Fast tumor-infiltrating lymphocytes (TILs): rapid manufacture of an adoptive cellular therapeutic from pleural infiltrating T cells for intrapleural administration.

IF 3.2 3区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY
Vera S Donnenberg, John Lister, Charlene L Briedenbaugh, Patrick L Wagner, David L Bartlett, Albert D Donnenberg
{"title":"Fast tumor-infiltrating lymphocytes (TILs): rapid manufacture of an adoptive cellular therapeutic from pleural infiltrating T cells for intrapleural administration.","authors":"Vera S Donnenberg, John Lister, Charlene L Briedenbaugh, Patrick L Wagner, David L Bartlett, Albert D Donnenberg","doi":"10.1016/j.jcyt.2025.06.011","DOIUrl":null,"url":null,"abstract":"<p><strong>Background aims: </strong>Cancers of many different tissue origins can metastasize to the pleura, a space with a unique immune environment that predisposes to aggressive tumor behavior and the development of effusions, an exudative leakage of serous fluid accompanied by an immune infiltrate. Effusions are drained therapeutically to relieve dyspnea, often several times per week. Characteristically, they contain 50-1000 × 10<sup>6</sup> viable pleural T cells (PITs), which can be reliably activated and expanded in culture, making them an ideal source for generation of a cellular therapeutic. We sought to determine the feasibility of a Good Manufacturing Practice-compliant, rapidly manufactured adoptive cellular therapeutic from pleural-infiltrating T cells and to determine the cytolytic activity against autologous tumor, cytokine secretion profile, and immune check point molecule (ICM) expression.</p><p><strong>Methods: </strong>Six products were generated from consecutively collected malignant pleural effusions (MPEs) drained from patients with breast (4) or non-small cell lung (2) cancer metastatic to the pleura. CD4+ and CD8+ cells were immunomagnetically selected, stimulated with anti-CD3/anti-CD28 and expanded in the presence of interleukin (IL)-7 and IL-15 (12.5 ng/mL each) using the Miltenyi CliniMACS Prodigy device. Cells were cultured for 8- 12 days. Cytokines were assayed in the MPE and in the culture medium before harvest using a multiplexed bead assay. Cytolytic activity of the final cellular product formulation against an autologous tumor was measured in a 4-hour killing assay by lactate dehydrogenase release. T-cell content, ICM expression and intracellular interferon ɣ were assessed by flow cytometry.</p><p><strong>Results: </strong>All MPEs successfully generated products containing 0.7 to 3.2 × 10<sup>9</sup> viable T cells. All final products showed no growth in bacterial or fungal cultures. T-cell purity was 98.3 ± 1.7% (mean, standard deviation), viability was 97.6 ± 1.7% and T-cell fold expansion was 14.3 ± 10.6. Twenty cytokines (excluding IL-7 and IL-15) were present in the culture supernatants at ≥10 pmol/L. These include granzyme B, interferon-ɣ, IL-13, perforin, granulocyte-macrophage colony-stimulating factor and tumor necrosis factor-α. In the final cellular product formulation, 69 ± 28% of CD4+ T cells, and 75 ± 27% of CD8+ T-cells produced interferon-ɣ without additional stimulation. ICM expression was well correlated between CD4+ and CD8+ T cells and was relatively low, with TIGIT (44.6 ± 10.9%) and programmed cell death protein 1(21.1 ± 6.7%) being the highest. All products evidenced cytolytic activity against an autologous tumor, with maximal lysis ranging from 19.4% to 100% and cytolytic indices ranging from 4.3 to 21.1.</p><p><strong>Conclusions: </strong>We conclude that Fast tumor-infiltrating lymphocytes (Fast TIL), an adoptive cellular therapeutic generated from drained MPEs, can be rapidly and reliably manufactured using the Prodigy system. The products have demonstrable in vitro effector activity against an autologous tumor. Expression of interferon-ɣ in the majority of cells, without accompanying elevated expression of ICM, suggests that the cells have not been exhausted during expansion. Based in part on the results presented here, the US Food and Drug Administration has issued Investigational New Drug status (IND # 30892) for the rapid manufacture of PIT-based cellular therapeutic (Fast TIL), paving the way for a first-in-human clinical trial (supported by CDMRP-TTSA CA230972). We plan to infuse this product intrapleurally, accompanied by low-dose intrapleural IL-2 with the expectation that infused cells will immediately encounter tumor antigens, continue to expand in the pleural space, and migrate to the peripheral circulation.</p>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":" ","pages":""},"PeriodicalIF":3.2000,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cytotherapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.jcyt.2025.06.011","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background aims: Cancers of many different tissue origins can metastasize to the pleura, a space with a unique immune environment that predisposes to aggressive tumor behavior and the development of effusions, an exudative leakage of serous fluid accompanied by an immune infiltrate. Effusions are drained therapeutically to relieve dyspnea, often several times per week. Characteristically, they contain 50-1000 × 106 viable pleural T cells (PITs), which can be reliably activated and expanded in culture, making them an ideal source for generation of a cellular therapeutic. We sought to determine the feasibility of a Good Manufacturing Practice-compliant, rapidly manufactured adoptive cellular therapeutic from pleural-infiltrating T cells and to determine the cytolytic activity against autologous tumor, cytokine secretion profile, and immune check point molecule (ICM) expression.

Methods: Six products were generated from consecutively collected malignant pleural effusions (MPEs) drained from patients with breast (4) or non-small cell lung (2) cancer metastatic to the pleura. CD4+ and CD8+ cells were immunomagnetically selected, stimulated with anti-CD3/anti-CD28 and expanded in the presence of interleukin (IL)-7 and IL-15 (12.5 ng/mL each) using the Miltenyi CliniMACS Prodigy device. Cells were cultured for 8- 12 days. Cytokines were assayed in the MPE and in the culture medium before harvest using a multiplexed bead assay. Cytolytic activity of the final cellular product formulation against an autologous tumor was measured in a 4-hour killing assay by lactate dehydrogenase release. T-cell content, ICM expression and intracellular interferon ɣ were assessed by flow cytometry.

Results: All MPEs successfully generated products containing 0.7 to 3.2 × 109 viable T cells. All final products showed no growth in bacterial or fungal cultures. T-cell purity was 98.3 ± 1.7% (mean, standard deviation), viability was 97.6 ± 1.7% and T-cell fold expansion was 14.3 ± 10.6. Twenty cytokines (excluding IL-7 and IL-15) were present in the culture supernatants at ≥10 pmol/L. These include granzyme B, interferon-ɣ, IL-13, perforin, granulocyte-macrophage colony-stimulating factor and tumor necrosis factor-α. In the final cellular product formulation, 69 ± 28% of CD4+ T cells, and 75 ± 27% of CD8+ T-cells produced interferon-ɣ without additional stimulation. ICM expression was well correlated between CD4+ and CD8+ T cells and was relatively low, with TIGIT (44.6 ± 10.9%) and programmed cell death protein 1(21.1 ± 6.7%) being the highest. All products evidenced cytolytic activity against an autologous tumor, with maximal lysis ranging from 19.4% to 100% and cytolytic indices ranging from 4.3 to 21.1.

Conclusions: We conclude that Fast tumor-infiltrating lymphocytes (Fast TIL), an adoptive cellular therapeutic generated from drained MPEs, can be rapidly and reliably manufactured using the Prodigy system. The products have demonstrable in vitro effector activity against an autologous tumor. Expression of interferon-ɣ in the majority of cells, without accompanying elevated expression of ICM, suggests that the cells have not been exhausted during expansion. Based in part on the results presented here, the US Food and Drug Administration has issued Investigational New Drug status (IND # 30892) for the rapid manufacture of PIT-based cellular therapeutic (Fast TIL), paving the way for a first-in-human clinical trial (supported by CDMRP-TTSA CA230972). We plan to infuse this product intrapleurally, accompanied by low-dose intrapleural IL-2 with the expectation that infused cells will immediately encounter tumor antigens, continue to expand in the pleural space, and migrate to the peripheral circulation.

快速肿瘤浸润淋巴细胞(TILs):从胸腔浸润T细胞快速制造一种用于胸腔内给药的过继细胞疗法。
背景目的:许多不同组织来源的癌症可以转移到胸膜,胸膜是一个具有独特免疫环境的空间,容易发生侵袭性肿瘤行为和积液的发展,浆液渗出性渗漏伴免疫浸润。引流积液治疗性地缓解呼吸困难,通常每周数次。其特点是,它们含有50-1000 × 106个活的胸膜T细胞(pit),可以在培养中可靠地激活和扩增,使其成为产生细胞治疗药物的理想来源。我们试图确定一种符合《药品生产质量管理规范》的、快速生产的胸膜浸润性T细胞过继细胞疗法的可行性,并确定其对自体肿瘤的细胞溶解活性、细胞因子分泌谱和免疫检查点分子(ICM)表达。方法:连续收集乳腺癌(4)或非小细胞肺癌(2)转移至胸膜的恶性胸腔积液(MPEs),生成6种产品。使用Miltenyi CliniMACS Prodigy装置免疫磁选择CD4+和CD8+细胞,用抗cd3 /抗cd28刺激,并在白细胞介素(IL)-7和IL-15(各12.5 ng/mL)存在下扩增。细胞培养8- 12天。细胞因子在收获前在MPE和培养基中使用多重头试验进行检测。通过乳酸脱氢酶释放,在4小时杀伤实验中测量最终细胞产品配方对自体肿瘤的细胞溶解活性。流式细胞术检测t细胞含量、ICM表达及细胞内干扰素γ。结果:所有MPEs均成功生成含有0.7 ~ 3.2 × 109个活T细胞的产物。所有最终产品在细菌或真菌培养中均未显示生长。t细胞纯度为98.3±1.7%(平均值,标准差),活力为97.6±1.7%,t细胞折叠扩增率为14.3±10.6。培养上清中有20种细胞因子(不包括IL-7和IL-15),浓度≥10 pmol/L。这些包括颗粒酶B、干扰素-α、IL-13、穿孔素、粒细胞-巨噬细胞集落刺激因子和肿瘤坏死因子-α。在最终的细胞产品配方中,69±28%的CD4+ T细胞和75±27%的CD8+ T细胞在没有额外刺激的情况下产生干扰素- α。ICM表达在CD4+和CD8+ T细胞间相关性较好,表达水平相对较低,其中TIGIT(44.6±10.9%)和程序性细胞死亡蛋白1(21.1±6.7%)表达最高。所有产品均显示出对自体肿瘤的细胞溶解活性,最大溶解范围为19.4%至100%,细胞溶解指数范围为4.3至21.1。结论:我们得出结论,快速肿瘤浸润淋巴细胞(Fast TIL)是一种过继细胞治疗方法,可以使用Prodigy系统快速可靠地制造出来。该产品对自体肿瘤具有明显的体外效应活性。干扰素- α在大多数细胞中的表达,没有伴随ICM的表达升高,表明细胞在扩增过程中没有被耗尽。部分基于本文提出的结果,美国食品和药物管理局(fda)已经为基于pit的细胞治疗(Fast TIL)的快速生产颁发了新药研究状态(IND # 30892),为首次人体临床试验铺平了道路(由CDMRP-TTSA CA230972支持)。我们计划在胸膜内灌注本品,同时注入低剂量的IL-2,期望注入的细胞能立即遇到肿瘤抗原,在胸膜间隙继续扩张,并向外周循环迁移。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Cytotherapy
Cytotherapy 医学-生物工程与应用微生物
CiteScore
6.30
自引率
4.40%
发文量
683
审稿时长
49 days
期刊介绍: The journal brings readers the latest developments in the fast moving field of cellular therapy in man. This includes cell therapy for cancer, immune disorders, inherited diseases, tissue repair and regenerative medicine. The journal covers the science, translational development and treatment with variety of cell types including hematopoietic stem cells, immune cells (dendritic cells, NK, cells, T cells, antigen presenting cells) mesenchymal stromal cells, adipose cells, nerve, muscle, vascular and endothelial cells, and induced pluripotential stem cells. We also welcome manuscripts on subcellular derivatives such as exosomes. A specific focus is on translational research that brings cell therapy to the clinic. Cytotherapy publishes original papers, reviews, position papers editorials, commentaries and letters to the editor. We welcome "Protocols in Cytotherapy" bringing standard operating procedure for production specific cell types for clinical use within the reach of the readership.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信