首页 > 最新文献

Regular and Young Investigator Award Abstracts最新文献

英文 中文
755 DEKA-1 a dose-finding Phase 1 trial: observing safety and biomarkers using DK210(EGFR) for inoperable locally advanced and/or metastatic EGFR+ tumors failing systemic therapy 755 DEKA-1是一项剂量发现的1期试验:观察DK210(EGFR)治疗无法手术的局部晚期和/或转移性EGFR+肿瘤的安全性和生物标志物
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0755
Alexander I Spira, Douglas Orr, Aurélien Marabelle, Elizabeth C Moser, Deb Kientop, John Mumm

Background

Both interleukin-2 (IL-2) and interleukin-10 (IL-10) have been extensively studied for their stimulatory function on T cells and their potential to obtain sustainable tumor control in renal, melanoma, lung, and pancreatic cancers as monotherapy. While approved, IL-2 exhibits significant toxicity in a high percentage of the patient population, limiting its widespread use. The significant efforts undertaken to uncouple IL-2 toxicity from its anti-tumor function have been unsuccessful and early phase clinical safety observed with PEGylated IL-10 was not met in a blinded Phase 3 trial. Deka Biosciences has engineered a novel molecule coupling wild-type IL-2 to a high affinity variant of Epstein Barr Viral (EBV) IL-10 via a scaffold that binds to epidermal growth factor receptors (EGFR). This patented molecule, named DK210 (EGFR), is targeted to EGFR expressing cells and demonstrated to be retained at high levels within the tumor microenvironment days after dosing. In addition to overlapping and non-redundant anti-tumor function, IL-10 reduces IL-2 mediated cytokine release syndrome (CRS) risks and inhibits IL-2 mediated T regulatory cell proliferation.

Methods

DK210 (EGFR) is being evaluated in an open-label, dose-escalation (Phase 1) study (BOIN design) with five (0.025–0.3 mg/kg) monotherapy dose levels. DK210 (EGFR) is home-administered via subcutaneous injection three times a week. The objectives of this study include evaluating the safety, CRS occurrence, pharmacokinetics, pharmacodynamic and predictive biomarkers, presence of anti-drug antibodies, and antitumor activity.

Results

As of June 22, 2023, three patients were enrolled, and are continuing DK210 (EGFR) treatment. Subjects improved clinically and no drug related toxicities, nor CRS were observed in any of the patients. One subject with pancreatic cancer achieved stable disease by RECIST 1.1.

Conclusions

DK210 (EGFR) couples wild-type IL-2 with a high affinity IL-10 and targets cytokines directly to the tumor microenvironment significantly changing Il-2s therapeutic window. Preliminary human data shows an encouraging safety and efficiency profile. The dose-escalation study is expected to be completed by the end of this year (NCT05704985).

Acknowledgements

The authors would like to thank all patients who participated in this study and their families, as well as all the investigators and site staff who made the study possible.

Trial Registration

Trial Registration www.clinicaltrials.gov; NCT05704985

Ethics Approval

The study was approved by NEXT Oncology, Salus IRB, approval number NXVIR22.60, on 14-Feb-2023 and by Mary Crowley Medical Research Center Institutional Review Board, approval number 23–08, on 21-Apr-2023.

Consent

Written informed consent was obtained from the patient for publication of this abstract and any accompanying images. A copy of the w
白细胞介素-2 (IL-2)和白细胞介素-10 (IL-10)对T细胞的刺激功能以及它们作为单一疗法在肾癌、黑色素瘤、肺癌和胰腺癌中获得持续肿瘤控制的潜力已经被广泛研究。虽然获得批准,但IL-2在很大比例的患者群体中显示出显著的毒性,限制了其广泛使用。将IL-2毒性与其抗肿瘤功能分离的重大努力尚未成功,PEGylated IL-10在盲法3期试验中未达到早期临床安全性。Deka Biosciences公司设计了一种新型分子,通过结合表皮生长因子受体(EGFR)的支架,将野生型IL-2与eb病毒(EBV) IL-10的高亲和力变体偶联。这种专利分子名为DK210 (EGFR),针对表达EGFR的细胞,并在给药后数天内在肿瘤微环境中保持高水平。除了重叠和非冗余的抗肿瘤功能外,IL-10还可以降低IL-2介导的细胞因子释放综合征(CRS)风险,抑制IL-2介导的T调节性细胞增殖。DK210 (EGFR)正在一项开放标签、剂量递增(1期)研究(BOIN设计)中进行评估,该研究采用5个(0.025-0.3 mg/kg)单药剂量水平。DK210 (EGFR)通过皮下注射每周在家给药三次。本研究的目的包括评估安全性、CRS发生、药代动力学、药效学和预测性生物标志物、抗药物抗体的存在和抗肿瘤活性。截至2023年6月22日,3名患者入组,并继续接受DK210 (EGFR)治疗。受试者临床改善,无药物相关毒性,未观察到任何患者的CRS。1例胰腺癌患者通过RECIST 1.1达到病情稳定。结论DK210 (EGFR)将野生型IL-2与高亲和力IL-10偶联,将细胞因子直接靶向肿瘤微环境,显著改变Il-2s的治疗窗口期。初步的人体数据显示出令人鼓舞的安全性和有效性。剂量递增研究预计将于今年年底完成(NCT05704985)。作者要感谢所有参与这项研究的患者及其家属,以及所有使这项研究成为可能的研究者和现场工作人员。试验注册试验注册www.clinicaltrials.gov;该研究于2023年2月14日获得NEXT Oncology (Salus IRB)批准,批准号为NXVIR22.60,并于2023年4月21日获得Mary Crowley Medical Research Center机构审查委员会批准,批准号为23-08。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。一份书面同意书副本可供本刊编辑审阅
{"title":"755 DEKA-1 a dose-finding Phase 1 trial: observing safety and biomarkers using DK2<sup>10</sup>(EGFR) for inoperable locally advanced and/or metastatic EGFR+ tumors failing systemic therapy","authors":"Alexander I Spira, Douglas Orr, Aurélien Marabelle, Elizabeth C Moser, Deb Kientop, John Mumm","doi":"10.1136/jitc-2023-sitc2023.0755","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0755","url":null,"abstract":"<h3>Background</h3> Both interleukin-2 (IL-2) and interleukin-10 (IL-10) have been extensively studied for their stimulatory function on T cells and their potential to obtain sustainable tumor control in renal, melanoma, lung, and pancreatic cancers as monotherapy. While approved, IL-2 exhibits significant toxicity in a high percentage of the patient population, limiting its widespread use. The significant efforts undertaken to uncouple IL-2 toxicity from its anti-tumor function have been unsuccessful and early phase clinical safety observed with PEGylated IL-10 was not met in a blinded Phase 3 trial. Deka Biosciences has engineered a novel molecule coupling wild-type IL-2 to a high affinity variant of Epstein Barr Viral (EBV) IL-10 via a scaffold that binds to epidermal growth factor receptors (EGFR). This patented molecule, named DK2<sup>10</sup> (EGFR), is targeted to EGFR expressing cells and demonstrated to be retained at high levels within the tumor microenvironment days after dosing. In addition to overlapping and non-redundant anti-tumor function, IL-10 reduces IL-2 mediated cytokine release syndrome (CRS) risks and inhibits IL-2 mediated T regulatory cell proliferation. <h3>Methods</h3> DK2<sup>10</sup> (EGFR) is being evaluated in an open-label, dose-escalation (Phase 1) study (BOIN design) with five (0.025–0.3 mg/kg) monotherapy dose levels. DK2<sup>10</sup> (EGFR) is home-administered via subcutaneous injection three times a week. The objectives of this study include evaluating the safety, CRS occurrence, pharmacokinetics, pharmacodynamic and predictive biomarkers, presence of anti-drug antibodies, and antitumor activity. <h3>Results</h3> As of June 22, 2023, three patients were enrolled, and are continuing DK2<sup>10</sup> (EGFR) treatment. Subjects improved clinically and no drug related toxicities, nor CRS were observed in any of the patients. One subject with pancreatic cancer achieved stable disease by RECIST 1.1. <h3>Conclusions</h3> DK2<sup>10</sup> (EGFR) couples wild-type IL-2 with a high affinity IL-10 and targets cytokines directly to the tumor microenvironment significantly changing Il-2s therapeutic window. Preliminary human data shows an encouraging safety and efficiency profile. The dose-escalation study is expected to be completed by the end of this year (NCT05704985). <h3>Acknowledgements</h3> The authors would like to thank all patients who participated in this study and their families, as well as all the investigators and site staff who made the study possible. <h3>Trial Registration</h3> Trial Registration www.clinicaltrials.gov; NCT05704985 <h3>Ethics Approval</h3> The study was approved by NEXT Oncology, Salus IRB, approval number NXVIR22.60, on 14-Feb-2023 and by Mary Crowley Medical Research Center Institutional Review Board, approval number 23–08, on 21-Apr-2023. <h3>Consent</h3> Written informed consent was obtained from the patient for publication of this abstract and any accompanying images. A copy of the w","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":"135162552","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
708 A phase 1/2 study of rinatabart sesutecan (PRO1184), a novel folate receptor alpha-directed antibody-drug conjugate, in patients with locally advanced and/or metastatic solid tumors 708 .一项针对局部晚期和/或转移性实体瘤患者的新型叶酸受体靶向抗体-药物偶联物——rinatabart sesutecan (PRO1184)的1/2期研究
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0708
Justin A Call, Ian Anderson, Ira Winer, Douglas Orr, Oladapo Yeku, Debra L Richardson, Jian Zhang, Elizabeth Lee, Gottfried Konecny, Ning Li, Sandip P Patel, Lin Wu, Jing Wang, Jun Zhang, Ying Cheng, Xiaohua Wu, Naomi Hunder, Lian Lu, Sharon Ma, Eric Song, Erika Hamilton

Background

Rinatabart sesutecan (Rina-S) is an antibody-drug conjugate (ADC) consisting of a human monoclonal antibody that selectively binds FRα, a novel cleavable hydrophilic linker, and a topoisomerase 1 inhibitor payload, exatecan. The hydrophilic linker confers superior physicochemical properties and pharmacokinetics compared to conventional linkers in preclinical models. Rina-S exerts robust antitumor activity in mouse xenograft models of multiple tumor types with high, moderate, and low FRα expression, consistent with the broad potency and bystander activity of the exatecan payload. Here we present emerging data from the first-in-human trial (NCT05579366).

Methods

PRO1184–001 is an ongoing, phase 1/2, open-label, dose escalation and expansion study. Eligible patients have locally advanced and/or metastatic/unresectable solid tumors, including epithelial ovarian cancer (EOC), endometrial cancer, non-small cell lung cancer (NSCLC), breast cancer, or mesothelioma. Patients must have measurable disease per the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1, or mRECIST 1.1 for pleural mesothelioma. FRα tumor expression levels were retrospectively tested using the Ventana immunohistochemistry FOLR1 assay. Primary objectives are to identify the maximum tolerated dose, recommended phase 2 dose, and evaluate safety and tolerability.

Results

As of 09 June 2023, 10 patients have been treated with Rina-S at 60 (n=3) and 120 (n=7) mg/m2. Tumor types included platinum-resistant/refractory EOC (n=5), endometrial cancer (n=2), NSCLC (n=2), and mesothelioma (n=1). Patients received a median of 4 (range, 1 to 9) prior treatments. Eight patients completed the DLT period and had post-baseline tumor assessments per RECIST. The other 2 patients, who had FRα-negative tumors, discontinued due to early clinical progression. Patients have received between 1 and 8 cycles and 5 remain on treatment. The most common treatment-related adverse events (AEs) were nausea (n=5), decreased white blood cell counts (n=3), and fatigue, decreased lymphocyte counts, and decreased neutrophil counts (n=2 each); most events were Grade 1 or 2. Treatment-related ≥ Grade 3 hematologic AEs were reported for 2 patients treated at 120 mg/m2. No ocular toxicity or interstitial lung disease was observed. No DLTs were observed. Antitumor activity was observed at both dose levels and in patients with high, medium, and low FRα expression, including an ongoing confirmed partial response in a patient with endometrial cancer and decreased tumor measurements in additional patients.

Conclusions

Emerging data suggest a promising safety profile for Rina-S, with most AEs being mild or moderate and consistent with findings in preclinical studies. Antitumor activity has been observed at well tolerated dose levels. Dose escalation continues.

Trial Registration

Clinicaltrials.gov NCT05579366

Ethics Approval

The study obt
Rinatabart sesutecan (Rina-S)是一种抗体-药物偶联物(ADC),由一种人单克隆抗体选择性结合FRα(一种新型可切割亲水连接体)和拓扑异构酶1抑制剂exatecan组成。在临床前模型中,与传统的连接剂相比,亲水性连接剂具有优越的物理化学性质和药代动力学。在FRα高、中、低表达的多种肿瘤类型的小鼠异种移植物模型中,Rina-S具有强大的抗肿瘤活性,与exatecan有效载荷的广泛效力和旁观者活性一致。在此,我们介绍来自首次人体试验(NCT05579366)的新数据。方法PRO1184-001是一项正在进行的1/2期、开放标签、剂量递增和扩展研究。符合条件的患者患有局部晚期和/或转移性/不可切除的实体肿瘤,包括上皮性卵巢癌(EOC)、子宫内膜癌、非小细胞肺癌(NSCLC)、乳腺癌或间皮瘤。根据实体瘤反应评价标准(RECIST) 1.1或胸膜间皮瘤反应评价标准(mRECIST) 1.1,患者必须有可测量的疾病。采用Ventana免疫组化FOLR1法回顾性检测FRα肿瘤表达水平。主要目的是确定最大耐受剂量,推荐的2期剂量,并评估安全性和耐受性。截至2023年6月9日,10例患者接受了60 (n=3)和120 (n=7) mg/m2的Rina-S治疗。肿瘤类型包括铂耐药/难治性EOC (n=5)、子宫内膜癌(n=2)、非小细胞肺癌(n=2)和间皮瘤(n=1)。患者先前接受的治疗中位数为4次(范围1至9次)。8名患者完成了DLT期,并根据RECIST进行了基线后肿瘤评估。另外2例fr α阴性肿瘤患者因早期临床进展而停药。患者接受了1至8个周期的治疗,其中5个仍在接受治疗。最常见的治疗相关不良事件(ae)是恶心(n=5)、白细胞计数减少(n=3)、疲劳、淋巴细胞计数减少和中性粒细胞计数减少(各n=2);大多数项目是1级或2级。在120 mg/m2治疗的2例患者中报告了治疗相关≥3级血液学ae。未观察到眼毒性或间质性肺疾病。未观察到dlt。在两种剂量水平和高、中、低FRα表达的患者中观察到抗肿瘤活性,包括在子宫内膜癌患者中持续证实的部分反应和其他患者的肿瘤测量降低。新出现的数据表明,Rina-S具有良好的安全性,大多数不良反应为轻度或中度,与临床前研究结果一致。在耐受良好的剂量水平下观察到抗肿瘤活性。剂量继续增加。试验注册Clinicaltrials.gov NCT05579366伦理批准该研究获得了WCG机构审查委员会的伦理批准;ID 20223552),参与者在参与前给予知情同意。
{"title":"708 A phase 1/2 study of rinatabart sesutecan (PRO1184), a novel folate receptor alpha-directed antibody-drug conjugate, in patients with locally advanced and/or metastatic solid tumors","authors":"Justin A Call, Ian Anderson, Ira Winer, Douglas Orr, Oladapo Yeku, Debra L Richardson, Jian Zhang, Elizabeth Lee, Gottfried Konecny, Ning Li, Sandip P Patel, Lin Wu, Jing Wang, Jun Zhang, Ying Cheng, Xiaohua Wu, Naomi Hunder, Lian Lu, Sharon Ma, Eric Song, Erika Hamilton","doi":"10.1136/jitc-2023-sitc2023.0708","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0708","url":null,"abstract":"<h3>Background</h3> Rinatabart sesutecan (Rina-S) is an antibody-drug conjugate (ADC) consisting of a human monoclonal antibody that selectively binds FRα, a novel cleavable hydrophilic linker, and a topoisomerase 1 inhibitor payload, exatecan. The hydrophilic linker confers superior physicochemical properties and pharmacokinetics compared to conventional linkers in preclinical models. Rina-S exerts robust antitumor activity in mouse xenograft models of multiple tumor types with high, moderate, and low FRα expression, consistent with the broad potency and bystander activity of the exatecan payload. Here we present emerging data from the first-in-human trial (NCT05579366). <h3>Methods</h3> PRO1184–001 is an ongoing, phase 1/2, open-label, dose escalation and expansion study. Eligible patients have locally advanced and/or metastatic/unresectable solid tumors, including epithelial ovarian cancer (EOC), endometrial cancer, non-small cell lung cancer (NSCLC), breast cancer, or mesothelioma. Patients must have measurable disease per the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1, or mRECIST 1.1 for pleural mesothelioma. FRα tumor expression levels were retrospectively tested using the Ventana immunohistochemistry FOLR1 assay. Primary objectives are to identify the maximum tolerated dose, recommended phase 2 dose, and evaluate safety and tolerability. <h3>Results</h3> As of 09 June 2023, 10 patients have been treated with Rina-S at 60 (n=3) and 120 (n=7) mg/m<sup>2</sup>. Tumor types included platinum-resistant/refractory EOC (n=5), endometrial cancer (n=2), NSCLC (n=2), and mesothelioma (n=1). Patients received a median of 4 (range, 1 to 9) prior treatments. Eight patients completed the DLT period and had post-baseline tumor assessments per RECIST. The other 2 patients, who had FRα-negative tumors, discontinued due to early clinical progression. Patients have received between 1 and 8 cycles and 5 remain on treatment. The most common treatment-related adverse events (AEs) were nausea (n=5), decreased white blood cell counts (n=3), and fatigue, decreased lymphocyte counts, and decreased neutrophil counts (n=2 each); most events were Grade 1 or 2. Treatment-related ≥ Grade 3 hematologic AEs were reported for 2 patients treated at 120 mg/m<sup>2</sup>. No ocular toxicity or interstitial lung disease was observed. No DLTs were observed. Antitumor activity was observed at both dose levels and in patients with high, medium, and low FRα expression, including an ongoing confirmed partial response in a patient with endometrial cancer and decreased tumor measurements in additional patients. <h3>Conclusions</h3> Emerging data suggest a promising safety profile for Rina-S, with most AEs being mild or moderate and consistent with findings in preclinical studies. Antitumor activity has been observed at well tolerated dose levels. Dose escalation continues. <h3>Trial Registration</h3> Clinicaltrials.gov NCT05579366 <h3>Ethics Approval</h3> The study obt","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":"135162554","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
947 The activation states of tumor-resident type 2 dendritic cells impact the strength of ovarian cancer immune responses 肿瘤驻留的2型树突状细胞的激活状态影响卵巢癌免疫应答的强度
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0947
Fiona Chatterjee, Vincent Butty, Emi A Lutz, K Dane Wittrup, Stefani Spranger

Background

Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States.1 To date, checkpoint blockade therapy (CBT) has failed to be effective in ovarian cancer.2 CBT efficacy typically requires a strong pre-existing tumor-specific T cell response.3 4 However, analysis of patient data indicates that tumor-infiltrating T cells in ovarian cancer are often poorly activated.5 Understanding the mechanisms governing this poor T cell activation could lead to novel ovarian cancer therapeutics.

Methods

We utilized a transplantable, syngeneic, murine ovarian cancer cell line driven by Ccne1OE p53-/-R172H Ak2OE and KrasG12V (CPAK).6 CPAK tumor cells were implanted intraperitoneally to model metastatic ovarian cancer or subcutaneously to model productive systemic immunity. CPAK cells were engineered to express the model CD8+ T cell antigen SIY to enable studies of tumor-specific T cells. CBT consisted of αCTLA-4 and αPD-L1 therapy. Immune cells were profiled using flow cytometry and single-cell RNA sequencing (scRNA-seq).

Results

Intraperitoneal (IP) CPAK-SIY tumors were unresponsive to CBT. However, mice bearing subcutaneous (SQ) CPAK-SIY tumors treated with CBT displayed delayed tumor growth compared to control animals. Flow cytometric analysis of tumor-infiltrating CD8+ T cells illustrated that while tumor-reactive T cells were activated in IP and SQ tumors, tumor-reactive T cells in IP tumors failed to upregulate high levels of effector molecules such as granzyme B. Unbiased analysis of dendritic cells (DCs) within IP tumors using scRNA-seq revealed a population of DCs that expressed CD103 and CD11b and a type 2 conventional DC (cDC2) gene signature. Gene signature analysis indicated these CD103+ CD11b+ double-positive DCs were a suppressive state of cDC2s induced by TGFb that reside in gut tissue and induce Tregs.7 Analysis of tumor-infiltrating DCs in IP tumors revealed that the proportion of double-positive DCs increased during tumor growth while the proportion of cDC2s decreased. We hypothesized that the polarization of cDC2s away from these suppressive double-positive DCs may improve ovarian cancer immunity. Previous work from our group demonstrated that cDC2s acquire an activation state characterized by an interferon-stimulated gene signature (ISG+ DCs) upon exposure to interferon-beta (IFNβ) and that ISG+ DCs are potent activators of CD8+ T cells.8 In our ovarian cancer model, addition of IFNβ to IP tumors enhanced tumor-specific T cell responses.

Conclusions

Metastatic ovarian tumors are refractory to CBT and are infiltrated by poorly activated CD8+ T cells. Our results suggest that the poor T cell activation is a consequence of the cDC2 activation state within the tumor
背景:卵巢癌是美国女性癌症相关死亡的第五大原因,迄今为止,检查点阻断疗法(CBT)对卵巢癌无效CBT的有效性通常需要一个强大的预先存在的肿瘤特异性T细胞反应。然而,对患者数据的分析表明,卵巢癌的肿瘤浸润性T细胞往往激活不良了解控制这种不良T细胞活化的机制可能会导致新的卵巢癌治疗方法。方法利用一种可移植的、同基因的小鼠卵巢癌细胞系,由Ccne1OE p53-/- r172h Ak2OE和KrasG12V (CPAK)驱动CPAK肿瘤细胞被腹腔植入以模拟转移性卵巢癌,或皮下植入以模拟产生性全身免疫。CPAK细胞被工程化表达模型CD8+ T细胞抗原SIY,使肿瘤特异性T细胞的研究成为可能。CBT包括αCTLA-4和αPD-L1治疗。利用流式细胞术和单细胞RNA测序(scRNA-seq)对免疫细胞进行分析。结果腹腔(IP) cpap - siy肿瘤对CBT无反应。然而,与对照动物相比,接受CBT治疗的皮下(SQ) cpap - siy肿瘤小鼠的肿瘤生长延迟。流式细胞术分析肿瘤浸润的CD8+ T细胞表明,虽然肿瘤反应性T细胞在IP和SQ肿瘤中被激活,但IP肿瘤中的肿瘤反应性T细胞未能上调高水平的效应分子,如颗粒酶b。使用scRNA-seq对IP肿瘤中的树突状细胞(DC)进行无偏分析,结果显示DC群体表达CD103和CD11b以及2型常规DC (cDC2)基因特征。基因标记分析表明,这些CD103+ CD11b+双阳性dc是TGFb诱导的cDC2s存在于肠道组织并诱导tregs的抑制状态。7对IP肿瘤中肿瘤浸润dc的分析显示,肿瘤生长过程中双阳性dc的比例增加,而cDC2s的比例下降。我们假设cDC2s远离这些抑制性双阳性dc的极化可能改善卵巢癌免疫。本小组之前的研究表明,cDC2s在暴露于干扰素- β (ifn - β)时获得以干扰素刺激基因特征(ISG+ DCs)为特征的激活状态,ISG+ DCs是CD8+ T细胞的有效激活剂在我们的卵巢癌模型中,在IP肿瘤中添加IFNβ增强了肿瘤特异性T细胞反应。结论卵巢转移性肿瘤CBT治疗难治性强,且有低活化CD8+ T细胞浸润。我们的研究结果表明,T细胞激活不良是肿瘤内cDC2激活状态的结果。我们要感谢麻省理工学院的比较医学系和科赫研究所的斯旺森生物技术核心设施。这项工作得到了突破癌症和玛格丽特A.坎宁安癌症研究免疫机制奖学金的支持。引用文献sigel RL, Miller KD, Wagle NS . Jemal A.癌症统计,2023。中华肿瘤杂志[J]; 2009; 33 (3): 417 - 418, doi:10.3322/caac.21763。坎达拉夫特LE, Odunsi K, Coukos G.卵巢癌的免疫治疗:我们还在那里吗?中华临床医学杂志,2019;37:2460-2471,doi:10.1200/JCO.19.00508。Tumeh PC等。PD-1阻断通过抑制适应性免疫抵抗诱导应答。Nature 2014; 515:568-571, doi:10.1038/nature13954。Cristescu R,等。基于PD-1检查点阻断免疫治疗的泛肿瘤基因组生物标志物。Science 2018;362, doi:10.1126/ Science。aar3593(2018)。巴斯克斯-加西亚等人。卵巢癌突变过程驱动位点特异性免疫逃避。Nature 2022; 612:778-786, doi:10.1038/s41586-022-05496-1。Iyer S,等。基因定义的同基因卵巢癌小鼠模型作为发现联合免疫治疗的工具。癌症发现2021;11:384-407,doi:10.1158/2159-8290.CD-20-0818。Bain CC等。TGFbetaR信号控制肠道中CD103(+)CD11b(+)树突状细胞的发育。学报,2017;8:620,doi:10.1038/s41467-017-00658-6。杜恩,等。I型干扰素激活MHC I类修饰的CD11b(+)常规树突状细胞,促进保护性抗肿瘤CD8(+) T细胞免疫。免疫2022;55:308-323 e309, doi:10.1016/ j.j immune .2021.10.020。所有小鼠实验均经麻省理工学院动物护理委员会(CAC) - PHS动物福利保证# D16-00078 (A3125-01)批准。
{"title":"947 The activation states of tumor-resident type 2 dendritic cells impact the strength of ovarian cancer immune responses","authors":"Fiona Chatterjee, Vincent Butty, Emi A Lutz, K Dane Wittrup, Stefani Spranger","doi":"10.1136/jitc-2023-sitc2023.0947","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0947","url":null,"abstract":"<h3>Background</h3> Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States.<sup>1</sup> To date, checkpoint blockade therapy (CBT) has failed to be effective in ovarian cancer.<sup>2</sup> CBT efficacy typically requires a strong pre-existing tumor-specific T cell response.<sup>3 4</sup> However, analysis of patient data indicates that tumor-infiltrating T cells in ovarian cancer are often poorly activated.<sup>5</sup> Understanding the mechanisms governing this poor T cell activation could lead to novel ovarian cancer therapeutics. <h3>Methods</h3> We utilized a transplantable, syngeneic, murine ovarian cancer cell line driven by Ccne1<sup>OE</sup> p53<sup>-/-R172H</sup> Ak2<sup>OE</sup> and Kras<sup>G12V</sup> (CPAK).<sup>6</sup> CPAK tumor cells were implanted intraperitoneally to model metastatic ovarian cancer or subcutaneously to model productive systemic immunity. CPAK cells were engineered to express the model CD8<sup>+</sup> T cell antigen SIY to enable studies of tumor-specific T cells. CBT consisted of αCTLA-4 and αPD-L1 therapy. Immune cells were profiled using flow cytometry and single-cell RNA sequencing (scRNA-seq). <h3>Results</h3> Intraperitoneal (IP) CPAK-SIY tumors were unresponsive to CBT. However, mice bearing subcutaneous (SQ) CPAK-SIY tumors treated with CBT displayed delayed tumor growth compared to control animals. Flow cytometric analysis of tumor-infiltrating CD8<sup>+</sup> T cells illustrated that while tumor-reactive T cells were activated in IP and SQ tumors, tumor-reactive T cells in IP tumors failed to upregulate high levels of effector molecules such as granzyme B. Unbiased analysis of dendritic cells (DCs) within IP tumors using scRNA-seq revealed a population of DCs that expressed CD103 and CD11b and a type 2 conventional DC (cDC2) gene signature. Gene signature analysis indicated these CD103<sup>+</sup> CD11b<sup>+</sup> double-positive DCs were a suppressive state of cDC2s induced by TGFb that reside in gut tissue and induce Tregs.<sup>7</sup> Analysis of tumor-infiltrating DCs in IP tumors revealed that the proportion of double-positive DCs increased during tumor growth while the proportion of cDC2s decreased. We hypothesized that the polarization of cDC2s away from these suppressive double-positive DCs may improve ovarian cancer immunity. Previous work from our group demonstrated that cDC2s acquire an activation state characterized by an interferon-stimulated gene signature (ISG<sup>+</sup> DCs) upon exposure to interferon-beta (IFNβ) and that ISG<sup>+</sup> DCs are potent activators of CD8<sup>+</sup> T cells.<sup>8</sup> In our ovarian cancer model, addition of IFNβ to IP tumors enhanced tumor-specific T cell responses. <h3>Conclusions</h3> Metastatic ovarian tumors are refractory to CBT and are infiltrated by poorly activated CD8<sup>+</sup> T cells. Our results suggest that the poor T cell activation is a consequence of the cDC2 activation state within the tumor","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":"135162561","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
1227 Oral inulin gel formulation modulates the gut microbiome and improves the safety and efficacy of immune checkpoint blockers 1227口服菊粉凝胶制剂可调节肠道微生物群,提高免疫检查点阻断剂的安全性和有效性
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.1227
Jin Xu, Kai Han, Xuehui Huang, James J Moon

Background

The dysregulated gut microbiota found in cancer patients is emerging as the new therapeutic target. Here, we have engineered inulin – a widely consumed dietary fiber – into an oral gel formulation to modulate the gut microbiota and the host immune responses. We show that inulin gel improves the safety and anti-tumor efficacy of immune checkpoint blockers (ICBs) in various murine tumor models.

Methods

We have optimized the scale-up production of inulin gel. In tumor-bearing mice, inulin gel was orally administered starting day 7 after tumor inoculation, while anti-PD-1 was intraperitoneally injected from day 10. The gut microbiota profile in fecal samples was examined by 16s rRNA gene sequencing, and the metabolites in feces and serum were tested by ion- or liquid-chromatography. Tumor-infiltrating lymphocytes were measured via flow cytometry. In addition, inulin gel was tested on the mouse ICBs-associated colitis model, where 3% dextran sulphate sodium (DSS) was supplied in the drinking water.

Results

After oral gavage in mice, inulin gel formulation was retained longer in the colon, thus increasing the cumulative inulin exposure and fermentation in the colon. Consequently, inulin gel increased the frequencies of Akkermensia as well as other commensal microbes known to modulate the systemic and colonic immune responses. Oral administration of inulin gel markedly augmented the antitumor efficacy of anti-PD-1 and anti-CTLA-4 ICBs in multiple tumor models, including CT26 colon carcinoma, B16F10 melanoma, and DSS-accelerated colon tumour model in CDX2-cre NLS-APCfl/fl mice. Notably, colitis is one of the most frequently observed immune-related adverse events (irAEs) associated with ICB therapy in the clinic. Our results showed that oral administration of inulin gel ameliorated DSS-induced, ICBs-exacerbated colitis, suggesting that inulin gel can also improve the safety profiles of ICB therapy. Metabolomics analysis revealed that inulin gel plus anti-PD-1 increased the concentrations of short-chain-fatty-acids, which promoted the differentiation of stem-like Tcf1+PD-1+CD8+ T cells, conferring long-lasting protection against tumor re-growth. Meanwhile, inulin gel plus anti-PD-1 decreased the concentrations of ATP and L-Phenylalanine that could exacerbate colitis. Toward the goal of initiating a human clinical study, we have optimized the scale-up manufacturing of inulin gel.

Conclusions

Orally administered inulin gel formulation normalizes the dysregulated gut microbiome, improves the host immune responses, and decreases the ICB-associated colitis. Based on this work, we are initiating a Phase I study to examine the impact of inulin gel consumption in healthy volunteers.

Acknowledgements

This work was supported by NIH (R01AI127070, R01CA210273, U01CA210152, R01DK108901, R01DE026728, R01DE030691, R01DE031951) and the University of Michigan Rogel Cancer
背景癌症患者肠道菌群失调正成为新的治疗靶点。在这里,我们将菊粉——一种广泛食用的膳食纤维——改造成一种口服凝胶配方,以调节肠道微生物群和宿主的免疫反应。我们发现菊糖凝胶在多种小鼠肿瘤模型中提高了免疫检查点阻断剂(ICBs)的安全性和抗肿瘤效果。方法对菊粉凝胶的规模化生产工艺进行优化。荷瘤小鼠在肿瘤接种后第7天口服菊糖凝胶,从第10天开始腹腔注射抗pd -1。采用16s rRNA基因测序检测粪便样品中的肠道微生物群,采用离子或液相色谱法检测粪便和血清中的代谢物。流式细胞术检测肿瘤浸润淋巴细胞。此外,在小鼠icbs相关性结肠炎模型上对菊糖凝胶进行了测试,其中在饮用水中提供3%葡聚糖硫酸钠(DSS)。结果小鼠灌胃后,菊粉凝胶制剂在结肠内滞留时间延长,从而增加了菊粉在结肠内的累积暴露和发酵。因此,菊粉凝胶增加了Akkermensia以及其他已知调节系统和结肠免疫反应的共生微生物的频率。在CDX2-cre NLS-APCfl/fl小鼠的多种肿瘤模型中,口服菊粉凝胶可显著增强抗pd -1和抗ctla -4 ICBs的抗肿瘤功效,包括CT26结肠癌、B16F10黑色素瘤和dss加速结肠癌模型。值得注意的是,结肠炎是临床中与ICB治疗相关的最常见的免疫相关不良事件(irAEs)之一。我们的研究结果表明,口服菊糖凝胶可以改善dss诱导的、icbs加重的结肠炎,这表明菊糖凝胶也可以提高ICB治疗的安全性。代谢组学分析显示,菊粉凝胶加抗PD-1增加了短链脂肪酸的浓度,促进了干细胞样Tcf1+PD-1+CD8+ T细胞的分化,赋予肿瘤再生长持久的保护作用。同时,菊粉凝胶加抗pd -1可降低可加重结肠炎的ATP和l -苯丙氨酸的浓度。针对启动人体临床研究的目标,我们优化了菊粉凝胶的规模化生产。结论口服菊糖凝胶制剂可使失调的肠道菌群恢复正常,改善宿主免疫反应,减少icb相关性结肠炎。基于这项工作,我们正在启动一项I期研究,以检查菊糖凝胶对健康志愿者的影响。本工作得到美国国立卫生研究院(R01AI127070, R01CA210273, U01CA210152, R01DK108901, R01DE026728, R01DE030691, R01DE031951)和密歇根大学Rogel癌症中心支持基金(P30CA46592)的支持。密歇根大学安娜堡分校是AAALAC国际认可的机构,所有在动物身上进行的工作都符合并得到了机构动物护理和使用委员会(IACUC)的批准。
{"title":"1227 Oral inulin gel formulation modulates the gut microbiome and improves the safety and efficacy of immune checkpoint blockers","authors":"Jin Xu, Kai Han, Xuehui Huang, James J Moon","doi":"10.1136/jitc-2023-sitc2023.1227","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.1227","url":null,"abstract":"<h3>Background</h3> The dysregulated gut microbiota found in cancer patients is emerging as the new therapeutic target. Here, we have engineered inulin – a widely consumed dietary fiber – into an oral gel formulation to modulate the gut microbiota and the host immune responses. We show that inulin gel improves the safety and anti-tumor efficacy of immune checkpoint blockers (ICBs) in various murine tumor models. <h3>Methods</h3> We have optimized the scale-up production of inulin gel. In tumor-bearing mice, inulin gel was orally administered starting day 7 after tumor inoculation, while anti-PD-1 was intraperitoneally injected from day 10. The gut microbiota profile in fecal samples was examined by 16s rRNA gene sequencing, and the metabolites in feces and serum were tested by ion- or liquid-chromatography. Tumor-infiltrating lymphocytes were measured via flow cytometry. In addition, inulin gel was tested on the mouse ICBs-associated colitis model, where 3% dextran sulphate sodium (DSS) was supplied in the drinking water. <h3>Results</h3> After oral gavage in mice, inulin gel formulation was retained longer in the colon, thus increasing the cumulative inulin exposure and fermentation in the colon. Consequently, inulin gel increased the frequencies of <i>Akkermensia</i> as well as other commensal microbes known to modulate the systemic and colonic immune responses. Oral administration of inulin gel markedly augmented the antitumor efficacy of anti-PD-1 and anti-CTLA-4 ICBs in multiple tumor models, including CT26 colon carcinoma, B16F10 melanoma, and DSS-accelerated colon tumour model in <i>CDX2-cre NLS-APC</i>fl/fl mice. Notably, colitis is one of the most frequently observed immune-related adverse events (irAEs) associated with ICB therapy in the clinic. Our results showed that oral administration of inulin gel ameliorated DSS-induced, ICBs-exacerbated colitis, suggesting that inulin gel can also improve the safety profiles of ICB therapy. Metabolomics analysis revealed that inulin gel plus anti-PD-1 increased the concentrations of short-chain-fatty-acids, which promoted the differentiation of stem-like Tcf1<sup>+</sup>PD-1<sup>+</sup>CD8<sup>+</sup> T cells, conferring long-lasting protection against tumor re-growth. Meanwhile, inulin gel plus anti-PD-1 decreased the concentrations of ATP and L-Phenylalanine that could exacerbate colitis. Toward the goal of initiating a human clinical study, we have optimized the scale-up manufacturing of inulin gel. <h3>Conclusions</h3> Orally administered inulin gel formulation normalizes the dysregulated gut microbiome, improves the host immune responses, and decreases the ICB-associated colitis. Based on this work, we are initiating a Phase I study to examine the impact of inulin gel consumption in healthy volunteers. <h3>Acknowledgements</h3> This work was supported by NIH (R01AI127070, R01CA210273, U01CA210152, R01DK108901, R01DE026728, R01DE030691, R01DE031951) and the University of Michigan Rogel Cancer","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":"135162804","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
330 Combination therapy with stem cell derived natural killer cells and monoclonal antibodies leads to potent ADCC through engagement of endogenouse CD16 330干细胞来源的自然杀伤细胞和单克隆抗体联合治疗通过内源性CD16导致有效的ADCC
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0330
Anna-Maria Georgoudaki, Nina Lamers-Kok, Amanda van Vliet, Didem Özkazanc, Denise Vodegel, Daniëlle Steenmans, Monica Raimo, Adil Duru, Jan Spanholtz

Background

Glycostem’s ex vivo expansion and differentiation method in a fully closed automated manufacturing platform (uNiK™), generates GTA002 (oNKord®), an ‘off-the-shelf’ allogeneic cryopreserved NK cell product derived from umbilical cord blood CD34+ hematopoietic stem cell progenitor cells, which is currently being tested in a Phase I/II clinical trial in AML, WiNK (NCT04632316). Safety and tolerability of a non-cryopreserved predecessor was demonstrated in an earlier Phase I trial in AML (Dolstra et al. 2017). One of the important outcomes of this study was the notable increase in the CD16 expression of infused NK cells. Thus, we next exploited the potential of further enhancing and focusing cryopreserved NK cell anti-tumor responses in an antigen (Ag)-specific manner via antibody-dependent cellular cytotoxicity (ADCC) in pre-clinical models of hematological and solid malignancies.

Methods

Similar to its predecessor non-cryopreserved NK cells, GTA002 significantly upregulated CD16 expression in vivo in immunodeficient NCG mice. This spurred the optimization of the culture process to upregulate CD16 expression in order to study the ADCC potential of GTA002 in vitro. ADCC was assessed against CD19+ and HER2+ targets at low effector-to-target (E:T) ratios by end-point flow cytometry assays as well as impedance- and live imaging- (2D & 3D) based real time analysis. Next, we engineered CD16-NK cells by introduction of a lentiviral transduction step to the uNiK™ platform, to evaluate the effect of CD19-targeted ADCC of NK cells expressing engineered or endogenous CD16. Furthermore, expression of important activating and inhibitory receptors and intracellular levels of TNF, IFNγ, perforin and granzyme B were measured by flow cytometry to investigate their role in efficient cytotoxicity of GTA002 cells. We detected simultaneous tumor targeting by GTA002, both via preserved innate NK cell responses as well as Ag-specific targeting via ADCC at low E:T ratios.

Results

Moreover, GTA002 cells were tested for their ability to mediate killing of an ovarian cancer cell line in the presence of an Fc-active monoclonal antibody (mAb) targeting a tumor associated antigen expressed by SKOV-3 cells. Impedance-based cytotoxicity assays revealed that GTA002 exerted potent ADCC upon CD16 engagement. Addition of cytokine support further enhanced both baseline cytotoxicity as well as ADCC, leading to complete eradication of the SKOV-3 tumor cells.

Conclusions

Overall, the enhancement of the inherent potency of GTA002 by harnessing ADCC through combination therapy with mAbs achieved efficient Ag-specific responses demonstrating the great potential of multimodal targeting against a variety of challenging cancers using a highly safe ‘off-the-shelf’ NK cell-based cellular therapeutic.
糖干公司在全封闭自动化制造平台(uNiK™)中的体外扩增和分化方法产生了GTA002 (oNKord®),这是一种“现成的”同种异体低温保存NK细胞产品,来源于脐带血CD34+造血干细胞祖细胞,目前正在AML的I/II期临床试验中进行测试,WiNK (NCT04632316)。在AML的早期I期试验中证实了非冷冻保存前体的安全性和耐受性(Dolstra et al. 2017)。本研究的一个重要结果是灌注NK细胞的CD16表达显著增加。因此,我们接下来利用抗体依赖性细胞毒性(ADCC)在血液学和实体恶性肿瘤的临床前模型中,以抗原(Ag)特异性的方式进一步增强和聚焦冷冻保存的NK细胞抗肿瘤反应。方法GTA002与前代非冷冻NK细胞相似,在免疫缺陷NCG小鼠体内显著上调CD16的表达。这促使我们优化培养过程,上调CD16的表达,以研究GTA002在体外的ADCC潜力。通过终点流式细胞术分析以及阻抗和实时成像,以低效靶比(E:T)评估ADCC对CD19+和HER2+靶标的作用(2D &基于3D的实时分析。接下来,我们通过将慢病毒转导步骤引入uNiK™平台来设计CD16-NK细胞,以评估表达工程化或内源性CD16的NK细胞的cd19靶向ADCC的效果。此外,通过流式细胞术检测重要的激活和抑制受体的表达以及细胞内TNF、IFNγ、穿孔素和颗粒酶B的水平,以研究它们在GTA002细胞有效细胞毒性中的作用。我们检测到GTA002同时靶向肿瘤,既通过保存的天然NK细胞反应,也通过低E:T比的ADCC靶向ag。此外,我们还测试了GTA002细胞在靶向SKOV-3细胞表达的肿瘤相关抗原的fc活性单克隆抗体(mAb)存在下介导卵巢癌细胞系杀伤的能力。基于阻抗的细胞毒性实验显示,GTA002对CD16结合具有有效的ADCC作用。细胞因子支持的加入进一步增强了基线细胞毒性和ADCC,从而完全根除SKOV-3肿瘤细胞。总的来说,利用ADCC与单克隆抗体联合治疗增强GTA002的固有效力,实现了高效的ag特异性应答,表明使用高度安全的“现成”NK细胞为基础的细胞治疗方法,多模式靶向治疗各种具有挑战性的癌症的巨大潜力。
{"title":"330 Combination therapy with stem cell derived natural killer cells and monoclonal antibodies leads to potent ADCC through engagement of endogenouse CD16","authors":"Anna-Maria Georgoudaki, Nina Lamers-Kok, Amanda van Vliet, Didem Özkazanc, Denise Vodegel, Daniëlle Steenmans, Monica Raimo, Adil Duru, Jan Spanholtz","doi":"10.1136/jitc-2023-sitc2023.0330","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0330","url":null,"abstract":"<h3>Background</h3> Glycostem’s ex vivo expansion and differentiation method in a fully closed automated manufacturing platform (uNiK™), generates GTA002 (oNKord®), an ‘off-the-shelf’ allogeneic cryopreserved NK cell product derived from umbilical cord blood CD34+ hematopoietic stem cell progenitor cells, which is currently being tested in a Phase I/II clinical trial in AML, WiNK (NCT04632316). Safety and tolerability of a non-cryopreserved predecessor was demonstrated in an earlier Phase I trial in AML (Dolstra et al. 2017). One of the important outcomes of this study was the notable increase in the CD16 expression of infused NK cells. Thus, we next exploited the potential of further enhancing and focusing cryopreserved NK cell anti-tumor responses in an antigen (Ag)-specific manner via antibody-dependent cellular cytotoxicity (ADCC) in pre-clinical models of hematological and solid malignancies. <h3>Methods</h3> Similar to its predecessor non-cryopreserved NK cells, GTA002 significantly upregulated CD16 expression in vivo in immunodeficient NCG mice. This spurred the optimization of the culture process to upregulate CD16 expression in order to study the ADCC potential of GTA002 in vitro. ADCC was assessed against CD19<sup>+</sup> and HER2<sup>+</sup> targets at low effector-to-target (E:T) ratios by end-point flow cytometry assays as well as impedance- and live imaging- (2D &amp; 3D) based real time analysis. Next, we engineered CD16-NK cells by introduction of a lentiviral transduction step to the uNiK™ platform, to evaluate the effect of CD19-targeted ADCC of NK cells expressing engineered or endogenous CD16. Furthermore, expression of important activating and inhibitory receptors and intracellular levels of TNF, IFNγ, perforin and granzyme B were measured by flow cytometry to investigate their role in efficient cytotoxicity of GTA002 cells. We detected simultaneous tumor targeting by GTA002, both via preserved innate NK cell responses as well as Ag-specific targeting via ADCC at low E:T ratios. <h3>Results</h3> Moreover, GTA002 cells were tested for their ability to mediate killing of an ovarian cancer cell line in the presence of an Fc-active monoclonal antibody (mAb) targeting a tumor associated antigen expressed by SKOV-3 cells. Impedance-based cytotoxicity assays revealed that GTA002 exerted potent ADCC upon CD16 engagement. Addition of cytokine support further enhanced both baseline cytotoxicity as well as ADCC, leading to complete eradication of the SKOV-3 tumor cells. <h3>Conclusions</h3> Overall, the enhancement of the inherent potency of GTA002 by harnessing ADCC through combination therapy with mAbs achieved efficient Ag-specific responses demonstrating the great potential of multimodal targeting against a variety of challenging cancers using a highly safe ‘off-the-shelf’ NK cell-based cellular therapeutic.","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":"135162991","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
403 Tumor-infiltrating lymphocytes (TIL) with inducible and membrane-bound IL-12 exhibit superior antitumor activityin vitro 具有诱导和膜结合IL-12的肿瘤浸润淋巴细胞(TIL)在体外表现出优越的抗肿瘤活性
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0403
Yongliang Zhang, Nathan Gilbert, Patrick Innamarato, Judy Fang, Hequn Yin

Background

TIL cell therapy has shown clinical benefit for patients with solid tumors.1 2 However, an immunosuppressive tumor microenvironment (TME) may abrogate the full potential of TIL cell therapy.3 The proinflammatory cytokine IL-12, known for its capability to increase IFN-γ production and promote type 1 immune responses, reshapes the TME and has a potential to augment antitumor activity. Here, we report genetic engineering of TIL with an inducible and membrane-bound IL-12, incorporated into a 22-day manufacturing process, which showed superior cytotoxic function in vitro.

Methods

Tumor tissue from several solid tumor types, including non-small cell lung, breast, head and neck, and ovarian cancers, was fragmented and cultured, followed by transduction with lentivirus containing a gene encoding membrane-bound IL-12 via an NFAT-promoter (TeIL-12), and a rapid expansion protocol (REP). Expression, biological function, and shedding of IL-12 molecule were assessed in vitro. Immune phenotype and in vitro cytotoxic activity of TeIL-12 gene-engineered TIL were examined in various assays.

Results

Lentiviral gene transfer resulted in TIL expressing IL-12 on their surface via a membrane anchor. This modification potentiated IL-12 receptor downstream signaling activation in a contact-dependent manner in assays using HEK-IL-12-Blue reporter cells. TeIL-12-TIL exhibited increased IFN-γ production and superior cytotoxicity in a KILR assay. An xCELLigence-based cytotoxicity assay further confirmed increased killing with TIL that were unstimulated or stimulated. Moreover, phenotype profiling revealed TeIL-12-TIL were less differentiated, with reduced expression of immune-inhibitory receptors and increased production of cytotoxic molecules associated with antitumor activity. IL-12 shedding in the co-culture supernatant was minimal.

Conclusions

The enhanced in vitro killing activity, combined with a less-differentiated phenotype of TIL with inducible and membrane-bound IL-12 suggests a potential for increased clinical efficacy. Further, minimal IL-12 shedding supports reduced potential for IL-12-associated toxicity.4 Together, these data support investigation of TeIL-12 in vivo and subsequent clinical development.

Acknowledgements

Editorial support was provided by Amanda Kelly (Iovance Biotherapeutics).

References

Chesney J, Lewis KD, Kluger H, et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C- 144–01 study. Journal for ImmunoTherapy of Cancer 2022;10:e005755. Schoenfeld AJ, Lee S, Paz-Ares L, et al. First phase 2 results of autologous tumor-infiltrating lymphocyte (TIL; LN-145) monotherapy in p
TIL细胞治疗已显示出对实体瘤患者的临床益处。然而,免疫抑制肿瘤微环境(TME)可能会使TIL细胞治疗的全部潜力失效促炎细胞因子IL-12以其增加IFN-γ产生和促进1型免疫反应的能力而闻名,重塑TME并具有增强抗肿瘤活性的潜力。在这里,我们报道了用一种可诱导和膜结合的IL-12进行TIL基因工程,并将其纳入22天的制造过程中,在体外显示出优越的细胞毒性功能。方法将非小细胞肺癌、乳腺癌、头颈癌和卵巢癌等多种类型实体瘤的肿瘤组织切片培养,然后用含有编码膜结合IL-12基因的慢病毒通过nfat启动子(TeIL-12)和快速扩增方案(REP)进行转导。体外观察IL-12分子的表达、生物学功能和脱落情况。采用多种方法检测TeIL-12基因工程TIL的免疫表型和体外细胞毒活性。结果慢病毒基因转移导致TIL通过膜锚在其表面表达IL-12。在使用HEK-IL-12-Blue报告细胞的实验中,这种修饰以接触依赖的方式增强了IL-12受体下游信号的激活。在KILR试验中,TeIL-12-TIL表现出增加的IFN-γ产生和优越的细胞毒性。基于xcelligence的细胞毒性试验进一步证实,未刺激或刺激的TIL杀伤增加。此外,表型分析显示TeIL-12-TIL分化程度较低,免疫抑制受体的表达减少,与抗肿瘤活性相关的细胞毒性分子的产生增加。共培养上清中IL-12的脱落极少。结论增强的体外杀伤活性,结合TIL与诱导和膜结合IL-12的低分化表型,表明有可能提高临床疗效。此外,少量的IL-12脱落支持降低IL-12相关毒性的可能性总之,这些数据支持TeIL-12的体内研究和随后的临床开发。编辑支持由Amanda Kelly (Iovance biotheraptics)提供。Chesney J, Lewis KD, Kluger H,等。lifileucel是一种一次性自体肿瘤浸润淋巴细胞(TIL)细胞疗法,用于免疫检查点抑制剂和靶向治疗进展后的晚期黑色素瘤患者的疗效和安全性:C- 144-01研究连续队列的汇总分析肿瘤免疫治疗杂志2022;10:e005755。肖恩菲尔德,李S, Paz-Ares L,等。自体肿瘤浸润淋巴细胞(TIL;LN-145单药治疗晚期免疫检查点抑制剂治疗的非小细胞肺癌(NSCLC)患者2021年SITC年会。张建军,张建军,张建军,等。肿瘤浸润淋巴细胞的过继细胞治疗:最新进展、挑战和未来方向。专家意见生物学报,2022;1-15。张丽,Morgan RA, Beane JD,等。肿瘤浸润淋巴细胞基因工程编码白介素-12用于转移性黑色素瘤的免疫治疗。中华肿瘤杂志,2015;21(2):588 - 588。伦理批准本研究由各试验点的机构审查委员会批准,并按照《赫尔辛基宣言》和国际协调会议的良好临床实践指南进行。所有患者均提供书面知情同意书。
{"title":"403 Tumor-infiltrating lymphocytes (TIL) with inducible and membrane-bound IL-12 exhibit superior antitumor activity<i>in vitro</i>","authors":"Yongliang Zhang, Nathan Gilbert, Patrick Innamarato, Judy Fang, Hequn Yin","doi":"10.1136/jitc-2023-sitc2023.0403","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0403","url":null,"abstract":"<h3>Background</h3> TIL cell therapy has shown clinical benefit for patients with solid tumors.<sup>1 2</sup> However, an immunosuppressive tumor microenvironment (TME) may abrogate the full potential of TIL cell therapy.<sup>3</sup> The proinflammatory cytokine IL-12, known for its capability to increase IFN-γ production and promote type 1 immune responses, reshapes the TME and has a potential to augment antitumor activity. Here, we report genetic engineering of TIL with an inducible and membrane-bound IL-12, incorporated into a 22-day manufacturing process, which showed superior cytotoxic function <i>in vitro</i>. <h3>Methods</h3> Tumor tissue from several solid tumor types, including non-small cell lung, breast, head and neck, and ovarian cancers, was fragmented and cultured, followed by transduction with lentivirus containing a gene encoding membrane-bound IL-12 via an NFAT-promoter (TeIL-12), and a rapid expansion protocol (REP). Expression, biological function, and shedding of IL-12 molecule were assessed <i>in vitro</i>. Immune phenotype and <i>in vitro</i> cytotoxic activity of TeIL-12 gene-engineered TIL were examined in various assays. <h3>Results</h3> Lentiviral gene transfer resulted in TIL expressing IL-12 on their surface via a membrane anchor. This modification potentiated IL-12 receptor downstream signaling activation in a contact-dependent manner in assays using HEK-IL-12-Blue reporter cells. TeIL-12-TIL exhibited increased IFN-γ production and superior cytotoxicity in a KILR assay. An xCELLigence-based cytotoxicity assay further confirmed increased killing with TIL that were unstimulated or stimulated. Moreover, phenotype profiling revealed TeIL-12-TIL were less differentiated, with reduced expression of immune-inhibitory receptors and increased production of cytotoxic molecules associated with antitumor activity. IL-12 shedding in the co-culture supernatant was minimal. <h3>Conclusions</h3> The enhanced <i>in vitro</i> killing activity, combined with a less-differentiated phenotype of TIL with inducible and membrane-bound IL-12 suggests a potential for increased clinical efficacy. Further, minimal IL-12 shedding supports reduced potential for IL-12-associated toxicity.<sup>4</sup> Together, these data support investigation of TeIL-12 <i>in vivo</i> and subsequent clinical development. <h3>Acknowledgements</h3> Editorial support was provided by Amanda Kelly (Iovance Biotherapeutics). <h3>References</h3> Chesney J, Lewis KD, Kluger H, <i>et al</i>. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C- 144–01 study. <i>Journal for ImmunoTherapy of Cancer</i> 2022;<b>10</b>:e005755. Schoenfeld AJ, Lee S, Paz-Ares L, <i>et al.</i> First phase 2 results of autologous tumor-infiltrating lymphocyte (TIL; LN-145) monotherapy in p","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":"135163179","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
222 The chick embryo chorioallantoic membrane (CAM) as a platform for assessing the in vivo efficacy of chimeric antigen receptor (CAR) T cell therapy in solid tumors 鸡胚绒毛尿囊膜(CAM)作为评价嵌合抗原受体(CAR) T细胞治疗实体瘤体内疗效的平台
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0222
Allison Nipper, Emilie AK Warren, Gabrielle Wells, Caroline Porter, Mariana Villanueva, Hsuan-Chen Liu, Hugo Villanueva, Masataka Suzuki, Andrew Sikora

Background

While chimeric antigen receptor T cell (CAR T) treatment has been efficacious in blood cancers, mirroring this success in solid tumors, like head and neck squamous cell carcinoma (HNSCC) has proven difficult due to the challenge of identifying and validating suitable target antigens. To rapidly model CAR-T treatment against HNSCC antigens, we have developed a robust, scalable model using fertilized chicken eggs to facilitate CAR-T screening against vascularized solid tumors. The fertilized chicken egg inner shell membrane, the chorioallantoic membrane (CAM), is a highly vascularized membrane which facilitates nutrient delivery and oxygen exchange for growth of the developing chick. The nutrient transfer capabilities of the CAM can be exploited to drive the formation of vascularized solid tumors which can be targeted by CAR T within one week of engraftment. In contrast to murine systems which can be costly, labor intensive, or use murine cells, this system quickly models the development of human cancers expressing human epitopes treated with human cell therapies.

Methods

HER2+ (FaDu and SCC-47) and HER2- (MDA-MB-468) cell lines with luciferase reporters were engrafted onto the CAM. Tumor growth was monitored by IVIS imaging of luciferase activity of viable tumor cells. Engrafted cells grew for three days during which tumors establish and incorporate into the CAM. Established tumors were treated with second generation HER2-directed CAR T with a CD28 costimulatory endodomain or an expanded T cell control which was not transduced with CARs. Viability of tumor cells was monitored through quantification of luciferase activity from engrafted cells before and following four days of CAR-T treatment. Histology was used to visualize tumor structure on the CAM and CAR-T infiltration of tumor tissue. Staining was scored by percent positive area and intensity using QuPath software.

Results

Luciferase activity of viable tumor cells was significantly reduced in CAR-T treated FaDu tumors (p<0.0001). Following tumor harvest, we observed a significant reduction in Ki67 staining for CAR-T treated tumors relative to T cell treatment alone for both SCC-47 and FaDu tumors (p=0.0121 and p=0.0176 respectively). We also observed CAR-T persistence in tissue and infiltration of tumors.

Conclusions

The CAM system supports the growth of solid tumors which can be targeted by immune cell infiltrates. These data support continued development of the chick CAM model as a candidate in vivo system for rapid, scalable screening of CAR T efficacy against human solid tumors.
虽然嵌合抗原受体T细胞(CAR - T)治疗在血癌中是有效的,但在实体肿瘤(如头颈部鳞状细胞癌(HNSCC))中复制这种成功被证明是困难的,因为识别和验证合适的靶抗原是一项挑战。为了快速模拟针对HNSCC抗原的CAR-T治疗,我们开发了一个强大的、可扩展的模型,使用受精卵来促进针对血管化实体瘤的CAR-T筛查。受精卵的内壳膜,即绒毛膜-尿囊膜(CAM),是一种高度血管化的膜,有利于发育中的小鸡生长所需的营养物质输送和氧气交换。CAM的营养转移能力可以被利用来驱动血管化实体肿瘤的形成,CAR - T可以在植入一周内靶向。与昂贵、劳动密集或使用小鼠细胞的小鼠系统相比,该系统可以快速模拟人类癌症的发展,表达人类细胞疗法治疗的人类表位。方法将带有荧光素酶报告细胞的HER2+ (FaDu和SCC-47)和HER2- (MDA-MB-468)细胞株移植到CAM上。通过活体肿瘤细胞荧光素酶活性的IVIS成像监测肿瘤生长。移植细胞生长三天,在此期间肿瘤形成并融入CAM。已确定的肿瘤用第二代her2定向CAR - T治疗,该CAR - T带有CD28共刺激内域或扩增的T细胞对照,不经CAR转导。在CAR-T治疗前后4天,通过对移植细胞荧光素酶活性的量化来监测肿瘤细胞的活力。用组织学方法观察肿瘤组织的CAM和CAR-T浸润情况。采用QuPath软件对染色进行阳性面积百分比和染色强度评分。结果CAR-T治疗FaDu肿瘤后,活细胞荧光素酶活性显著降低(p < 0.01;0.0001)。肿瘤收获后,我们观察到CAR-T治疗肿瘤的Ki67染色明显降低,相对于单独T细胞治疗SCC-47和FaDu肿瘤(p=0.0121和p=0.0176分别)。我们还观察到CAR-T在组织中的持久性和肿瘤的浸润。结论CAM系统支持实体瘤的生长,可被免疫细胞浸润靶向。这些数据支持继续发展小鸡CAM模型作为候选体内系统,用于快速、可扩展地筛选CAR - T对人类实体肿瘤的疗效。
{"title":"222 The chick embryo chorioallantoic membrane (CAM) as a platform for assessing the in vivo efficacy of chimeric antigen receptor (CAR) T cell therapy in solid tumors","authors":"Allison Nipper, Emilie AK Warren, Gabrielle Wells, Caroline Porter, Mariana Villanueva, Hsuan-Chen Liu, Hugo Villanueva, Masataka Suzuki, Andrew Sikora","doi":"10.1136/jitc-2023-sitc2023.0222","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0222","url":null,"abstract":"<h3>Background</h3> While chimeric antigen receptor T cell (CAR T) treatment has been efficacious in blood cancers, mirroring this success in solid tumors, like head and neck squamous cell carcinoma (HNSCC) has proven difficult due to the challenge of identifying and validating suitable target antigens. To rapidly model CAR-T treatment against HNSCC antigens, we have developed a robust, scalable model using fertilized chicken eggs to facilitate CAR-T screening against vascularized solid tumors. The fertilized chicken egg inner shell membrane, the chorioallantoic membrane (CAM), is a highly vascularized membrane which facilitates nutrient delivery and oxygen exchange for growth of the developing chick. The nutrient transfer capabilities of the CAM can be exploited to drive the formation of vascularized solid tumors which can be targeted by CAR T within one week of engraftment. In contrast to murine systems which can be costly, labor intensive, or use murine cells, this system quickly models the development of human cancers expressing human epitopes treated with human cell therapies. <h3>Methods</h3> HER2+ (FaDu and SCC-47) and HER2- (MDA-MB-468) cell lines with luciferase reporters were engrafted onto the CAM. Tumor growth was monitored by IVIS imaging of luciferase activity of viable tumor cells. Engrafted cells grew for three days during which tumors establish and incorporate into the CAM. Established tumors were treated with second generation HER2-directed CAR T with a CD28 costimulatory endodomain or an expanded T cell control which was not transduced with CARs. Viability of tumor cells was monitored through quantification of luciferase activity from engrafted cells before and following four days of CAR-T treatment. Histology was used to visualize tumor structure on the CAM and CAR-T infiltration of tumor tissue. Staining was scored by percent positive area and intensity using QuPath software. <h3>Results</h3> Luciferase activity of viable tumor cells was significantly reduced in CAR-T treated FaDu tumors (p<0.0001). Following tumor harvest, we observed a significant reduction in Ki67 staining for CAR-T treated tumors relative to T cell treatment alone for both SCC-47 and FaDu tumors (p=0.0121 and p=0.0176 respectively). We also observed CAR-T persistence in tissue and infiltration of tumors. <h3>Conclusions</h3> The CAM system supports the growth of solid tumors which can be targeted by immune cell infiltrates. These data support continued development of the chick CAM model as a candidate in vivo system for rapid, scalable screening of CAR T efficacy against human solid tumors.","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":"135163181","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
297 Improvement of CAR T-cell performance by simultaneous downregulation of multiple co-inhibitory receptors 297通过同时下调多个共抑制受体改善CAR - t细胞性能
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0297
Matteo Rossi, Fanny Huberty, Thuy Nguyen, Jerome Marijsse, Celine Jacques-Hespel, Eytan Breman

Background

Co-inhibitory receptors, such as PD-1 and LAG-3, have a crucial role in regulating T-cell activity, as their expression on the cell surface upon chronic T-cell activation is associated with T-cell exhaustion. Immunotherapies directed against co-inhibitory receptors exhibited unprecedented efficacy in several cancer indications. However, many patients do not respond to these therapies and some tumor types remain largely refractory. A key determinant of CAR T-cell failure against solid tumors is T-cell exhaustion induced by checkpoint inhibitors on cancer cells. The coordinated targeting of different co-inhibitory receptors may help overcome these hurdles. Here we show that the simultaneous downregulation of co-inhibitory receptors PD-1, LAG-3, TIM-3 and TIGIT endows CAR T-cells with superior efficacy.

Methods

A microRNA (miRNA)-based short hairpin RNA (shRNA) platform was developed, to allow for the tunable modulation of multiple target genes simultaneously. The platform was equipped with shRNA-derived guide sequences (shGuides) targeting PD-1, LAG-3, TIM-3 and TIGIT, and combined with an anti-CD19 CAR. The shRNA platform-equipped CAR T-cells were challenged with target cancer cells expressing the co-inhibitory receptors’ ligands. The impact of the simultaneous downregulation of the four co-inhibitory receptors was assessed in vitro by monitoring T-cell activation (via surface activation markers and cytokine secretion), killing activity, and persistence (via repeated challenges with the target cells).

Results

The miRNA-derived shRNA platform targeting PD-1, LAG-3, TIM-3 and TIGIT led to a significant downregulation of the four co-inhibitory receptors in the CAR T-cells. Upon activation by the target cancer cells, the simultaneous knock-down of the four genes enhanced cytokine secretion compared to CAR T-cells not engineered with the shRNA platform. Moreover, the CAR T-cells carrying the quadruple knock-down showed superior killing ability, increased persistence and prolonged activity when repeatedly challenged in sequential cycles with the target cancer cells.

Conclusions

These data validate our technology for the effective introduction of multiple functionally relevant edits in CAR T-cells. Co-inhibitory receptors concomitantly contribute at regulating T-cell responses, making it difficult to inhibit or knock-out multiple receptors together. With our approach we proved the feasibility of the simultaneous knock-down of PD-1, LAG-3, TIM-3 and TIGIT. Moreover, we show increased performance of the engineered CAR T-cells, suggesting that the simultaneous downregulation of the four co-inhibitory receptors may improve the antitumor activity of adoptive cell therapy. This strategy may empower the CAR T-cells to successfully target tumors reliant on high expression of immune checkpoint molecules and therefore often refractory to immunotherapy, such as solid tumors.
共抑制受体,如PD-1和LAG-3,在调节t细胞活性方面起着至关重要的作用,因为它们在慢性t细胞活化时在细胞表面的表达与t细胞衰竭有关。针对共抑制受体的免疫疗法在几种癌症适应症中显示出前所未有的疗效。然而,许多患者对这些治疗没有反应,一些肿瘤类型仍然是难治性的。CAR - t细胞对抗实体肿瘤失败的一个关键决定因素是癌细胞检查点抑制剂诱导的t细胞衰竭。不同共抑制受体的协同靶向可能有助于克服这些障碍。本研究表明,同时下调共抑制受体PD-1、LAG-3、TIM-3和TIGIT使CAR - t细胞具有优越的疗效。方法建立基于microRNA (miRNA)的短发夹RNA (shRNA)平台,可同时调控多个靶基因。该平台配备了shrna衍生的靶向PD-1、LAG-3、TIM-3和TIGIT的引导序列(shGuides),并与抗cd19 CAR结合。用表达共抑制受体配体的靶癌细胞挑战配备shRNA平台的CAR - t细胞。通过监测t细胞活化(通过表面活化标记物和细胞因子分泌)、杀伤活性和持久性(通过反复挑战靶细胞),在体外评估四种共抑制受体同时下调的影响。结果靶向PD-1、LAG-3、TIM-3和TIGIT的mirna衍生shRNA平台导致CAR - t细胞中这四种共抑制受体的显著下调。在被目标癌细胞激活后,与未使用shRNA平台进行工程改造的CAR - t细胞相比,同时敲除这四种基因可以增强细胞因子的分泌。此外,携带四重基因敲除的CAR - t细胞在连续周期中与目标癌细胞反复挑战时,表现出卓越的杀伤能力,增强的持久性和延长的活性。这些数据验证了我们在CAR - t细胞中有效引入多种功能相关编辑的技术。共抑制受体同时参与调节t细胞反应,使其难以同时抑制或敲除多个受体。通过我们的方法,我们证明了同时敲除PD-1、LAG-3、TIM-3和TIGIT的可行性。此外,我们发现工程化CAR - t细胞的性能有所提高,这表明同时下调四种共抑制受体可能会提高过继细胞治疗的抗肿瘤活性。这种策略可能使CAR - t细胞能够成功靶向依赖于免疫检查点分子高表达的肿瘤,因此通常难以免疫治疗,如实体瘤。
{"title":"297 Improvement of CAR T-cell performance by simultaneous downregulation of multiple co-inhibitory receptors","authors":"Matteo Rossi, Fanny Huberty, Thuy Nguyen, Jerome Marijsse, Celine Jacques-Hespel, Eytan Breman","doi":"10.1136/jitc-2023-sitc2023.0297","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0297","url":null,"abstract":"<h3>Background</h3> Co-inhibitory receptors, such as PD-1 and LAG-3, have a crucial role in regulating T-cell activity, as their expression on the cell surface upon chronic T-cell activation is associated with T-cell exhaustion. Immunotherapies directed against co-inhibitory receptors exhibited unprecedented efficacy in several cancer indications. However, many patients do not respond to these therapies and some tumor types remain largely refractory. A key determinant of CAR T-cell failure against solid tumors is T-cell exhaustion induced by checkpoint inhibitors on cancer cells. The coordinated targeting of different co-inhibitory receptors may help overcome these hurdles. Here we show that the simultaneous downregulation of co-inhibitory receptors PD-1, LAG-3, TIM-3 and TIGIT endows CAR T-cells with superior efficacy. <h3>Methods</h3> A microRNA (miRNA)-based short hairpin RNA (shRNA) platform was developed, to allow for the tunable modulation of multiple target genes simultaneously. The platform was equipped with shRNA-derived guide sequences (shGuides) targeting PD-1, LAG-3, TIM-3 and TIGIT, and combined with an anti-CD19 CAR. The shRNA platform-equipped CAR T-cells were challenged with target cancer cells expressing the co-inhibitory receptors’ ligands. The impact of the simultaneous downregulation of the four co-inhibitory receptors was assessed <i>in vitro</i> by monitoring T-cell activation (via surface activation markers and cytokine secretion), killing activity, and persistence (via repeated challenges with the target cells). <h3>Results</h3> The miRNA-derived shRNA platform targeting PD-1, LAG-3, TIM-3 and TIGIT led to a significant downregulation of the four co-inhibitory receptors in the CAR T-cells. Upon activation by the target cancer cells, the simultaneous knock-down of the four genes enhanced cytokine secretion compared to CAR T-cells not engineered with the shRNA platform. Moreover, the CAR T-cells carrying the quadruple knock-down showed superior killing ability, increased persistence and prolonged activity when repeatedly challenged in sequential cycles with the target cancer cells. <h3>Conclusions</h3> These data validate our technology for the effective introduction of multiple functionally relevant edits in CAR T-cells. Co-inhibitory receptors concomitantly contribute at regulating T-cell responses, making it difficult to inhibit or knock-out multiple receptors together. With our approach we proved the feasibility of the simultaneous knock-down of PD-1, LAG-3, TIM-3 and TIGIT. Moreover, we show increased performance of the engineered CAR T-cells, suggesting that the simultaneous downregulation of the four co-inhibitory receptors may improve the antitumor activity of adoptive cell therapy. This strategy may empower the CAR T-cells to successfully target tumors reliant on high expression of immune checkpoint molecules and therefore often refractory to immunotherapy, such as solid tumors.","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":"135163191","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
355 Directing a high avidity KRAS G12D-specific TCR engineered with a CD8αβ co-receptor and chimeric cytokine receptor using non-viral knock-in enhances anti-tumor responses 355利用非病毒敲入介导CD8αβ共受体和嵌合细胞因子受体工程的高亲和度KRAS g12d特异性TCR增强抗肿瘤反应
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0355
Allison P Drain, Nicholas Rouillard, Nathaniel Swanson, Santosh Narayan, Tyler Warner, Nicole Danek, Ken Gareau, Cheryl Black, James Parsons, Anthony Thomas, Jinsheng Liang, Luhua Shen, Tanya Tetrault, Vince Nguyen, Iqraa Priyata, Sarah Vidyasagar, Joshua Francis, Xingyue He, Patrick J Browne, Rebecca C Lamothe, Meghan D Storlie, Gregory J Cost, Thomas M Schmitt, Philip D Greenberg, Smita S Chandran, Christopher A Klebanoff, Ankit Gupta, Damien Hallet, Gary Shapiro, Kim Nguyen, Loic Vincent

Background

Adoptive T cell therapy has demonstrated clinical activity in a subset of patients with solid tumors; however, consistent responses will require further optimization. T cell receptor (TCR)-engineered T cells recognize peptides derived from intracellular and surface proteins presented in the context of MHC class I. Immunologic targeting of recurrently mutated oncogenic drivers, such as KRAS, overcomes many of the major obstacles of this modality because the resulting epitope is: 1) tumor-specific, 2) essential for cancer cell fitness, and 3) derived from a stably expressed non-self protein. AFNT-212 is a next-generation engineered T cell therapy that uses non-viral targeted knock-in (KI) at the TCRα constant (TRAC) locus to express a multi-cistronic cassette that includes 1) a high-affinity TCR specific for KRASG12D mutation, 2) a CD8αβ coreceptor, and 3) a chimeric cytokine receptor.

Methods

Human CD4+ and CD8+ T cells were genetically engineered by a novel CRISPR-Cas nuclease and gRNAs targeting TRAC and the TCRβ constant (TRBC) genes allowing for knock-out of the endogenous TCR loci and simultaneous integration of the non-viral plasmid-based transgene cassette. Engineered T cells were assessed for specificity and potency, including activation, proliferation, and cytotoxicity, against KRAS G12D peptide presented by HLA-A*11:01 and a panel of KRAS G12D-expressing tumor cell lines. In vitro safety studies were performed along with in vivo efficacy studies in multiple human xenograft models.

Results

Engineered primary T cells showed specific recognition of KRAS G12D peptide, demonstrated cytotoxicity against endogenously expressing HLA-A*11:01+/KRAS G12D+ cell lines in tumor cell re-challenge assays in vitro, and mediated robust anti-tumor activity in vivo. Inclusion of the chimeric cytokine receptor allowed for a more potent anti-tumor response stemming from improved T cell expansion and resistance to exhaustion. No off-target liabilities were identified upon co-incubation of AFNT-212 with all possible peptides in the human proteome matching the xScan-defined epitope recognition motif for the TCR, demonstrating specificity. Gene editing safety evaluation did not reveal any off-target activity for the CRISPR-Cas nucleases and engineered T cells did not show cytokine-independent proliferation, collectively supporting a favorable pre-clinical safety profile for AFNT-212.

Conclusions

We report a novel TCR gene therapy approach targeting mutant KRAS G12D-expressing tumors with a coordinated CD4/CD8 T cell response that has a promising efficacy and safety profile. Our work supports the planned clinical development of AFNT-212 as a novel non-viral KI TCR-engineered T cell therapy for KRAS-mutant solid tumors.
过继性T细胞疗法已经证明对一部分实体瘤患者具有临床活性;然而,一致的响应需要进一步优化。T细胞受体(TCR)工程T细胞识别来自MHC i类背景下的细胞内和表面蛋白的肽。针对复发突变的致癌驱动因子(如KRAS)的免疫靶向克服了这种模式的许多主要障碍,因为所产生的表位是:1)肿瘤特异性,2)癌细胞适应性必需的,3)来源于稳定表达的非自身蛋白。AFNT-212是一种新一代工程化T细胞疗法,它使用TCRα常数(TRAC)位点的非病毒靶向敲入(KI)来表达一个多顺反子盒,其中包括1)KRASG12D突变的高亲和力TCR, 2) CD8αβ辅助受体,以及3)嵌合细胞因子受体。方法利用一种新的CRISPR-Cas核酸酶和靶向TRAC和TCRβ常数(TRBC)基因的gRNAs对人CD4+和CD8+ T细胞进行基因工程改造,从而敲除内源性TCR位点并同时整合基于非病毒质粒的转基因盒。通过HLA-A*11:01和一组表达KRAS G12D的肿瘤细胞系,评估工程T细胞对KRAS G12D肽的特异性和效力,包括活化、增殖和细胞毒性。在多种人类异种移植模型中进行了体外安全性研究和体内有效性研究。结果经工程改造的原代T细胞对KRAS G12D肽具有特异性识别能力,在体外肿瘤细胞再攻毒实验中对内源性表达HLA-A*11:01+/KRAS G12D+的细胞系显示出细胞毒性,并介导了较强的体内抗肿瘤活性。嵌合细胞因子受体的包含允许更有效的抗肿瘤反应,源于改善T细胞扩增和抵抗衰竭。在将AFNT-212与与xscan定义的TCR表位识别基序匹配的所有可能的人类蛋白质组肽共孵育时,没有发现脱靶性,证明了特异性。基因编辑安全性评估没有显示CRISPR-Cas核酸酶的任何脱靶活性,工程T细胞没有显示细胞因子不依赖性增殖,共同支持AFNT-212良好的临床前安全性。我们报道了一种新的TCR基因治疗方法,靶向表达KRAS g12d的突变肿瘤,具有协调的CD4/CD8 T细胞反应,具有良好的疗效和安全性。我们的工作支持AFNT-212作为一种新的非病毒性KI tcr工程T细胞治疗kras突变实体瘤的计划临床开发。
{"title":"355 Directing a high avidity KRAS G12D-specific TCR engineered with a CD8αβ co-receptor and chimeric cytokine receptor using non-viral knock-in enhances anti-tumor responses","authors":"Allison P Drain, Nicholas Rouillard, Nathaniel Swanson, Santosh Narayan, Tyler Warner, Nicole Danek, Ken Gareau, Cheryl Black, James Parsons, Anthony Thomas, Jinsheng Liang, Luhua Shen, Tanya Tetrault, Vince Nguyen, Iqraa Priyata, Sarah Vidyasagar, Joshua Francis, Xingyue He, Patrick J Browne, Rebecca C Lamothe, Meghan D Storlie, Gregory J Cost, Thomas M Schmitt, Philip D Greenberg, Smita S Chandran, Christopher A Klebanoff, Ankit Gupta, Damien Hallet, Gary Shapiro, Kim Nguyen, Loic Vincent","doi":"10.1136/jitc-2023-sitc2023.0355","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0355","url":null,"abstract":"<h3>Background</h3> Adoptive T cell therapy has demonstrated clinical activity in a subset of patients with solid tumors; however, consistent responses will require further optimization. T cell receptor (TCR)-engineered T cells recognize peptides derived from intracellular and surface proteins presented in the context of MHC class I. Immunologic targeting of recurrently mutated oncogenic drivers, such as KRAS, overcomes many of the major obstacles of this modality because the resulting epitope is: 1) tumor-specific, 2) essential for cancer cell fitness, and 3) derived from a stably expressed non-self protein. AFNT-212 is a next-generation engineered T cell therapy that uses non-viral targeted knock-in (KI) at the TCRα constant (<i>TRAC</i>) locus to express a multi-cistronic cassette that includes 1) a high-affinity TCR specific for KRAS<sub>G12D</sub> mutation, 2) a CD8αβ coreceptor, and 3) a chimeric cytokine receptor. <h3>Methods</h3> Human CD4<sup>+</sup> and CD8<sup>+</sup> T cells were genetically engineered by a novel CRISPR-Cas nuclease and gRNAs targeting <i>TRAC</i> and the TCRβ constant (<i>TRBC</i>) genes allowing for knock-out of the endogenous TCR loci and simultaneous integration of the non-viral plasmid-based transgene cassette. Engineered T cells were assessed for specificity and potency, including activation, proliferation, and cytotoxicity, against KRAS G12D peptide presented by HLA-A*11:01 and a panel of KRAS G12D-expressing tumor cell lines. <i>In vitro</i> safety studies were performed along with <i>in vivo</i> efficacy studies in multiple human xenograft models. <h3>Results</h3> Engineered primary T cells showed specific recognition of KRAS G12D peptide, demonstrated cytotoxicity against endogenously expressing HLA-A*11:01<sup>+</sup>/KRAS G12D<sup>+</sup> cell lines in tumor cell re-challenge assays <i>in vitro</i>, and mediated robust anti-tumor activity <i>in vivo</i>. Inclusion of the chimeric cytokine receptor allowed for a more potent anti-tumor response stemming from improved T cell expansion and resistance to exhaustion. No off-target liabilities were identified upon co-incubation of AFNT-212 with all possible peptides in the human proteome matching the xScan-defined epitope recognition motif for the TCR, demonstrating specificity. Gene editing safety evaluation did not reveal any off-target activity for the CRISPR-Cas nucleases and engineered T cells did not show cytokine-independent proliferation, collectively supporting a favorable pre-clinical safety profile for AFNT-212. <h3>Conclusions</h3> We report a novel TCR gene therapy approach targeting mutant KRAS G12D-expressing tumors with a coordinated CD4/CD8 T cell response that has a promising efficacy and safety profile. Our work supports the planned clinical development of AFNT-212 as a novel non-viral KI TCR-engineered T cell therapy for KRAS-mutant solid tumors.","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":"135163192","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
742 Phase 1/2 study of the bispecific 4–1BB and PD-L1 antibody INBRX-105 alone and in combination with pembrolizumab in select solid tumors 742双特异性4-1BB和PD-L1抗体INBRX-105单独和联合pembrolizumab在选定实体瘤中的1/2期研究
Pub Date : 2023-11-01 DOI: 10.1136/jitc-2023-sitc2023.0742
Jong Chul Park, David Berz, Manish Sharma, Erminia Massarelli, Ralph J Hauke, Frank Yung-Chin Tsai, David Hong, Neal Akhave, Justin A Call, Jennifer Carlisle, Rachel E Sanborn, Naomi B Haas, John Hamm, D Ross Camidge, Alexander I Spira, Vasily Andrianov, Brianne O’Neill, Heather Kinkead, Anthony W Tolcher

Background

4–1BB is a costimulatory receptor upregulated on tumor-infiltrating lymphocytes. 4–1BB signaling promotes T-cell proliferation and activation and decreases T-cell exhaustion. 4–1BB agonists have shown promising antitumor activity but have been limited by hepatotoxicity resulting from systemic 4–1BB activation. INBRX-105 is a 4–1BB×PD-L1 bispecific antibody designed to localize 4–1BB costimulatory signaling to PD-L1-rich environments. INBRX-105 consists of 2 agonistic 4–1BB single-domain antibodies (sdAbs) and 2 PD-L1 sdAbs, which allow anchoring to PD-L1. Cross-linking of PD-L1 to 4–1BB by INBRX-105 leads to conditional 4–1BB activation at sites of high PD-L1 expression, potentially limiting toxicities associated with prior 4–1BB agonists. In mouse tumor models, an INBRX-105 surrogate (INBRX-105-a) exhibited antitumor efficacy and increased T-cell frequency in the tumor microenvironment.1 Robust proliferation of CD8+ T effector memory cells was observed intratumorally and in peripheral blood. INBRX-105-a plus a PD-1 antagonist resulted in greater inhibition of tumor growth. We describe a phase 1/2 study (NCT03809624) of INBRX-105 alone and in combination with pembrolizumab in patients with solid tumors.

Methods

This open-label, 4-part study of INBRX-105±pembrolizumab in locally advanced unresectable/metastatic solid tumors (N≈300) is enrolling in the US (figure 1). Dose escalation (part 1, INBRX-105 single agent; part 3, combination) was completed. INBRX-105 is being assessed in dose-expansion cohorts (part 2, INBRX-105 single agent; part 4, combination). Patients naive to 4–1BB agonists with disease that progressed despite standard therapy or who have no alternative treatment options (except checkpoint inhibitor [CPI]-naive cohorts) were included. Part 2a (recommended phase 2 dose [RP2D] expansion) consists of 4 cohorts: non-small cell lung cancer (NSCLC; PD-L1+), melanoma/solid tumors, head and neck squamous cell carcinoma (HNSCC), and other solid tumors (PD-L1+). Part 2b includes PD-L1-high HNSCC (including nasopharyngeal carcinoma). All part 2 cohorts, except melanoma (noncutaneous)/solid tumors, were CPI relapsed/refractory. PD-L1+ NSCLC (part 2a) and PD-L1-high HNSCC (part 2b) cohorts were opened based on encouraging early single-agent activity. Cohorts are enrolling and complete data readouts are forthcoming. Part 4 consists of CPI-relapsed/refractory cohorts (PD-L1+ NSCLC, cutaneous melanoma, PD-L1+ HNSCC, microsatellite instability-/tumor mutation burden-high or mismatch repair-deficient solid tumors) and CPI-naive cohorts (PDL1+ NSCLC and HNSCC). PD-L1 immunohistochemistry scores are required in parts 2 and 4, with threshold scores defined per protocol. Primary objectives are safety and determination of the maximum tolerated dose and/or RP2D of INBRX-105 as monotherapy and with pembrolizumab. Secondary objectives include pharmacokinetics, immunogenicity, and preliminary antitumor activi
背景4-1BB是一种在肿瘤浸润淋巴细胞上上调的共刺激受体。4-1BB信号传导促进t细胞增殖和活化,减少t细胞衰竭。4-1BB激动剂显示出有希望的抗肿瘤活性,但由于全身4-1BB激活引起的肝毒性而受到限制。INBRX-105是一种4-1BB×PD-L1双特异性抗体,旨在将4-1BB共刺激信号定位到富含pd - l1的环境中。INBRX-105由2个激动性4-1BB单域抗体(sabs)和2个PD-L1单域抗体组成,可锚定PD-L1。INBRX-105将PD-L1与4-1BB交联,导致PD-L1高表达位点有条件的4-1BB激活,潜在地限制了先前4-1BB激动剂相关的毒性。在小鼠肿瘤模型中,INBRX-105替代物(INBRX-105-a)表现出抗肿瘤功效,并增加了肿瘤微环境中的t细胞频率在瘤内和外周血中观察到CD8+ T效应记忆细胞的强劲增殖。INBRX-105-a加PD-1拮抗剂对肿瘤生长有更大的抑制作用。我们描述了在实体瘤患者中单独使用INBRX-105和联合使用pembrolizumab的1/2期研究(NCT03809624)。方法:这项开放标签、4部分的INBRX-105±pembrolizumab治疗局部晚期不可切除/转移性实体瘤(N≈300)的研究正在美国进行(图1)。第3部分(组合)已经完成。INBRX-105正在剂量扩展队列中进行评估(第2部分,INBRX-105单药;第4部分,组合)。首次使用4-1BB激动剂的患者,尽管接受标准治疗,但病情进展或没有其他治疗方案(检查点抑制剂[CPI]初始队列除外)。2a部分(推荐的2期剂量[RP2D]扩大)包括4个队列:非小细胞肺癌(NSCLC;PD-L1+)、黑色素瘤/实体瘤、头颈部鳞状细胞癌(HNSCC)和其他实体瘤(PD-L1+)。2b部分包括pd - l1高的HNSCC(包括鼻咽癌)。除黑色素瘤(非皮肤)/实体瘤外,所有第2部分队列均为CPI复发/难治性。PD-L1+ NSCLC (2a部分)和PD-L1高HNSCC (2b部分)队列基于鼓励早期单药活性而开放。队列正在登记,完整的数据即将公布。第4部分包括cpi复发/难治队列(PD-L1+ NSCLC、皮肤黑色素瘤、PD-L1+ HNSCC、微卫星不稳定性/肿瘤突变负担高或错配修复缺陷的实体肿瘤)和cpi初始队列(PDL1+ NSCLC和HNSCC)。第2部分和第4部分需要PD-L1免疫组织化学评分,每个方案定义阈值评分。主要目标是安全性和确定INBRX-105作为单药和派姆单抗的最大耐受剂量和/或RP2D。次要目标包括药代动力学、免疫原性和RECIST的初步抗肿瘤活性。金凯德,马塞多,张国华,等。与双特异性PD-L1条件4-1BB激动剂抗肿瘤活性相关的关键药代动力学和药效学参数[J]中华肿瘤杂志,2013;抽象的12。本研究方案由各参与机构的机构审查委员会审查并批准;所有患者均提供书面知情同意书。
{"title":"742 Phase 1/2 study of the bispecific 4–1BB and PD-L1 antibody INBRX-105 alone and in combination with pembrolizumab in select solid tumors","authors":"Jong Chul Park, David Berz, Manish Sharma, Erminia Massarelli, Ralph J Hauke, Frank Yung-Chin Tsai, David Hong, Neal Akhave, Justin A Call, Jennifer Carlisle, Rachel E Sanborn, Naomi B Haas, John Hamm, D Ross Camidge, Alexander I Spira, Vasily Andrianov, Brianne O’Neill, Heather Kinkead, Anthony W Tolcher","doi":"10.1136/jitc-2023-sitc2023.0742","DOIUrl":"https://doi.org/10.1136/jitc-2023-sitc2023.0742","url":null,"abstract":"<h3>Background</h3> 4–1BB is a costimulatory receptor upregulated on tumor-infiltrating lymphocytes. 4–1BB signaling promotes T-cell proliferation and activation and decreases T-cell exhaustion. 4–1BB agonists have shown promising antitumor activity but have been limited by hepatotoxicity resulting from systemic 4–1BB activation. INBRX-105 is a 4–1BB×PD-L1 bispecific antibody designed to localize 4–1BB costimulatory signaling to PD-L1-rich environments. INBRX-105 consists of 2 agonistic 4–1BB single-domain antibodies (sdAbs) and 2 PD-L1 sdAbs, which allow anchoring to PD-L1. Cross-linking of PD-L1 to 4–1BB by INBRX-105 leads to conditional 4–1BB activation at sites of high PD-L1 expression, potentially limiting toxicities associated with prior 4–1BB agonists. In mouse tumor models, an INBRX-105 surrogate (INBRX-105-a) exhibited antitumor efficacy and increased T-cell frequency in the tumor microenvironment.<sup>1</sup> Robust proliferation of CD8<sup>+</sup> T effector memory cells was observed intratumorally and in peripheral blood. INBRX-105-a plus a PD-1 antagonist resulted in greater inhibition of tumor growth. We describe a phase 1/2 study (NCT03809624) of INBRX-105 alone and in combination with pembrolizumab in patients with solid tumors. <h3>Methods</h3> This open-label, 4-part study of INBRX-105±pembrolizumab in locally advanced unresectable/metastatic solid tumors (N≈300) is enrolling in the US (figure 1). Dose escalation (part 1, INBRX-105 single agent; part 3, combination) was completed. INBRX-105 is being assessed in dose-expansion cohorts (part 2, INBRX-105 single agent; part 4, combination). Patients naive to 4–1BB agonists with disease that progressed despite standard therapy or who have no alternative treatment options (except checkpoint inhibitor [CPI]-naive cohorts) were included. Part 2a (recommended phase 2 dose [RP2D] expansion) consists of 4 cohorts: non-small cell lung cancer (NSCLC; PD-L1+), melanoma/solid tumors, head and neck squamous cell carcinoma (HNSCC), and other solid tumors (PD-L1+). Part 2b includes PD-L1-high HNSCC (including nasopharyngeal carcinoma). All part 2 cohorts, except melanoma (noncutaneous)/solid tumors, were CPI relapsed/refractory. PD-L1+ NSCLC (part 2a) and PD-L1-high HNSCC (part 2b) cohorts were opened based on encouraging early single-agent activity. Cohorts are enrolling and complete data readouts are forthcoming. Part 4 consists of CPI-relapsed/refractory cohorts (PD-L1+ NSCLC, cutaneous melanoma, PD-L1+ HNSCC, microsatellite instability-/tumor mutation burden-high or mismatch repair-deficient solid tumors) and CPI-naive cohorts (PDL1+ NSCLC and HNSCC). PD-L1 immunohistochemistry scores are required in parts 2 and 4, with threshold scores defined per protocol. Primary objectives are safety and determination of the maximum tolerated dose and/or RP2D of INBRX-105 as monotherapy and with pembrolizumab. Secondary objectives include pharmacokinetics, immunogenicity, and preliminary antitumor activi","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":"135163211","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