首页 > 最新文献

Regular and Young Investigator Award Abstracts最新文献

英文 中文
1304 Robust machine learning (ML) approach for screening microbiome ecosystem therapies (MET) drug candidates in combination with immune checkpoint inhibitors 1304:结合免疫检查点抑制剂筛选微生物组生态系统疗法(MET)候选药物的鲁棒机器学习(ML)方法
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1304
Emmanuel Prestat, Elsa Schalck, Antoine Bonnefoy, Antoine Sabourin, Cyrielle Gasc, Carole Schwintner, Nathalie Corvaia

Background

Studies for the last 8 years have connected gut microbiome composition to ICI efficacy in cancer therapy, including pilot studies1 2 demonstrating that FMT from responders to non-responders can improve response rates. However, interstudy inconsistencies were observed in microbiome signature findings3 also confirmed when reprocessing internally raw data from multiple studies. It remains critical to tackle this heterogeneity to learn from stable patterns and develop a robust and reliable drug candidate screening algorithm. Hence, MaaT Pharma developed an AI framework to train models from microbiota Whole Metagenome Sequencing (WMS) datasets that predict the responder status to ICIs. We focused on performance and robustness, which was achieved by monitoring the AUC as a standard approach, and precision to control for false positive rate and to emphasize the positive classification criticality in a drug candidate selection approach.

Methods

We collected baseline WMS datasets from 10 cohorts in 3 ICI indications: melanoma, non-small cell lung cancer and renal cell carcinoma, along with clinical evaluation of ICI treatment. Those datasets were processed by gutPrint® MgRunner software, before being included in the AI framework. About 70 experiments were conducted within a Leave-One-Dataset-Out cross-validation scheme. Various factors such as taxonomic or functional inputs, dataset bias correction, data augmentation approaches, ML algorithms and data representation methods, were tested to select the top ones. Finally, a model was refit with the best performing parameters on the entire dataset, and applied to score MaaT Pharma mono-donor and healthy-pooled-donors-derived drug substances (DS).

Results

The best performing experiment provided models based on the XGBoost algorithm with AUCs ranging from 0.52 to 0.73 depending on the left-out cohort (average AUC = 0.65), and a precision that ranges between 0.55 and 0.81 (average precision = 0.65). Those results outperform melanoma-centered study with a comparable method.4 Despite the diverse data sources and indications, the multi-indication approach surpassed the mono-indication (melanoma) training approach for predictions related to melanoma patients. Considering the scoring of DS derived from healthy donors, 73% of mono-donors and 91% of healthy-pooled-donors-derived DS were classified as ‘Responder-like’.

Conclusions

Present study highlights the significance of dataset size in ICI microbiota models and presents a methodology to enhance the performances of a multi-cohort-based ML approach. Conditioned to the performances we obtained, the healthy-pooled-donors-derived DS harbor a considerable ratio (91%) of ‘ICI Responder-like’, significantly higher than the mono-donor stools (73%) suggesting that pooled ecosystems from healthy donors could better convert ICI-non responders into responders.

References

过去8年的研究已经将肠道微生物组组成与癌症治疗中的ICI疗效联系起来,包括初步研究表明,从应答者到无应答者的FMT可以提高应答率。然而,当重新处理来自多个研究的内部原始数据时,在微生物组特征发现中观察到的研究间不一致也得到了证实。解决这种异质性,从稳定的模式中学习,并开发一种强大而可靠的候选药物筛选算法仍然是至关重要的。因此,MaaT Pharma开发了一个人工智能框架来训练来自微生物群全宏基因组测序(WMS)数据集的模型,以预测对ICIs的反应状态。我们关注的是性能和鲁棒性,这是通过监测AUC作为标准方法来实现的,以及精确控制假阳性率和强调候选药物选择方法中的阳性分类临界性。方法:我们收集了来自10个队列的基线WMS数据集,包括3种ICI适应症:黑色素瘤、非小细胞肺癌和肾细胞癌,以及ICI治疗的临床评估。这些数据集在被纳入人工智能框架之前,由gutPrint®MgRunner软件处理。在Leave-One-Dataset-Out交叉验证方案下进行了大约70个实验。我们测试了各种因素,如分类或功能输入、数据集偏差校正、数据增强方法、ML算法和数据表示方法,以选择最重要的因素。最后,利用整个数据集上表现最好的参数重构模型,并应用于MaaT Pharma单供体和健康池供体衍生原料药(DS)的评分。结果基于XGBoost算法的模型,根据遗漏队列的不同,AUC范围为0.52 ~ 0.73(平均AUC = 0.65),精度范围为0.55 ~ 0.81(平均精度= 0.65)。这些结果优于用可比方法进行的以黑色素瘤为中心的研究尽管有不同的数据来源和适应症,但在黑色素瘤患者相关预测方面,多适应症方法优于单适应症(黑色素瘤)训练方法。考虑到来自健康供者的DS评分,73%的单一供者和91%的健康汇集供者衍生的DS被归类为“类似响应者”。本研究强调了ICI微生物群模型中数据集大小的重要性,并提出了一种方法来提高基于多队列的机器学习方法的性能。根据我们获得的表现,健康供者衍生的DS具有相当大的“ICI响应样”比例(91%),显著高于单一供者粪便(73%),这表明来自健康供者的汇集生态系统可以更好地将ICI无应答者转化为应答者。D Davar, et .“粪便微生物群移植克服了黑色素瘤患者对抗pd -1治疗的耐药性,”Science, 2021年2月;371(6529):595-602,doi: 10.1126/ Science .abf3363。EN Baruch等。“粪便微生物群移植促进免疫治疗难治性黑色素瘤患者的反应,”科学,2020年12月;371(6529):602-609,doi: 10.1126/ Science .abb5920。S Wojciechowski, et .“机器学习在解锁微生物群潜力的道路上,促进免疫检查点治疗,”国际医学微生物学杂志,2022年10月;312(7):151560,2022年10月,doi: 10.1016/ j.j jmm.2022.151560。KA Lee,等。“跨队列肠道微生物组与晚期黑色素瘤免疫检查点抑制剂反应的关联”,《中华医学杂志》,2022;28(3):535-544,doi: 10.1038/ s41591-022-01695-5。
{"title":"1304 Robust machine learning (ML) approach for screening microbiome ecosystem therapies (MET) drug candidates in combination with immune checkpoint inhibitors","authors":"Emmanuel Prestat, Elsa Schalck, Antoine Bonnefoy, Antoine Sabourin, Cyrielle Gasc, Carole Schwintner, Nathalie Corvaia","doi":"10.1136/jitc-2023-sitc2023.1304","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1304","url":null,"abstract":"<h3>Background</h3> Studies for the last 8 years have connected gut microbiome composition to ICI efficacy in cancer therapy, including pilot studies<sup>1 2</sup> demonstrating that FMT from responders to non-responders can improve response rates. However, interstudy inconsistencies were observed in microbiome signature findings<sup>3</sup> also confirmed when reprocessing internally raw data from multiple studies. It remains critical to tackle this heterogeneity to learn from stable patterns and develop a robust and reliable drug candidate screening algorithm. Hence, MaaT Pharma developed an AI framework to train models from microbiota Whole Metagenome Sequencing (WMS) datasets that predict the responder status to ICIs. We focused on performance and robustness, which was achieved by monitoring the <i>AUC</i> as a standard approach, and <i>precision</i> to control for false positive rate and to emphasize the positive classification criticality in a drug candidate selection approach. <h3>Methods</h3> We collected baseline WMS datasets from 10 cohorts in 3 ICI indications: melanoma, non-small cell lung cancer and renal cell carcinoma, along with clinical evaluation of ICI treatment. Those datasets were processed by gutPrint® <i>MgRunner</i> software, before being included in the AI framework. About 70 experiments were conducted within a Leave-One-Dataset-Out cross-validation scheme. Various factors such as taxonomic or functional inputs, dataset bias correction, data augmentation approaches, ML algorithms and data representation methods, were tested to select the top ones. Finally, a model was refit with the best performing parameters on the entire dataset, and applied to score MaaT Pharma mono-donor and healthy-pooled-donors-derived drug substances (DS). <h3>Results</h3> The best performing experiment provided models based on the XGBoost algorithm with AUCs ranging from 0.52 to 0.73 depending on the left-out cohort (average AUC = 0.65), and a precision that ranges between 0.55 and 0.81 (average precision = 0.65). Those results outperform melanoma-centered study with a comparable method.<sup>4</sup> Despite the diverse data sources and indications, the multi-indication approach surpassed the mono-indication (melanoma) training approach for predictions related to melanoma patients. Considering the scoring of DS derived from healthy donors, 73% of mono-donors and 91% of healthy-pooled-donors-derived DS were classified as ‘Responder-like’. <h3>Conclusions</h3> Present study highlights the significance of dataset size in ICI microbiota models and presents a methodology to enhance the performances of a multi-cohort-based ML approach. Conditioned to the performances we obtained, the healthy-pooled-donors-derived DS harbor a considerable ratio (91%) of ‘ICI Responder-like’, significantly higher than the mono-donor stools (73%) suggesting that pooled ecosystems from healthy donors could better convert ICI-non responders into responders. <h3>References</h3","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":"135161127","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
644 A phase 1 dose escalation study of GCC19CART – A novel CoupledCAR® therapy for subjects with metastatic colorectal cancer GCC19CART -一种用于转移性结直肠癌患者的新型CoupledCAR®疗法的1期剂量递增研究
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0644
Naifei Chen, Chengfei Pu, Lingling Zhao, Ning Li, Chang Wang, Yusheng Huang, Suxia Luo, Xun Li, Zhenzhou Yang, Jun Bie, Ruihong Zhu, Xi Huang, Haiyang Tang, Tingting Liang, Yizhuo Wang, Beibei Jia, Dongqi Chen, Victor Lu, Zhao Wu, Yongping Song, Lei Xiao, Jiuwei Cui

Background

GCC19CART, the first clinical candidate from the CoupledCAR® solid tumor platform, targets guanylate cyclase-C (GCC) which is expressed in colorectal cancers. CoupledCAR utilizes multiple vectors to make both solid tumor targeting CAR-T and CD19 CAR-T in a single manufacturing step. An investigator-initiated dose escalation trial in China for patients with relapsed or refractory metastatic colorectal cancer (R/R mCRC) is reported here.

Methods

Subjects are screened for GCC expression by immunohistochemistry. Eligible subjects undergo leukapheresis, a single dose of lymphodepleting chemotherapy (fludarabine 30mg/m2 and cyclophosphamide 300mg/m2) 3 days prior to infusion, and then administration of a single infusion of GCC19CART at one of two preassigned doses: 1x106 or 2x106 CAR T-cells/kg. Endpoints are safety and preliminary evidence of efficacy as determined by CT or PET/CT per RECIST 1.1 or PERCIST 1.0. All responses were confirmed by an independent third-party imaging contract research organization (CRO).

Results

13 subjects have been enrolled to dose level 1 (1x106 cells/kg) and 8 subjects have been enrolled to dose level 2 (2x106 cells/kg). The most common adverse events were cytokine release syndrome (CRS) in 21/21 subjects (Grade 1 19/21 (90.48%) or Grade 2 2/21 (9.52%)) and diarrhea in 21/21 subjects (Grade 1 6/21 (28.57%) Grade 2 5/21 (23.81%) Grade 3 9/21 (42.86%) or Grade 4 1/21 (4.76%)). All patients with grade 3 and higher side effects were well managed. Immune effector cell-associated neurotoxicity syndrome(ICANS) was observed in 2/21 (9.52%) subjects at Grade 3 or 4 and resolved with corticosteroids. The combined overall response rate (ORR) for both dose levels was 28.6% (6/21). For dose level 1, the overall response rate (ORR) per RECIST 1.1 was 15.4% (2/13). Two subjects demonstrated a partial response (PR) while 3 additional subjects had partial metabolic response (PMR) on PET/CT with stable disease (SD) or progressive disease (PD) per RECIST 1.1. For dose level 2, The ORR per RECIST 1.1 was 50% (4/8). 4 subjects demonstrated a PR (3 at month 1, 1 at month 3 after being SD at month 1) and 2 additional subjects had PMR on PET/CT with SD per RECIST 1.1.

Conclusions

Preliminary results demonstrate that GCC19CART has meaningful dose-dependent clinical activity and an acceptable safety profile in relapsed or refractory metastatic colorectal cancer. This trial is ongoing and updated data will be presented. A Phase 1 trial of GCC19CART in the US has opened for accrual and is expected to enroll patients in mid-2022.
GCC19CART是CoupledCAR®实体肿瘤平台的首个临床候选药物,靶向结直肠癌中表达的鸟苷酸环化酶c (guanyate cyclase-C, GCC)。CoupledCAR利用多个载体在一个制造步骤中制造出靶向CAR-T和CD19 CAR-T的实体肿瘤。在中国,研究者发起了一项针对复发或难治性转移性结直肠癌(R/R mCRC)患者的剂量递增试验。方法采用免疫组化方法筛选受试者GCC表达情况。符合条件的受试者在输注前3天进行白细胞清除,单剂量淋巴细胞消耗化疗(氟达拉平30mg/m2和环磷酰胺300mg/m2),然后以两种预先指定剂量之一(1x106或2x106 CAR - t细胞/kg)单次输注GCC19CART。终点是由CT或PET/CT根据RECIST 1.1或PERCIST 1.0确定的安全性和有效性的初步证据。所有回复均由独立的第三方成像合同研究组织(CRO)确认。结果入组剂量1 (1x106细胞/kg) 13例,入组剂量2 (2x106细胞/kg) 8例。21/21患者中最常见的不良事件为细胞因子释放综合征(CRS)(1级19/21(90.48%)或2级2/21(9.52%))和腹泻(1级6/21(28.57%)、2级5/21(23.81%)、3级9/21(42.86%)或4级1/21(4.76%))。所有3级及以上副作用的患者都得到了良好的管理。免疫效应细胞相关神经毒性综合征(ICANS)出现在2/21(9.52%)的3级或4级患者中,并通过皮质类固醇治疗。两种剂量水平的综合总有效率(ORR)为28.6%(6/21)。对于剂量水平1,根据RECIST 1.1的总缓解率(ORR)为15.4%(2/13)。根据RECIST 1.1, 2名患者在PET/CT上表现出部分缓解(PR),另外3名患者在疾病稳定(SD)或进展(PD)时表现出部分代谢缓解(PMR)。对于剂量水平2,根据RECIST 1.1的ORR为50%(4/8)。4名受试者表现出PR(3名在第1个月,1名在第1个月被SD后的第3个月),另外2名受试者在PET/CT上显示PMR, SD符合RECIST 1.1。初步结果表明,GCC19CART在复发或难治性转移性结直肠癌中具有显著的剂量依赖性临床活性和可接受的安全性。该试验正在进行中,将提供最新数据。GCC19CART的1期临床试验已在美国开放,预计将于2022年中期入组患者。
{"title":"644 A phase 1 dose escalation study of GCC19CART – A novel CoupledCAR® therapy for subjects with metastatic colorectal cancer","authors":"Naifei Chen, Chengfei Pu, Lingling Zhao, Ning Li, Chang Wang, Yusheng Huang, Suxia Luo, Xun Li, Zhenzhou Yang, Jun Bie, Ruihong Zhu, Xi Huang, Haiyang Tang, Tingting Liang, Yizhuo Wang, Beibei Jia, Dongqi Chen, Victor Lu, Zhao Wu, Yongping Song, Lei Xiao, Jiuwei Cui","doi":"10.1136/jitc-2023-sitc2023.0644","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0644","url":null,"abstract":"<h3>Background</h3> GCC19CART, the first clinical candidate from the CoupledCAR® solid tumor platform, targets guanylate cyclase-C (GCC) which is expressed in colorectal cancers. CoupledCAR utilizes multiple vectors to make both solid tumor targeting CAR-T and CD19 CAR-T in a single manufacturing step. An investigator-initiated dose escalation trial in China for patients with relapsed or refractory metastatic colorectal cancer (R/R mCRC) is reported here. <h3>Methods</h3> Subjects are screened for GCC expression by immunohistochemistry. Eligible subjects undergo leukapheresis, a single dose of lymphodepleting chemotherapy (fludarabine 30mg/m<sup>2</sup> and cyclophosphamide 300mg/m<sup>2</sup>) 3 days prior to infusion, and then administration of a single infusion of GCC19CART at one of two preassigned doses: 1x10<sup>6</sup> or 2x10<sup>6</sup> CAR T-cells/kg. Endpoints are safety and preliminary evidence of efficacy as determined by CT or PET/CT per RECIST 1.1 or PERCIST 1.0. All responses were confirmed by an independent third-party imaging contract research organization (CRO). <h3>Results</h3> 13 subjects have been enrolled to dose level 1 (1x10<sup>6</sup> cells/kg) and 8 subjects have been enrolled to dose level 2 (2x10<sup>6</sup> cells/kg). The most common adverse events were cytokine release syndrome (CRS) in 21/21 subjects (Grade 1 19/21 (90.48%) or Grade 2 2/21 (9.52%)) and diarrhea in 21/21 subjects (Grade 1 6/21 (28.57%) Grade 2 5/21 (23.81%) Grade 3 9/21 (42.86%) or Grade 4 1/21 (4.76%)). All patients with grade 3 and higher side effects were well managed. Immune effector cell-associated neurotoxicity syndrome(ICANS) was observed in 2/21 (9.52%) subjects at Grade 3 or 4 and resolved with corticosteroids. The combined overall response rate (ORR) for both dose levels was 28.6% (6/21). For dose level 1, the overall response rate (ORR) per RECIST 1.1 was 15.4% (2/13). Two subjects demonstrated a partial response (PR) while 3 additional subjects had partial metabolic response (PMR) on PET/CT with stable disease (SD) or progressive disease (PD) per RECIST 1.1. For dose level 2, The ORR per RECIST 1.1 was 50% (4/8). 4 subjects demonstrated a PR (3 at month 1, 1 at month 3 after being SD at month 1) and 2 additional subjects had PMR on PET/CT with SD per RECIST 1.1. <h3>Conclusions</h3> Preliminary results demonstrate that GCC19CART has meaningful dose-dependent clinical activity and an acceptable safety profile in relapsed or refractory metastatic colorectal cancer. This trial is ongoing and updated data will be presented. A Phase 1 trial of GCC19CART in the US has opened for accrual and is expected to enroll patients in mid-2022.","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":"135161136","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
74 a-TIGIT mAb belrestotug in combination with anti-PD1 induces an immunocompetent tumor microenvironment (TME) a-TIGIT mAb与抗pd1联合诱导免疫活性肿瘤微环境(TME)
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0074
Julia Cuende, Stephanie Ma, Iain Welsby, Nicolas Rosewick, Noemie Wald, Anais Vezzu, Marjorie Mercier, Paola Tieppo, Bart Claes, Yvonne McGrath, Olivier De Henau, Gregory Driessens, Maurizio Ceppi, Marion Libouban

Background

T cell immunoglobulin and ITIM domain (TIGIT) is an immune checkpoint inhibitor expressed mainly on NK and T cell populations. Antagonist a-TIGIT mAbs in combination with a-PD(L)-1 demonstrated clinical proof of concept in 1L NSCLC.1 2 Belrestotug (previously known as EOS-448 or GSK4428859A) is an antagonistic anti-TIGIT human immunoglobulin G1 (hIgG1) antibody, designed to prevent ligand binding and to engage Fc gamma receptors (FcγR), resulting in a multifaceted mode of action: (i) activate effector T cells and NK cells (ii) modulate antigen-presenting cells, and (iii) deplete suppressive regulatory T cells (Tregs) as well as terminally exhausted CD8 T cells that express the highest levels of TIGIT.3–5 In a first-in-human trial, belrestotug demonstrated a good tolerability profile with early signs of efficacy6 and we reported for the first time intratumoral TIGIT target engagement in the patient tumors.5 Belrestotug is currently being tested in combination with anti-PD1 in solid tumors.

Results

Pharmacodynamic assessment made by flow cytometry in the blood of patients treated with belrestotug in monotherapy and in combination with anti-PD1 (pembrolizumab or dostarlimab) showed (i) increased proportion of proliferating memory CD8+ T and NK cells during the first treatment cycle, (ii) sustained depletion of immunosuppressive Tregs, and (iii) decreased proportion of TIGIT high CD8+ T cells. We demonstrated the terminally exhausted phenotype of the TIGIT high CD8+ T cells targeted by belrestotug by isolating peripheral blood mononuclear cells (PBMCs) from treatment naïve cancer patients. Tregs in the tumor microenvironment (TME) hinder effective tumor immune response and are mainly localized in the stromal area. Stromal Tregs co-expressing several immune checkpoint inhibitors, including TIGIT, have a high immunosuppressive profile and its density is associated with poor clinical outcome.7 We investigated the functional effect of belrestotug combined with anti-PD1 on the TME using a multiplex immunofluorescence (mIF) panel. We observed a decrease of immunosuppressive TIGIT+ Tregs and PD1+ Tregs in the stroma, as well as a spatial reorganization.

Conclusions

Overall, our data suggests a mechanism whereby belrestotug in combination with anti-PD1 induces a more immunocompetent TME driven by immunosuppressive Treg depletion. These data support the clinical development of the doublet therapy to enhance an anti-tumor immune response.

References

Cho BC, et al. Abstract #LBA2 ESMO-IO; 2021 Jonhson ARC-7 ASCO Dec 2022 Yu X, et al, Nature, 2008 Preillon J, et al. Mol Cancer Ther, 2021. Cuende, et al. AACR 2022 Van den Mooter TF, et al. Abstract #CT118 AACR; 2021 Devi-Marulkar, et al, comms Bio, 2022
T细胞免疫球蛋白和ITIM结构域(TIGIT)是一种主要表达于NK细胞和T细胞群的免疫检查点抑制剂。Belrestotug(以前称为os -448或GSK4428859A)是一种抗tigit的人类免疫球蛋白G1 (hIgG1)抗体,旨在阻止配体结合并参与Fcγ受体(Fcγ r),产生多方面的作用模式:(1)激活效应T细胞和NK细胞;(2)调节抗原呈递细胞;(3)消耗表达最高水平TIGIT的抑制性调节性T细胞(treg)以及最终耗尽的CD8 T细胞。3 - 5在首次人体试验中,belrestotug显示出良好的耐受性,具有早期疗效迹象6,我们首次报道了肿瘤患者的肿瘤内TIGIT靶点参与Belrestotug目前正在与抗pd1联合治疗实体肿瘤。结果通过流式细胞术对贝莱妥托单药和联合抗pd1(派姆单抗或多司单抗)治疗患者血液中的药效学评估显示:(1)在第一个治疗周期内增殖记忆性CD8+ T细胞和NK细胞比例增加,(2)免疫抑制Tregs持续耗竭,(3)TIGIT高CD8+ T细胞比例下降。我们通过分离治疗naïve癌症患者的外周血单个核细胞(PBMCs),证明了belrestotug靶向的TIGIT高CD8+ T细胞的终耗竭表型。肿瘤微环境(tumor microenvironment, TME)中的Tregs主要局限于间质区,阻碍肿瘤有效的免疫应答。基质Tregs共表达几种免疫检查点抑制剂,包括TIGIT,具有高免疫抑制谱,其密度与不良临床结果相关我们采用多重免疫荧光(mIF)技术研究了belrestotug联合抗pd1对TME的功能影响。我们观察到基质中免疫抑制的TIGIT+ Tregs和PD1+ Tregs减少,以及空间重组。总的来说,我们的数据表明了一种机制,即belrestotug联合抗pd1诱导由免疫抑制性Treg消耗驱动的更具免疫活性的TME。这些数据支持双重疗法的临床发展,以增强抗肿瘤免疫反应。参考文献Cho BC等。#LBA2 ESMO-IO;2021 johnson ARC-7 ASCO Dec 2022 Yu X等,Nature, 2008 Preillon J等。Mol巨蟹座,2021年。Cuende等人。[2]范登·缪特等。摘要#CT118 AACR;2021 Devi-Marulkar等,通讯生物,2022
{"title":"74 a-TIGIT mAb belrestotug in combination with anti-PD1 induces an immunocompetent tumor microenvironment (TME)","authors":"Julia Cuende, Stephanie Ma, Iain Welsby, Nicolas Rosewick, Noemie Wald, Anais Vezzu, Marjorie Mercier, Paola Tieppo, Bart Claes, Yvonne McGrath, Olivier De Henau, Gregory Driessens, Maurizio Ceppi, Marion Libouban","doi":"10.1136/jitc-2023-sitc2023.0074","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0074","url":null,"abstract":"<h3>Background</h3> T cell immunoglobulin and ITIM domain (TIGIT) is an immune checkpoint inhibitor expressed mainly on NK and T cell populations. Antagonist a-TIGIT mAbs in combination with a-PD(L)-1 demonstrated clinical proof of concept in 1L NSCLC.<sup>1 2</sup> Belrestotug (previously known as EOS-448 or GSK4428859A) is an antagonistic anti-TIGIT human immunoglobulin G1 (hIgG1) antibody, designed to prevent ligand binding and to engage Fc gamma receptors (FcγR), resulting in a multifaceted mode of action: (i) activate effector T cells and NK cells (ii) modulate antigen-presenting cells, and (iii) deplete suppressive regulatory T cells (Tregs) as well as terminally exhausted CD8 T cells that express the highest levels of TIGIT.<sup>3–5</sup> In a first-in-human trial, belrestotug demonstrated a good tolerability profile with early signs of efficacy<sup>6</sup> and we reported for the first time intratumoral TIGIT target engagement in the patient tumors.<sup>5</sup> Belrestotug is currently being tested in combination with anti-PD1 in solid tumors. <h3>Results</h3> Pharmacodynamic assessment made by flow cytometry in the blood of patients treated with belrestotug in monotherapy and in combination with anti-PD1 (pembrolizumab or dostarlimab) showed (i) increased proportion of proliferating memory CD8+ T and NK cells during the first treatment cycle, (ii) sustained depletion of immunosuppressive Tregs, and (iii) decreased proportion of TIGIT high CD8+ T cells. We demonstrated the terminally exhausted phenotype of the TIGIT high CD8+ T cells targeted by belrestotug by isolating peripheral blood mononuclear cells (PBMCs) from treatment naïve cancer patients. Tregs in the tumor microenvironment (TME) hinder effective tumor immune response and are mainly localized in the stromal area. Stromal Tregs co-expressing several immune checkpoint inhibitors, including TIGIT, have a high immunosuppressive profile and its density is associated with poor clinical outcome.<sup>7</sup> We investigated the functional effect of belrestotug combined with anti-PD1 on the TME using a multiplex immunofluorescence (mIF) panel. We observed a decrease of immunosuppressive TIGIT+ Tregs and PD1+ Tregs in the stroma, as well as a spatial reorganization. <h3>Conclusions</h3> Overall, our data suggests a mechanism whereby belrestotug in combination with anti-PD1 induces a more immunocompetent TME driven by immunosuppressive Treg depletion. These data support the clinical development of the doublet therapy to enhance an anti-tumor immune response. <h3>References</h3> Cho BC, <i>et al.</i> Abstract #LBA2 ESMO-IO; 2021 Jonhson ARC-7 ASCO Dec 2022 Yu X, <i>et al</i>, <i>Nature</i>, 2008 Preillon J, <i>et al. Mol Cancer Ther</i>, 2021. Cuende, <i>et al. AACR</i> 2022 Van den Mooter TF, <i>et al.</i> Abstract #CT118 AACR; 2021 Devi-Marulkar, <i>et al</i>, <i>comms Bio</i>, 2022","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":"135161142","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
1422 Th1-specific TROP2 vaccine (AST-07X) elicits strong antigen-specific T cell responsesin vitro 1422 th1特异性TROP2疫苗(AST-07X)在体外引起强烈的抗原特异性T细胞反应
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1422
JinHo Kang, Hyo-Hyun Park, Jin Kyeong Choi, Eunkyo Joung, Hun Jung

Background

Th-Vac® discovery platform, consists of module 1 (Immunoinformatics-based in-silico) and module 2 (in-vitro and vaccine efficacy evaluation) under the comprehensive immunologic algorithms, is aimed to identify antigen-specific MHC class II epitopes for CD4+ T cell with optimal binding affinity and promiscuity across multiple alleles. In a therapeutic cancer area, Th1-specific epitopes prediction followed by cancer vaccine discovery is fully applicable.1–3 Therefore, an immunological mode of action of cancer vaccine powered by Th-Vac® discovery platform is to generate the inflammatory T-cell immunity (type 1 immunity) against tumor antigens with enough potency to overcome either the absence of a T-cell immune response to the tumor or a preexisting immune tolerant response which tends to be the immune-suppressing (type 2 immunity) phenomenon. TROP2 protein is a transmembrane glycoprotein encoded by the Tacstd2 gene. It is known to be overexpressed on the surface of various epithelial cancer cells, including prostate cancer, colorectal cancer, pancreatic cancer, and ovarian cancer, although the role of TROP2 protein in cancer cell growth and proliferation is not well understood.4 In this study, Th1-specific TROP2 epitopes were precisely identified via module 1 and 2 of Th-Vac® discovery platform.

Methods

MHC class II binding epitope predictions were conducted thru engaging common human MHC class II alleles in module 1 of Th-Vac® discovery platform, and MHC class II-specific peptide sequences (up to 15 mers) were initially selected based on the rank order of the predicted binding affinity which are representing potential immunogenic hot spots to MHC class II. Each peptides were synthesized to be stepping into in-vitro immunologic assessment (module 2 of Th-Vac® discovery platform). Th1-specific immunologic response was evaluated with ELISpot and/or FACS analysis.

Results

In a module 1 of Th-Vac® discovery platform, 10 sequences (consists of 15 mers) were finally predicted as potential epitopes that have a high-affinity to MHC class II. In module 2 (2a for in-vitro and 2b for in-vivo immunologic evaluation), four epitopes out of 10 epitopes showed only strong Th1 immune responses without type 2 immunity in both ELISpot and FACS analysis.

Conclusions

TROP2 epitopes that were identified in the Th-Vac® discovery platform has been developed as a promising ‘off-the-shelf’ type Th1-specific therapeutic cancer vaccine (AST-07X). Additionally, the Th-Vac® platform were fully validated in terms of performance and its application would be expanded beyond a cancer vaccine.

References

Watt WC, Cecil DL, Disis ML. Selection of epitopes from self-antigens for eliciting Th2 or Th1 activity in the treatment of autoimmune disease or cancer. Semin Immunopath
Th-Vac®发现平台由综合免疫学算法下的模块1(基于免疫信息学的芯片)和模块2(体外和疫苗功效评估)组成,旨在鉴定CD4+ T细胞抗原特异性MHC II类表位,具有最佳的结合亲和力和多个等位基因的混杂性。在癌症治疗领域,预测th1特异性表位,然后发现癌症疫苗是完全适用的。1 - 3因此,由Th-Vac®发现平台驱动的癌症疫苗的免疫作用模式是产生针对肿瘤抗原的炎性t细胞免疫(1型免疫),其效力足以克服对肿瘤缺乏t细胞免疫反应或预先存在的免疫耐受反应,这往往是免疫抑制(2型免疫)现象。TROP2蛋白是一种由Tacstd2基因编码的跨膜糖蛋白。虽然TROP2蛋白在癌细胞生长和增殖中的作用尚不清楚,但已知它在各种上皮癌细胞表面过表达,包括前列腺癌、结直肠癌、胰腺癌和卵巢癌在本研究中,通过Th-Vac®发现平台的模块1和2精确地鉴定了th1特异性TROP2表位。方法在Th-Vac®发现平台模块1中,通过参与人类常见MHC II类等位基因进行MHC II类结合表位预测,并根据预测结合亲和力的等级顺序初步选择MHC II类特异性肽序列(最长15 mers),这些序列代表MHC II类潜在的免疫原性热点。每个肽被合成后进入体外免疫评估(Th-Vac®发现平台模块2)。采用ELISpot和/或FACS分析评估th1特异性免疫应答。结果在Th-Vac®发现平台的模块1中,最终预测了10个序列(由15个mers组成)作为与MHC II类高亲和力的潜在表位。在模块2中(2a用于体外免疫评估,2b用于体内免疫评估),ELISpot和FACS分析显示,10个表位中有4个表位仅表现出强烈的Th1免疫应答,没有2型免疫。在Th-Vac®发现平台中鉴定的TROP2表位已被开发为有前景的“现成”型th1特异性治疗性癌症疫苗(AST-07X)。此外,Th-Vac®平台在性能方面得到了充分验证,其应用范围将扩展到癌症疫苗之外。Watt WC, Cecil DL, Disis ML.从自身抗原中选择诱导Th2或Th1活性的表位在自身免疫性疾病或癌症的治疗中。免疫学杂志,2017;39(3):245-253。Disis ML, Watt WC, Cecil DL。临床有效癌症疫苗的Th1表位选择肿瘤免疫学。2014年12月13日;3(9):e954971。Park KH, Gad E, Goodell V, Dang Y, Wild T, Higgins D, Fintak P, Childs J, Dela Rosa C, Disis ML.胰岛素样生长因子结合蛋白-2在乳腺癌免疫调节中的作用。王晓东,王晓东,王晓东,等。Trop2基因在肿瘤组织中的表达及其临床意义。中华肿瘤杂志,2008,28(2):349 - 349。的基因,癌症,2015;6(3 - 4)。
{"title":"1422 Th1-specific TROP2 vaccine (AST-07X) elicits strong antigen-specific T cell responses<i>in vitro</i>","authors":"JinHo Kang, Hyo-Hyun Park, Jin Kyeong Choi, Eunkyo Joung, Hun Jung","doi":"10.1136/jitc-2023-sitc2023.1422","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1422","url":null,"abstract":"<h3>Background</h3> Th-Vac<sup>®</sup> discovery platform, consists of module 1 (Immunoinformatics-based <i>in-silico</i>) and module 2 (<i>in-vitro</i> and vaccine efficacy evaluation) under the comprehensive immunologic algorithms, is aimed to identify antigen-specific MHC class II epitopes for CD4<sup>+</sup> T cell with optimal binding affinity and promiscuity across multiple alleles. In a therapeutic cancer area, Th1-specific epitopes prediction followed by cancer vaccine discovery is fully applicable.<sup>1–3</sup> Therefore, an immunological mode of action of cancer vaccine powered by Th-Vac<sup>®</sup> discovery platform is to generate the inflammatory T-cell immunity (type 1 immunity) against tumor antigens with enough potency to overcome either the absence of a T-cell immune response to the tumor or a preexisting immune tolerant response which tends to be the immune-suppressing (type 2 immunity) phenomenon. TROP2 protein is a transmembrane glycoprotein encoded by the Tacstd2 gene. It is known to be overexpressed on the surface of various epithelial cancer cells, including prostate cancer, colorectal cancer, pancreatic cancer, and ovarian cancer, although the role of TROP2 protein in cancer cell growth and proliferation is not well understood.<sup>4</sup> In this study, Th1-specific TROP2 epitopes were precisely identified via module 1 and 2 of Th-Vac<sup>®</sup> discovery platform. <h3>Methods</h3> MHC class II binding epitope predictions were conducted thru engaging common human MHC class II alleles in module 1 of Th-Vac<sup>®</sup> discovery platform, and MHC class II-specific peptide sequences (up to 15 mers) were initially selected based on the rank order of the predicted binding affinity which are representing potential immunogenic hot spots to MHC class II. Each peptides were synthesized to be stepping into <i>in-vitro</i> immunologic assessment (module 2 of Th-Vac<sup>®</sup> discovery platform). Th1-specific immunologic response was evaluated with ELISpot and/or FACS analysis. <h3>Results</h3> In a module 1 of Th-Vac<sup>®</sup> discovery platform, 10 sequences (consists of 15 mers) were finally predicted as potential epitopes that have a high-affinity to MHC class II. In module 2 (2a for <i>in-vitro</i> and 2b for <i>in-vivo</i> immunologic evaluation), four epitopes out of 10 epitopes showed only strong Th1 immune responses without type 2 immunity in both ELISpot and FACS analysis. <h3>Conclusions</h3> TROP2 epitopes that were identified in the Th-Vac<sup>®</sup> discovery platform has been developed as a promising ‘off-the-shelf’ type Th1-specific therapeutic cancer vaccine (AST-07X). Additionally, the Th-Vac<sup>®</sup> platform were fully validated in terms of performance and its application would be expanded beyond a cancer vaccine. <h3>References</h3> Watt WC, Cecil DL, Disis ML. Selection of epitopes from self-antigens for eliciting Th2 or Th1 activity in the treatment of autoimmune disease or cancer. <i>Semin Immunopath","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":"135161268","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
1411 Feasibility, safety, and efficacy of personal vaccines consisting of autologous dendritic cells loadedex vivowith autologous tumor antigens from self-renewing cancer cells 1411自体树突状细胞携带来自自我更新癌细胞的自体肿瘤抗原组成的个人疫苗的可行性、安全性和有效性
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1411
Robert O Dillman, Gabriel I Nistor, Hans S Keirstead

Background

Personal therapeutic cancer vaccines consisting of autologous dendritic cells (DC) loaded ex vivo with autologous tumor antigens (ATA) derived from cells that are self-renewing in culture (cancer-initiating cells, stem cells) have been clinically tested for more than 20 years. The current study addressed feasibility, safety, and efficacy of the DC-ATA approach.

Methods

ATA, as irradiated tumor cells (ITC) or ITC lysates, were derived from short-term tumor cell lines established from resected cancer tissue. Peripheral blood monocytes (MC) were collected by leukapheresis; MC were differentiated into DC by culturing with interleukin-4 and granulocyte-macrophage colony-stimulating-factor (GM-CSF). Cryopreserved doses were thawed, suspended in 500 mcg GM-CSF, and injected at weeks 1, 2, 3, 8, 12, 16, 20, and 24. Data was derived from clinical trials conducted during 2000–2023 in patients with metastatic renal, hepatocellular, and ovary cancers, melanoma, and glioblastoma (GBM). Key end-points were DC-ATA manufacturing success rates, treatment emergent adverse events (TEAE), objective response rates, progression-free survival (PFS) and overall survival (OS).

Results

The success rate for establishing tumor cell lines in proprietary stem cell media was 173/178 (97.2%) including 80/82 GBM, 73/75 ovary, 17/18 liver, 3/3 other. During 2002–2023, leukapheresis procedures yielded sufficient monocytes in 218/223 patients (97.8%) including 74/74 melanoma, 72/74 GBM, 50/53 ovary, 11/11 renal cell, 8/8 hepatoma, and 3/3 other. Of 187 treated patients, no one discontinued DC-ATA because of TEAE; there were no grade 4 TEAE. Most common TEAE were mild to moderate, self-limited local injection site reactions and flu-like symptoms. There was no difference in TEAE frequency or grade in a blinded, randomized trial of DC-ATA vs MC in ovary patients. Immune responses were more favorable for DC-ATA vs ITC in metastatic melanoma, and for DC-ATA vs MC in advanced ovary cancer. During treatment there were no objective responses among 30 patients with measurable metastatic disease, but 3 (10%) had delayed, durable, complete remissions (2 renal, 1 melanoma). In GBM PFS of 10.4 months was 50% longer than in standard treatment arms of six randomized trials. In metastatic melanoma DC-ATA was associated with better OS compared to historical treatment with ITC vaccine (median 60 vs 20.4 months, p<0.001), and better OS compared to ITC in a randomized trial (43.4 vs 20.5 months with a 70% reduction in death p=0.0053)

Conclusions

This DC-ATA approach is feasible and reproducible across tumor types, treatment is well-tolerated, and there is efficacy in some patients. Additional investigation is warranted.

Trial Registration

Clinicaltrials.gov: NCT00014131, NCT00012064, NCT00436930, NCT00331526, NCT03400917

Ethics Approval

All clinical studies obtained approvals from local ethics committees or institutional review bo
个人治疗性癌症疫苗由自体树突状细胞(DC)在体外装载来自培养中自我更新的细胞(癌起始细胞、干细胞)的自体肿瘤抗原(ATA)组成,已经进行了20多年的临床试验。目前的研究探讨了DC-ATA方法的可行性、安全性和有效性。方法ATA作为辐照肿瘤细胞(ITC)或ITC裂解物,来源于切除肿瘤组织建立的短期肿瘤细胞系。白细胞分离法采集外周血单核细胞(MC);通过白细胞介素-4和粒细胞-巨噬细胞集落刺激因子(GM-CSF)培养将MC分化为DC。冷冻保存的剂量解冻,悬浮在500 mcg GM-CSF中,并在第1、2、3、8、12、16、20和24周注射。数据来源于2000-2023年期间在转移性肾癌、肝细胞癌和卵巢癌、黑色素瘤和胶质母细胞瘤(GBM)患者中进行的临床试验。主要终点为DC-ATA制造成功率、治疗紧急不良事件(TEAE)、客观缓解率、无进展生存期(PFS)和总生存期(OS)。结果在专有干细胞培养基中建立肿瘤细胞系的成功率为173/178(97.2%),其中GBM 80/82,卵巢73/75,肝脏17/18,其他3/3。2002-2023年期间,218/223例患者(97.8%)的白细胞分离术获得了足够的单核细胞,包括74/74例黑色素瘤、72/74例GBM、50/53例卵巢、11/11例肾细胞、8/8例肝癌和3/3其他。在187名接受治疗的患者中,没有人因为TEAE而停用DC-ATA;无4级TEAE。最常见的TEAE是轻至中度、自限性局部注射部位反应和流感样症状。在一项针对卵巢患者的DC-ATA和MC的盲法随机试验中,TEAE的频率和级别没有差异。在转移性黑色素瘤中DC-ATA和ITC的免疫应答更有利,在晚期卵巢癌中DC-ATA和MC的免疫应答更有利。在治疗期间,30例可测量的转移性疾病患者没有客观反应,但3例(10%)有延迟,持久,完全缓解(2例肾脏,1例黑色素瘤)。在6个随机试验中,GBM患者的PFS为10.4个月,比标准治疗组长50%。在转移性黑色素瘤中,与ITC疫苗的历史治疗相比,DC-ATA具有更好的OS(中位60个月vs 20.4个月,p<0.001),在一项随机试验中,与ITC相比,DC-ATA具有更好的OS(43.4个月vs 20.5个月,死亡率降低70% p=0.0053)。结论DC-ATA方法在不同肿瘤类型中是可行的,可重复的,治疗耐受性良好,并且在一些患者中有效。有必要进行进一步调查。临床试验注册Clinicaltrials.gov: NCT00014131、NCT00012064、NCT00436930、NCT00331526、NCT03400917伦理批准所有临床研究均获得当地伦理委员会或机构审查委员会的批准,所有参与者均给予书面知情同意。
{"title":"1411 Feasibility, safety, and efficacy of personal vaccines consisting of autologous dendritic cells loaded<i>ex vivo</i>with autologous tumor antigens from self-renewing cancer cells","authors":"Robert O Dillman, Gabriel I Nistor, Hans S Keirstead","doi":"10.1136/jitc-2023-sitc2023.1411","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1411","url":null,"abstract":"<h3>Background</h3> Personal therapeutic cancer vaccines consisting of autologous dendritic cells (DC) loaded <i>ex vivo</i> with autologous tumor antigens (ATA) derived from cells that are self-renewing in culture (cancer-initiating cells, stem cells) have been clinically tested for more than 20 years. The current study addressed feasibility, safety, and efficacy of the DC-ATA approach. <h3>Methods</h3> ATA, as irradiated tumor cells (ITC) or ITC lysates, were derived from short-term tumor cell lines established from resected cancer tissue. Peripheral blood monocytes (MC) were collected by leukapheresis; MC were differentiated into DC by culturing with interleukin-4 and granulocyte-macrophage colony-stimulating-factor (GM-CSF). Cryopreserved doses were thawed, suspended in 500 mcg GM-CSF, and injected at weeks 1, 2, 3, 8, 12, 16, 20, and 24. Data was derived from clinical trials conducted during 2000–2023 in patients with metastatic renal, hepatocellular, and ovary cancers, melanoma, and glioblastoma (GBM). Key end-points were DC-ATA manufacturing success rates, treatment emergent adverse events (TEAE), objective response rates, progression-free survival (PFS) and overall survival (OS). <h3>Results</h3> The success rate for establishing tumor cell lines in proprietary stem cell media was 173/178 (97.2%) including 80/82 GBM, 73/75 ovary, 17/18 liver, 3/3 other. During 2002–2023, leukapheresis procedures yielded sufficient monocytes in 218/223 patients (97.8%) including 74/74 melanoma, 72/74 GBM, 50/53 ovary, 11/11 renal cell, 8/8 hepatoma, and 3/3 other. Of 187 treated patients, no one discontinued DC-ATA because of TEAE; there were no grade 4 TEAE. Most common TEAE were mild to moderate, self-limited local injection site reactions and flu-like symptoms. There was no difference in TEAE frequency or grade in a blinded, randomized trial of DC-ATA vs MC in ovary patients. Immune responses were more favorable for DC-ATA vs ITC in metastatic melanoma, and for DC-ATA vs MC in advanced ovary cancer. During treatment there were no objective responses among 30 patients with measurable metastatic disease, but 3 (10%) had delayed, durable, complete remissions (2 renal, 1 melanoma). In GBM PFS of 10.4 months was 50% longer than in standard treatment arms of six randomized trials. In metastatic melanoma DC-ATA was associated with better OS compared to historical treatment with ITC vaccine (median 60 vs 20.4 months, p<0.001), and better OS compared to ITC in a randomized trial (43.4 vs 20.5 months with a 70% reduction in death p=0.0053) <h3>Conclusions</h3> This DC-ATA approach is feasible and reproducible across tumor types, treatment is well-tolerated, and there is efficacy in some patients. Additional investigation is warranted. <h3>Trial Registration</h3> Clinicaltrials.gov: NCT00014131, NCT00012064, NCT00436930, NCT00331526, NCT03400917 <h3>Ethics Approval</h3> All clinical studies obtained approvals from local ethics committees or institutional review bo","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":"135161272","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
418 The development of ‘off-the-shelf’ manufacturing strategies of iPSC-based gamma-delta T cells 基于ipsc的γ - δ T细胞的“现成”制造策略的发展
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0418
Yanjie Li, Lei Ding, Jixue Li, Mariska Ter Haak, Kate Rochlin, Lawrence Lamb

Background

Gamma-delta (γδ) T cells are depleted during cancer progression resulting in the progressive loss of anti-cancer activity. Elevated numbers of γδ T cells are associated with greater survival outcomes in both hematopoietic and solid malignancies. Induced pluripotent stem cell (iPSC) derived γδ T cells could address the therapeutic challenges of multiple allogeneic γδ T cell infusions as iPSCs possess nearly unlimited self-renewal and multi-lineage differentiation potential. These can be genetically modified, selected, and propagated to provide a source of potentially ‘off-the-shelf’ immune cells.

Methods

Precursor cells obtained from healthy volunteer donors were reprogrammed into iPSCs using non-integrating Yamanaka factors. A feeder-free multi-step strategy was used to differentiate iPSCs, leading to the generation of Vδ1+ γδ T cells. Characterization of the Vδ1+ T cell product included multiplex genomic PCR assays and Sanger sequencing to examine the rearrangement of the TCRγ and TCRδ gene loci, and G-band karyotype analysis. Pluripotent markers (Tra-1–60, OCT3/4 & SSEA4), HPC markers (CD34, CD43), γδ T cell surface markers (CD3, γδ TCR, CD4, CD8, CD16, CD56), effector memory markers (CD45RA, CD27), natural cytotoxicity receptors (NKG2D) were identified using multiparameter flow cytometry. T cell function was determined by flow cytometric cytotoxicity assays against K562, OLM13, U87MG, OVSAHO, OVCAR-3, KURAMUCHI targets at increasing Effector to Target (E:T) ratios. Th1/2/17 cytokine release was determined following PMA/ionomycin stimulation and LEGENDplex™ bead-based immunoassays.

Results

We generated Vδ1T-iPSC lines (iVδ1T) identified as Vγ5-to-Jγ1/2 and Vδ1-to-Jδ1 recombination. One iPSC line showed normal karyotype with 99% cells expressing OCT3/4 & SSEA4. The differentiation process generated 70+ million iVδ1T cells from 3 million iPSCs expressing γδ T cell markers CD45, CD3, Vδ1-TCR, CD16, CD56, NKG2D, CD45RA, and CD27. Cytokine release following PMA/ionomycin stimulation showed increases of at least 50x for Granzyme A, 300x for IFN-γ, 1400x for TNF-α, ~10 to 20x for Granzyme B, ~5 to 10x for Perforin, ~6x for Granulysin. IL-6 was not detected either before or after stimulation, and IL17A was at low concentration. At a 16:1 E:T ratio, preliminary data shows that Vδ1+ γδ T cells killed K562 (CML) 95.7%; MOLM13 (AML) 60.3%; U87MG (glioblastoma) 70.3%; and ovarian cancer lines OVSAHO 57.1%, OVCAR-3 69.6%, and KURAMUCHI 55.1%.

Conclusions

We generated Vδ1+ iPSC derived γδ T cells with effector cytokine phenotype and low risk for cytokine release syndrome. Robust cytotoxic activity was seen across a variety of cancer cell lines, potentially providing an off-the-shelf platform for allogeneic cell therapy.
γ - δ (γδ) T细胞在癌症进展过程中被耗尽,导致抗癌活性逐渐丧失。在造血和实体恶性肿瘤中,γδ T细胞数量的升高与更高的生存结果相关。诱导多能干细胞(iPSC)衍生的γδ T细胞具有几乎无限的自我更新和多谱系分化潜力,可以解决多种异体γδ T细胞输注的治疗挑战。这些细胞可以经过基因改造、筛选和繁殖,以提供潜在的“现成的”免疫细胞来源。方法利用非整合Yamanaka因子将健康志愿者供体的前体细胞重组为iPSCs。采用无喂食多步策略分化iPSCs,导致Vδ1+ γδ T细胞的产生。Vδ1+ T细胞产物的鉴定包括多重基因组PCR测定和Sanger测序,以检查TCRγ和TCRδ基因位点的重排,以及g带核型分析。多能标记(Tra-1-60, OCT3/4 &采用多参数流式细胞术对SSEA4、HPC标记物(CD34、CD43)、γδ T细胞表面标记物(CD3、γδ TCR、CD4、CD8、CD16、CD56)、效应记忆标记物(CD45RA、CD27)、天然细胞毒性受体(NKG2D)进行鉴定。流式细胞术检测T细胞对K562、OLM13、U87MG、OVSAHO、OVCAR-3、KURAMUCHI靶点的细胞毒性,提高靶效比(E:T)。在PMA/离子霉素刺激和LEGENDplex™微球免疫分析后,检测Th1/2/17细胞因子释放。结果获得了Vδ1T-iPSC系(iVδ1T),鉴定为v γ5- j γ1/2和v δ1- j δ1重组系。一个iPSC细胞系显示正常核型,99%的细胞表达OCT3/4和amp;SSEA4。分化过程从300万个iPSCs中产生了7000多万个表达γδ T细胞标记物CD45、CD3、Vδ1-TCR、CD16、CD56、NKG2D、CD45RA和CD27的iVδ1T细胞。PMA/离子霉素刺激后的细胞因子释放显示颗粒酶A至少增加50倍,IFN-γ增加300倍,TNF-α增加1400倍,颗粒酶B增加10 ~ 20倍,穿孔素增加5 ~10倍,颗粒酶增加6倍。刺激前后均未检测到IL-6, il - 17a浓度较低。在16:1的E:T比下,初步数据显示,Vδ1+ γδ T细胞杀伤K562 (CML) 95.7%;Molm13 (aml) 60.3%;U87MG(胶质母细胞瘤)70.3%;卵巢癌系OVSAHO 57.1%, OVCAR-3 69.6%, KURAMUCHI 55.1%。结论我们获得了Vδ1+ iPSC衍生的γδ T细胞,具有效应细胞因子表型,细胞因子释放综合征风险低。在多种癌细胞系中发现了强大的细胞毒活性,可能为同种异体细胞治疗提供现成的平台。
{"title":"418 The development of ‘off-the-shelf’ manufacturing strategies of iPSC-based gamma-delta T cells","authors":"Yanjie Li, Lei Ding, Jixue Li, Mariska Ter Haak, Kate Rochlin, Lawrence Lamb","doi":"10.1136/jitc-2023-sitc2023.0418","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0418","url":null,"abstract":"<h3>Background</h3> Gamma-delta (γδ) T cells are depleted during cancer progression resulting in the progressive loss of anti-cancer activity. Elevated numbers of γδ T cells are associated with greater survival outcomes in both hematopoietic and solid malignancies. Induced pluripotent stem cell (iPSC) derived γδ T cells could address the therapeutic challenges of multiple allogeneic γδ T cell infusions as iPSCs possess nearly unlimited self-renewal and multi-lineage differentiation potential. These can be genetically modified, selected, and propagated to provide a source of potentially ‘off-the-shelf’ immune cells. <h3>Methods</h3> Precursor cells obtained from healthy volunteer donors were reprogrammed into iPSCs using non-integrating Yamanaka factors. A feeder-free multi-step strategy was used to differentiate iPSCs, leading to the generation of Vδ1+ γδ T cells. Characterization of the Vδ1+ T cell product included multiplex genomic PCR assays and Sanger sequencing to examine the rearrangement of the TCRγ and TCRδ gene loci, and G-band karyotype analysis. Pluripotent markers (Tra-1–60, OCT3/4 &amp; SSEA4), HPC markers (CD34, CD43), γδ T cell surface markers (CD3, γδ TCR, CD4, CD8, CD16, CD56), effector memory markers (CD45RA, CD27), natural cytotoxicity receptors (NKG2D) were identified using multiparameter flow cytometry. T cell function was determined by flow cytometric cytotoxicity assays against K562, OLM13, U87MG, OVSAHO, OVCAR-3, KURAMUCHI targets at increasing Effector to Target (E:T) ratios. Th1/2/17 cytokine release was determined following PMA/ionomycin stimulation and LEGENDplex™ bead-based immunoassays. <h3>Results</h3> We generated Vδ1T-iPSC lines (iVδ1T) identified as Vγ5-to-Jγ1/2 and Vδ1-to-Jδ1 recombination. One iPSC line showed normal karyotype with 99% cells expressing OCT3/4 &amp; SSEA4. The differentiation process generated 70+ million iVδ1T cells from 3 million iPSCs expressing γδ T cell markers CD45, CD3, Vδ1-TCR, CD16, CD56, NKG2D, CD45RA, and CD27. Cytokine release following PMA/ionomycin stimulation showed increases of at least 50x for Granzyme A, 300x for IFN-γ, 1400x for TNF-α, ~10 to 20x for Granzyme B, ~5 to 10x for Perforin, ~6x for Granulysin. IL-6 was not detected either before or after stimulation, and IL17A was at low concentration. At a 16:1 E:T ratio, preliminary data shows that Vδ1+ γδ T cells killed K562 (CML) 95.7%; MOLM13 (AML) 60.3%; U87MG (glioblastoma) 70.3%; and ovarian cancer lines OVSAHO 57.1%, OVCAR-3 69.6%, and KURAMUCHI 55.1%. <h3>Conclusions</h3> We generated Vδ1+ iPSC derived γδ T cells with effector cytokine phenotype and low risk for cytokine release syndrome. Robust cytotoxic activity was seen across a variety of cancer cell lines, potentially providing an off-the-shelf platform for allogeneic cell therapy.","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":"135161285","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
69 A novel protease-free method for the co-detection of RNA and protein biomarkers using the RNAscope™ technology 使用RNAscope™技术,一种新的无蛋白酶方法用于RNA和蛋白质生物标志物的共同检测
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0069
Laetitia Chatelain, Anji Bei, Nancy George, Ge-Ah Kim, Sonali Deshpande, Steve Zhou, Li-Chong Wang, Maithreyan Srinivasan

Background

Spatial biology methods are increasingly used for the characterization of complex tissue microenvironments, the understanding of which can shed light on fundamental biological mechanisms and better inform development of targeted therapeutics. RNAscope™ in situ hybridization (ISH) technology, capable of highly sensitive single-molecule RNA detection, can be combined with immunohistochemistry (IHC) or immunofluorescence (IF) for the co-detection of clinically relevant biomarkers on the same slide with morphological context. This application is especially important in immuno-oncology research to profile immune cell populations using protein markers and characterize their activation states by detecting cytokine and chemokine expression with RNA. However, RNAscope necessitates the use of proteases to digest RNA-associated proteins and facilitate probe access to RNA targets, which can negatively impact epitopes targeted by some antibodies. Previously, we developed the Integrated Co-detection Workflow (ICW) to partially solve this problem, with the fixation of the primary antibody-target complex prior to the protease application. While ICW rescues signal for many previously incompatible antibodies, proteases can still adversely impact some of the primary antibody-target complex. To address this deficiency, we have developed a novel RNA-protein co-detection workflow that eliminates the need for protease, resulting in high detection sensitivities for both protein and RNA markers.

Methods

To maintain the same RNA detection sensitivity without the use of proteases, we formulated a new protease-free pretreatment buffer to replace the existing protease step within the current workflow which allows adequate accessibility of RNAscope™ probes to the target RNAs. Following this protease-free pretreatment, tissue specimens were assayed with RNAscope Multiplex to detect RNA species in the tissue, followed by standard IF staining to co-detect protein biomarkers. Antibodies which previously exhibited degraded protein signal in both sequential ISH-IF and ICW were tested in the new protocol.

Results

Here, we present results from FFPE human tissues to co-detect several protease-sensitive antibodies, including degranulating cytotoxic lymphocyte marker CD107a along with human house-keeping genes TBP, POLR2A and PPIB. The protease-free RNAscope co-detection workflow restored the protein staining pattern at nominal antibody concentrations used for IHC while maintaining mRNA dot counts for TBP, POLR2A and PPIB, indicating minimal impact of protease-free pretreatment buffer on both RNA and protein signal.

Conclusions

The protease-free RNAscope co-detection workflow will serve as a powerful multi-omics staining technique for a wider range of antibodies by enabling visualization of RNA-protein co-detection for the comprehensive profiling of tissue microenvironments, facilitating fast
空间生物学方法越来越多地用于复杂组织微环境的表征,对其的理解可以揭示基本的生物学机制,并更好地为靶向治疗的发展提供信息。RNAscope™原位杂交(ISH)技术能够进行高灵敏度的单分子RNA检测,可以与免疫组织化学(IHC)或免疫荧光(IF)相结合,在具有形态学背景的同一张载玻片上共同检测临床相关的生物标志物。这一应用在免疫肿瘤学研究中尤为重要,它利用蛋白质标记来描绘免疫细胞群体,并通过RNA检测细胞因子和趋化因子的表达来表征它们的激活状态。然而,RNAscope需要使用蛋白酶来消化RNA相关蛋白,并促进探针进入RNA靶标,这可能会对一些抗体靶向的表位产生负面影响。以前,我们开发了集成协同检测工作流(ICW)来部分解决这个问题,在蛋白酶应用之前固定初级抗体-靶标复合物。虽然ICW可以挽救许多先前不相容的抗体的信号,但蛋白酶仍然可以对一些主要抗体-靶标复合物产生不利影响。为了解决这一缺陷,我们开发了一种新的RNA-蛋白质共检测工作流程,消除了对蛋白酶的需求,从而对蛋白质和RNA标记物都具有高检测灵敏度。为了在不使用蛋白酶的情况下保持相同的RNA检测灵敏度,我们制定了一种新的无蛋白酶预处理缓冲液,以取代当前工作流程中现有的蛋白酶步骤,使RNAscope™探针能够充分接近目标RNA。在这种无蛋白酶预处理之后,组织标本用RNAscope Multiplex检测组织中的RNA种类,然后用标准IF染色来共同检测蛋白质生物标志物。先前在顺序ISH-IF和ICW中表现出降解蛋白信号的抗体在新方案中进行了测试。在这里,我们展示了从FFPE人组织中共同检测几种蛋白酶敏感抗体的结果,包括脱颗粒细胞毒性淋巴细胞标志物CD107a以及人类看家基因TBP, POLR2A和PPIB。无蛋白酶RNAscope共检测工作流程恢复了用于免疫组化的标准抗体浓度下的蛋白质染色模式,同时维持了TBP、POLR2A和PPIB的mRNA点计数,表明无蛋白酶预处理缓冲液对RNA和蛋白质信号的影响最小。无蛋白酶RNAscope共检测工作流程将成为一种强大的多组学染色技术,可用于更广泛的抗体,使rna -蛋白共检测可视化,从而全面分析组织微环境,促进治疗方法发现的更快突破。
{"title":"69 A novel protease-free method for the co-detection of RNA and protein biomarkers using the RNAscope™ technology","authors":"Laetitia Chatelain, Anji Bei, Nancy George, Ge-Ah Kim, Sonali Deshpande, Steve Zhou, Li-Chong Wang, Maithreyan Srinivasan","doi":"10.1136/jitc-2023-sitc2023.0069","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0069","url":null,"abstract":"<h3>Background</h3> Spatial biology methods are increasingly used for the characterization of complex tissue microenvironments, the understanding of which can shed light on fundamental biological mechanisms and better inform development of targeted therapeutics. RNAscope™ <i>in situ</i> hybridization (ISH) technology, capable of highly sensitive single-molecule RNA detection, can be combined with immunohistochemistry (IHC) or immunofluorescence (IF) for the co-detection of clinically relevant biomarkers on the same slide with morphological context. This application is especially important in immuno-oncology research to profile immune cell populations using protein markers and characterize their activation states by detecting cytokine and chemokine expression with RNA. However, RNAscope necessitates the use of proteases to digest RNA-associated proteins and facilitate probe access to RNA targets, which can negatively impact epitopes targeted by some antibodies. Previously, we developed the Integrated Co-detection Workflow (ICW) to partially solve this problem, with the fixation of the primary antibody-target complex prior to the protease application. While ICW rescues signal for many previously incompatible antibodies, proteases can still adversely impact some of the primary antibody-target complex. To address this deficiency, we have developed a novel RNA-protein co-detection workflow that eliminates the need for protease, resulting in high detection sensitivities for both protein and RNA markers. <h3>Methods</h3> To maintain the same RNA detection sensitivity without the use of proteases, we formulated a new protease-free pretreatment buffer to replace the existing protease step within the current workflow which allows adequate accessibility of RNAscope™ probes to the target RNAs. Following this protease-free pretreatment, tissue specimens were assayed with RNAscope Multiplex to detect RNA species in the tissue, followed by standard IF staining to co-detect protein biomarkers. Antibodies which previously exhibited degraded protein signal in both sequential ISH-IF and ICW were tested in the new protocol. <h3>Results</h3> Here, we present results from FFPE human tissues to co-detect several protease-sensitive antibodies, including degranulating cytotoxic lymphocyte marker CD107a along with human house-keeping genes <i>TBP</i>, <i>POLR2A</i> and <i>PPIB</i>. The protease-free RNAscope co-detection workflow restored the protein staining pattern at nominal antibody concentrations used for IHC while maintaining mRNA dot counts for <i>TBP</i>, <i>POLR2A</i> and <i>PPIB</i>, indicating minimal impact of protease-free pretreatment buffer on both RNA and protein signal. <h3>Conclusions</h3> The protease-free RNAscope co-detection workflow will serve as a powerful multi-omics staining technique for a wider range of antibodies by enabling visualization of RNA-protein co-detection for the comprehensive profiling of tissue microenvironments, facilitating fast","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":"135161288","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
47 Baseline blood DNA methylation-based immune profiles and tumor mutational burden predict survival outcomes in anti-PD-1 treated head and neck cancer patient 基线血液DNA甲基化免疫谱和肿瘤突变负担预测抗pd -1治疗的头颈癌患者的生存结果
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0047
Ze Zhang, Kartik Sehgal, Keisuke Shirai, Rondi Butler, John Wiencke, Devin Koestler, Geat Ramush, Min Kyung Lee, Annette Molinaro, Hannah Stolrow, Lucas A Salas, Robert Haddad, Karl Kelsey, Brock Christensen

Background

Immune checkpoint inhibitors (ICIs) are approved to treat patients with recurrent/metastatic head and neck squamous cell carcinoma (HNSCC). However, currently approved biomarkers are limited due to the heterogeneity and availability of tumor samples. The development of peripheral biomarkers offers an alternative noninvasive approach to assess immunotherapy response. DNA methylation-based immune cell deconvolution provides opportunities for developing blood-based biomarkers to predict immunotherapy response outcomes in HNSCC.

Methods

Our study is an ongoing prospective multi-center study aimed at identifying blood DNA methylation biomarkers of therapy response in patients with HNSCC undergoing standard-of-care, FDA-approved ICIs. Blood was drawn prior to immunotherapy initiation. DNA isolated from these samples underwent methylation profiling using the Illumina EPIC microarray. Peripheral blood immune profiles were generated using cellular deconvolution.1 69 HNSCC patients with anti-PD-1 monotherapy were included (figure 1). 47 patients’ tumor samples were sequenced to evaluate tumor mutational burden (TMB) (figure 2). We investigated 48 immune variables and TMB for relation with progression-free survival (PFS) and overall survival (OS) using Cox proportional-hazard models adjusted for age, sex, and a marker for corticosteroid exposure.2 12 primary immune cell proportions and TMB were investigated for interaction. A p-value < 0.05 was used as the cut-off for statistical significance.

Results

In 69 patients with HNSCC who received anti-PD-1 therapy, neutrophil proportion, monocyte count, and total B cell count were associated with worse PFS outcomes while CD4T memory cell count and total T cell count were associated with better PFS outcomes (figure 3). Regarding OS, NLR, neutrophil proportion, total naïve lymphocyte proportion, monocyte count, CD4T naïve percentage, CD4T naïve to memory ratio, and T regulatory cell percentage were found to be associated with poorer survival while total CD4 T cell count, total CD4 T cell proportion, CD4 T memory cell proportion, total T cell count, lymphocyte to monocyte ratio, and total lymphocyte proportion were found to be associated with better outcomes (figure 4). Higher TMB was found to be associated with better survival outcomes. TMB exhibits significant interaction with peripheral monocyte proportion. TMB is a better predictor of survival in individuals with a lower level of monocyte proportion (figure 5).

Conclusions

DNA methylation-based immune profiling in peripheral blood at baseline identifies clinically relevant biomarkers of benefit from ICIs. Our results demonstrate the potential of new blood DNA methylation-based biomarkers to predict immunotherapy response prior to the initial treatment, and connected peripheral immune profile with TMB through their interactive impact on survival outcomes.

References

Salas LA, Zhang Z, Ko
免疫检查点抑制剂(ICIs)被批准用于治疗复发/转移性头颈部鳞状细胞癌(HNSCC)患者。然而,由于肿瘤样本的异质性和可获得性,目前批准的生物标志物受到限制。外周生物标志物的发展为评估免疫治疗反应提供了一种非侵入性的替代方法。基于DNA甲基化的免疫细胞反褶积为开发基于血液的生物标志物来预测HNSCC的免疫治疗反应结果提供了机会。方法:本研究是一项正在进行的前瞻性多中心研究,旨在确定接受fda批准的标准治疗ICIs的HNSCC患者的血液DNA甲基化生物标志物。在免疫治疗开始前抽血。从这些样品中分离的DNA使用Illumina EPIC微阵列进行甲基化分析。使用细胞反褶积生成外周血免疫谱。1 69例接受抗pd -1单药治疗的HNSCC患者被纳入研究(图1)。47例患者的肿瘤样本被测序以评估肿瘤突变负担(TMB)(图2)。我们使用Cox比例风险模型,对年龄、性别和皮质类固醇暴露标志物进行调整,研究了48个免疫变量和TMB与无进展生存期(PFS)和总生存期(OS)的关系。研究了12个原代免疫细胞比例与TMB的相互作用。p值<统计学意义以0.05为截止值。结果在69例接受抗pd -1治疗的HNSCC患者中,中性粒细胞比例、单核细胞计数和总B细胞计数与较差的PFS结果相关,而CD4T记忆细胞计数和总T细胞计数与较好的PFS结果相关(图3)。对于OS、NLR、中性粒细胞比例、总naïve淋巴细胞比例、单核细胞计数、CD4T naïve百分比、CD4T naïve与记忆比,和T调节细胞百分比被发现与较差的生存率相关,而总CD4 T细胞计数、总CD4 T细胞比例、CD4 T记忆细胞比例、总T细胞计数、淋巴细胞/单核细胞比例和总淋巴细胞比例被发现与较好的预后相关(图4)。较高的TMB被发现与较好的生存结果相关。TMB与外周单核细胞比例有显著的相互作用。在单核细胞比例较低的个体中,TMB是更好的生存预测指标(图5)。结论:基线时外周血中基于DNA甲基化的免疫谱确定了ICIs获益的临床相关生物标志物。我们的研究结果证明了新的基于血液DNA甲基化的生物标志物在初始治疗前预测免疫治疗反应的潜力,并通过它们对生存结果的相互影响将外周免疫谱与TMB联系起来。参考文献Salas LA, Zhang Z, Koestler DC, Butler RA, Hansen HM, Molinaro AM, Wiencke JK, Kelsey KT, Christensen BC。使用DNA甲基化增强外周血细胞反褶积,用于高分辨率免疫谱分析。学报,2022;13:761。Wiencke JK, Molinaro AM, Warrier G, Rice T, Clarke J, Taylor JW, wresch M, Hansen H, McCoy L, Tang E,等。DNA甲基化作为糖皮质激素反应和胶质瘤生存的药效学标志物。自然科学学报(英文版);2022;13:5505。本研究由达特茅斯癌症中心IRB (STUDY02001227)、布朗大学IRB(1901002321)和丹娜-法伯癌症研究所IRB(18-548)批准。
{"title":"47 Baseline blood DNA methylation-based immune profiles and tumor mutational burden predict survival outcomes in anti-PD-1 treated head and neck cancer patient","authors":"Ze Zhang, Kartik Sehgal, Keisuke Shirai, Rondi Butler, John Wiencke, Devin Koestler, Geat Ramush, Min Kyung Lee, Annette Molinaro, Hannah Stolrow, Lucas A Salas, Robert Haddad, Karl Kelsey, Brock Christensen","doi":"10.1136/jitc-2023-sitc2023.0047","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0047","url":null,"abstract":"<h3>Background</h3> Immune checkpoint inhibitors (ICIs) are approved to treat patients with recurrent/metastatic head and neck squamous cell carcinoma (HNSCC). However, currently approved biomarkers are limited due to the heterogeneity and availability of tumor samples. The development of peripheral biomarkers offers an alternative noninvasive approach to assess immunotherapy response. DNA methylation-based immune cell deconvolution provides opportunities for developing blood-based biomarkers to predict immunotherapy response outcomes in HNSCC. <h3>Methods</h3> Our study is an ongoing prospective multi-center study aimed at identifying blood DNA methylation biomarkers of therapy response in patients with HNSCC undergoing standard-of-care, FDA-approved ICIs. Blood was drawn prior to immunotherapy initiation. DNA isolated from these samples underwent methylation profiling using the Illumina EPIC microarray. Peripheral blood immune profiles were generated using cellular deconvolution.<sup>1</sup> 69 HNSCC patients with anti-PD-1 monotherapy were included (figure 1). 47 patients’ tumor samples were sequenced to evaluate tumor mutational burden (TMB) (figure 2). We investigated 48 immune variables and TMB for relation with progression-free survival (PFS) and overall survival (OS) using Cox proportional-hazard models adjusted for age, sex, and a marker for corticosteroid exposure.<sup>2</sup> 12 primary immune cell proportions and TMB were investigated for interaction. A p-value < 0.05 was used as the cut-off for statistical significance. <h3>Results</h3> In 69 patients with HNSCC who received anti-PD-1 therapy, neutrophil proportion, monocyte count, and total B cell count were associated with worse PFS outcomes while CD4T memory cell count and total T cell count were associated with better PFS outcomes (figure 3). Regarding OS, NLR, neutrophil proportion, total naïve lymphocyte proportion, monocyte count, CD4T naïve percentage, CD4T naïve to memory ratio, and T regulatory cell percentage were found to be associated with poorer survival while total CD4 T cell count, total CD4 T cell proportion, CD4 T memory cell proportion, total T cell count, lymphocyte to monocyte ratio, and total lymphocyte proportion were found to be associated with better outcomes (figure 4). Higher TMB was found to be associated with better survival outcomes. TMB exhibits significant interaction with peripheral monocyte proportion. TMB is a better predictor of survival in individuals with a lower level of monocyte proportion (figure 5). <h3>Conclusions</h3> DNA methylation-based immune profiling in peripheral blood at baseline identifies clinically relevant biomarkers of benefit from ICIs. Our results demonstrate the potential of new blood DNA methylation-based biomarkers to predict immunotherapy response prior to the initial treatment, and connected peripheral immune profile with TMB through their interactive impact on survival outcomes. <h3>References</h3> Salas LA, Zhang Z, Ko","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":"135161298","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
1466 Galectin-1 orchestrates a hierarchical tumor to stromal extracellular vesicles system that fosters immune suppression 1466 .半乳糖凝集素-1协调了肿瘤到间质细胞外囊泡系统的分级,促进免疫抑制
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1466
Camila A Bach, Ada Blidner, Ramiro Perrotta, Joaquin P Merlo, Mariana Salatino, Gabriel Rabinovich

Background

Extracellular vesicles (EVs) suppress effector cells and activate immunosuppressive cells in the ‘escape phase’ of tumor immunoediting. Tumor derived EVs contribute to the differentiation and expansion of immunosuppressive cell subsets. Bone marrow-derived cells (BMDCs) can uptake EVs released by 4T1 -a triple-negative breast cancer cell line- and induce myeloid-derived suppressor cells (MDSCs).1 Immunosuppressive myeloid cells expansion can also be promoted by their own EV in an autocrine manner. Moreover, EVs secreted by immunosuppressive myeloid cells, may inherit their parental functions.2 Nevertheless, the molecular circuits and mediators of tumor microenvironment EV release remain uncertain.

Methods

We generated immunosuppressive myeloid cells by culturing mouse BMDCs with GM-CSF for three days in the presence of Gal1. Additionally, we cultured 4T1 WT and Lgals1 knocked out cells. We purified Small EVs by size exclusion chromatography from conditioned medium of immunosuppressive myeloid cells (control and Gal1-treated) or 4T1 cells (WT or Gal1 KO). We further purified 4T1 EVs with CD63+ beads, a tumoral EV marker. ELISA, flow cytometry, miRNA sequencing, proteomic and metabolomic analyses verified EV identity. We performed functional assays by co-culturing tumoral or myeloid EVs with BMDC, activated T or B cells.

Results

When exposed to EVs from 4T1 cells in vitro, BMDCs switched their differentiation pathway to a M-MDSCs phenotype (CD11b+Ly6G-Ly6Chi) and showed higher immune checkpoint molecules expression, including PD-L1 (p=0.005) and VISTA (p=0.003). However, when exposed to tumoral EVs lacking Gal1, BMDCs reinforced a PMN-MDSCs phenotype (CD11b+Ly6G+Ly6Clo). In contrast with Gal1+ EVs, tumoral EVs lacking Gal1 failed to inhibit CD4+ and CD8+ T and B cell proliferation and activation. Gal1 is both in the cargo and corona of 4T1 EVs as revealed by flow cytometry and ELISA (p=0.0032). Moreover, EV production inhibition with GW4869 in 4T1 cells decreased Gal1 levels secreted (p=0.037). To analyze EV-derived Gal1 contribution as an MDSC autocrine signal, we added EVs from Gal1-treated immunosuppressive myeloid cells to new BMDCs and observed CD11b+ cells expansion and higher VISTA expression on cells with M-MDSCs phenotype. Moreover, these EVs showed greater T cell-suppressive capacity and has different protein, metabolic, and miRNA cargo compared to control EVs.

Conclusions

Here, we propose that Gal1+ EVs released by tumoral cells interact with myeloid cells and potentiate their immunosuppressive properties, including the release of EV with autocrine and paracrine functions. Targeting Gal1 as a molecular mechanism of EV-mediated tumor development may help overcome therapy resistance.

References

Xiang X, Poliakov A, Liu C, Liu Y, D
细胞外囊泡(EVs)在肿瘤免疫编辑的“逃逸期”抑制效应细胞并激活免疫抑制细胞。肿瘤源性ev有助于免疫抑制细胞亚群的分化和扩增。骨髓源性细胞(bmdc)可以摄取由4T1(三阴性乳腺癌细胞系)释放的ev,并诱导髓源性抑制细胞(MDSCs) 1免疫抑制性骨髓细胞的扩张也可以通过自身的EV以自分泌的方式促进。此外,免疫抑制髓细胞分泌的ev可能继承了亲代的功能然而,肿瘤微环境EV释放的分子通路和介质仍不确定。方法在Gal1存在下,用GM-CSF培养小鼠BMDCs 3 d,产生免疫抑制的骨髓细胞。此外,我们还培养了4T1 WT和Lgals1敲除的细胞。我们从免疫抑制骨髓细胞(对照和Gal1处理)或4T1细胞(WT或Gal1 KO)的条件培养基中纯化了小ev。我们进一步用CD63+珠(一种肿瘤EV标记物)纯化了4T1 EV。ELISA、流式细胞术、miRNA测序、蛋白质组学和代谢组学分析证实了EV的身份。我们通过将肿瘤或髓系EVs与BMDC、活化的T细胞或B细胞共培养进行了功能测定。结果当体外暴露于4T1细胞的EVs时,bmdscs将其分化途径转换为M-MDSCs表型(CD11b+Ly6G-Ly6Chi),并表现出更高的免疫检查点分子表达,包括PD-L1 (p=0.005)和VISTA (p=0.003)。然而,当暴露于缺乏Gal1的肿瘤ev时,BMDCs增强了PMN-MDSCs表型(CD11b+Ly6G+Ly6Clo)。与Gal1+ ev相比,缺乏Gal1的肿瘤ev不能抑制CD4+和CD8+ T细胞和B细胞的增殖和活化。流式细胞术和酶联免疫吸附试验显示,4T1型EVs的货物和冠状细胞中均存在Gal1 (p=0.0032)。此外,GW4869抑制4T1细胞的EV产生可降低Gal1分泌水平(p=0.037)。为了分析ev来源的Gal1作为MDSC自分泌信号的贡献,我们将来自Gal1处理的免疫抑制骨髓细胞的ev添加到新的bmscs中,观察到CD11b+细胞在M-MDSCs表型细胞上的扩增和更高的VISTA表达。此外,与对照电动汽车相比,这些电动汽车表现出更强的T细胞抑制能力,并且具有不同的蛋白质、代谢和miRNA载货。结论:肿瘤细胞释放的Gal1+ EV与髓系细胞相互作用,增强其免疫抑制特性,包括释放具有自分泌和旁分泌功能的EV。靶向Gal1作为ev介导的肿瘤发展的分子机制可能有助于克服治疗耐药性。向祥,Poliakov A,刘超,刘勇,邓志斌,王军,程忠,沙世伟,王广军,张磊,Grizzle WE, Moble J,张洪柱。肿瘤外泌体诱导髓源性抑制细胞的研究。中华医学杂志,2009;24(4):391 - 391。Rashid MH, Borin TF。mscs来源的外泌体在肿瘤微环境中的免疫抑制作用。2021;45:1171-1181。
{"title":"1466 Galectin-1 orchestrates a hierarchical tumor to stromal extracellular vesicles system that fosters immune suppression","authors":"Camila A Bach, Ada Blidner, Ramiro Perrotta, Joaquin P Merlo, Mariana Salatino, Gabriel Rabinovich","doi":"10.1136/jitc-2023-sitc2023.1466","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1466","url":null,"abstract":"<h3>Background</h3> Extracellular vesicles (EVs) suppress effector cells and activate immunosuppressive cells in the ‘escape phase’ of tumor immunoediting. Tumor derived EVs contribute to the differentiation and expansion of immunosuppressive cell subsets. Bone marrow-derived cells (BMDCs) can uptake EVs released by 4T1 -a triple-negative breast cancer cell line- and induce myeloid-derived suppressor cells (MDSCs).<sup>1</sup> Immunosuppressive myeloid cells expansion can also be promoted by their own EV in an autocrine manner. Moreover, EVs secreted by immunosuppressive myeloid cells, may inherit their parental functions.<sup>2</sup> Nevertheless, the molecular circuits and mediators of tumor microenvironment EV release remain uncertain. <h3>Methods</h3> We generated immunosuppressive myeloid cells by culturing mouse BMDCs with GM-CSF for three days in the presence of Gal1. Additionally, we cultured 4T1 WT and <i>Lgals1</i> knocked out cells. We purified Small EVs by size exclusion chromatography from conditioned medium of immunosuppressive myeloid cells (control and Gal1-treated) or 4T1 cells (WT or Gal1 KO). We further purified 4T1 EVs with CD63+ beads, a tumoral EV marker. ELISA, flow cytometry, miRNA sequencing, proteomic and metabolomic analyses verified EV identity. We performed functional assays by co-culturing tumoral or myeloid EVs with BMDC, activated T or B cells. <h3>Results</h3> When exposed to EVs from 4T1 cells <i>in vitro</i>, BMDCs switched their differentiation pathway to a M-MDSCs phenotype (CD11b<sup>+</sup>Ly6G<sup>-</sup>Ly6C<sup>hi</sup>) and showed higher immune checkpoint molecules expression, including PD-L1 (p=0.005) and VISTA (p=0.003). However, when exposed to tumoral EVs lacking Gal1, BMDCs reinforced a PMN-MDSCs phenotype (CD11b<sup>+</sup>Ly6G<sup>+</sup>Ly6C<sup>lo</sup>). In contrast with Gal1<sup>+</sup> EVs, tumoral EVs lacking Gal1 failed to inhibit CD4<sup>+</sup> and CD8<sup>+</sup> T and B cell proliferation and activation. Gal1 is both in the cargo and corona of 4T1 EVs as revealed by flow cytometry and ELISA (p=0.0032). Moreover, EV production inhibition with GW4869 in 4T1 cells decreased Gal1 levels secreted (p=0.037). To analyze EV-derived Gal1 contribution as an MDSC autocrine signal, we added EVs from Gal1-treated immunosuppressive myeloid cells to new BMDCs and observed CD11b<sup>+</sup> cells expansion and higher VISTA expression on cells with M-MDSCs phenotype. Moreover, these EVs showed greater T cell-suppressive capacity and has different protein, metabolic, and miRNA cargo compared to control EVs. <h3>Conclusions</h3> Here, we propose that Gal1<sup>+</sup> EVs released by tumoral cells interact with myeloid cells and potentiate their immunosuppressive properties, including the release of EV with autocrine and paracrine functions. Targeting Gal1 as a molecular mechanism of EV-mediated tumor development may help overcome therapy resistance. <h3>References</h3> Xiang X, Poliakov A, Liu C, Liu Y, D","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":"135161402","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
1397 CDR404, an antibody-based bispecific & bivalent T-cell engager targeted against MAGE-A4, for Squamous Non-Small Cell Lung Cancer (SQ-NSCLC) 1397 CDR404是一种基于抗体的双特异性抗体。靶向MAGE-A4的二价t细胞接合剂治疗鳞状非小细胞肺癌(SQ-NSCLC)
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1397
Melissa Vrohlings, Stephanie Jungmichel, Ivana Tosevski, Alessio Vantellini, Philip Knobel, Nadia Sanchez, Elizabeth Ross, Marian Van Kerckhoven, Giorgia Giacomazzi, Maria Liivrand, Reija Hieta, Nicholas Dupuis, Dieter Rondas, Pamela Swatkowski, Daniel Lenherr-Frey, Swethajit Biswas, Gilberto Lopes, Leonardo Borras

Background

Squamous non-small cell lung cancer (SQ-NSCLC) is the 2nd most common type of lung cancer. Given the paucity of actionable oncogene drivers, and lack of efficacy from multiple therapies in the Lung-MAP trial, there is a high unmet need in SQ-NSCLC to develop effective 2nd-line immunotherapies for patients with disease progression after immune checkpoint inhibitors (ICI). The melanoma antigen gene A4 (MAGE-A4) is exclusively expressed in cancer and absent in somatic tissues. MAGE-A4-derived peptides presented on HLA molecules at the cell surface recently emerged as a novel therapeutic opportunity. Thus, the two key objectives of this study were to: 1). Evaluate MAGE-A4 expression in human SQ-NSCLC; 2). Demonstrate the anti-cancer activity of CDR404, an antibody-based bispecific and bivalent T-cell engager targeted against MAGE-A4230–239 peptide in vitro and in vivo xenograft models of SQ-NSCLC.

Methods

MAGE-A4 mRNA prevalence and expression in SQ-NSCLC was analyzed using the TCGA database (https://www.cancer.gov/tcga). Protein expression of MAGE-A4 was confirmed using immunohistochemistry (IHC) in fifty FFPE human SQ-NSCLC samples (clone E7O1U). CDR404 target cell killing in the presence of human PBMCs was assessed using the human SQ-NSCLC cell line NCI-H1703. HLA-A*02:01+MAGE-A4neg cancer cells were used as controls. To exclude reactivity of CDR404 in healthy tissues, HLA-A*02:01+ primary cells presenting peptides with high MAGE-A4 similarity were co-cultured with human PBMCs. In vivo activity of CDR404 in SQ-NSCLC was evaluated with an NCI-H1703 xenograft model in NSG mice.

Results

SQ-NSCLC had the highest MAGE-A4 mRNA expression levels among solid cancers in the TCGA database. IHC showed positive MAGE-A4 staining in 28/50 (56%) of SQ-NSCLC samples. In vitro, CDR404 showed efficient target cell lysis across all effector-to-target ratios tested. Similarly, simultaneous target engagement and resulting synapse formation induced T cell activation and secretion of cytolytic molecules in an effector-to-target ratio-dependent fashion. No reactivity was observed using co-cultured HLA-A*02:01+MAGE-A4neg cancer cells. Lack of T cell activation/cytolytic molecule release in the presence of HLA-A*02:01+ primary cells confirmed the specificity profile of CDR404. In vivo, treatment with four different doses of CDR404 induced complete tumor regression in the SQ-NSCLC NCI-H1703 xenograft model.

Conclusions

The high MAGE-A4 expression levels and the highly specific anti-cancer cell activity of CDR404 make it a highly attractive immunotherapy for development post-progression on ICI for patients with HLA-A*02:01+ SQ-NSCLC. A multi-tumor phase 1 trial of CDR404, including SQ-NSCLC, is expected to begin in 2024 with prospective patient selection for both HLA-A*02:01 and tumor MAGE-A4.

鳞状非小细胞肺癌(SQ-NSCLC)是第二常见的肺癌类型。鉴于在Lung-MAP试验中缺乏可操作的癌基因驱动因素,以及多种治疗缺乏疗效,SQ-NSCLC在使用免疫检查点抑制剂(ICI)后疾病进展的患者开发有效的二线免疫疗法的需求仍未得到满足。黑色素瘤抗原基因A4 (MAGE-A4)仅在癌症中表达,在体细胞组织中不存在。mage - a4衍生肽在细胞表面HLA分子上呈现,最近成为一种新的治疗机会。因此,本研究的两个关键目标是:1)评估MAGE-A4在人类SQ-NSCLC中的表达;2).在SQ-NSCLC的体外和体内异种移植模型中,验证了针对MAGE-A4230-239肽的基于抗体的双特异性和双价t细胞参与器CDR404的抗癌活性。方法采用TCGA数据库(https://www.cancer.gov/tcga)分析MAGE-A4 mRNA在SQ-NSCLC中的表达及流行情况。50例FFPE人SQ-NSCLC(克隆E7O1U)中MAGE-A4蛋白的表达经免疫组化(IHC)证实。使用人SQ-NSCLC细胞系NCI-H1703评估CDR404靶细胞在人PBMCs存在下的杀伤作用。以HLA-A*02:01+ mage - a4阴性癌细胞为对照。为了排除CDR404在健康组织中的反应性,我们将HLA-A*02:01+原代细胞与人PBMCs共培养,这些原代细胞的多肽具有较高的MAGE-A4相似性。采用NSG小鼠NCI-H1703异种移植模型评价CDR404在SQ-NSCLC中的体内活性。结果在TCGA数据库的实体癌中,SQ-NSCLC的MAGE-A4 mRNA表达水平最高。在28/50(56%)的SQ-NSCLC样本中,免疫组化显示MAGE-A4阳性。在体外,CDR404在所有效靶比测试中都显示出有效的靶细胞裂解。同样,同时的靶标接合和由此产生的突触形成诱导T细胞以效应靶比依赖的方式激活和分泌细胞溶解分子。共培养HLA-A*02:01+ mage - a4阴性癌细胞无反应性。在HLA-A*02:01+原代细胞存在时,缺乏T细胞活化/细胞溶解分子释放,证实了CDR404的特异性。在体内,四种不同剂量的CDR404治疗在SQ-NSCLC NCI-H1703异种移植模型中诱导肿瘤完全消退。结论CDR404具有MAGE-A4高表达水平和高特异性的抗癌活性,是一种极具吸引力的治疗进展后ICI的HLA-A*02:01+ SQ-NSCLC的免疫疗法。CDR404包括SQ-NSCLC在内的多肿瘤1期临床试验预计将于2024年开始,前瞻性患者选择HLA-A*02:01和肿瘤MAGE-A4。伦理批准动物研究是按照《实验动物护理和使用指南》的建议进行的,涉及约束、饲养、外科手术、饲料和液体调节以及兽医护理。动物护理和使用计划得到了国际实验动物护理评估和认证协会(AAALAC)的认可,该协会确保符合公认的实验室动物护理和使用标准。
{"title":"1397 CDR404, an antibody-based bispecific &amp; bivalent T-cell engager targeted against MAGE-A4, for Squamous Non-Small Cell Lung Cancer (SQ-NSCLC)","authors":"Melissa Vrohlings, Stephanie Jungmichel, Ivana Tosevski, Alessio Vantellini, Philip Knobel, Nadia Sanchez, Elizabeth Ross, Marian Van Kerckhoven, Giorgia Giacomazzi, Maria Liivrand, Reija Hieta, Nicholas Dupuis, Dieter Rondas, Pamela Swatkowski, Daniel Lenherr-Frey, Swethajit Biswas, Gilberto Lopes, Leonardo Borras","doi":"10.1136/jitc-2023-sitc2023.1397","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1397","url":null,"abstract":"<h3>Background</h3> Squamous non-small cell lung cancer (SQ-NSCLC) is the 2<sup>nd</sup> most common type of lung cancer. Given the paucity of actionable oncogene drivers, and lack of efficacy from multiple therapies in the Lung-MAP trial, there is a high unmet need in SQ-NSCLC to develop effective 2<sup>nd</sup>-line immunotherapies for patients with disease progression after immune checkpoint inhibitors (ICI). The melanoma antigen gene A4 (MAGE-A4) is exclusively expressed in cancer and absent in somatic tissues. MAGE-A4-derived peptides presented on HLA molecules at the cell surface recently emerged as a novel therapeutic opportunity. Thus, the two key objectives of this study were to: 1). Evaluate MAGE-A4 expression in human SQ-NSCLC; 2). Demonstrate the anti-cancer activity of CDR404, an antibody-based bispecific and bivalent T-cell engager targeted against MAGE-A4<sub>230–239</sub> peptide <i>in vitro</i> and <i>in vivo</i> xenograft models of SQ-NSCLC. <h3>Methods</h3> MAGE-A4 mRNA prevalence and expression in SQ-NSCLC was analyzed using the TCGA database (https://www.cancer.gov/tcga). Protein expression of MAGE-A4 was confirmed using immunohistochemistry (IHC) in fifty FFPE human SQ-NSCLC samples (clone E7O1U). CDR404 target cell killing in the presence of human PBMCs was assessed using the human SQ-NSCLC cell line NCI-H1703. HLA-A*02:01<sup>+</sup>MAGE-A4<sup>neg</sup> cancer cells were used as controls. To exclude reactivity of CDR404 in healthy tissues, HLA-A*02:01<sup>+</sup> primary cells presenting peptides with high MAGE-A4 similarity were co-cultured with human PBMCs. <i>In vivo</i> activity of CDR404 in SQ-NSCLC was evaluated with an NCI-H1703 xenograft model in NSG mice. <h3>Results</h3> SQ-NSCLC had the highest MAGE-A4 mRNA expression levels among solid cancers in the TCGA database. IHC showed positive MAGE-A4 staining in 28/50 (56%) of SQ-NSCLC samples. <i>In vitro</i>, CDR404 showed efficient target cell lysis across all effector-to-target ratios tested. Similarly, simultaneous target engagement and resulting synapse formation induced T cell activation and secretion of cytolytic molecules in an effector-to-target ratio-dependent fashion. No reactivity was observed using co-cultured HLA-A*02:01<sup>+</sup>MAGE-A4<sup>neg</sup> cancer cells. Lack of T cell activation/cytolytic molecule release in the presence of HLA-A*02:01<sup>+</sup> primary cells confirmed the specificity profile of CDR404. <i>In vivo</i>, treatment with four different doses of CDR404 induced complete tumor regression in the SQ-NSCLC NCI-H1703 xenograft model. <h3>Conclusions</h3> The high MAGE-A4 expression levels and the highly specific anti-cancer cell activity of CDR404 make it a highly attractive immunotherapy for development post-progression on ICI for patients with HLA-A*02:01+ SQ-NSCLC. A multi-tumor phase 1 trial of CDR404, including SQ-NSCLC, is expected to begin in 2024 with prospective patient selection for both HLA-A*02:01 and tumor MAGE-A4. <h3","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":"135161403","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
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