isaacs, Timothy Reddy, Smita Nair, Scott Antonia, Charles Gersbach

Background

The clinical response to adoptive T cell therapies is strongly associated with transcriptional and epigenetic state. Thus, technologies to discover regulators of T cell gene networks and their corresponding phenotypes have great potential to improve the efficacy of T cell therapies.

Methods

We developed pooled CRISPR screening approaches with compact epigenome editors to systematically profile the effects of activation and repression of 120 transcription factors and epigenetic modifiers on human CD8 T cell state.

Results

CRISPR interference and activation screens nominated known and novel regulators of T cell phenotypes with BATF3 emerging as a high confidence gene in both screens. We found that BATF3 overexpression promoted specific features of memory T cells such as increased IL7R expression and glycolytic capacity, while attenuating gene programs associated with cytotoxicity, regulatory T cell function, and T cell exhaustion. In the context of chronic antigen stimulation, BATF3 overexpression countered phenotypic and epigenetic signatures of T cell exhaustion. For example, only 13% of BATF3 engineered T cells co-expressed canonical exhaustion markers (LAG3, TIM3, TIGIT), whereas 65% of wild type T cell co-expressed all three markers after multiple rounds of antigen stimulation. CAR T cells overexpressing BATF3 significantly outperformed control CAR T cells in both in vitro and in vivo tumor models. Moreover, we found that BATF3 programmed a transcriptional profile that correlated with positive clinical response to adoptive T cell therapy. Finally, we performed CRISPR knockout screens with and without BATF3 overexpression to define co-factors and downstream factors of BATF3, as well as other therapeutic targets.

Conclusions

BATF3 overexpression markedly enhanced the therapeutic potential of CD8 T cells in both in vitro and in vivo tumor models. The compact size of BATF3 could seamlessly integrate into current manufacturing processes of FDA-approved adoptive T cell therapies, which all use lentivirus to deliver the CAR construct to donor T cells. To our knowledge, this work is the first example that combines overexpression of a specific transcription factor with a transcription factor wide knockout screen to dissect co-factors and downstream factors and highlights the power of this approach. These screens pointed to a model where BATF3 interacts with JUNB and IRF4 to regulate gene expression and illuminated several other novel targets for further investigation.
背景过继性T细胞治疗的临床反应与转录和表观遗传状态密切相关。因此,发现T细胞基因网络的调节因子及其相应表型的技术对提高T细胞治疗的疗效具有很大的潜力。方法采用紧凑的表观基因组编辑器开发了CRISPR筛选方法,系统地分析了120个转录因子和表观遗传修饰因子的激活和抑制对人CD8 T细胞状态的影响。结果CRISPR干扰和激活筛选提名了已知的和新的T细胞表型调节因子,其中BATF3在两种筛选中都作为高置信度基因出现。我们发现BATF3过表达促进了记忆性T细胞的特定特征,如IL7R表达和糖酵解能力的增加,同时减弱了与细胞毒性、调节性T细胞功能和T细胞衰竭相关的基因程序。在慢性抗原刺激的背景下,BATF3过表达对抗T细胞衰竭的表型和表观遗传特征。例如,只有13%的BATF3工程T细胞共表达典型耗竭标志物(LAG3, TIM3, TIGIT),而65%的野生型T细胞在多轮抗原刺激后共表达所有三种标志物。在体外和体内肿瘤模型中,过表达BATF3的CAR - T细胞都明显优于对照CAR - T细胞。此外,我们发现BATF3编程的转录谱与对过继T细胞治疗的阳性临床反应相关。最后,我们进行了有和没有BATF3过表达的CRISPR敲除筛选,以确定BATF3的辅助因子和下游因子,以及其他治疗靶点。结论BATF3过表达可显著增强CD8 T细胞在体内和体外肿瘤模型中的治疗潜力。BATF3的紧凑尺寸可以无缝集成到fda批准的过继性T细胞疗法的当前制造过程中,这些疗法都使用慢病毒将CAR结构体递送到供体T细胞中。据我们所知,这项工作是第一个将特定转录因子的过表达与转录因子的广泛敲除筛选相结合来解剖辅助因子和下游因子的例子,并突出了这种方法的力量。这些筛选指出了BATF3与JUNB和IRF4相互作用以调节基因表达的模型,并阐明了其他几个有待进一步研究的新靶点。
{"title":"1220 Orthogonal CRISPR screens to identify transcriptional and epigenetic regulators of human CD8 T cell function","authors":"Sean McCutcheon, Adam Swartz, Michael Brown, Alejandro Barrera, Christian McRoberts Amador, Keith Siklenka, Lucas Humayun, James Isaacs, Timothy Reddy, Smita Nair, Scott Antonia, Charles Gersbach","doi":"10.1136/jitc-2023-sitc2023.1220","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1220","url":null,"abstract":"<h3>Background</h3> The clinical response to adoptive T cell therapies is strongly associated with transcriptional and epigenetic state. Thus, technologies to discover regulators of T cell gene networks and their corresponding phenotypes have great potential to improve the efficacy of T cell therapies. <h3>Methods</h3> We developed pooled CRISPR screening approaches with compact epigenome editors to systematically profile the effects of activation and repression of 120 transcription factors and epigenetic modifiers on human CD8 T cell state. <h3>Results</h3> CRISPR interference and activation screens nominated known and novel regulators of T cell phenotypes with BATF3 emerging as a high confidence gene in both screens. We found that BATF3 overexpression promoted specific features of memory T cells such as increased IL7R expression and glycolytic capacity, while attenuating gene programs associated with cytotoxicity, regulatory T cell function, and T cell exhaustion. In the context of chronic antigen stimulation, BATF3 overexpression countered phenotypic and epigenetic signatures of T cell exhaustion. For example, only 13% of BATF3 engineered T cells co-expressed canonical exhaustion markers (LAG3, TIM3, TIGIT), whereas 65% of wild type T cell co-expressed all three markers after multiple rounds of antigen stimulation. CAR T cells overexpressing BATF3 significantly outperformed control CAR T cells in both in vitro and in vivo tumor models. Moreover, we found that BATF3 programmed a transcriptional profile that correlated with positive clinical response to adoptive T cell therapy. Finally, we performed CRISPR knockout screens with and without BATF3 overexpression to define co-factors and downstream factors of BATF3, as well as other therapeutic targets. <h3>Conclusions</h3> BATF3 overexpression markedly enhanced the therapeutic potential of CD8 T cells in both in vitro and in vivo tumor models. The compact size of BATF3 could seamlessly integrate into current manufacturing processes of FDA-approved adoptive T cell therapies, which all use lentivirus to deliver the CAR construct to donor T cells. To our knowledge, this work is the first example that combines overexpression of a specific transcription factor with a transcription factor wide knockout screen to dissect co-factors and downstream factors and highlights the power of this approach. These screens pointed to a model where BATF3 interacts with JUNB and IRF4 to regulate gene expression and illuminated several other novel targets for further investigation.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135161894","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
1122 Vidutolimod, an immunostimulatory virus-like particle, reduces proliferation but enhances the activation of tumor-specific T cells Vidutolimod是一种免疫刺激病毒样颗粒,可减少增殖,但增强肿瘤特异性T细胞的活化
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1122
Travis D Fischer, Caitlin D Lemke-Miltner, George J Weiner

Background

One approach to enhancing the anti-tumor T cell response is to alter the tumor microenvironment (TME) through intratumoral injection (IT) of immunostimulatory agents such as Vidutolimod (Vidu). Vidu is a virus-like particle (VLP) composed of a TLR9 agonist (CpG-A, known as G10) encapsulated by the Qβ bacteriophage capsid. IT Vidu shows considerable promise in early phase clinical trials. The immune response to Vidu is initiated by induction of IFNa production by pDCs within the TME. This effect is dependent on coating of Vidu with antibodies against the Qβ capsid. This is followed by a series of changes in the TME that ultimately result in an enhanced anti-tumor T cell response. Mouse models have shown that the efficacy of IT Vidu depends on the presence of both CD4+ and CD8+ T cells. Most studies to date exploring the impact of Vidu on T cells have focused on the overall T cell population.

Methods

The current studies were designed to further assess the complex mechanisms by which Vidu induces an anti-tumor T cell response through use of the well-established OT-1 mouse model that allows for analysis of the tumor-specific T cell population. OT-1 mice contain CD8+ T cells with a transgenic TCR that recognizes the ovalbumin (OVA) peptide SIINFEKL sequence (OVA257–264) presented on MHC Class I. Prior to culture, OT-1 splenocytes were labeled with CellTrace Violet in order to monitor proliferation over time. OT-1 splenocytes were then cultured with EL4 cells (an OVA-negative T lymphoblast cell line) or E.G7-OVA (OVA-expressing EL4 derivative cells).

Results

Minimal proliferation or evidence of T cell activation was seen when OT-1 CD8+ T cells were cultured with EL4 cells regardless of the addition of Vidu and anti-Qβ antibodies. OT-1 CD8+ T cells cultured with E.G7-OVA cells showed both proliferation and activation as indicated by increased intracellular IFNy and surface PD-1. Addition of Vidu and anti-Qb antibody reduced OT-1 CD8+ proliferation but enhanced production of IFNγ and expression of PD-1. The increase in IFNγ and PD-1 expression was strongest in the dividing OT-1 CD8+ T cell population. Preliminary results of ongoing in vivo studies are consistent with these results.

Conclusions

Vidu reduces proliferation but enhances phenotypic markers of activation expressed by tumor-specific CD8+ T cells (OT-1 cells) when co-cultured with cells expressing OVA, their target antigen. Markers of activation are most notable in dividing OT-1 CD8+ T cells.

Ethics Approval

Mouse studies were approved and performed according to guidelines established by the University of Iowa Institutional Animal Care and Use Committee (IACUC) under the approved Protocol #1011236.
增强抗肿瘤T细胞反应的一种方法是通过肿瘤内注射免疫刺激剂如维多利莫(Vidutolimod, Vidu)来改变肿瘤微环境(TME)。Vidu是一种病毒样颗粒(VLP),由被Qβ噬菌体衣壳包裹的TLR9激动剂(CpG-A,称为G10)组成。IT Vidu在早期临床试验中显示出相当大的希望。对Vidu的免疫应答是通过TME内pDCs诱导IFNa产生而启动的。这种作用依赖于针对Qβ衣壳的抗体包被Vidu。随后,TME发生了一系列变化,最终导致抗肿瘤T细胞反应增强。小鼠模型表明,IT Vidu的功效取决于CD4+和CD8+ T细胞的存在。迄今为止,大多数探索Vidu对T细胞影响的研究都集中在整个T细胞群上。目前的研究旨在通过使用成熟的OT-1小鼠模型进一步评估Vidu诱导抗肿瘤T细胞反应的复杂机制,该模型允许分析肿瘤特异性T细胞群。OT-1小鼠含有CD8+ T细胞,其转基因TCR可识别MHC类上呈现的卵白蛋白(OVA)肽SIINFEKL序列(OVA257-264)。在培养之前,OT-1脾细胞用CellTrace Violet标记,以监测随时间的增殖情况。然后将OT-1脾细胞与EL4细胞(一种ova阴性的T淋巴母细胞系)或E.G7-OVA(表达ova的EL4衍生细胞)一起培养。结果无论是否添加Vidu和抗qβ抗体,将OT-1 CD8+ T细胞与EL4细胞一起培养时,T细胞增殖最小,T细胞活化的证据也很少。与E.G7-OVA细胞一起培养的OT-1 CD8+ T细胞增殖和活化,细胞内IFNy和表面PD-1均增加。Vidu和抗qb抗体的加入降低了OT-1 CD8+的增殖,但增强了IFNγ的产生和PD-1的表达。在OT-1 CD8+ T细胞群中,IFNγ和PD-1表达的增加最为明显。正在进行的体内研究的初步结果与这些结果一致。结论:当与表达OVA的靶抗原细胞共培养时,Vidu可抑制肿瘤特异性CD8+ T细胞(OT-1细胞)的增殖,但可增强其表达的活化表型标志物。活化标记在OT-1 CD8+ T细胞分裂中最为显著。小鼠研究是根据爱荷华大学机构动物护理和使用委员会(IACUC)根据批准的第1011236号协议制定的指导方针批准和进行的。
{"title":"1122 Vidutolimod, an immunostimulatory virus-like particle, reduces proliferation but enhances the activation of tumor-specific T cells","authors":"Travis D Fischer, Caitlin D Lemke-Miltner, George J Weiner","doi":"10.1136/jitc-2023-sitc2023.1122","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1122","url":null,"abstract":"<h3>Background</h3> One approach to enhancing the anti-tumor T cell response is to alter the tumor microenvironment (TME) through intratumoral injection (IT) of immunostimulatory agents such as Vidutolimod (<b>Vidu</b>). Vidu is a virus-like particle (VLP) composed of a TLR9 agonist (CpG-A, known as G10) encapsulated by the Qβ bacteriophage capsid. IT Vidu shows considerable promise in early phase clinical trials. The immune response to Vidu is initiated by induction of IFNa production by pDCs within the TME. This effect is dependent on coating of Vidu with antibodies against the Qβ capsid. This is followed by a series of changes in the TME that ultimately result in an enhanced anti-tumor T cell response. Mouse models have shown that the efficacy of IT Vidu depends on the presence of both CD4+ and CD8+ T cells. Most studies to date exploring the impact of Vidu on T cells have focused on the overall T cell population. <h3>Methods</h3> The current studies were designed to further assess the complex mechanisms by which Vidu induces an anti-tumor T cell response through use of the well-established OT-1 mouse model that allows for analysis of the tumor-specific T cell population. OT-1 mice contain CD8+ T cells with a transgenic TCR that recognizes the ovalbumin (OVA) peptide SIINFEKL sequence (OVA257–264) presented on MHC Class I. Prior to culture, OT-1 splenocytes were labeled with CellTrace Violet in order to monitor proliferation over time. OT-1 splenocytes were then cultured with EL4 cells (an OVA-negative T lymphoblast cell line) or E.G7-OVA (OVA-expressing EL4 derivative cells). <h3>Results</h3> Minimal proliferation or evidence of T cell activation was seen when OT-1 CD8+ T cells were cultured with EL4 cells regardless of the addition of Vidu and anti-Qβ antibodies. OT-1 CD8+ T cells cultured with E.G7-OVA cells showed both proliferation and activation as indicated by increased intracellular IFNy and surface PD-1. Addition of Vidu and anti-Qb antibody reduced OT-1 CD8+ proliferation but enhanced production of IFNγ and expression of PD-1. The increase in IFNγ and PD-1 expression was strongest in the dividing OT-1 CD8+ T cell population. Preliminary results of ongoing <i>in vivo</i> studies are consistent with these results. <h3>Conclusions</h3> Vidu reduces proliferation but enhances phenotypic markers of activation expressed by tumor-specific CD8+ T cells (OT-1 cells) when co-cultured with cells expressing OVA, their target antigen. Markers of activation are most notable in dividing OT-1 CD8+ T cells. <h3>Ethics Approval</h3> Mouse studies were approved and performed according to guidelines established by the University of Iowa Institutional Animal Care and Use Committee (IACUC) under the approved Protocol #1011236.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135161895","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
799 Combination intraperitoneal chemoimmunotherapy triggers a T-cell chemotactic locoregional response in patients with recurrent platinum-sensitive ovarian cancer 799腹腔化疗免疫联合治疗可引发复发性铂敏感卵巢癌患者的t细胞局部趋化反应
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0799
Mackenzy Radolec, Brian Orr, Lixin Zhang, Mary Strange, Syed Zaidi, Robert Edwards, Anda Vlad

Background

The increased prevalence of CD8+ tumor-infiltrating lymphocytes (TIL) in the tumor microenvironment (TME) correlates with improved outcomes in patients with epithelial ovarian cancer (EOC). We hypothesize that a combination of intraperitoneal (IP) chemotherapy (via immunogenic cell death-inducing cisplatin) with dual agent immunotherapy using IV pembrolizumab (anti-PD1) and IP rintatolimod (dsRNA and TLR-3 agonist) promotes increased T cell chemotaxis and cytolytic function, for improved clinical outcomes.

Methods

We have performed translational studies focused on the immune TME, using a longitudinal collection of biospecimens, including plasma, PBMC, IP washes and tumor tissue. The samples were obtained from patients treated in a phase II, investigator- initiated trial (NCT03734692) that tests the efficacy/safety of IP cisplatin/IP rintatolimod/IV pembrolizumab administered in 6 cycles, three weeks apart. Serial collection of biologic samples includes aspiration of peritoneal resident cells (IP washes) before and after each treatment, at each of the 6 cycles. RNA sequencing of IP wash cells was performed using the Novogene platform. Additionally, the MesoScale Delivery (MSD) platform was used to profile 20 biomarkers in the peritoneal samples throughout treatment.

Results

Sequential sampling of the intraperitoneal cavity showed an increase in cellularity immediately after treatment consistent with an ‘acute’ pro-inflammatory reaction. RNA sequencing data showed a significant upregulation acutely in genes associated with anti-tumor immunity (STAT1/STAT2 and downstream targets), T lymphotactic chemokines (CXCL9, 10, 11), and TH1 type response (IFNgamma, Tbet) (p<0.05) all of which are important for T lymphotaxis and function via TCR engagement with cognate tumor antigens. Gene Set Enrichment Analysis demonstrates an acute enrichment in Interferon α response as well as the Interferon γ response (figure 1). MSD measurements in IP washes demonstrated an acute increase in granzyme B, perforin, TNF alpha, CXCL9, 10 and 11, IFN gamma, and IL-15 after treatment (p<0.05) (figure 2). Longitudinal data revealed a progressive increase in CXCL9, 10 and 11, as well as perforin, IFN gamma and TNF alpha from baseline levels at cycle 1 to cycle 6, suggestive of a sustained, ‘chronic’ response during treatment.

Conclusions

Analysis of the locoregional immune environment taken from patients receiving this novel, triple drug combination has demonstrated an acute and persistent increase in biomarkers associated with T cell chemotaxis and T cell function. Ongoing chip cytometry profiling of both IP wash cells and tumor samples will further elucidate the treatment induced changes in various innate and adaptive immune cell types in the TME.

Acknowledgements

Acknowledgments: Pembrolizumab was provided by MERCK and Rintatolimod was provided by AIM Immunotech. This study was funded in part by the National Institutes o
背景:肿瘤微环境(TME)中CD8+肿瘤浸润淋巴细胞(TIL)的增加与上皮性卵巢癌(EOC)患者预后的改善相关。我们假设腹腔内(IP)化疗(通过免疫原性细胞死亡诱导顺铂)与使用IV派姆单抗(抗pd1)和IP rintatolmod (dsRNA和TLR-3激动剂)的双药免疫治疗相结合,可以促进T细胞趋化性和细胞溶解功能的增加,从而改善临床结果。方法:我们对免疫TME进行了转化研究,使用纵向收集的生物标本,包括血浆、PBMC、IP洗涤和肿瘤组织。这些样本来自于研究者发起的II期试验(NCT03734692),该试验测试了顺铂/ rintatolimod/IV派姆单抗的有效性/安全性,分6个周期,间隔3周给药。连续收集生物样本,包括在每次治疗前后,在每6个周期中抽取腹膜驻留细胞(IP洗涤)。利用Novogene平台对IP洗涤细胞进行RNA测序。此外,在整个治疗过程中,MesoScale Delivery (MSD)平台用于分析腹膜样品中的20种生物标志物。结果连续的腹腔取样显示,治疗后细胞数量立即增加,与“急性”促炎反应一致。RNA测序数据显示,与抗肿瘤免疫(STAT1/STAT2和下游靶标)、T淋巴趋化因子(CXCL9、10,11)和TH1型反应(IFNgamma、Tbet)相关的基因显著上调(p<0.05),所有这些基因通过TCR与同源肿瘤抗原结合,对T淋巴趋向性和功能都很重要。基因集富集分析表明急性浓缩以及干扰素γ干扰素α响应响应(图1),并测量IP洗granzyme B,表现出了极大的增加穿孔素、肿瘤坏死因子α,CXCL9, 10和11,干扰素γ,在治疗后和IL-15(术中;0.05)(图2)。纵向数据显示CXCL9逐渐增加,10号和11号,以及穿孔素、干扰素γ和肿瘤坏死因子α从基线水平6周期1周期,持续的暗示,治疗期间的“慢性”反应。对接受这种新型三联药物联合治疗的患者的局部免疫环境分析表明,与T细胞趋化性和T细胞功能相关的生物标志物急性和持续增加。正在进行的IP洗涤细胞和肿瘤样本的芯片细胞分析将进一步阐明治疗诱导的TME中各种先天和适应性免疫细胞类型的变化。致谢:Pembrolizumab由MERCK提供,Rintatolimod由AIM Immunotech提供。本研究部分由美国国立卫生研究院奖P01CA234212以及Magee妇女辅助机构资助。试验注册编号NCT03734692
{"title":"799 Combination intraperitoneal chemoimmunotherapy triggers a T-cell chemotactic locoregional response in patients with recurrent platinum-sensitive ovarian cancer","authors":"Mackenzy Radolec, Brian Orr, Lixin Zhang, Mary Strange, Syed Zaidi, Robert Edwards, Anda Vlad","doi":"10.1136/jitc-2023-sitc2023.0799","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0799","url":null,"abstract":"<h3>Background</h3> The increased prevalence of CD8+ tumor-infiltrating lymphocytes (TIL) in the tumor microenvironment (TME) correlates with improved outcomes in patients with epithelial ovarian cancer (EOC). We hypothesize that a combination of intraperitoneal (IP) chemotherapy (via immunogenic cell death-inducing cisplatin) with dual agent immunotherapy using IV pembrolizumab (anti-PD1) and IP rintatolimod (dsRNA and TLR-3 agonist) promotes increased T cell chemotaxis and cytolytic function, for improved clinical outcomes. <h3>Methods</h3> We have performed translational studies focused on the immune TME, using a longitudinal collection of biospecimens, including plasma, PBMC, IP washes and tumor tissue. The samples were obtained from patients treated in a phase II, investigator- initiated trial (NCT03734692) that tests the efficacy/safety of IP cisplatin/IP rintatolimod/IV pembrolizumab administered in 6 cycles, three weeks apart. Serial collection of biologic samples includes aspiration of peritoneal resident cells (IP washes) before and after each treatment, at each of the 6 cycles. RNA sequencing of IP wash cells was performed using the Novogene platform. Additionally, the MesoScale Delivery (MSD) platform was used to profile 20 biomarkers in the peritoneal samples throughout treatment. <h3>Results</h3> Sequential sampling of the intraperitoneal cavity showed an increase in cellularity immediately after treatment consistent with an ‘acute’ pro-inflammatory reaction. RNA sequencing data showed a significant upregulation acutely in genes associated with anti-tumor immunity (STAT1/STAT2 and downstream targets), T lymphotactic chemokines (CXCL9, 10, 11), and TH1 type response (IFNgamma, Tbet) (p<0.05) all of which are important for T lymphotaxis and function via TCR engagement with cognate tumor antigens. Gene Set Enrichment Analysis demonstrates an acute enrichment in Interferon α response as well as the Interferon γ response (figure 1). MSD measurements in IP washes demonstrated an acute increase in granzyme B, perforin, TNF alpha, CXCL9, 10 and 11, IFN gamma, and IL-15 after treatment (p<0.05) (figure 2). Longitudinal data revealed a progressive increase in CXCL9, 10 and 11, as well as perforin, IFN gamma and TNF alpha from baseline levels at cycle 1 to cycle 6, suggestive of a sustained, ‘chronic’ response during treatment. <h3>Conclusions</h3> Analysis of the locoregional immune environment taken from patients receiving this novel, triple drug combination has demonstrated an acute and persistent increase in biomarkers associated with T cell chemotaxis and T cell function. Ongoing chip cytometry profiling of both IP wash cells and tumor samples will further elucidate the treatment induced changes in various innate and adaptive immune cell types in the TME. <h3>Acknowledgements</h3> Acknowledgments: Pembrolizumab was provided by MERCK and Rintatolimod was provided by AIM Immunotech. This study was funded in part by the National Institutes o","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135161903","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
48 Blood-based peripheral T cell cytotoxicity assay in predicting response to immune checkpoint inhibitors: a US pilot study 基于血液的外周血T细胞毒性测定预测免疫检查点抑制剂的反应:美国的一项试点研究
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0048
Rajesh Nair, Jonathon Woo, Suzanna Lee, Shumei Kato, Michele Baltay, Yali Li, Kota Iwahori, Junichi Akatsuka, Srinath Sampath, Christian Schmedt, Srihari Sampath

Background

Immunotherapy with Immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment over the last decade. Despite the phenomenal success of ICIs, the clinical response is confined to a small subset of patients. Conventional biomarkers such as PDL1 expression and Tumor Mutational Burden (TMB) rely on invasive biopsies and remain inadequate in predicting clinical benefits for patients. Therefore, there is an urgent need to develop better noninvasive biomarkers to predict the response to ICIs and to identify patients for whom these therapies are both safe and effective. A novel blood-based functional assay, peripheral T cell cytotoxicity (PeriCyto), has previously been shown to accurately predict the clinical response for advanced non-small cell lung cancer (NSCLC) in Japanese patients.1 2 The present study aimed to assess clinical feasibility of PeriCyto in predicting the response to ICIs in a US patient cohort.

Methods

Prospective samples (n=13) were obtained from patients with diverse solid tumors prior to treatment with ICIs either as monotherapy or in combination with chemotherapy/targeted therapy. Peripheral Blood Mononuclear Cells (PBMCs) isolated from whole blood were cocultured with U251 cells in the presence of an EphA2/CD3 Bispecific T cell Engager antibody (BiTE). After 48hrs, peripheral T cell cytotoxicity was measured using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay (MTS) assay. PBMCs from a healthy donor with an established cytotoxicity score served as an internal positive control for the assay.

Results

The study evaluated the positive predictive value (PPV), negative predictive value (NPV), sensitivity and specificity of PeriCyto in 13 patients with diverse set of cancers. As previously published1 2 the assay was able to accurately predict the clinical response in the subgroup of patients with advanced NSCLC (4/4). Combining data from patients with all cancer types, the PPV was ~70% and NPV was determined to be 100% (n=13). The assay identified all patients who subsequently responded to ICIs (sensitivity 100%), whereas specificity was 50%. The latter was driven by incorrect prediction of positive treatment response in tumor types traditionally known to be immunologically ‘cold’, raising the possibility that these non-responding histologies may reflect tumor types wherein peripheral T cells are activatable but remain absent or suppressed in the TME.

Conclusions

Overall, these early pilot findings indicate that PeriCyto can help to identify patients who may benefit from ICIs. Limitations include small sample size and single site design. Future efforts will focus on expansion of this patient cohort and extending followup to further assess PeriCyto predictive value.

Acknowledgements

We would like to thank the patients and healthy donors who participated in the study.

References

Iwahori K, S
在过去的十年中,免疫检查点抑制剂(ICIs)的免疫治疗已经彻底改变了癌症治疗。尽管ICIs取得了显著的成功,但临床反应仅限于一小部分患者。传统的生物标志物,如PDL1表达和肿瘤突变负荷(TMB)依赖于侵入性活检,在预测患者的临床获益方面仍然不足。因此,迫切需要开发更好的无创生物标志物来预测对ICIs的反应,并确定这些治疗既安全又有效的患者。一种新的基于血液的功能测定,外周T细胞毒性(PeriCyto),先前已被证明可以准确预测日本晚期非小细胞肺癌(NSCLC)患者的临床反应。本研究旨在评估美国患者队列中使用PeriCyto预测ICIs疗效的临床可行性。方法前瞻性样本(n=13)来自不同类型的实体瘤患者,在接受ICIs单独治疗或联合化疗/靶向治疗之前。在EphA2/CD3双特异性T细胞接合抗体(BiTE)存在下,将全血外周血单核细胞(PBMCs)与U251细胞共培养。48h后,采用3-(4,5-二甲基噻唑-2-基)-5-(3-羧基甲氧基苯基)-2-(4-磺苯基)- 2h -四氮唑法(MTS)测定外周T细胞毒性。来自健康供体的pbmc具有确定的细胞毒性评分,可作为该试验的内部阳性对照。结果本研究评估了13例不同类型肿瘤患者的PeriCyto阳性预测值(PPV)、阴性预测值(NPV)、敏感性和特异性。正如先前发表的12,该检测能够准确预测晚期NSCLC患者亚组的临床反应(4/4)。结合所有癌症类型患者的数据,确定PPV为~70%,NPV为100% (n=13)。该检测确定了所有随后对ICIs有反应的患者(敏感性100%),而特异性为50%。后者是由于对传统上已知的免疫“冷”肿瘤类型的阳性治疗反应的错误预测所驱动的,这提高了这些无反应组织学可能反映肿瘤类型的可能性,其中外周T细胞是可激活的,但在TME中仍然缺失或抑制。总的来说,这些早期的试验结果表明,PeriCyto可以帮助识别可能受益于ici的患者。限制包括小样本量和单一站点设计。未来的工作将集中在扩大患者队列和延长随访时间,以进一步评估PeriCyto的预测价值。我们要感谢参与这项研究的患者和健康捐赠者。Iwahori K, Shintani Y, Funaki S, Yamamoto Y, Matsumoto M, Yoshida T, Morimoto-Okazawa A, Kawashima A, Sato E, Gottschalk S, Okumura M, Kumanogoh A, Wada H.外周T细胞毒性预测T细胞在肿瘤微环境中的功能。科学通报2019;(9):2636。Iwahori K, Uenami T, Yano Y, Ueda T, Tone M, neto Y, Suga Y, Fukushima K, Shiroyama T, Miyake K, Koyama S, Hirata H, nagatomi, Kida H, Mori M, Takeda Y, kumangoh A, Wada H。外周血T细胞细胞毒性预测晚期非小细胞癌患者抗PD-1治疗的疗效。科学通报2022;(12):17461。本研究经UCSD伦理委员会批准,批准号:UCSD 130794。本摘要及任何随附图片的发表均已获得患者的书面知情同意。
{"title":"48 Blood-based peripheral T cell cytotoxicity assay in predicting response to immune checkpoint inhibitors: a US pilot study","authors":"Rajesh Nair, Jonathon Woo, Suzanna Lee, Shumei Kato, Michele Baltay, Yali Li, Kota Iwahori, Junichi Akatsuka, Srinath Sampath, Christian Schmedt, Srihari Sampath","doi":"10.1136/jitc-2023-sitc2023.0048","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0048","url":null,"abstract":"<h3>Background</h3> Immunotherapy with Immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment over the last decade. Despite the phenomenal success of ICIs, the clinical response is confined to a small subset of patients. Conventional biomarkers such as PDL1 expression and Tumor Mutational Burden (TMB) rely on invasive biopsies and remain inadequate in predicting clinical benefits for patients. Therefore, there is an urgent need to develop better noninvasive biomarkers to predict the response to ICIs and to identify patients for whom these therapies are both safe and effective. A novel blood-based functional assay, peripheral T cell cytotoxicity (PeriCyto), has previously been shown to accurately predict the clinical response for advanced non-small cell lung cancer (NSCLC) in Japanese patients.<sup>1 2</sup> The present study aimed to assess clinical feasibility of PeriCyto in predicting the response to ICIs in a US patient cohort. <h3>Methods</h3> Prospective samples (n=13) were obtained from patients with diverse solid tumors prior to treatment with ICIs either as monotherapy or in combination with chemotherapy/targeted therapy. Peripheral Blood Mononuclear Cells (PBMCs) isolated from whole blood were cocultured with U251 cells in the presence of an EphA2/CD3 Bispecific T cell Engager antibody (BiTE). After 48hrs, peripheral T cell cytotoxicity was measured using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay (MTS) assay. PBMCs from a healthy donor with an established cytotoxicity score served as an internal positive control for the assay. <h3>Results</h3> The study evaluated the positive predictive value (PPV), negative predictive value (NPV), sensitivity and specificity of PeriCyto in 13 patients with diverse set of cancers. As previously published<sup>1 2</sup> the assay was able to accurately predict the clinical response in the subgroup of patients with advanced NSCLC (4/4). Combining data from patients with all cancer types, the PPV was ~70% and NPV was determined to be 100% (n=13). The assay identified all patients who subsequently responded to ICIs (sensitivity 100%), whereas specificity was 50%. The latter was driven by incorrect prediction of positive treatment response in tumor types traditionally known to be immunologically ‘cold’, raising the possibility that these non-responding histologies may reflect tumor types wherein peripheral T cells are activatable but remain absent or suppressed in the TME. <h3>Conclusions</h3> Overall, these early pilot findings indicate that PeriCyto can help to identify patients who may benefit from ICIs. Limitations include small sample size and single site design. Future efforts will focus on expansion of this patient cohort and extending followup to further assess PeriCyto predictive value. <h3>Acknowledgements</h3> We would like to thank the patients and healthy donors who participated in the study. <h3>References</h3> Iwahori K, S","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135161917","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
1465 Multiplex imaging identifies unique immunophenotypic and spatial characteristics associated with response to immune checkpoint inhibitors (ICIs) in metastatic urothelial cancer (mUC) 1465多重成像识别转移性尿路上皮癌(mUC)中与免疫检查点抑制剂(ICIs)应答相关的独特免疫表型和空间特征
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1465
Jonathan Anker, John-William Sidhom, Guray Akturk, Sudeh Izadmehr, Justin David, Saurabh Gupta, Seunghee Kim-Schulze, Padmanee Sharma, Sacha Gnjatic, Matthew Galsky

Background

ICIs increase survival in mUC1 but only a subset (~15–25%) of patients experience durable disease control.2 Differences in the tumor microenvironment (TME) might underlie such differential responses. However, the complex network of cellular interactions within the TME that associate with response and resistance to ICIs remains underexplored.

Methods

Multiplex Immunohistochemical Consecutive Staining on a Single Slide (MICSSS)3 was performed on UC specimens (N=40) from CheckMate 2754 prior to treatment with nivolumab. 9 immunohistochemical stains (PD-L1, CD8, CD3, pan-cytokeratin, fibronectin, CD68, FAP, DC-LAMP,5 CD11b) were sequentially performed on a single slide per patient. Image processing, whole slide annotation (median 464,554 cells/slide), and intra- and extra-tumoral compartment training, were performed using QuPath (figure 1).6 Responders (CR, PR) and non-responders (SD, PD) were defined per RECIST v1.1. Immunophenotypic designations of ‘inflamed’, ‘excluded’, and ‘desert’ were defined via tumor margin CD8 analysis.7 Lymphoid aggregates were identified morphologically with dense CD3 positivity. Single-cell spatial analysis was performed defining neighborhoods as the 25 nearest neighboring cells.

Results

TME characterization demonstrated inter-tumoral heterogeneity, both in the intra- and extra-tumoral compartments (figure 2). Responders contained >2-fold increased intra-tumoral CD8 cells, though no cell types were significantly altered in comparison to non-responders. In contrast, extra-tumoral CD3, CD8, CD3CD8-, DC-LAMP (PD-L1- and PD-L1+), and PD-L1+ CD11b cells were significantly enriched in responders (figure 3). Inflamed tumors were more prevalent and excluded/desert tumors less prevalent in responders, with inflamed tumors containing increased intra-tumoral T cell and DC-LAMP infiltration. There were no significant differences in infiltrate composition between inflamed responders and inflamed non-responders, while excluded/desert responders demonstrated enrichment for extra-tumoral DC-LAMP cells (PD-L1- and PD-L1+) and intra-tumoral PD-L1- DC-LAMP cells as compared to excluded/desert non-responders (figure 4). Responder tumors also contained an increased density of lymphoid aggregates, which were found in closer proximity to tumor regions, were associated with increased survival, and were comprised of a greater degree of DC-LAMP cells (figure 5). Spatial analysis of extra-tumoral cells identified a unique immune and tumor-enriched PD-L1+ neighborhood (cluster 0) predominant in responders, and a distinct CD11b and tumor-based neighborhood devoid of PD-L1 and other immune cells (cluster 2) predominant in non-responders (figure 6).

Conclusions

Multiplex immunohistochemistry identified unique immunophenotypic and spatial TME features specific to mUC responders to ICI. Both increased infiltration and the geogra
背景:ICIs可增加mUC1患者的生存,但只有一小部分(约15-25%)患者能获得持久的疾病控制肿瘤微环境(TME)的差异可能是这种差异反应的基础。然而,TME内与ICIs反应和耐药性相关的细胞相互作用的复杂网络仍未得到充分研究。方法对来自CheckMate 2754的UC标本(N=40)在接受纳武单抗治疗前进行多重免疫组化连续染色(MICSSS)。9个免疫组化染色(PD-L1、CD8、CD3、泛细胞角蛋白、纤维连接蛋白、CD68、FAP、DC-LAMP、5个CD11b)依次在每位患者的一张载玻片上进行。使用QuPath进行图像处理、整张幻灯片注释(平均464,554个细胞/张)以及瘤内和瘤外隔室训练(图1)根据RECIST v1.1定义应答者(CR, PR)和无应答者(SD, PD)。通过肿瘤边缘CD8分析确定“炎症”、“排除”和“沙漠”的免疫表型形态学上发现淋巴样聚集体具有密集的CD3阳性。进行单细胞空间分析,将邻居定义为最近的25个相邻细胞。结果TME表征显示肿瘤间的异质性,无论是在肿瘤内还是肿瘤外(图2)。应答者的肿瘤内CD8细胞增加了2倍,尽管与无应答者相比,细胞类型没有显著改变。相比之下,肿瘤外CD3、CD8、CD3CD8-、DC-LAMP (PD-L1-和PD-L1+)和PD-L1+ CD11b细胞在应答者中显著富集(图3)。炎症性肿瘤更普遍,而在应答者中,排除/沙漠肿瘤的发生率较低,炎症性肿瘤中肿瘤内T细胞和DC-LAMP浸润增加。炎症反应者和炎症无反应者之间浸润成分没有显著差异,而与排除/沙漠无反应者相比,排除/沙漠反应者显示肿瘤外DC-LAMP细胞(PD-L1-和PD-L1+)和肿瘤内PD-L1- DC-LAMP细胞富集(图4)。反应性肿瘤还含有更接近肿瘤区域的淋巴样聚集体密度增加,这与生存率增加有关。并且由更大程度的DC-LAMP细胞组成(图5)。肿瘤外细胞的空间分析发现,在应答者中主要存在一个独特的免疫和肿瘤富集的PD-L1+邻域(簇0),而在无应答者中主要存在一个独特的CD11b和基于肿瘤的邻域,缺乏PD-L1和其他免疫细胞(簇2)(图6)。多重免疫组织化学鉴定出了ICI的mUC应答者特有的独特免疫表型和空间TME特征。T细胞、树突状细胞和PD-L1阳性的浸润和地理分布的增加,特别是在肿瘤外腔室,可能是识别ICI应答者的关键。参考文献bellmont J, de Wit R, Vaughn DJ, Fradet Y, Lee J-L, Fong L,等。派姆单抗作为晚期尿路上皮癌的二线治疗。新英格兰医学杂志。2017;376:1015-26。Galsky MD, Arija JÁA, Bamias A, Davis ID, Santis MD, Kikuchi E等。Atezolizumab联合或不联合化疗治疗转移性尿路上皮癌(IMvigor130):一项多中心、随机、安慰剂对照的3期试验。《柳叶刀》,2020;395:1547-57。Akturk G, Sweeney R, Remark R, Merad M, Gnjatic S.单张切片上的多重免疫组化连续染色(MICSSS):用于高维组织分析的多重显色免疫组化试验。[j] .生物医学工程学报。2010;29(5):497 - 519。李建军,李建军,李建军,等。Nivolumab用于铂治疗后转移性尿路上皮癌(CheckMate 275):一项多中心、单组、2期试验。中华医学杂志。2017;18:312-22。Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J,等。保守的树突细胞调控程序限制了抗肿瘤免疫。大自然。2020;580:257 - 62。Bankhead P, Loughrey MB, Fernández JA, Dombrowski Y, mcat DG, Dunne PD,等。QuPath:用于数字病理图像分析的开源软件。科学通报,2017;7:16878。Braun DA, Hou Y, Bakouny Z, official M, Sant’Angelo M, Forman J,等。体细胞改变和免疫浸润的相互作用调节了晚期透明细胞肾细胞癌对PD-1阻断的反应。中华医学杂志。2020;26:9 9 - 18。初始临床试验NCT02387996获得了适当的伦理批准,并按照国际协调会议定义的良好临床实践指南进行。
{"title":"1465 Multiplex imaging identifies unique immunophenotypic and spatial characteristics associated with response to immune checkpoint inhibitors (ICIs) in metastatic urothelial cancer (mUC)","authors":"Jonathan Anker, John-William Sidhom, Guray Akturk, Sudeh Izadmehr, Justin David, Saurabh Gupta, Seunghee Kim-Schulze, Padmanee Sharma, Sacha Gnjatic, Matthew Galsky","doi":"10.1136/jitc-2023-sitc2023.1465","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1465","url":null,"abstract":"<h3>Background</h3> ICIs increase survival in mUC<sup>1</sup> but only a subset (~15–25%) of patients experience durable disease control.<sup>2</sup> Differences in the tumor microenvironment (TME) might underlie such differential responses. However, the complex network of cellular interactions within the TME that associate with response and resistance to ICIs remains underexplored. <h3>Methods</h3> Multiplex Immunohistochemical Consecutive Staining on a Single Slide (MICSSS)<sup>3</sup> was performed on UC specimens (N=40) from CheckMate 275<sup>4</sup> prior to treatment with nivolumab. 9 immunohistochemical stains (PD-L1, CD8, CD3, pan-cytokeratin, fibronectin, CD68, FAP, DC-LAMP,<sup>5</sup> CD11b) were sequentially performed on a single slide per patient. Image processing, whole slide annotation (median 464,554 cells/slide), and intra- and extra-tumoral compartment training, were performed using QuPath (figure 1).<sup>6</sup> Responders (CR, PR) and non-responders (SD, PD) were defined per RECIST v1.1. Immunophenotypic designations of ‘inflamed’, ‘excluded’, and ‘desert’ were defined via tumor margin CD8 analysis.<sup>7</sup> Lymphoid aggregates were identified morphologically with dense CD3 positivity. Single-cell spatial analysis was performed defining neighborhoods as the 25 nearest neighboring cells. <h3>Results</h3> TME characterization demonstrated inter-tumoral heterogeneity, both in the intra- and extra-tumoral compartments (figure 2). Responders contained &gt;2-fold increased intra-tumoral CD8 cells, though no cell types were significantly altered in comparison to non-responders. In contrast, extra-tumoral CD3, CD8, CD3CD8-, DC-LAMP (PD-L1- and PD-L1+), and PD-L1+ CD11b cells were significantly enriched in responders (figure 3). Inflamed tumors were more prevalent and excluded/desert tumors less prevalent in responders, with inflamed tumors containing increased intra-tumoral T cell and DC-LAMP infiltration. There were no significant differences in infiltrate composition between inflamed responders and inflamed non-responders, while excluded/desert responders demonstrated enrichment for extra-tumoral DC-LAMP cells (PD-L1- and PD-L1+) and intra-tumoral PD-L1- DC-LAMP cells as compared to excluded/desert non-responders (figure 4). Responder tumors also contained an increased density of lymphoid aggregates, which were found in closer proximity to tumor regions, were associated with increased survival, and were comprised of a greater degree of DC-LAMP cells (figure 5). Spatial analysis of extra-tumoral cells identified a unique immune and tumor-enriched PD-L1+ neighborhood (cluster 0) predominant in responders, and a distinct CD11b and tumor-based neighborhood devoid of PD-L1 and other immune cells (cluster 2) predominant in non-responders (figure 6). <h3>Conclusions</h3> Multiplex immunohistochemistry identified unique immunophenotypic and spatial TME features specific to mUC responders to ICI. Both increased infiltration and the geogra","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135162078","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
1164 BSA01, a bispecific antibody-drug conjugate targeting EGFR and membrane-bound MUC-1-C, exhibits anti-tumor efficacyin vivo 1164 BSA01是一种双特异性抗体-药物偶联物,靶向EGFR和膜结合的MUC-1-C,在体内具有抗肿瘤作用
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1164
Yifu Zhang, Chengzhang Shang, Anqi Wang, Jia Zhang, Yuji Liu, Hao Li, Xiaopeng Li, Gao An, W Frank An, Chaoshe Guo, Yi Yang

Background

EGFR and the surface glycoprotein MUC-1 are commonly co-expressed in several malignancies, including esophageal squamous cell carcinomas, non-small cell lung cancers (NSCLC), and triple-negative breast cancers. To overcome limitations of current EGFR- and MUC-1-targeting therapies, we generated a novel bispecific antibody-drug conjugate conjugated with monomethyl auristatin E, BSA01, which targets both antigens. EGFR and MUC-1 antibodies were identified using RenLite® fully human common light chain mice and further evaluated for efficacy and specificity in vitro and in vivo.

Methods

Parental MUC1 antibody was tested in a binding competition assay alongside a benchmark identifying cleaved MUC-1.1 Surface plasmon resonance and flow cytometry was employed to determine the affinity and species cross-reactivity of anti-MUC1. Binding avidity was assessed by flow cytometry. Internalization and cytotoxicity were assessed by Incucyte® live cell imaging. The specificity of BSA01 was assessed by comparing the cytotoxicity of tumor cells and neonatal Human Epidermal Keratinocytes (HEKn). The efficacy of BSA01 in preventing growth of cell line-derived (CDX) and patient-derived xenograft (PDX) tumors in vivo was subsequently evaluated.

Results

Binding competition assays indicate that the parental MUC1 antibody of BSA01 binds to MUC1-C*, which remains membrane-bound after cleavage. The affinity of the anti-MUC1 antibody was similar to human and cynomolgus monkey antigens. BSA01 BsAbs bound EGFR+MUC-1+ cell lines (HCC827 and HCC70) with stronger avidity than a single-positive cell line (A431). The internalization activity of BSA01 BsAbs was superior to its monovalent parental antibodies. BSA01 was able to effectively induce cytotoxicity in vitro, while only marginally affecting human normal cells that express low levels of MUC1 and EGFR. Notably, BSA01 showed superior anti-tumor efficacy when compared with benchmark ADCs in CDX and pancreatic PDX models in vivo, In NSCLC PDX tumors, BSA01 performed similar to MUC-1 benchmark.

Conclusions

We generated a novel bispecific ADC targeting EGFR and MUC-1. The MUC-1 arm of BSA01 binds to the cleaved MUC1-C*protein, which remains membrane bound on tumor cells. BSA01 exhibits strong affinity and internalization activity in vitro, while also demonstrating a good safety profile. Moreover, BSA01 shows superior anti-tumor efficacy to benchmarks in certain in vivo PDX models evaluated.

Reference

Wu G, Kim D, Kim JN, Park S, Maharjan S, Koh H, Moon K, Lee Y, Kwon HJ. A Mucin1 C-terminal Subunit-directed Monoclonal Antibody Targets Overexpressed Mucin1 in Breast Cancer. Theranostics, 2018;8(1):78–91. https://doi.org/10.7150/thno.21278

Ethics Approval

All animal studies were reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) of Biocytogen Beijing Co., Ltd.
EGFR和表面糖蛋白MUC-1通常在几种恶性肿瘤中共表达,包括食管鳞状细胞癌、非小细胞肺癌(NSCLC)和三阴性乳腺癌。为了克服目前EGFR-和muc -1靶向治疗的局限性,我们产生了一种新的双特异性抗体-药物偶联物,与单甲基耳素E偶联,BSA01,可靶向两种抗原。使用RenLite®全人普通轻链小鼠鉴定EGFR和MUC-1抗体,并进一步评估体外和体内的功效和特异性。方法采用结合竞争法检测亲本MUC1抗体和鉴定裂解MUC1 -1.1的基准抗体,采用表面等离子体共振和流式细胞术检测抗MUC1的亲和力和种间反应性。通过流式细胞术评估结合亲和度。采用Incucyte®活细胞成像技术评估内化和细胞毒性。通过比较肿瘤细胞和新生儿人表皮角质形成细胞(HEKn)的细胞毒性来评估BSA01的特异性。随后评估了BSA01在体内阻止细胞系来源(CDX)和患者来源的异种移植(PDX)肿瘤生长的功效。结果BSA01的亲本MUC1抗体与MUC1- c *结合,MUC1- c *在裂解后仍保持膜结合。muc1抗体的亲和力与人和食蟹猴抗原相似。BSA01 BsAbs结合EGFR+MUC-1+细胞系(HCC827和HCC70)的亲和力比单阳性细胞系(A431)强。BSA01 bsab的内化活性优于其单价亲本抗体。BSA01能够在体外有效诱导细胞毒性,而仅轻微影响表达低水平MUC1和EGFR的人正常细胞。值得注意的是,与基准adc相比,BSA01在体内CDX和胰腺PDX模型中表现出更强的抗肿瘤功效,在NSCLC PDX肿瘤中,BSA01的抗肿瘤效果与MUC-1相似。我们生成了一种新的靶向EGFR和MUC-1的双特异性ADC。BSA01的muc1臂与裂解的MUC1-C*蛋白结合,MUC1-C*蛋白在肿瘤细胞上保持膜结合。BSA01在体外具有较强的亲和力和内化活性,同时也具有良好的安全性。此外,BSA01在某些体内PDX模型中显示出优于基准的抗肿瘤功效。参考文献吴刚,Kim D, Kim JN, Park S, Maharjan S, Koh H, Moon K, Lee Y, Kwon HJ。Mucin1 c端亚基定向单克隆抗体靶向乳腺癌中过表达的Mucin1开展,2018;8(1):78 - 91。https://doi.org/10.7150/thno.21278伦理批准所有动物研究均由北京生物细胞原有限公司机构动物护理和使用委员会(IACUC)审查和批准。
{"title":"1164 BSA01, a bispecific antibody-drug conjugate targeting EGFR and membrane-bound MUC-1-C, exhibits anti-tumor efficacy<i>in vivo</i>","authors":"Yifu Zhang, Chengzhang Shang, Anqi Wang, Jia Zhang, Yuji Liu, Hao Li, Xiaopeng Li, Gao An, W Frank An, Chaoshe Guo, Yi Yang","doi":"10.1136/jitc-2023-sitc2023.1164","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1164","url":null,"abstract":"<h3>Background</h3> EGFR and the surface glycoprotein MUC-1 are commonly co-expressed in several malignancies, including esophageal squamous cell carcinomas, non-small cell lung cancers (NSCLC), and triple-negative breast cancers. To overcome limitations of current EGFR- and MUC-1-targeting therapies, we generated a novel bispecific antibody-drug conjugate conjugated with monomethyl auristatin E, BSA01, which targets both antigens. EGFR and MUC-1 antibodies were identified using RenLite® fully human common light chain mice and further evaluated for efficacy and specificity <i>in vitro</i> and <i>in vivo.</i> <h3>Methods</h3> Parental MUC1 antibody was tested in a binding competition assay alongside a benchmark identifying cleaved MUC-1.<sup>1</sup> Surface plasmon resonance and flow cytometry was employed to determine the affinity and species cross-reactivity of anti-MUC1. Binding avidity was assessed by flow cytometry. Internalization and cytotoxicity were assessed by Incucyte® live cell imaging. The specificity of BSA01 was assessed by comparing the cytotoxicity of tumor cells and neonatal Human Epidermal Keratinocytes (HEKn). The efficacy of BSA01 in preventing growth of cell line-derived (CDX) and patient-derived xenograft (PDX) tumors <i>in vivo</i> was subsequently evaluated. <h3>Results</h3> Binding competition assays indicate that the parental MUC1 antibody of BSA01 binds to MUC1-C*, which remains membrane-bound after cleavage. The affinity of the anti-MUC1 antibody was similar to human and cynomolgus monkey antigens. BSA01 BsAbs bound EGFR+MUC-1+ cell lines (HCC827 and HCC70) with stronger avidity than a single-positive cell line (A431). The internalization activity of BSA01 BsAbs was superior to its monovalent parental antibodies. BSA01 was able to effectively induce cytotoxicity <i>in vitro</i>, while only marginally affecting human normal cells that express low levels of MUC1 and EGFR. Notably, BSA01 showed superior anti-tumor efficacy when compared with benchmark ADCs in CDX and pancreatic PDX models <i>in vivo</i>, In NSCLC PDX tumors, BSA01 performed similar to MUC-1 benchmark. <h3>Conclusions</h3> We generated a novel bispecific ADC targeting EGFR and MUC-1. The MUC-1 arm of BSA01 binds to the cleaved MUC1-C*protein, which remains membrane bound on tumor cells. BSA01 exhibits strong affinity and internalization activity <i>in vitro</i>, while also demonstrating a good safety profile. Moreover, BSA01 shows superior anti-tumor efficacy to benchmarks in certain <i>in vivo</i> PDX models evaluated. <h3>Reference</h3> Wu G, Kim D, Kim JN, Park S, Maharjan S, Koh H, Moon K, Lee Y, Kwon HJ. A Mucin1 C-terminal Subunit-directed Monoclonal Antibody Targets Overexpressed Mucin1 in Breast Cancer. <i>Theranostics</i>, 2018;<b>8</b>(1):78–91. https://doi.org/10.7150/thno.21278 <h3>Ethics Approval</h3> All animal studies were reviewed and approved by the Institutional Animal Care and Use Committee (IACUC) of Biocytogen Beijing Co., Ltd.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135162090","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
5 Severely immunodeficient NOG-EXL mice allow for humanization and development of a human glioblastoma-derived tumor microenvironment 5 .严重免疫缺陷的NOG-EXL小鼠允许人胶质母细胞瘤源性肿瘤微环境的人源化和发展
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0005
Zev Binder, Lamia Lamrani, Anthony Secreto, Nicholas Skuli, Moriah Jacobson, Charles-Antoine Assenmacher, Elinor Willis, Enrico Radaelli, Donald O’Rourke, Cecile Alanio, Maclean Nasrallah

Background

Cellular immunotherapy has resulted in tremendous therapeutic advances across liquid tumors. However, for solid tumors, including glioblastoma (GBM), significant limitations still exist in successfully translating immune-based adoptive cell therapeutics from the bench to the clinic. The majority of preclinical in vivo GBM models either use human cell lines grown in immunodeficient animals or syngeneic tumors grown in immunocompetent animals. Both approaches lack critical immune components of the tumor microenvironment (TME), thus inherently limiting the ability to adequately model immunotherapies.

Methods

Here, we demonstrate the ability of hematopoietic stem cells (HSCs) from a GBM patient to successfully engraft in the severely immunodeficient NOG-EXL mouse strain and generate a patient-derived immune system. Mouse brains, with and without tumors, were evaluated by immunohistochemistry and flow cytometry.

Results

Intraosseous injections of a low number (10,000) of CD34+ HSCs, obtained from a bone marrow draw from the patient at the time of tumor resection, resulted in an average of 20% human CD45+ cells in peripheral blood 17 weeks post-transplant. GBM organoids, established from the same patient, were implanted intracranially at Week 18 and allowed to grow for 5 weeks without significant clinical evidence of graft-vs-host disease or macrophage activation syndrome. Immunohistochemical and flow cytometry analyses of mouse brains, with and without tumors, revealed infiltration of human-derived, CD45+/CD33+ cells, in proximity to the tumor engraftment site.

Conclusions

These results demonstrate the ability of patient-obtained CD34+ HSCs to engraft and form a TME in severely immunodeficient mice. Subsequent work will focus on the differences between autologous and allogeneic humanized tumor-bearing mice in the context of the originating tumor. Additional future work will focus on testing cellular therapies directed towards the relevant areas of the autologous model.
细胞免疫疗法在液体肿瘤治疗方面取得了巨大的进展。然而,对于实体肿瘤,包括胶质母细胞瘤(GBM),在成功地将基于免疫的过继细胞疗法从实验室转化为临床方面仍然存在显著的局限性。大多数临床前体内GBM模型要么使用在免疫缺陷动物中生长的人类细胞系,要么使用在免疫正常动物中生长的同基因肿瘤。这两种方法都缺乏肿瘤微环境(TME)的关键免疫成分,因此固有地限制了充分模拟免疫治疗的能力。方法在此,我们证明了来自GBM患者的造血干细胞(hsc)能够成功植入严重免疫缺陷的NOG-EXL小鼠品系,并产生患者源性免疫系统。采用免疫组织化学和流式细胞术对小鼠脑内有无肿瘤进行评价。结果骨内注射少量(10,000)CD34+造血干细胞,从肿瘤切除时的患者骨髓中提取,移植后17周外周血中平均有20%的人CD45+细胞。从同一患者建立的GBM类器官在第18周植入颅内,并允许生长5周,无明显的移植物抗宿主病或巨噬细胞激活综合征的临床证据。免疫组织化学和流式细胞术分析显示,肿瘤植入部位附近有人源性CD45+/CD33+细胞浸润。结论这些结果表明患者获得的CD34+造血干细胞能够在严重免疫缺陷小鼠中移植并形成TME。后续的工作将集中在自体和异体人源化荷瘤小鼠在原发肿瘤背景下的差异。未来的额外工作将集中于测试针对自体模型相关区域的细胞疗法。
{"title":"5 Severely immunodeficient NOG-EXL mice allow for humanization and development of a human glioblastoma-derived tumor microenvironment","authors":"Zev Binder, Lamia Lamrani, Anthony Secreto, Nicholas Skuli, Moriah Jacobson, Charles-Antoine Assenmacher, Elinor Willis, Enrico Radaelli, Donald O’Rourke, Cecile Alanio, Maclean Nasrallah","doi":"10.1136/jitc-2023-sitc2023.0005","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0005","url":null,"abstract":"<h3>Background</h3> Cellular immunotherapy has resulted in tremendous therapeutic advances across liquid tumors. However, for solid tumors, including glioblastoma (GBM), significant limitations still exist in successfully translating immune-based adoptive cell therapeutics from the bench to the clinic. The majority of preclinical <i>in vivo</i> GBM models either use human cell lines grown in immunodeficient animals or syngeneic tumors grown in immunocompetent animals. Both approaches lack critical immune components of the tumor microenvironment (TME), thus inherently limiting the ability to adequately model immunotherapies. <h3>Methods</h3> Here, we demonstrate the ability of hematopoietic stem cells (HSCs) from a GBM patient to successfully engraft in the severely immunodeficient NOG-EXL mouse strain and generate a patient-derived immune system. Mouse brains, with and without tumors, were evaluated by immunohistochemistry and flow cytometry. <h3>Results</h3> Intraosseous injections of a low number (10,000) of CD34+ HSCs, obtained from a bone marrow draw from the patient at the time of tumor resection, resulted in an average of 20% human CD45+ cells in peripheral blood 17 weeks post-transplant. GBM organoids, established from the same patient, were implanted intracranially at Week 18 and allowed to grow for 5 weeks without significant clinical evidence of graft-vs-host disease or macrophage activation syndrome. Immunohistochemical and flow cytometry analyses of mouse brains, with and without tumors, revealed infiltration of human-derived, CD45+/CD33+ cells, in proximity to the tumor engraftment site. <h3>Conclusions</h3> These results demonstrate the ability of patient-obtained CD34+ HSCs to engraft and form a TME in severely immunodeficient mice. Subsequent work will focus on the differences between autologous and allogeneic humanized tumor-bearing mice in the context of the originating tumor. Additional future work will focus on testing cellular therapies directed towards the relevant areas of the autologous model.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135162099","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
1106 Molecular mechanisms of immunogenic cell death driven by PT-112 PT-112驱动免疫原性细胞死亡的分子机制
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1106
Emma Guilbaud, Takahiro Yamazaki, Maria Congenie, Christina Yim, Tyler Ames, Lorenzo Galluzzi

Background

PT-112 is a novel immunogenic small molecule1 under Phase II clinical development for cancer therapy.2–8 Besides mediating cytostatic and cytotoxic effects in numerous human and mouse cancer cells, PT-112 elicits various danger signals that are linked to immunogenic cell death (ICD) such as calreticulin exposure, as well as ATP and HMGB1 secretion.1 9–11 Accordingly, mouse cancer cells succumbing to PT-112 in vitro efficiently protect immunocompetent, tumor-naïve mice from challenge with living cancer cells of the same type.1 9 Moreover, PT-112 synergizes with PD-1 or PD-L1 blockade to control mouse tumors developing in immunologically competent hosts.1 9 This work focuses on elucidating the underlying mechanisms of PT-112-induced ICD.

Methods

We harnessed a panel of human and mouse cell lines optionally engineered to lack specific genes involved in mitochondrial apoptosis (namely, Bcl2, Bax and Bak1) coupled with flow cytometry, immunoblotting, RT-PCR, immunofluorescence microscopy and clonogenic assays to determine the impact of reticular and mitochondrial events on the established ability of PT-112 to kill malignant cells in an immunogenic manner.1

Results

In line with previous findings,10–13 PT-112 elicited eukaryotic translation initiation factor 2 subunit alpha (EIF2S1, best known as eIF2α) phosphorylation and mitochondrial dysfunction in malignant cells, a process that was accompanied by the release of interferogenic mitochondrial DNA (mtDNA)14 in the cytoplasm of PT-112 treated cells, and was differentially affected by the deletion of Bcl2, Bax, Bak1, or Bax plus Bak1. Similarly, the lack of Bcl2, Bax, Bak1, or Bax plus Bak1 had a differential impact on the ability of PT-112 to elicit early signs of mitochondrial apoptosis including reactive oxygen species (ROS) generation, mitochondrial transmembrane potential loss and ultimately plasma membrane permeabilization.

Conclusions

ER stress and mitochondrial dysfunction appear to underlie the ability of PT-112 to drive ICD, the integrated stress response, and viral mimicry. This is in line with the well-established connections between ER stress and cytoplasmic nucleic acid sensing, which are pristine mechanisms of antiviral defense in mammalian cells, with the capacity of dying cells to emit immunostimulatory signals.15 Whether PT-112-driven stress also shifts the antigenic properties of cancer cells as a consequence of the accumulation of non-mutational neoantigens16 remains to be determined. Despite these and other open questions, PT-112 stands out as a powerful immunotherapeutic agent with promising clinical activity in patients with a variety of tumors1 under Phase II clinical development for cancer therapy.2–8

Reference

PT-112是一种新的免疫原性小分子,目前正处于癌症治疗的II期临床开发阶段。2-8除了介导许多人类和小鼠癌细胞的细胞抑制和细胞毒性作用外,PT-112还引发与免疫原性细胞死亡(ICD)相关的各种危险信号,如钙网蛋白暴露,以及ATP和HMGB1的分泌。因此,在体外屈服于PT-112的小鼠癌细胞有效地保护免疫能力强的tumor-naïve小鼠免受同类型活癌细胞的攻击。此外,PT-112与PD-1或PD-L1抑制剂协同作用,在免疫能力强的宿主中控制小鼠肿瘤的发展。这项工作的重点是阐明pt -112诱导ICD的潜在机制。方法:我们利用一组人类和小鼠细胞系,选择性地工程化以缺乏与线粒体凋亡相关的特定基因(即Bcl2, Bax和Bak1),结合流式细胞术,免疫印迹,RT-PCR,免疫荧光显微镜和克隆测定来确定网状和线粒体事件对PT-112以免疫原性方式杀死恶性细胞的既定能力的影响结果与先前的研究结果一致,10-13 PT-112在恶性细胞中引发真核翻译起始因子2亚单位α (EIF2S1,即eIF2α)磷酸化和线粒体功能障碍,这一过程伴随着在PT-112处理的细胞细胞质中释放干扰源线粒体DNA (mtDNA)14,并受到Bcl2、Bax、Bak1或Bax + Bak1缺失的不同影响。同样,缺乏Bcl2、Bax、Bak1或Bax + Bak1对PT-112引发线粒体凋亡的早期迹象的能力有不同的影响,包括活性氧(ROS)的产生、线粒体跨膜电位损失和最终的质膜通透性。结论内质网应激和线粒体功能障碍可能是PT-112驱动ICD、综合应激反应和病毒模仿能力的基础。这与内质网应激与细胞质核酸感知之间建立的良好联系是一致的,内质网应激与细胞质核酸感知是哺乳动物细胞中抗病毒防御的原始机制,具有死亡细胞发射免疫刺激信号的能力pt -112驱动的应激是否也会由于非突变新抗原的积累而改变癌细胞的抗原特性仍有待确定。尽管存在这些和其他悬而未决的问题,PT-112作为一种强大的免疫治疗剂脱颖而出,在癌症治疗的II期临床开发中,对各种肿瘤患者具有良好的临床活性。Yamazaki,等。在小鼠肿瘤模型中,PT-112诱导免疫原性细胞死亡并与免疫检查点阻断剂协同作用。张建军,张建军,张建军,等。肿瘤免疫学杂志。2020;9(1):1721810一种新型焦磷酸盐-铂免疫原性细胞死亡诱导剂PT-112在晚期实体肿瘤中的I期研究。EClinicalMedicine。2022; 49:101430。Bryce AH,等。新型免疫原性细胞死亡诱导剂PT-112联合PD-L1抑制剂avelumab治疗转移性去势抵抗性前列腺癌(mCRPC)患者的1b期研究临床肿瘤杂志,2021;39(15 -增刊):e17025-e17025。doi: 10.1200 / JCO.2021.39.15_suppl。[17025]王晓东。PT-112在复发或难治性多发性骨髓瘤患者中的一期剂量递增研究血液。2020;136(增刊1):9-10。Karp DD等。新型免疫原性细胞死亡(ICD)诱导剂PT-112加PD-L1抑制剂avelumab治疗实体肿瘤的Ib期剂量递增研究中华肿瘤学杂志,2020;31:5708。Imbimbo M等。一项针对晚期非小细胞肺癌(NSCLC)患者的新型免疫原性细胞死亡(ICD)诱导剂PT-112加avelumab (PAVE)的IIa期研究。免疫肿瘤学与技术,2022;16:100237。Swift S,等。胸腺上皮肿瘤(TETs)患者的2期概念验证研究中PT-112的初步疗效、安全性和免疫调节作用。临床肿瘤学杂志,2023;41(16 -增刊):e20647-e20647。Bryce AH,等。免疫原性细胞死亡诱导剂PT-112在转移性去势抵抗性前列腺癌患者中的2期研究临床肿瘤杂志,2023;41(6_supl):TPS292-TPS292。杨建军,张建军,张建军,等。免疫原性细胞凋亡的研究进展。癌症免疫学研究,2019;7(2增刊):B199-B199。Soler-Agesta R,等。PT-112诱导人前列腺癌细胞系线粒体应激和免疫原性细胞死亡。中国癌症杂志,2022;32(增刊):1115-1115。Soler-Agesta R,等。靶向线粒体缺陷肿瘤细胞的PT-112诱导线粒体应激和免疫原性细胞死亡癌症(巴塞尔)。2022; 14(16): 3851。Soler-Agesta R,等。
{"title":"1106 Molecular mechanisms of immunogenic cell death driven by PT-112","authors":"Emma Guilbaud, Takahiro Yamazaki, Maria Congenie, Christina Yim, Tyler Ames, Lorenzo Galluzzi","doi":"10.1136/jitc-2023-sitc2023.1106","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1106","url":null,"abstract":"<h3>Background</h3> PT-112 is a novel immunogenic small molecule<sup>1</sup> under Phase II clinical development for cancer therapy.<sup>2–8</sup> Besides mediating cytostatic and cytotoxic effects in numerous human and mouse cancer cells, PT-112 elicits various danger signals that are linked to immunogenic cell death (ICD) such as calreticulin exposure, as well as ATP and HMGB1 secretion.<sup>1 9–11</sup> Accordingly, mouse cancer cells succumbing to PT-112 <i>in vitro</i> efficiently protect immunocompetent, tumor-naïve mice from challenge with living cancer cells of the same type.<sup>1 9</sup> Moreover, PT-112 synergizes with PD-1 or PD-L1 blockade to control mouse tumors developing in immunologically competent hosts.<sup>1 9</sup> This work focuses on elucidating the underlying mechanisms of PT-112-induced ICD. <h3>Methods</h3> We harnessed a panel of human and mouse cell lines optionally engineered to lack specific genes involved in mitochondrial apoptosis (namely, <i>Bcl2, Bax</i> and <i>Bak1</i>) coupled with flow cytometry, immunoblotting, RT-PCR, immunofluorescence microscopy and clonogenic assays to determine the impact of reticular and mitochondrial events on the established ability of PT-112 to kill malignant cells in an immunogenic manner.<sup>1</sup> <h3>Results</h3> In line with previous findings,<sup>10–13</sup> PT-112 elicited eukaryotic translation initiation factor 2 subunit alpha (EIF2S1, best known as eIF2α) phosphorylation and mitochondrial dysfunction in malignant cells, a process that was accompanied by the release of interferogenic mitochondrial DNA (mtDNA)<sup>14</sup> in the cytoplasm of PT-112 treated cells, and was differentially affected by the deletion of <i>Bcl2</i>, <i>Bax</i>, <i>Bak1</i>, or <i>Bax</i> plus <i>Bak1</i>. Similarly, the lack of <i>Bcl2</i>, <i>Bax</i>, <i>Bak1</i>, or <i>Bax</i> plus <i>Bak1</i> had a differential impact on the ability of PT-112 to elicit early signs of mitochondrial apoptosis including reactive oxygen species (ROS) generation, mitochondrial transmembrane potential loss and ultimately plasma membrane permeabilization. <h3>Conclusions</h3> ER stress and mitochondrial dysfunction appear to underlie the ability of PT-112 to drive ICD, the integrated stress response, and viral mimicry. This is in line with the well-established connections between ER stress and cytoplasmic nucleic acid sensing, which are pristine mechanisms of antiviral defense in mammalian cells, with the capacity of dying cells to emit immunostimulatory signals.<sup>15</sup> Whether PT-112-driven stress also shifts the antigenic properties of cancer cells as a consequence of the accumulation of non-mutational neoantigens<sup>16</sup> remains to be determined. Despite these and other open questions, PT-112 stands out as a powerful immunotherapeutic agent with promising clinical activity in patients with a variety of tumors<sup>1</sup> under Phase II clinical development for cancer therapy.<sup>2–8</sup> <h3>Reference","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135162314","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
359 Developing human placental CD34-derived natural killer cells with high affinity and cleavage resistant CD16 and secreted IL-15 (CYNK-201) for cancer immunotherapy 359 .开发具有高亲和力和抗切割CD16和分泌IL-15 (CYNK-201)的人胎盘cd34来源的自然杀伤细胞用于癌症免疫治疗
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0359
Marina Gergues, Yuechao Zhao, Irene Raitman, Shengchen Lin, Valentina Rousseva, Siyara Kilcoyne, Adrian Kilcoyne, Robert Hariri, Shuyang He, Lin Kang

Background

Natural killer (NK) cells can display antibody dependent cellular cytotoxicity (ADCC) activity against tumor cells via the CD16 Fc receptor in combination with tumor specific antibodies. IL-15 is important for NK cell survival, proliferation and function. Celularity is developing human placental CD34+-derived, cryopreserved, off-the-shelf, allogenic NK cells (CYNK-201) with high IgG binding affinity, protease cleavage resistant CD16 and secreted IL-15 for cancer treatment. Here we report the preclinical efficacy results of CYNK-201 against tumor cells.

Methods

CYNK-201 cells were generated by transduction of human placental CD34 cells with lentivirus vector expressing CD16 and IL-15, followed by culture expansion in the presence of cytokines. Transgene expression and phenotype of CYNK-201 cells were characterized by flow cytometry. The concentration of secreted IL-15 by CYNK-201 was measured by a human IL-15 ELISA kit. ADCC activity of CYNK-201 against HER2+ gastric cancer cell line NCI-N87 and CD20+ Burkitts lymphoma cell line Daudi was assessed in combination with trastuzumab and rituximab, respectively. In vivo persistence and efficacy of CYNK-201 were assessed using NSG mice and NCI-N87 xenografted NSG mice, respectively.

Results

CYNK-201 was generated from multiple placental CD34+ donors (n=5) with 91.8±1.1% CD56+CD3- , 78.9± 9.77% CD16 expression, 3274± 1605 fold expansion and the average of 203±1.1 pg/mL secreted IL-15. While 2h PMAi treatment resulted in 61.6±11.3% CD16 cleavage on non-transduced (NT)-CYNK cells, no cleavage was observed from CYNK-201 demonstrating CD16 shedding resistance. CYNK-201 showed increased expression of activation markers including CD11a, NKG2D, CD226 and NKp30 as compared to the NT-CYNK cells. CYNK-201 displayed enhanced in vitro ADCC against NCI-N87 and Daudi as compared to that of NT-CYNK cells. At an E:T ratio of 1:1, CYNK-201 elicited increased ADCC against NCI-N87 with trastuzumab compared to that of NT-CYNK cells, with 65.6±15.1% vs. 52.0±9.1% at 4 hours. At an E:T ratio of 2:1, CYNK-201 showed higher cytotoxicity against Daudi in combination with rituximab compared to that of NT-CYNK with 49.0±18.8% vs. 31.8±9.9%. Post 14 days of infusion, CYNK-201 cells were detectable in NSG mice, with significantly higher abundance than that of NT-CYNK cells plus recombinant IL-15. This enhanced persistence of CYNK-201 led to significant tumor reduction in NCI-N87 tumor model in combination with trastuzumab.

Conclusions

Our results demonstrated enhanced in vitro ADCC, in vivo persistence and anti-tumor activity of CYNK-201. Further development of CYNK-201 is warranted.
自然杀伤细胞(NK)可以通过cd16fc受体与肿瘤特异性抗体结合,对肿瘤细胞显示抗体依赖的细胞毒性(ADCC)活性。IL-15对NK细胞的存活、增殖和功能起重要作用。cellulil正在开发人类胎盘CD34+衍生的、冷冻保存的、现成的、具有高IgG结合亲和力、抗蛋白酶裂解CD16和分泌IL-15的同种异体NK细胞(CYNK-201),用于癌症治疗。在此,我们报告了CYNK-201对肿瘤细胞的临床前疗效结果。方法用表达CD16和IL-15的慢病毒载体将人胎盘CD34细胞转导成CYNK-201细胞,在细胞因子作用下培养扩增。采用流式细胞术对cyck -201细胞的转基因表达和表型进行了表征。采用人IL-15 ELISA试剂盒检测cyck -201分泌IL-15的浓度。分别联合曲妥珠单抗和利妥昔单抗评估CYNK-201对HER2+胃癌细胞系NCI-N87和CD20+伯基茨淋巴瘤细胞系Daudi的ADCC活性。采用NSG小鼠和NCI-N87异种移植NSG小鼠分别评估CYNK-201的体内持久性和有效性。结果CYNK-201来源于5个CD34+胎盘供体,CD56+CD3-表达91.8±1.1%,CD16表达78.9±9.77%,扩增3274±1605倍,IL-15平均分泌203±1.1 pg/mL。PMAi处理2h后,非转导(NT)-CYNK细胞的CD16切割率为61.6±11.3%,而CYNK-201细胞未观察到CD16脱落抗性。与NT-CYNK细胞相比,CYNK-201细胞活化标记CD11a、NKG2D、CD226和NKp30的表达增加。与NT-CYNK细胞相比,CYNK-201细胞对NCI-N87和Daudi的ADCC增强。在E:T比为1:1时,与NT-CYNK细胞相比,曲妥珠单抗作用下CYNK-201细胞对NCI-N87的ADCC增加,4小时时为65.6±15.1%比52.0±9.1%。在E:T比为2:1时,CYNK-201对Daudi联合利妥昔单抗的细胞毒性较NT-CYNK高,分别为49.0±18.8%和31.8±9.9%。注射14 d后,NSG小鼠体内检测到CYNK-201细胞,其丰度显著高于NT-CYNK细胞加重组IL-15。这种增强的CYNK-201持久性导致NCI-N87肿瘤模型与曲妥珠单抗联合显著降低肿瘤。结论CYNK-201体外ADCC、体内持久性和抗肿瘤活性增强。有必要进一步开发CYNK-201。
{"title":"359 Developing human placental CD34-derived natural killer cells with high affinity and cleavage resistant CD16 and secreted IL-15 (CYNK-201) for cancer immunotherapy","authors":"Marina Gergues, Yuechao Zhao, Irene Raitman, Shengchen Lin, Valentina Rousseva, Siyara Kilcoyne, Adrian Kilcoyne, Robert Hariri, Shuyang He, Lin Kang","doi":"10.1136/jitc-2023-sitc2023.0359","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0359","url":null,"abstract":"<h3>Background</h3> Natural killer (NK) cells can display antibody dependent cellular cytotoxicity (ADCC) activity against tumor cells via the CD16 Fc receptor in combination with tumor specific antibodies. IL-15 is important for NK cell survival, proliferation and function. Celularity is developing human placental CD34<sup>+</sup>-derived, cryopreserved, off-the-shelf, allogenic NK cells (CYNK-201) with high IgG binding affinity, protease cleavage resistant CD16 and secreted IL-15 for cancer treatment. Here we report the preclinical efficacy results of CYNK-201 against tumor cells. <h3>Methods</h3> CYNK-201 cells were generated by transduction of human placental CD34 cells with lentivirus vector expressing CD16 and IL-15, followed by culture expansion in the presence of cytokines. Transgene expression and phenotype of CYNK-201 cells were characterized by flow cytometry. The concentration of secreted IL-15 by CYNK-201 was measured by a human IL-15 ELISA kit. ADCC activity of CYNK-201 against HER2<sup>+</sup> gastric cancer cell line NCI-N87 and CD20<sup>+</sup> Burkitts lymphoma cell line Daudi was assessed in combination with trastuzumab and rituximab, respectively. <i>In vivo</i> persistence and efficacy of CYNK-201 were assessed using NSG mice and NCI-N87 xenografted NSG mice, respectively. <h3>Results</h3> CYNK-201 was generated from multiple placental CD34<sup>+</sup> donors (n=5) with 91.8±1.1% CD56<sup>+</sup>CD3<sup>-</sup> , 78.9± 9.77% CD16 expression, 3274± 1605 fold expansion and the average of 203±1.1 pg/mL secreted IL-15. While 2h PMAi treatment resulted in 61.6±11.3% CD16 cleavage on non-transduced (NT)-CYNK cells, no cleavage was observed from CYNK-201 demonstrating CD16 shedding resistance. CYNK-201 showed increased expression of activation markers including CD11a, NKG2D, CD226 and NKp30 as compared to the NT-CYNK cells. CYNK-201 displayed enhanced <i>in vitro</i> ADCC against NCI-N87 and Daudi as compared to that of NT-CYNK cells. At an E:T ratio of 1:1, CYNK-201 elicited increased ADCC against NCI-N87 with trastuzumab compared to that of NT-CYNK cells, with 65.6±15.1% vs. 52.0±9.1% at 4 hours. At an E:T ratio of 2:1, CYNK-201 showed higher cytotoxicity against Daudi in combination with rituximab compared to that of NT-CYNK with 49.0±18.8% vs. 31.8±9.9%. Post 14 days of infusion, CYNK-201 cells were detectable in NSG mice, with significantly higher abundance than that of NT-CYNK cells plus recombinant IL-15. This enhanced persistence of CYNK-201 led to significant tumor reduction in NCI-N87 tumor model in combination with trastuzumab. <h3>Conclusions</h3> Our results demonstrated enhanced <i>in vitro</i> ADCC, <i>in vivo</i> persistence and anti-tumor activity of CYNK-201. Further development of CYNK-201 is warranted.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135162326","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
501 AZD2796, a myeloid checkpoint inhibitor targeting LILRB2 that promotes pro-inflammatory responses by macrophages and enhances T cell anti-tumor activity 501 AZD2796,一种靶向LILRB2的髓细胞检查点抑制剂,可促进巨噬细胞的促炎反应,增强T细胞抗肿瘤活性
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0501
Des C Jones, Lorraine Irving, Becki Dudley, Marcin Wolny, Seraina Blümli, Ellie Chatzopoulou, Alan Sandercock, Georgina Bowyer, Stacy Pryts, Kathy Mulgrew, Simon Dovedi, Fernanda Arnaldez, Mark Cobbold

Background

Inhibition of T cell immune checkpoints has revolutionized cancer therapy. However, responses are not uniformly positive, and many patients develop resistance. Myeloid cells are an abundant component of the tumor microenvironment able to promote cancer progression and suppress immune responses. Consequently, targeting these cells may be an avenue to optimize immune-based cancer therapies. LILRB2 is an inhibitory member of the leukocyte immunoglobulin-like receptor (LILR) family that is expressed on the surface of myeloid cells, including key suppressive myeloid subsets such as macrophages and myeloid-derived suppressor cells (MDSCs) found in tumors. LILRB2 signalling contributes to the suppressive phenotype of myeloid cells by inhibiting the activity of pro-inflammatory signalling pathways.

Methods

AZD2796 is a fully humanised monoclonal antibody that binds LILRB2. AZD2796 was assessed for target specificity, binding affinity and antagonism of ligand binding. The ability of AZD2796 to enhance proinflammatory responses of macrophages was determined by the release of pro inflammatory cytokines from monocyte derived macrophages in vitro, and enhancement of tumor cell lysis by T cells when co-cultured in the presence of macrophages. The anti-tumor effect of AZD2796 was explored in vivo using two xenograft models of human cancer in humanised mice.

Results

AZD2796 is highly specific to LILRB2 and does not bind other members of the LILR family. It binds LILRB2 with high affinity and blocks binding of LILRB2 to its major histocompatibility complex (MHC) class I ligands. In functional assays, AZD2796 enhanced the production of the proinflammatory cytokines TNF-α and GM-CSF from human macrophages stimulated with CD40L, while reducing the production of VEGF, an important driver of angiogenesis. In addition, AZD2796 increased tumor cell killing by T cells when co-cultured with macrophages. In vivo, AZD2796 significantly reduced tumor growth rate of NCI-H358 lung cancer cells and SK-MEL-5 melanoma cancer cells in humanised mice.

Conclusions

AZD2796 is a high affinity anti-LILRB2 monoclonal antibody that promotes pro-inflammatory responses by macrophages and enhances anti-tumor activity of T cells. Our pre-clinical data support the potential of AZD2796 as an anti-cancer therapy with opportunities to combine with T-cell-based therapeutics.

Ethics Approval

All animal studies are run under the Institutional Animal Care and Use Committee (IACUC) approved in vivo protocol number AUP-22–17. AstraZeneca’s US site IACUC committee oversees the specific use of animals by conducting a formal review of the animal use, ethics and protocols and grants approval prior to the work commencing.
抑制T细胞免疫检查点已经彻底改变了癌症治疗。然而,反应并不都是积极的,许多患者会产生耐药性。骨髓细胞是肿瘤微环境的丰富组成部分,能够促进癌症进展并抑制免疫反应。因此,靶向这些细胞可能是优化基于免疫的癌症治疗的途径。LILRB2是白细胞免疫球蛋白样受体(LILR)家族的抑制性成员,在髓细胞表面表达,包括肿瘤中发现的巨噬细胞和髓源性抑制细胞(MDSCs)等关键抑制性髓细胞亚群。LILRB2信号通过抑制促炎信号通路的活性,有助于髓细胞的抑制性表型。方法AZD2796是一种结合LILRB2的全人源化单克隆抗体。评估AZD2796的靶特异性、结合亲和力和配体结合的拮抗性。AZD2796增强巨噬细胞促炎反应的能力是通过体外单核细胞源性巨噬细胞释放促炎细胞因子,以及在巨噬细胞存在下共培养时T细胞对肿瘤细胞溶解的增强来确定的。采用两种人源化小鼠肿瘤异种移植模型,探讨AZD2796的体内抗肿瘤作用。结果AZD2796对LILRB2具有高度特异性,不结合LILR家族的其他成员。它以高亲和力结合LILRB2,并阻断LILRB2与其主要组织相容性复合体(MHC) I类配体的结合。在功能分析中,AZD2796增强了CD40L刺激的人巨噬细胞中促炎细胞因子TNF-α和GM-CSF的产生,同时降低了血管生成的重要驱动因子VEGF的产生。此外,AZD2796与巨噬细胞共培养可提高T细胞对肿瘤细胞的杀伤作用。在体内,AZD2796显著降低人源化小鼠NCI-H358肺癌细胞和SK-MEL-5黑色素瘤癌细胞的肿瘤生长速度。结论AZD2796是一种高亲和力的抗lilrb2单克隆抗体,可促进巨噬细胞的促炎反应,增强T细胞的抗肿瘤活性。我们的临床前数据支持AZD2796作为抗癌疗法的潜力,并有机会与基于t细胞的疗法结合。所有动物研究均在机构动物护理和使用委员会(IACUC)批准的体内试验方案编号为up -22 - 17下进行。阿斯利康在美国的IACUC委员会通过对动物使用、伦理和协议进行正式审查,并在工作开始前批准批准,监督动物的具体使用。
{"title":"501 AZD2796, a myeloid checkpoint inhibitor targeting LILRB2 that promotes pro-inflammatory responses by macrophages and enhances T cell anti-tumor activity","authors":"Des C Jones, Lorraine Irving, Becki Dudley, Marcin Wolny, Seraina Blümli, Ellie Chatzopoulou, Alan Sandercock, Georgina Bowyer, Stacy Pryts, Kathy Mulgrew, Simon Dovedi, Fernanda Arnaldez, Mark Cobbold","doi":"10.1136/jitc-2023-sitc2023.0501","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0501","url":null,"abstract":"<h3>Background</h3> Inhibition of T cell immune checkpoints has revolutionized cancer therapy. However, responses are not uniformly positive, and many patients develop resistance. Myeloid cells are an abundant component of the tumor microenvironment able to promote cancer progression and suppress immune responses. Consequently, targeting these cells may be an avenue to optimize immune-based cancer therapies. LILRB2 is an inhibitory member of the leukocyte immunoglobulin-like receptor (LILR) family that is expressed on the surface of myeloid cells, including key suppressive myeloid subsets such as macrophages and myeloid-derived suppressor cells (MDSCs) found in tumors. LILRB2 signalling contributes to the suppressive phenotype of myeloid cells by inhibiting the activity of pro-inflammatory signalling pathways. <h3>Methods</h3> AZD2796 is a fully humanised monoclonal antibody that binds LILRB2. AZD2796 was assessed for target specificity, binding affinity and antagonism of ligand binding. The ability of AZD2796 to enhance proinflammatory responses of macrophages was determined by the release of pro inflammatory cytokines from monocyte derived macrophages in vitro, and enhancement of tumor cell lysis by T cells when co-cultured in the presence of macrophages. The anti-tumor effect of AZD2796 was explored <i>in vivo</i> using two xenograft models of human cancer in humanised mice. <h3>Results</h3> AZD2796 is highly specific to LILRB2 and does not bind other members of the LILR family. It binds LILRB2 with high affinity and blocks binding of LILRB2 to its major histocompatibility complex (MHC) class I ligands. In functional assays, AZD2796 enhanced the production of the proinflammatory cytokines TNF-α and GM-CSF from human macrophages stimulated with CD40L, while reducing the production of VEGF, an important driver of angiogenesis. In addition, AZD2796 increased tumor cell killing by T cells when co-cultured with macrophages. <i>In vivo</i>, AZD2796 significantly reduced tumor growth rate of NCI-H358 lung cancer cells and SK-MEL-5 melanoma cancer cells in humanised mice. <h3>Conclusions</h3> AZD2796 is a high affinity anti-LILRB2 monoclonal antibody that promotes pro-inflammatory responses by macrophages and enhances anti-tumor activity of T cells. Our pre-clinical data support the potential of AZD2796 as an anti-cancer therapy with opportunities to combine with T-cell-based therapeutics. <h3>Ethics Approval</h3> All animal studies are run under the Institutional Animal Care and Use Committee (IACUC) approved in vivo protocol number AUP-22–17. AstraZeneca’s US site IACUC committee oversees the specific use of animals by conducting a formal review of the animal use, ethics and protocols and grants approval prior to the work commencing.","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135162545","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
Regular and Young Investigator Award Abstracts
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