首页 > 最新文献

Molecular Therapy Oncolytics最新文献

英文 中文
Controlled release of enhanced cross-hybrid IgGA Fc PD-L1 inhibitors using oncolytic adenoviruses. 利用溶瘤腺病毒控制释放增强型交叉杂交IgGA Fc PD-L1抑制剂。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2023-03-16 DOI: 10.1016/j.omto.2023.01.006
Firas Hamdan, Michaela Feodoroff, Salvatore Russo, Manlio Fusciello, Sara Feola, Jacopo Chiaro, Gabriella Antignani, Francesca Greco, Jeanette Leusen, Erkko Ylösmäki, Mikaela Grönholm, Vincenzo Cerullo

Immune checkpoint inhibitors have clinical success in prolonging the life of many cancer patients. However, only a minority of patients benefit from such therapy, calling for further improvements. Currently, most PD-L1 checkpoint inhibitors in the clinic do not elicit Fc effector mechanisms that would substantially increase their efficacy. To gain potency and circumvent off-target effects, we previously designed an oncolytic adenovirus (Ad-Cab) expressing an Fc fusion peptide against PD-L1 on a cross-hybrid immunoglobulin GA (IgGA) Fc. Ad-Cab elicited antibody effector mechanisms of IgG1 and IgA, which led to higher tumor killing compared with each isotype alone and with clinically approved PD-L1 checkpoint inhibitors. In this study, we further improved the therapy to increase the IgG1 Fc effector mechanisms of the IgGA Fc fusion peptide (Ad-Cab FT) by adding four somatic mutations that increase natural killer (NK) cell activation. Ad-Cab FT was shown to work better at lower concentrations compared with Ad-Cab in vitro and in vivo and to have better tumor- and myeloid-derived suppressor cell killing, likely because of higher NK cell activation. Additionally, the biodistribution of the Fc fusion peptide demonstrated targeted release in the tumor microenvironment with minimal or no leakage to the peripheral blood and organs in mice. These data demonstrate effective and safe use of Ad-Cab FT, bidding for further clinical investigation.

免疫检查点抑制剂在延长许多癌症患者的生命方面取得了临床成功。然而,只有少数患者从这种治疗中受益,这需要进一步的改进。目前,临床上的大多数PD-L1检查点抑制剂都不会引发Fc效应机制,从而大大提高其疗效。为了获得效力并避免脱靶效应,我们先前设计了一种溶瘤腺病毒(Ad-Cab),在交叉杂交免疫球蛋白GA (IgGA) Fc上表达针对PD-L1的Fc融合肽。Ad-Cab引发了IgG1和IgA的抗体效应机制,与单独使用每种同型和临床批准的PD-L1检查点抑制剂相比,这导致更高的肿瘤杀伤。在这项研究中,我们进一步改进了治疗方法,通过添加四种体细胞突变来增加IgGA Fc融合肽(Ad-Cab FT)的IgG1 Fc效应机制,从而增加自然杀伤细胞(NK)的活化。在体外和体内,与Ad-Cab相比,Ad-Cab FT在较低浓度下工作更好,并且具有更好的肿瘤和髓源性抑制细胞杀伤,可能是因为更高的NK细胞活化。此外,Fc融合肽的生物分布表明在肿瘤微环境中有靶向释放,很少或没有泄漏到小鼠外周血和器官。这些数据证明了Ad-Cab FT的有效和安全使用,需要进一步的临床研究。
{"title":"Controlled release of enhanced cross-hybrid IgGA Fc PD-L1 inhibitors using oncolytic adenoviruses.","authors":"Firas Hamdan,&nbsp;Michaela Feodoroff,&nbsp;Salvatore Russo,&nbsp;Manlio Fusciello,&nbsp;Sara Feola,&nbsp;Jacopo Chiaro,&nbsp;Gabriella Antignani,&nbsp;Francesca Greco,&nbsp;Jeanette Leusen,&nbsp;Erkko Ylösmäki,&nbsp;Mikaela Grönholm,&nbsp;Vincenzo Cerullo","doi":"10.1016/j.omto.2023.01.006","DOIUrl":"https://doi.org/10.1016/j.omto.2023.01.006","url":null,"abstract":"<p><p>Immune checkpoint inhibitors have clinical success in prolonging the life of many cancer patients. However, only a minority of patients benefit from such therapy, calling for further improvements. Currently, most PD-L1 checkpoint inhibitors in the clinic do not elicit Fc effector mechanisms that would substantially increase their efficacy. To gain potency and circumvent off-target effects, we previously designed an oncolytic adenovirus (Ad-Cab) expressing an Fc fusion peptide against PD-L1 on a cross-hybrid immunoglobulin GA (IgGA) Fc. Ad-Cab elicited antibody effector mechanisms of IgG1 and IgA, which led to higher tumor killing compared with each isotype alone and with clinically approved PD-L1 checkpoint inhibitors. In this study, we further improved the therapy to increase the IgG1 Fc effector mechanisms of the IgGA Fc fusion peptide (Ad-Cab FT) by adding four somatic mutations that increase natural killer (NK) cell activation. Ad-Cab FT was shown to work better at lower concentrations compared with Ad-Cab <i>in vitro</i> and <i>in vivo</i> and to have better tumor- and myeloid-derived suppressor cell killing, likely because of higher NK cell activation. Additionally, the biodistribution of the Fc fusion peptide demonstrated targeted release in the tumor microenvironment with minimal or no leakage to the peripheral blood and organs in mice. These data demonstrate effective and safe use of Ad-Cab FT, bidding for further clinical investigation.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2023-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/9e/30/main.PMC9995465.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9102590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Multi-modal efficacy of a chimeric vesiculovirus expressing the Morreton glycoprotein in sarcoma. 表达莫勒顿糖蛋白的嵌合囊状病毒对肉瘤的多模式疗效。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2023-03-01 eCollection Date: 2023-06-15 DOI: 10.1016/j.omto.2023.02.009
Chelsae R Watters, Oumar Barro, Natalie M Elliott, Yumei Zhou, Musa Gabere, Elizabeth Raupach, Alexander T Baker, Michael T Barrett, Kenneth H Buetow, Bertram Jacobs, Mahesh Seetharam, Mitesh J Borad, Bolni Marius Nagalo

Vesiculoviruses are attractive oncolytic virus platforms due to their rapid replication, appreciable transgene capacity, broad tropism, limited preexisting immunity, and tumor selectivity through type I interferon response defects in malignant cells. We developed a synthetic chimeric virus (VMG) expressing the glycoprotein (G) from Morreton virus (MorV) and utilizing the remaining structural genes from vesicular stomatitis virus (VSV). VMG exhibited in vitro efficacy by inducing oncolysis in a broad range of sarcoma subtypes across multiple species. Notably, all cell lines tested showed the ability of VMG to yield productive infection with rapid replication kinetics and induction of apoptosis. Furthermore, pilot safety evaluations of VMG in immunocompetent, non-tumor-bearing mice showed an absence of toxicity with intranasal doses as high as 1e10 50% tissue culture infectious dose (TCID50)/kg. Locoregional administration of VMG in vivo resulted in tumor reduction in an immunodeficient Ewing sarcoma xenograft at doses as low as 2e5 TCID50. In a murine syngeneic fibrosarcoma model, while no tumor inhibition was achieved with VMG, there was a robust induction of CD8+ T cells within the tumor. The studies described herein establish the promising potential for VMG to be used as a novel oncolytic virotherapy platform with anticancer effects in sarcoma.

膀胱病毒具有快速复制、可观的转基因能力、广泛的趋向性、有限的原有免疫力以及通过恶性细胞的 I 型干扰素反应缺陷产生的肿瘤选择性,因此是一种极具吸引力的溶瘤病毒平台。我们开发了一种合成嵌合病毒(VMG),它表达了莫勒顿病毒(MorV)的糖蛋白(G),并利用了水泡性口炎病毒(VSV)的其余结构基因。VMG 对多种亚型肉瘤都有体外诱导溶瘤的功效。值得注意的是,所有接受测试的细胞系都显示,VMG 能够通过快速复制动力学和诱导细胞凋亡产生有成效的感染。此外,在免疫功能正常的非肿瘤小鼠体内进行的 VMG 试验安全性评估显示,鼻内剂量高达 1e10 50%组织培养感染剂量(TCID50)/kg 时无毒性。在体内局部注射 VMG 可使免疫缺陷的尤文肉瘤异种移植中的肿瘤缩小,剂量低至 2e5 TCID50。在小鼠合成纤维肉瘤模型中,虽然 VMG 没有抑制肿瘤,但却能在肿瘤内强力诱导 CD8+ T 细胞。本文所述的研究为 VMG 作为一种新型溶瘤病毒治疗平台在肉瘤中发挥抗癌作用奠定了良好的基础。
{"title":"Multi-modal efficacy of a chimeric vesiculovirus expressing the Morreton glycoprotein in sarcoma.","authors":"Chelsae R Watters, Oumar Barro, Natalie M Elliott, Yumei Zhou, Musa Gabere, Elizabeth Raupach, Alexander T Baker, Michael T Barrett, Kenneth H Buetow, Bertram Jacobs, Mahesh Seetharam, Mitesh J Borad, Bolni Marius Nagalo","doi":"10.1016/j.omto.2023.02.009","DOIUrl":"10.1016/j.omto.2023.02.009","url":null,"abstract":"<p><p>Vesiculoviruses are attractive oncolytic virus platforms due to their rapid replication, appreciable transgene capacity, broad tropism, limited preexisting immunity, and tumor selectivity through type I interferon response defects in malignant cells. We developed a synthetic chimeric virus (VMG) expressing the glycoprotein (G) from Morreton virus (MorV) and utilizing the remaining structural genes from vesicular stomatitis virus (VSV). VMG exhibited <i>in vitro</i> efficacy by inducing oncolysis in a broad range of sarcoma subtypes across multiple species. Notably, all cell lines tested showed the ability of VMG to yield productive infection with rapid replication kinetics and induction of apoptosis. Furthermore, pilot safety evaluations of VMG in immunocompetent, non-tumor-bearing mice showed an absence of toxicity with intranasal doses as high as 1e10 50% tissue culture infectious dose (TCID<sub>50</sub>)/kg. Locoregional administration of VMG <i>in vivo</i> resulted in tumor reduction in an immunodeficient Ewing sarcoma xenograft at doses as low as 2e5 TCID<sub>50</sub>. In a murine syngeneic fibrosarcoma model, while no tumor inhibition was achieved with VMG, there was a robust induction of CD8+ T cells within the tumor. The studies described herein establish the promising potential for VMG to be used as a novel oncolytic virotherapy platform with anticancer effects in sarcoma.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/71/f4/main.PMC10033453.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9245945","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multivalent in vivo delivery of DNA-encoded bispecific T cell engagers effectively controls heterogeneous GBM tumors and mitigates immune escape. DNA 编码的双特异性 T 细胞吞噬因子的多价体内递送可有效控制异质性 GBM 肿瘤并减轻免疫逃逸。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2023-02-16 eCollection Date: 2023-03-16 DOI: 10.1016/j.omto.2023.02.004
Daniel H Park, Kevin Liaw, Pratik Bhojnagarwala, Xizhou Zhu, Jihae Choi, Ali R Ali, Devivasha Bordoloi, Ebony N Gary, Ryan P O'Connell, Abhijeet Kulkarni, Diana Guimet, Trevor Smith, Alfredo Perales-Puchalt, Ami Patel, David B Weiner

Glioblastoma multiforme (GBM) is among the most difficult cancers to treat with a 5-year survival rate less than 5%. An immunotherapeutic vaccine approach targeting GBM-specific antigen, EGFRvIII, previously demonstrated important clinical impact. However, immune escape variants were reported in the trial, suggesting that multivalent approaches targeting GBM-associated antigens may be of importance. Here we focused on multivalent in vivo delivery of synthetic DNA-encoded bispecific T cell engagers (DBTEs) targeting two GBM-associated antigens, EGFRvIII and HER2. We designed and optimized an EGFRvIII-DBTE that induced T cell-mediated cytotoxicity against EGFRvIII-expressing tumor cells. In vivo delivery in a single administration of EGFRvIII-DBTE resulted in durable expression over several months in NSG mice and potent tumor control and clearance in both peripheral and orthotopic animal models of GBM. Next, we combined delivery of EGFRvIII-DBTEs with an HER2-targeting DBTE to treat heterogeneous GBM tumors. In vivo delivery of dual DBTEs targeting these two GBM-associated antigens exhibited enhanced tumor control and clearance in a heterogeneous orthotopic GBM challenge, while treatment with single-target DBTE ultimately allowed for tumor escape. These studies support that combined delivery of DBTEs, targeting both EGFRvIII and HER2, can potentially improve outcomes of GBM immunotherapy, and such multivalent approaches deserve additional study.

多形性胶质母细胞瘤(GBM)是最难治疗的癌症之一,5 年生存率不到 5%。一种针对 GBM 特异性抗原 EGFRvIII 的免疫治疗疫苗方法曾显示出重要的临床效果。然而,试验中出现了免疫逃逸变异,这表明针对 GBM 相关抗原的多价方法可能具有重要意义。在这里,我们重点研究了针对两种 GBM 相关抗原(表皮生长因子受体阻断因子 vIII 和 HER2)的合成 DNA 编码双特异性 T 细胞吞噬因子(DBTE)的多价体内递送。我们设计并优化了一种 EGFRvIII-DBTE,它能诱导 T 细胞介导的细胞毒性,对抗表达 EGFRvIII 的肿瘤细胞。一次性体内给药 EGFRvIII-DBTE 可在 NSG 小鼠体内持续表达数月,并在外周和正位 GBM 动物模型中有效控制和清除肿瘤。接下来,我们将 EGFRvIII-DBTE 与 HER2 靶向 DBTE 结合起来,治疗异质性 GBM 肿瘤。针对这两种GBM相关抗原的双DBTE体内给药在异质性正位GBM挑战中显示出更强的肿瘤控制和清除能力,而单靶点DBTE治疗最终允许肿瘤逃逸。这些研究证明,同时靶向表皮生长因子受体vIII和HER2的DBTEs联合给药有可能改善GBM免疫疗法的效果,这种多价方法值得进一步研究。
{"title":"Multivalent <i>in vivo</i> delivery of DNA-encoded bispecific T cell engagers effectively controls heterogeneous GBM tumors and mitigates immune escape.","authors":"Daniel H Park, Kevin Liaw, Pratik Bhojnagarwala, Xizhou Zhu, Jihae Choi, Ali R Ali, Devivasha Bordoloi, Ebony N Gary, Ryan P O'Connell, Abhijeet Kulkarni, Diana Guimet, Trevor Smith, Alfredo Perales-Puchalt, Ami Patel, David B Weiner","doi":"10.1016/j.omto.2023.02.004","DOIUrl":"10.1016/j.omto.2023.02.004","url":null,"abstract":"<p><p>Glioblastoma multiforme (GBM) is among the most difficult cancers to treat with a 5-year survival rate less than 5%. An immunotherapeutic vaccine approach targeting GBM-specific antigen, EGFRvIII, previously demonstrated important clinical impact. However, immune escape variants were reported in the trial, suggesting that multivalent approaches targeting GBM-associated antigens may be of importance. Here we focused on multivalent <i>in vivo</i> delivery of synthetic DNA-encoded bispecific T cell engagers (DBTEs) targeting two GBM-associated antigens, EGFRvIII and HER2. We designed and optimized an EGFRvIII-DBTE that induced T cell-mediated cytotoxicity against EGFRvIII-expressing tumor cells. <i>In vivo</i> delivery in a single administration of EGFRvIII-DBTE resulted in durable expression over several months in NSG mice and potent tumor control and clearance in both peripheral and orthotopic animal models of GBM. Next, we combined delivery of EGFRvIII-DBTEs with an HER2-targeting DBTE to treat heterogeneous GBM tumors. <i>In vivo</i> delivery of dual DBTEs targeting these two GBM-associated antigens exhibited enhanced tumor control and clearance in a heterogeneous orthotopic GBM challenge, while treatment with single-target DBTE ultimately allowed for tumor escape. These studies support that combined delivery of DBTEs, targeting both EGFRvIII and HER2, can potentially improve outcomes of GBM immunotherapy, and such multivalent approaches deserve additional study.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2023-02-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10006507/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9114261","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reprogramming the tumor microenvironment leverages CD8+ T cell responses to a shared tumor/self antigen in ovarian cancer. 重编程肿瘤微环境可利用 CD8+ T 细胞对卵巢癌中肿瘤/自身共同抗原的反应。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2023-02-09 eCollection Date: 2023-03-16 DOI: 10.1016/j.omto.2023.02.002
Anna Mistarz, Marta Winkler, Sebastiano Battaglia, Song Liu, Alan Hutson, Hanna Rokita, Andrea Gambotto, Kunle O Odunsi, Prashant K Singh, A J Robert McGray, Jianmin Wang, Danuta Kozbor

Tumor antigen-driven responses to weakly immunogenic self-antigens and neoantigens directly affect treatment efficacy following immunotherapy. Using orthotopically grown SV40 T antigen+ ovarian carcinoma in antigen-naive wild-type or TgMISIIR-TAg-Low transgenic mice expressing SV40 T antigen as a self-antigen, we investigated the impact of CXCR4-antagonist-armed oncolytic virotherapy on tumor progression and antitumor immunity. Immunostaining and single-cell RNA sequencing analyses of the peritoneal tumor microenvironment of untreated tumors in syngeneic wild-type mice revealed the presence of SV40 T antigen-specific CD8+ T cells, a balanced M1/M2 transcriptomic signature of tumor-associated macrophages, and immunostimulatory cancer-associated fibroblasts. This contrasted with polarized M2 tumor-associated macrophages, immunosuppressive cancer-associated fibroblasts, and poor immune activation in TgMISIIR-TAg-Low mice. Intraperitoneal delivery of CXCR4-antagonist-armed oncolytic vaccinia virus led to nearly complete depletion of cancer-associated fibroblasts, M1 polarization of macrophages, and generation of SV40 T antigen-specific CD8+ T cells in transgenic mice. Cell depletion studies revealed that the therapeutic effect of armed oncolytic virotherapy was dependent primarily on CD8+ cells. These results demonstrate that targeting the interaction between immunosuppressive cancer-associated fibroblasts and macrophages in the tolerogenic tumor microenvironment by CXCR4-A-armed oncolytic virotherapy induces tumor/self-specific CD8+ T cell responses and consequently increases therapeutic efficacy in an immunocompetent ovarian cancer model.

肿瘤抗原驱动的对弱免疫原性自身抗原和新抗原的反应会直接影响免疫疗法的疗效。我们利用在无抗原的野生型小鼠或表达 SV40 T 抗原作为自身抗原的 TgMISIIR-TAg-Low 转基因小鼠体内直立生长的 SV40 T 抗原+卵巢癌,研究了 CXCR4-拮抗剂武装的溶瘤病毒疗法对肿瘤进展和抗肿瘤免疫的影响。免疫染色和单细胞 RNA 测序分析显示,共生野生型小鼠腹膜肿瘤微环境中存在 SV40 T 抗原特异性 CD8+ T 细胞、肿瘤相关巨噬细胞的 M1/M2 平衡转录组特征以及免疫刺激性癌症相关成纤维细胞。这与 TgMISIIR-TAg-Low 小鼠极化的 M2 肿瘤相关巨噬细胞、免疫抑制性癌症相关成纤维细胞和较差的免疫激活形成鲜明对比。通过腹腔注射 CXCR4 拮抗剂武装的溶瘤疫苗病毒,转基因小鼠体内的癌相关成纤维细胞几乎完全耗竭,巨噬细胞极化为 M1,并产生 SV40 T 抗原特异性 CD8+ T 细胞。细胞耗竭研究表明,武装溶瘤病毒疗法的治疗效果主要取决于 CD8+ 细胞。这些结果表明,在免疫功能正常的卵巢癌模型中,CXCR4-A-武装溶瘤病毒疗法能针对耐受性肿瘤微环境中免疫抑制性癌症相关成纤维细胞和巨噬细胞之间的相互作用,诱导肿瘤/自身特异性CD8+ T细胞应答,从而提高疗效。
{"title":"Reprogramming the tumor microenvironment leverages CD8<sup>+</sup> T cell responses to a shared tumor/self antigen in ovarian cancer.","authors":"Anna Mistarz, Marta Winkler, Sebastiano Battaglia, Song Liu, Alan Hutson, Hanna Rokita, Andrea Gambotto, Kunle O Odunsi, Prashant K Singh, A J Robert McGray, Jianmin Wang, Danuta Kozbor","doi":"10.1016/j.omto.2023.02.002","DOIUrl":"10.1016/j.omto.2023.02.002","url":null,"abstract":"<p><p>Tumor antigen-driven responses to weakly immunogenic self-antigens and neoantigens directly affect treatment efficacy following immunotherapy. Using orthotopically grown SV40 T antigen<sup>+</sup> ovarian carcinoma in antigen-naive wild-type or Tg<i>MISIIR-TAg-Low</i> transgenic mice expressing SV40 T antigen as a self-antigen, we investigated the impact of CXCR4-antagonist-armed oncolytic virotherapy on tumor progression and antitumor immunity. Immunostaining and single-cell RNA sequencing analyses of the peritoneal tumor microenvironment of untreated tumors in syngeneic wild-type mice revealed the presence of SV40 T antigen-specific CD8<sup>+</sup> T cells, a balanced M1/M2 transcriptomic signature of tumor-associated macrophages, and immunostimulatory cancer-associated fibroblasts. This contrasted with polarized M2 tumor-associated macrophages, immunosuppressive cancer-associated fibroblasts, and poor immune activation in Tg<i>MISIIR-TAg-Low</i> mice. Intraperitoneal delivery of CXCR4-antagonist-armed oncolytic vaccinia virus led to nearly complete depletion of cancer-associated fibroblasts, M1 polarization of macrophages, and generation of SV40 T antigen-specific CD8<sup>+</sup> T cells in transgenic mice. Cell depletion studies revealed that the therapeutic effect of armed oncolytic virotherapy was dependent primarily on CD8<sup>+</sup> cells. These results demonstrate that targeting the interaction between immunosuppressive cancer-associated fibroblasts and macrophages in the tolerogenic tumor microenvironment by CXCR4-A-armed oncolytic virotherapy induces tumor/self-specific CD8<sup>+</sup> T cell responses and consequently increases therapeutic efficacy in an immunocompetent ovarian cancer model.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2023-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9982455/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9759551","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
esRAGE-expressing oHSV enhances anti-tumor efficacy by inhibition of endothelial cell activation. 表达 esRAGE 的 oHSV 可通过抑制内皮细胞活化提高抗肿瘤疗效。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2023-01-16 eCollection Date: 2023-03-16 DOI: 10.1016/j.omto.2023.01.003
Jessica Swanner, Ji Seon Shim, Kimberly A Rivera-Caraballo, Karina Vázquez-Arreguín, Bangxing Hong, Alberto J Bueso-Perez, Tae Jin Lee, Yeshavanth Kumar Banasavadi-Siddegowda, Balveen Kaur, Ji Young Yoo

High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) molecule that plays an important role in inflammation and tumorigenesis. Receptor for advanced glycation end products (RAGE) is one of the major receptors to which extracellular HMGB1 binds to mediate its activity. RAGE is highly expressed on the endothelial cells (ECs) and regulates endothelial permeability during inflammation. Here, we introduced the endogenous secretory form of RAGE (esRAGE) as a decoy receptor for RAGE ligands into an oncolytic herpes simplex virus 1 (oHSV) (OVesRAGE), which, upon release, can function to block RAGE signaling. OVesRAGE significantly decreased phosphorylation of MEK1/2 and Erk and increased cleaved PARP in glioblastoma (GBM) cells in vitro and in vivo. oHSV-infected GBM cells co-cultured with ECs were used to test OVesRAGE effect on EC activation, vessel leakiness, virus replication, and tumor cell killing. OVesRAGE could effectively secrete esRAGE and rescue virus-induced EC migration and activation. Reduced EC activation facilitated virus replication in tumor cells when co-cultured with ECs. Finally, OVesRAGE significantly enhanced therapeutic efficacy in GBM-bearing mice. Collectively, our data demonstrate that HMGB1-RAGE signaling could be a promising target and that its inhibition is a feasible approach to improve the efficacy of oHSV therapy.

高迁移率基团框 1(HMGB1)是一种损伤相关分子模式(DAMP)分子,在炎症和肿瘤发生中发挥着重要作用。高级糖化终产物受体(RAGE)是细胞外 HMGB1 与之结合以介导其活性的主要受体之一。RAGE 在内皮细胞(EC)上高度表达,并在炎症过程中调节内皮的通透性。在这里,我们将 RAGE 的内源性分泌形式(esRAGE)作为 RAGE 配体的诱饵受体引入溶瘤性单纯疱疹病毒 1(oHSV)(OVesRAGE)。OVesRAGE 在体外和体内都能显著降低胶质母细胞瘤(GBM)细胞中 MEK1/2 和 Erk 的磷酸化,并增加 PARP 的裂解。OVesRAGE能有效分泌esRAGE,挽救病毒诱导的EC迁移和活化。当肿瘤细胞与心血管细胞共培养时,心血管细胞活化程度的降低有利于病毒在肿瘤细胞中的复制。最后,OVesRAGE 能显著提高对患 GBM 小鼠的疗效。总之,我们的数据表明,HMGB1-RAGE 信号转导可能是一个很有前景的靶点,抑制它是提高 oHSV 治疗效果的可行方法。
{"title":"esRAGE-expressing oHSV enhances anti-tumor efficacy by inhibition of endothelial cell activation.","authors":"Jessica Swanner, Ji Seon Shim, Kimberly A Rivera-Caraballo, Karina Vázquez-Arreguín, Bangxing Hong, Alberto J Bueso-Perez, Tae Jin Lee, Yeshavanth Kumar Banasavadi-Siddegowda, Balveen Kaur, Ji Young Yoo","doi":"10.1016/j.omto.2023.01.003","DOIUrl":"10.1016/j.omto.2023.01.003","url":null,"abstract":"<p><p>High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) molecule that plays an important role in inflammation and tumorigenesis. Receptor for advanced glycation end products (RAGE) is one of the major receptors to which extracellular HMGB1 binds to mediate its activity. RAGE is highly expressed on the endothelial cells (ECs) and regulates endothelial permeability during inflammation. Here, we introduced the endogenous secretory form of RAGE (esRAGE) as a decoy receptor for RAGE ligands into an oncolytic herpes simplex virus 1 (oHSV) (OVesRAGE), which, upon release, can function to block RAGE signaling. OVesRAGE significantly decreased phosphorylation of MEK1/2 and Erk and increased cleaved PARP in glioblastoma (GBM) cells <i>in vitro</i> and <i>in vivo</i>. oHSV-infected GBM cells co-cultured with ECs were used to test OVesRAGE effect on EC activation, vessel leakiness, virus replication, and tumor cell killing. OVesRAGE could effectively secrete esRAGE and rescue virus-induced EC migration and activation. Reduced EC activation facilitated virus replication in tumor cells when co-cultured with ECs. Finally, OVesRAGE significantly enhanced therapeutic efficacy in GBM-bearing mice. Collectively, our data demonstrate that HMGB1-RAGE signaling could be a promising target and that its inhibition is a feasible approach to improve the efficacy of oHSV therapy.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2023-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9918391/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9203029","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development and comparison of 68Ga/18F/64Cu-labeled nanobody tracers probing Claudin18.2. 探测claud18.2的68Ga/18F/ 64cu标记纳米体示踪剂的研制与比较
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2022-12-15 DOI: 10.1016/j.omto.2022.11.003
Weijun Wei, Di Zhang, You Zhang, Lianghua Li, Yuchen Jin, Shuxian An, Chun Lv, Haitao Zhao, Cheng Wang, Yanshan Huang, Jiali Dong, Gang Huang, Jianjun Liu

Claudin 18.2 (CLDN18.2) is an emerging target for the treatment of gastric cancers. We aim to develop tracers to image the expression of CLDN18.2. A humanized nanobody targeting CLDN18.2 (clone hu19V3) was produced and labeled with 68Ga, 64Cu, and 18F. The tracers were investigated in subcutaneous and metastatic models established using two different mouse types (nude and Balb/c mice) and two different cell lines (CHO-CLDN18.2 and CT26-CLDN18.2). Gastric cancer patient-derived xenograft (PDX) models were further established for validation experiments. Three novel CLDN18.2-targeted tracers (i.e., [68Ga]Ga-NOTA-hu19V3, [64Cu]Cu-NOTA-hu19V3, and [18F]F-hu19V3) were developed with good radiochemical yields and excellent radiochemical purities. [68Ga]Ga-NOTA-hu19V3 immuno-positron emission tomography (immunoPET) rapidly delineated subcutaneous CHO-CLDN18.2 lesions and CT26-CLDN18.2 tumors, as well as showing excellent diagnostic value in PDX models naturally expressing CLDN18.2. While [68Ga]Ga-NOTA-hu19V3 had high kidney accumulation, [64Cu]Cu-NOTA-hu19V3 showed reduced kidney accumulation and improved image contrast at late time points. Moreover, [18F]F-hu19V3 was developed via click chemistry reaction under mild conditions and precisely disseminated CHO-CLDN18.2 lesions in the lungs. Furthermore, region of interest analysis, biodistribution study, and histopathological staining results correlated well with the in vivo imaging results. Taken together, immunoPET imaging with the three tracers can reliably visualize CLDN18.2 expression.

Claudin 18.2 (CLDN18.2)是一个新兴的胃癌治疗靶点。我们的目标是开发示踪剂来成像CLDN18.2的表达。制备了靶向CLDN18.2的人源化纳米体(克隆hu19V3),并用68Ga、64Cu和18F标记。该示踪剂在两种不同小鼠类型(裸小鼠和Balb/c小鼠)和两种不同细胞系(CHO-CLDN18.2和CT26-CLDN18.2)建立的皮下和转移模型中进行了研究。进一步建立胃癌患者源性异种移植瘤(PDX)模型进行验证实验。开发了三种新型cldn18.2靶向示踪剂(即[68Ga]Ga-NOTA-hu19V3, [64Cu]Cu-NOTA-hu19V3和[18F]F-hu19V3),具有良好的放射化学产率和优异的放射化学纯度。[68Ga]Ga-NOTA-hu19V3免疫正电子发射断层扫描(immunoPET)快速描绘皮下CHO-CLDN18.2病变和CT26-CLDN18.2肿瘤,并在自然表达CLDN18.2的PDX模型中显示出良好的诊断价值。[68Ga]Ga-NOTA-hu19V3肾脏堆积较多,[64Cu]Cu-NOTA-hu19V3肾脏堆积较少,后期图像对比度提高。此外,[18F]F-hu19V3在温和条件下通过点击化学反应产生,并在肺部精确播散CHO-CLDN18.2病变。此外,兴趣区分析、生物分布研究和组织病理学染色结果与体内成像结果具有良好的相关性。综上所述,三种示踪剂的免疫pet成像可以可靠地显示CLDN18.2的表达。
{"title":"Development and comparison of <sup>68</sup>Ga/<sup>18</sup>F/<sup>64</sup>Cu-labeled nanobody tracers probing Claudin18.2.","authors":"Weijun Wei,&nbsp;Di Zhang,&nbsp;You Zhang,&nbsp;Lianghua Li,&nbsp;Yuchen Jin,&nbsp;Shuxian An,&nbsp;Chun Lv,&nbsp;Haitao Zhao,&nbsp;Cheng Wang,&nbsp;Yanshan Huang,&nbsp;Jiali Dong,&nbsp;Gang Huang,&nbsp;Jianjun Liu","doi":"10.1016/j.omto.2022.11.003","DOIUrl":"https://doi.org/10.1016/j.omto.2022.11.003","url":null,"abstract":"<p><p>Claudin 18.2 (CLDN18.2) is an emerging target for the treatment of gastric cancers. We aim to develop tracers to image the expression of CLDN18.2. A humanized nanobody targeting CLDN18.2 (clone hu19V3) was produced and labeled with <sup>68</sup>Ga, <sup>64</sup>Cu, and <sup>18</sup>F. The tracers were investigated in subcutaneous and metastatic models established using two different mouse types (nude and Balb/c mice) and two different cell lines (CHO-CLDN18.2 and CT26-CLDN18.2). Gastric cancer patient-derived xenograft (PDX) models were further established for validation experiments. Three novel CLDN18.2-targeted tracers (i.e., [<sup>68</sup>Ga]Ga-NOTA-hu19V3, [<sup>64</sup>Cu]Cu-NOTA-hu19V3, and [<sup>18</sup>F]F-hu19V3) were developed with good radiochemical yields and excellent radiochemical purities. [<sup>68</sup>Ga]Ga-NOTA-hu19V3 immuno-positron emission tomography (immunoPET) rapidly delineated subcutaneous CHO-CLDN18.2 lesions and CT26-CLDN18.2 tumors, as well as showing excellent diagnostic value in PDX models naturally expressing CLDN18.2. While [<sup>68</sup>Ga]Ga-NOTA-hu19V3 had high kidney accumulation, [<sup>64</sup>Cu]Cu-NOTA-hu19V3 showed reduced kidney accumulation and improved image contrast at late time points. Moreover, [<sup>18</sup>F]F-hu19V3 was developed via click chemistry reaction under mild conditions and precisely disseminated CHO-CLDN18.2 lesions in the lungs. Furthermore, region of interest analysis, biodistribution study, and histopathological staining results correlated well with the <i>in vivo</i> imaging results. Taken together, immunoPET imaging with the three tracers can reliably visualize CLDN18.2 expression.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2022-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/1a/51/main.PMC9747674.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10497609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Oncolytic Urabe mumps virus: A promising virotherapy for triple-negative breast cancer. 溶瘤性乌拉伯腮腺炎病毒:一种有前途的三阴性乳腺癌病毒疗法。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2022-12-15 DOI: 10.1016/j.omto.2022.11.002
Marshall D Behrens, Robert J Stiles, Gennett M Pike, Laura A Sikkink, Yongxian Zhuang, Jia Yu, Liewei Wang, Judy C Boughey, Matthew P Goetz, Mark J Federspiel

Historically, the clinical utility of oncolytic virotherapy as a treatment for a wide range of cancer types was first demonstrated by three pilot human clinical trials conducted in Japan in the 1970s and 1980s using a wild-type Urabe mumps virus (MuV) clinical isolate. Using a sample of the actual original oncolytic Urabe MuV clinical trial virus stock (MuV-U-Japan) used in these Japanese clinical trials, we found that MuV-U-Japan consisted of a wide variety of very closely related Urabe MuVs that differed by an average of only three amino acids. Two MuV-U-Japan isolates, MuV-UA and MuV-UC, potently killed a panel of established human breast cancer cell lines in vitro, significantly extended survival of nude mice with human triple-negative breast cancer (TNBC) MDA-MB-231 tumor xenografts in vivo, and demonstrated significant killing activity against breast cancer patient-derived xenograft (PDX) cell lines grown as 3D organoids, including PDXs from patients resistant to anthracycline- and taxane-based chemotherapy. We also report success in developing a large-scale MuV-U production and purification process suitable for supporting Investigational New Drug applications for clinical trials. This study demonstrates the suitability of the MuV-UC virus for translation to modern clinical trials for treating patients with TNBC.

从历史上看,溶瘤病毒疗法作为一种治疗多种癌症类型的临床应用,最早是在20世纪70年代和80年代在日本使用野型Urabe腮腺炎病毒(MuV)临床分离物进行的三次试点人体临床试验中得到证实的。使用这些日本临床试验中使用的实际原始溶瘤性Urabe MuV临床试验病毒库存(MuV- u - japan)的样本,我们发现MuV- u - japan由各种非常密切相关的Urabe MuV组成,平均只有三个氨基酸不同。两株MuV-U-Japan分离株MuV-UA和MuV-UC在体外有效地杀死了一组已建立的人乳腺癌细胞系,在体内显著延长了人三阴性乳腺癌(TNBC) MDA-MB-231肿瘤异种移植物裸鼠的存活时间,并对生长为3D类器官的乳腺癌患者来源的异种移植物(PDX)细胞系显示出显著的杀伤活性,包括对蒽环类和紫杉烷类化疗耐药的患者的PDX。我们还成功开发了大规模的MuV-U生产和纯化工艺,适用于支持临床试验新药申请。这项研究证明了MuV-UC病毒用于治疗TNBC患者的现代临床试验的适用性。
{"title":"Oncolytic Urabe mumps virus: A promising virotherapy for triple-negative breast cancer.","authors":"Marshall D Behrens,&nbsp;Robert J Stiles,&nbsp;Gennett M Pike,&nbsp;Laura A Sikkink,&nbsp;Yongxian Zhuang,&nbsp;Jia Yu,&nbsp;Liewei Wang,&nbsp;Judy C Boughey,&nbsp;Matthew P Goetz,&nbsp;Mark J Federspiel","doi":"10.1016/j.omto.2022.11.002","DOIUrl":"https://doi.org/10.1016/j.omto.2022.11.002","url":null,"abstract":"<p><p>Historically, the clinical utility of oncolytic virotherapy as a treatment for a wide range of cancer types was first demonstrated by three pilot human clinical trials conducted in Japan in the 1970s and 1980s using a wild-type Urabe mumps virus (MuV) clinical isolate. Using a sample of the actual original oncolytic Urabe MuV clinical trial virus stock (MuV-U-Japan) used in these Japanese clinical trials, we found that MuV-U-Japan consisted of a wide variety of very closely related Urabe MuVs that differed by an average of only three amino acids. Two MuV-U-Japan isolates, MuV-UA and MuV-UC, potently killed a panel of established human breast cancer cell lines <i>in vitro</i>, significantly extended survival of nude mice with human triple-negative breast cancer (TNBC) MDA-MB-231 tumor xenografts <i>in vivo</i>, and demonstrated significant killing activity against breast cancer patient-derived xenograft (PDX) cell lines grown as 3D organoids, including PDXs from patients resistant to anthracycline- and taxane-based chemotherapy. We also report success in developing a large-scale MuV-U production and purification process suitable for supporting Investigational New Drug applications for clinical trials. This study demonstrates the suitability of the MuV-UC virus for translation to modern clinical trials for treating patients with TNBC.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2022-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/5d/95/main.PMC9703006.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10702301","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 4
A library of cancer testis specific T cell receptors for T cell receptor gene therapy. 癌症睾丸特异性T细胞受体库,用于T细胞受体基因治疗。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2022-12-02 eCollection Date: 2023-03-16 DOI: 10.1016/j.omto.2022.11.007
Marije A J de Rooij, Dennis F G Remst, Dirk M van der Steen, Anne K Wouters, Renate S Hagedoorn, Michel G D Kester, Miranda H Meeuwsen, Tassilo L A Wachsmann, Arnoud H de Ru, Peter A van Veelen, Els M E Verdegaal, J H Frederik Falkenburg, Mirjam H M Heemskerk

To increase the number of cancer patients that can be treated with T cell receptor (TCR) gene therapy, we aimed to identify a set of high-affinity cancer-specific TCRs targeting different melanoma-associated antigens (MAGEs). In this study, peptides derived from MAGE genes with tumor-specific expression pattern were identified by human leukocyte antigen (HLA) peptidomics. Next, peptide-HLA tetramers were generated, and used to sort MAGE-specific CD8+ T cell clones from the allogeneic (allo) HLA repertoire of healthy donors. To evaluate the clinical potential, most potent TCRs were sequenced, transferred into peripheral blood-derived CD8+ T cells, and tested for antitumor efficacy. In total we identified, seven MAGE-specific TCRs that effectively target MAGE-A1, MAGE-A3, MAGE-A6, and MAGE-A9 in the context of HLA-A∗01:01, -A∗02:01, -A∗03:01, -B∗07:02, -B∗35:01, or -C∗07:02. TCR gene transfer into CD8⁺ T cells resulted in efficient reactivity against a variety of different tumor types, while no cross-reactivity was detected. In addition, major in vivo antitumor effects of MAGE-A1 specific TCR engineered CD8⁺ T cells were observed in the orthotopic xenograft model for established multiple myeloma. The identification of seven MAGE-specific TCRs expands the pool of cancer patients eligible for TCR gene therapy and increases possibilities for personalized TCR gene therapy.

为了增加可以接受T细胞受体(TCR)基因治疗的癌症患者的数量,我们旨在鉴定一组针对不同黑色素瘤相关抗原(MAGE)的高亲和力癌症特异性TCR。在本研究中,通过人类白细胞抗原(HLA)肽组学鉴定了具有肿瘤特异性表达模式的MAGE基因衍生的肽。接下来,产生肽HLA四聚体,并用于从健康供体的异基因(allo)HLA库中分选MAGE特异性CD8+T细胞克隆。为了评估临床潜力,对最有效的TCR进行测序,转移到外周血来源的CD8+T细胞中,并测试其抗肿瘤功效。我们总共确定了7个MAGE特异性TCR,它们在HLA-A*01:01、-A*02:01、-A*03:01、-B*07:02、-B*35:01或-C*07:02中有效靶向MAGE-A1、MAGE-A3、MAGE-A6和MAGE-A9。TCR基因转染CD8⁺ T细胞对各种不同的肿瘤类型产生有效的反应性,而没有检测到交叉反应性。此外,MAGE-A1特异性TCR工程化CD8的主要体内抗肿瘤作用⁺ 在已建立的多发性骨髓瘤的原位异种移植物模型中观察到T细胞。七种MAGE特异性TCR的鉴定扩大了有资格接受TCR基因治疗的癌症患者的范围,并增加了个性化TCR基因疗法的可能性。
{"title":"A library of cancer testis specific T cell receptors for T cell receptor gene therapy.","authors":"Marije A J de Rooij,&nbsp;Dennis F G Remst,&nbsp;Dirk M van der Steen,&nbsp;Anne K Wouters,&nbsp;Renate S Hagedoorn,&nbsp;Michel G D Kester,&nbsp;Miranda H Meeuwsen,&nbsp;Tassilo L A Wachsmann,&nbsp;Arnoud H de Ru,&nbsp;Peter A van Veelen,&nbsp;Els M E Verdegaal,&nbsp;J H Frederik Falkenburg,&nbsp;Mirjam H M Heemskerk","doi":"10.1016/j.omto.2022.11.007","DOIUrl":"10.1016/j.omto.2022.11.007","url":null,"abstract":"<p><p>To increase the number of cancer patients that can be treated with T cell receptor (TCR) gene therapy, we aimed to identify a set of high-affinity cancer-specific TCRs targeting different melanoma-associated antigens (MAGEs). In this study, peptides derived from <i>MAGE</i> genes with tumor-specific expression pattern were identified by human leukocyte antigen (HLA) peptidomics. Next, peptide-HLA tetramers were generated, and used to sort MAGE-specific CD8<sup>+</sup> T cell clones from the allogeneic (allo) HLA repertoire of healthy donors. To evaluate the clinical potential, most potent TCRs were sequenced, transferred into peripheral blood-derived CD8<sup>+</sup> T cells, and tested for antitumor efficacy. In total we identified, seven MAGE-specific TCRs that effectively target MAGE-A1, MAGE-A3, MAGE-A6, and MAGE-A9 in the context of HLA-A∗01:01, -A∗02:01, -A∗03:01, -B∗07:02, -B∗35:01, or -C∗07:02. TCR gene transfer into CD8⁺ T cells resulted in efficient reactivity against a variety of different tumor types, while no cross-reactivity was detected. In addition, major <i>in vivo</i> antitumor effects of MAGE-A1 specific TCR engineered CD8⁺ T cells were observed in the orthotopic xenograft model for established multiple myeloma. The identification of seven MAGE-specific TCRs expands the pool of cancer patients eligible for TCR gene therapy and increases possibilities for personalized TCR gene therapy.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2022-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9792401/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10833445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 4
Polycytidine tract deletion from microRNA-detargeted oncolytic Mengovirus optimizes the therapeutic index in a murine multiple myeloma model. 在小鼠多发性骨髓瘤模型中,微RNA靶向溶瘤孟病毒的多胞苷道缺失优化了治疗指数。
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2022-12-02 eCollection Date: 2023-03-16 DOI: 10.1016/j.omto.2022.11.006
Velia Penza, Justin W Maroun, Rebecca A Nace, Autumn J Schulze, Stephen J Russell

Mengovirus is an oncolytic picornavirus whose broad host range allows for testing in immunocompetent cancer models. Two pathogenicity-ablating approaches, polycytidine (polyC) tract truncation and microRNA (miRNA) targets insertion, eliminated the risk of encephalomyocarditis. To investigate whether a polyC truncated, miRNA-detargeted oncolytic Mengovirus might be boosted, we partially or fully rebuilt the polyC tract into the 5' noncoding region (NCR) of polyC-deleted (MC0) oncolytic constructs (NC) carrying miRNA target (miRT) insertions to eliminate cardiac/muscular (miR-133b and miR-208a) and neuronal (miR-124) tropisms. PolyC-reconstituted viruses (MC24-NC and MC37-NC) replicated in vitro and showed the expected tropism restrictions, but reduced cytotoxicity and miRT deletions were frequently observed. In the MPC-11 immune competent mouse plasmacytoma model, both intratumoral and systemic administration of MC0-NC led to faster tumor responses than MC24-NC or MC37-NC, with combined durable complete response rates of 75%, 0.5%, and 30%, respectively. Secondary viremia was higher following MC0-NC versus MC24-NC or MC37-NC therapy. Sequence analysis of virus progeny from treated mice revealed a high prevalence of miRT sequences loss among MC24- and MC37- viral genomes, but not in MC0-NC. Overall, MC0-NC was capable of stably retaining miRT sites and provided a more effective treatment and is therefore our lead Mengovirus candidate for clinical translation.

蒙哥马利病毒是一种溶瘤型皮卡病毒,其广泛的宿主范围允许在免疫功能正常的癌症模型中进行测试。聚胞苷(polyC)道截断和微RNA(miRNA)靶标插入这两种致病性消除方法消除了脑心肌炎的风险。为了研究polyC截短、miRNA靶向的溶瘤孟病毒是否会增强毒性,我们在携带miRNA靶点(miRT)插入物的polyC缺失(MC0)溶瘤构建体(NC)的5'非编码区(NCR)中部分或全部重建了polyC束,以消除心脏/肌肉(miR-133b和miR-208a)和神经元(miR-124)的致病性。PolyC重组病毒(MC24-NC和MC37-NC)可在体外复制,并显示出预期的趋向性限制,但细胞毒性降低,而且经常观察到miRT缺失。在 MPC-11 免疫功能正常的小鼠浆细胞瘤模型中,MC0-NC 的瘤内和全身给药均可导致比 MC24-NC 或 MC37-NC 更快的肿瘤反应,综合持久完全反应率分别为 75%、0.5% 和 30%。与 MC24-NC 或 MC37-NC 相比,MC0-NC 治疗后的继发性病毒血症更高。对接受治疗的小鼠的病毒后代进行的序列分析表明,在MC24-NC和MC37-NC病毒基因组中,miRT序列丢失的发生率很高,而在MC0-NC中则没有。总之,MC0-NC 能够稳定地保留 miRT 位点,并提供更有效的治疗,因此是我们用于临床转化的主要候选孟病毒。
{"title":"Polycytidine tract deletion from microRNA-detargeted oncolytic Mengovirus optimizes the therapeutic index in a murine multiple myeloma model.","authors":"Velia Penza, Justin W Maroun, Rebecca A Nace, Autumn J Schulze, Stephen J Russell","doi":"10.1016/j.omto.2022.11.006","DOIUrl":"10.1016/j.omto.2022.11.006","url":null,"abstract":"<p><p>Mengovirus is an oncolytic picornavirus whose broad host range allows for testing in immunocompetent cancer models. Two pathogenicity-ablating approaches, polycytidine (polyC) tract truncation and microRNA (miRNA) targets insertion, eliminated the risk of encephalomyocarditis. To investigate whether a polyC truncated, miRNA-detargeted oncolytic Mengovirus might be boosted, we partially or fully rebuilt the polyC tract into the 5' noncoding region (NCR) of polyC-deleted (MC<sub>0</sub>) oncolytic constructs (NC) carrying miRNA target (miRT) insertions to eliminate cardiac/muscular (miR-133b and miR-208a) and neuronal (miR-124) tropisms. PolyC-reconstituted viruses (MC<sub>24</sub>-NC and MC<sub>37</sub>-NC) replicated <i>in vitro</i> and showed the expected tropism restrictions, but reduced cytotoxicity and miRT deletions were frequently observed. In the MPC-11 immune competent mouse plasmacytoma model, both intratumoral and systemic administration of MC<sub>0</sub>-NC led to faster tumor responses than MC<sub>24</sub>-NC or MC<sub>37</sub>-NC, with combined durable complete response rates of 75%, 0.5%, and 30%, respectively. Secondary viremia was higher following MC<sub>0</sub>-NC versus MC<sub>24</sub>-NC or MC<sub>37</sub>-NC therapy. Sequence analysis of virus progeny from treated mice revealed a high prevalence of miRT sequences loss among MC<sub>24</sub>- and MC<sub>37</sub>- viral genomes, but not in MC<sub>0</sub>-NC. Overall, MC<sub>0</sub>-NC was capable of stably retaining miRT sites and provided a more effective treatment and is therefore our lead Mengovirus candidate for clinical translation.</p>","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2022-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/7a/f2/main.PMC9800256.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10513363","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Thank you to our 2022 reviewers 感谢我们2022年的评审
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2022-11-25 DOI: 10.1016/j.omto.2022.11.005
{"title":"Thank you to our 2022 reviewers","authors":"","doi":"10.1016/j.omto.2022.11.005","DOIUrl":"https://doi.org/10.1016/j.omto.2022.11.005","url":null,"abstract":"","PeriodicalId":18869,"journal":{"name":"Molecular Therapy Oncolytics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2022-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76347229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","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
Molecular Therapy Oncolytics
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