首页 > 最新文献

Oncoimmunology最新文献

英文 中文
Immunogenic cell death inducers and PD-1 blockade as neoadjuvant therapy for rectal cancer. 将免疫细胞死亡诱导剂和 PD-1 阻断剂作为直肠癌的新辅助疗法。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-17 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2416558
Yan Wang, Liwei Zhao, Zhen Zhang, Peng Liu

Immuno-oncological cancer management is shifting to neoadjuvant treatments. In patients with gastrointestinal cancers, particularly locally advanced rectal cancer, neoadjuvant chemoimmunotherapy often induce complete responses, hence avoiding surgical intervention. Recent clinical trials indicate that combinations of oxaliplatin-based chemotherapy and PD-1/PD-L1-targeting immunotherapy can be safely administered before surgery with curative intent.

免疫肿瘤治疗正转向新辅助治疗。对于胃肠道癌症患者,尤其是局部晚期直肠癌患者,新辅助化疗免疫疗法往往能诱导完全反应,从而避免手术干预。最近的临床试验表明,以奥沙利铂为基础的化疗和PD-1/PD-L1靶向免疫疗法的组合可以在手术前安全进行,并达到治愈目的。
{"title":"Immunogenic cell death inducers and PD-1 blockade as neoadjuvant therapy for rectal cancer.","authors":"Yan Wang, Liwei Zhao, Zhen Zhang, Peng Liu","doi":"10.1080/2162402X.2024.2416558","DOIUrl":"10.1080/2162402X.2024.2416558","url":null,"abstract":"<p><p>Immuno-oncological cancer management is shifting to neoadjuvant treatments. In patients with gastrointestinal cancers, particularly locally advanced rectal cancer, neoadjuvant chemoimmunotherapy often induce complete responses, hence avoiding surgical intervention. Recent clinical trials indicate that combinations of oxaliplatin-based chemotherapy and PD-1/PD-L1-targeting immunotherapy can be safely administered before surgery with curative intent.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2416558"},"PeriodicalIF":6.5,"publicationDate":"2024-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487966/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142478268","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
Trial watch: anticancer vaccination with dendritic cells. 试验观察:树突状细胞抗癌疫苗。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-09 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2412876
Francisca Borges, Raquel S Laureano, Isaure Vanmeerbeek, Jenny Sprooten, Octavie Demeulenaere, Jannes Govaerts, Lisa Kinget, Saurabh Saraswat, Benoit Beuselinck, Steven De Vleeschouwer, Paul Clement, Frederik De Smet, Rüdiger V Sorg, Angeliki Datsi, Nathalie Vigneron, Stefan Naulaerts, Abhishek D Garg

Dendritic cells (DCs) are critical players at the intersection of innate and adaptive immunity, making them ideal candidates for anticancer vaccine development. DC-based immunotherapies typically involve isolating patient-derived DCs, pulsing them with tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs), and utilizing maturation cocktails to ensure their effective activation. These matured DCs are then reinfused to elicit tumor-specific T-cell responses. While this approach has demonstrated the ability to generate potent immune responses, its clinical efficacy has been limited due to the immunosuppressive tumor microenvironment. Recent efforts have focused on enhancing the immunogenicity of DC-based vaccines, particularly through combination therapies with T cell-targeting immunotherapies. This Trial Watch summarizes recent advances in DC-based cancer treatments, including the development of new preclinical and clinical strategies, and discusses the future potential of DC-based vaccines in the evolving landscape of immuno-oncology.

树突状细胞(DC)是先天性免疫和适应性免疫交叉点上的关键角色,因此是开发抗癌疫苗的理想候选者。基于树突状细胞的免疫疗法通常包括分离患者来源的树突状细胞,用肿瘤相关抗原(TAAs)或肿瘤特异性抗原(TSAs)对其进行脉冲处理,并利用成熟鸡尾酒确保其有效活化。然后再回输这些成熟的 DCs,以激发肿瘤特异性 T 细胞反应。虽然这种方法已证明能产生有效的免疫反应,但由于肿瘤微环境具有免疫抑制作用,其临床疗效一直受到限制。最近的研究重点是增强基于 DC 的疫苗的免疫原性,特别是通过与 T 细胞靶向免疫疗法的联合疗法。本试验观察总结了基于直流电的癌症治疗的最新进展,包括新的临床前和临床策略的开发,并讨论了基于直流电的疫苗在不断发展的免疫肿瘤学领域的未来潜力。
{"title":"Trial watch: anticancer vaccination with dendritic cells.","authors":"Francisca Borges, Raquel S Laureano, Isaure Vanmeerbeek, Jenny Sprooten, Octavie Demeulenaere, Jannes Govaerts, Lisa Kinget, Saurabh Saraswat, Benoit Beuselinck, Steven De Vleeschouwer, Paul Clement, Frederik De Smet, Rüdiger V Sorg, Angeliki Datsi, Nathalie Vigneron, Stefan Naulaerts, Abhishek D Garg","doi":"10.1080/2162402X.2024.2412876","DOIUrl":"https://doi.org/10.1080/2162402X.2024.2412876","url":null,"abstract":"<p><p>Dendritic cells (DCs) are critical players at the intersection of innate and adaptive immunity, making them ideal candidates for anticancer vaccine development. DC-based immunotherapies typically involve isolating patient-derived DCs, pulsing them with tumor-associated antigens (TAAs) or tumor-specific antigens (TSAs), and utilizing maturation cocktails to ensure their effective activation. These matured DCs are then reinfused to elicit tumor-specific T-cell responses. While this approach has demonstrated the ability to generate potent immune responses, its clinical efficacy has been limited due to the immunosuppressive tumor microenvironment. Recent efforts have focused on enhancing the immunogenicity of DC-based vaccines, particularly through combination therapies with T cell-targeting immunotherapies. This Trial Watch summarizes recent advances in DC-based cancer treatments, including the development of new preclinical and clinical strategies, and discusses the future potential of DC-based vaccines in the evolving landscape of immuno-oncology.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2412876"},"PeriodicalIF":6.5,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11469433/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142478270","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
The landscape of T-cell engagers for the treatment of follicular lymphoma. 治疗滤泡性淋巴瘤的 T 细胞激活剂的前景。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-08 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2412869
Alfredo Rivas-Delgado, Ivan Landego, Lorenzo Falchi

Follicular lymphoma (FL), the second most common subtype of non-Hodgkin lymphoma, relies on interactions with immune elements in the tumor microenvironment, including T-follicular helper cells and follicular dendritic cells, for its survival and progression. Despite its initial responsiveness to chemoimmunotherapy, FL is generally considered incurable. Strategies to improve immune-mediated control of FL could significantly benefit this population, particularly as it includes many elderly and comorbid patients. Immune cell engagers, especially bispecific antibodies (BsAbs), are crucial in targeting FL by bridging tumor and effector cells, thereby triggering T-cell activation and cytotoxic killing. CD3 × CD20 BsAbs have shown the most promise in clinical development for B-NHL patients, with structural variations affecting their target affinity and potency. This review summarizes the current clinical trials of BsAbs for relapsed/refractory FL, highlighting the approval of some agents, their role in first-line treatment or combination therapies, their toxicity profiles, and the future of this therapeutic approach compared to other immune cell therapies.

滤泡淋巴瘤(FL)是非霍奇金淋巴瘤中第二常见的亚型,它的生存和发展依赖于与肿瘤微环境中免疫元素的相互作用,包括T-滤泡辅助细胞和滤泡树突状细胞。尽管 FL 最初对化学免疫疗法有反应,但一般认为它是无法治愈的。改善 FL 免疫介导控制的策略可使这一人群受益匪浅,尤其是其中包括许多老年和合并症患者。免疫细胞诱导剂,尤其是双特异性抗体(BsAbs),通过连接肿瘤和效应细胞,从而引发T细胞活化和细胞毒性杀伤,在靶向治疗FL方面至关重要。CD3 × CD20 BsAbs在B-NHL患者的临床开发中显示出最大的前景,其结构变化会影响其靶向亲和力和效力。本综述总结了目前针对复发/难治 FL 的 BsAbs 临床试验,重点介绍了一些药物的批准情况、它们在一线治疗或联合治疗中的作用、它们的毒性特征以及与其他免疫细胞疗法相比这种治疗方法的前景。
{"title":"The landscape of T-cell engagers for the treatment of follicular lymphoma.","authors":"Alfredo Rivas-Delgado, Ivan Landego, Lorenzo Falchi","doi":"10.1080/2162402X.2024.2412869","DOIUrl":"10.1080/2162402X.2024.2412869","url":null,"abstract":"<p><p>Follicular lymphoma (FL), the second most common subtype of non-Hodgkin lymphoma, relies on interactions with immune elements in the tumor microenvironment, including T-follicular helper cells and follicular dendritic cells, for its survival and progression. Despite its initial responsiveness to chemoimmunotherapy, FL is generally considered incurable. Strategies to improve immune-mediated control of FL could significantly benefit this population, particularly as it includes many elderly and comorbid patients. Immune cell engagers, especially bispecific antibodies (BsAbs), are crucial in targeting FL by bridging tumor and effector cells, thereby triggering T-cell activation and cytotoxic killing. CD3 × CD20 BsAbs have shown the most promise in clinical development for B-NHL patients, with structural variations affecting their target affinity and potency. This review summarizes the current clinical trials of BsAbs for relapsed/refractory FL, highlighting the approval of some agents, their role in first-line treatment or combination therapies, their toxicity profiles, and the future of this therapeutic approach compared to other immune cell therapies.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2412869"},"PeriodicalIF":6.5,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11468044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142478269","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
Benzodiazepines compromise the outcome of cancer immunotherapy. 苯二氮卓类药物会影响癌症免疫疗法的效果。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-07 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2413719
Léa Montégut, Lisa Derosa, Meriem Messaoudene, Hui Chen, Flavia Lambertucci, Bertrand Routy, Laurence Zitvogel, Isabelle Martins, Guido Kroemer

Acyl CoA binding protein (ACBP, which is encoded by diazepam binding inhibitor, DBI) acts on the gamma-amino butyric acid (GABA) receptor type A via a specific binding site that is shared by diazepam and other benzodiazepines. Both ACBP/DBI and benzodiazepines act as positive allosteric modulators, hence increasing GABA effects on this receptor. Recently, we found that ACBP/DBI acts as an endogenous immunosuppressor, meaning that its antibody-mediated neutralization has immunostimulatory effects and enhances the efficacy of immunotherapy and chemoimmunotherapy in mouse models. Driven by these considerations, we investigated whether diazepam administration in mice would reverse the beneficial effects of ACBP/DBI neutralization on cancer chemoimmunotherapy. Indeed, diazepam abolished the therapeutic of anti-ACBP/DBI antibodies, supporting the idea that diazepam exerts immunosuppressive properties. Of note, treatment with benzodiazepines was associated with poor clinical responses to chemoimmunotherapy in patients with non-small cell lung cancer (NSCLC) as compared to individuals not receiving any psychotropic drugs. Medication with other psychotropic drugs than benzodiazepines did not compromise the outcome of chemoimmunotherapy, indicating that this immunosuppressive effect was benzodiazepine specific. We conclude that benzodiazepines may confer systemic immunosuppression. This hypothesis requires further epidemiological and clinical confirmation.

Acyl CoA 结合蛋白(ACBP,由地西泮结合抑制剂 DBI 编码)通过地西泮和其他苯二氮卓类药物共享的一个特定结合位点作用于γ-氨基丁酸(GABA)受体 A 型。ACBP/DBI 和苯二氮卓类药物都是正异位调节剂,因此能增强 GABA 对该受体的作用。最近,我们发现 ACBP/DBI 是一种内源性免疫抑制剂,这意味着其抗体介导的中和具有免疫刺激作用,并能增强小鼠模型中免疫疗法和化学免疫疗法的疗效。基于这些考虑,我们研究了在小鼠体内施用地西泮是否会逆转 ACBP/DBI 中和对癌症化学免疫疗法的有利影响。事实上,地西泮可消除抗 ACBP/DBI 抗体的治疗作用,这支持了地西泮具有免疫抑制特性的观点。值得注意的是,与未接受任何精神药物治疗的患者相比,接受苯二氮卓类药物治疗的非小细胞肺癌(NSCLC)患者对化学免疫疗法的临床反应较差。除苯二氮卓类药物外,使用其他精神药物也不会影响化学免疫疗法的效果,这表明这种免疫抑制效应是苯二氮卓类的特异性效应。我们的结论是,苯二氮卓类药物可能会导致全身性免疫抑制。这一假设需要进一步的流行病学和临床证实。
{"title":"Benzodiazepines compromise the outcome of cancer immunotherapy.","authors":"Léa Montégut, Lisa Derosa, Meriem Messaoudene, Hui Chen, Flavia Lambertucci, Bertrand Routy, Laurence Zitvogel, Isabelle Martins, Guido Kroemer","doi":"10.1080/2162402X.2024.2413719","DOIUrl":"https://doi.org/10.1080/2162402X.2024.2413719","url":null,"abstract":"<p><p>Acyl CoA binding protein (ACBP, which is encoded by <i>diazepam binding inhibitor</i>, <i>DBI</i>) acts on the gamma-amino butyric acid (GABA) receptor type A via a specific binding site that is shared by diazepam and other benzodiazepines. Both ACBP/DBI and benzodiazepines act as positive allosteric modulators, hence increasing GABA effects on this receptor. Recently, we found that ACBP/DBI acts as an endogenous immunosuppressor, meaning that its antibody-mediated neutralization has immunostimulatory effects and enhances the efficacy of immunotherapy and chemoimmunotherapy in mouse models. Driven by these considerations, we investigated whether diazepam administration in mice would reverse the beneficial effects of ACBP/DBI neutralization on cancer chemoimmunotherapy. Indeed, diazepam abolished the therapeutic of anti-ACBP/DBI antibodies, supporting the idea that diazepam exerts immunosuppressive properties. Of note, treatment with benzodiazepines was associated with poor clinical responses to chemoimmunotherapy in patients with non-small cell lung cancer (NSCLC) as compared to individuals not receiving any psychotropic drugs. Medication with other psychotropic drugs than benzodiazepines did not compromise the outcome of chemoimmunotherapy, indicating that this immunosuppressive effect was benzodiazepine specific. We conclude that benzodiazepines may confer systemic immunosuppression. This hypothesis requires further epidemiological and clinical confirmation.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2413719"},"PeriodicalIF":6.5,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459736/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394376","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
Acyl CoA binding protein (ACBP): an autophagy checkpoint that can be targeted for improving cancer immunosurveillance. 酰基辅酶A结合蛋白(ACBP):自噬检查点,可作为改善癌症免疫监视的靶点。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-07 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2413200
Léa Montégut, Isabelle Martins, Guido Kroemer

Acyl CoA binding protein (ACBP) encoded by DBI is a tissue hormone that limits autophagy in multiple cell types, hence acting as an extracellular autophagy checkpoint. We recently reported in Molecular Cancer that monoclonal antibodies neutralizing ACBP improve immunosurveillance of breast and lung carcinomas. Moreover, ACBP neutralization improves the outcome of neoadjuvant chemoimmunotherapy with PD-1 blockade in preclinical models.

由DBI编码的酰基辅酶A结合蛋白(ACBP)是一种组织激素,可限制多种类型细胞的自噬,从而起到细胞外自噬检查点的作用。我们最近在《分子癌症》(Molecular Cancer)杂志上报道,中和 ACBP 的单克隆抗体能改善乳腺癌和肺癌的免疫监视。此外,在临床前模型中,中和 ACBP 可改善 PD-1 阻断新辅助化疗免疫疗法的疗效。
{"title":"Acyl CoA binding protein (ACBP): an autophagy checkpoint that can be targeted for improving cancer immunosurveillance.","authors":"Léa Montégut, Isabelle Martins, Guido Kroemer","doi":"10.1080/2162402X.2024.2413200","DOIUrl":"10.1080/2162402X.2024.2413200","url":null,"abstract":"<p><p>Acyl CoA binding protein (ACBP) encoded by <i>DBI</i> is a tissue hormone that limits autophagy in multiple cell types, hence acting as an extracellular autophagy checkpoint. We recently reported in <i>Molecular Cancer</i> that monoclonal antibodies neutralizing ACBP improve immunosurveillance of breast and lung carcinomas. Moreover, ACBP neutralization improves the outcome of neoadjuvant chemoimmunotherapy with PD-1 blockade in preclinical models.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2413200"},"PeriodicalIF":6.5,"publicationDate":"2024-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11459760/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394375","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
Elimination of cDC1 cells by regulatory T cells jeopardizes cancer immunotherapy. 调节性 T 细胞消灭 cDC1 细胞会危及癌症免疫疗法。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-04 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2412874
Peng Liu, Liwei Zhao, Guido Kroemer, Oliver Kepp

Recent findings revealed that neoantigen-specific cytotoxic type 1 regulatory T (TR1) CD4 T cells can subvert cancer immunotherapy by killing type 1 conventional dendritic cells (cDC1s) that present tumor antigens bound to MHC class II. This underlines the importance of cDC1s for eliciting anticancer immunity but poses a novel clinical challenge.

最近的研究结果表明,新抗原特异性细胞毒性1型调节性T(TR1)CD4 T细胞可以通过杀伤呈现与MHC II类结合的肿瘤抗原的1型常规树突状细胞(cDC1s)来颠覆癌症免疫疗法。这凸显了 cDC1s 在激发抗癌免疫力方面的重要性,但也带来了新的临床挑战。
{"title":"Elimination of cDC1 cells by regulatory T cells jeopardizes cancer immunotherapy.","authors":"Peng Liu, Liwei Zhao, Guido Kroemer, Oliver Kepp","doi":"10.1080/2162402X.2024.2412874","DOIUrl":"10.1080/2162402X.2024.2412874","url":null,"abstract":"<p><p>Recent findings revealed that neoantigen-specific cytotoxic type 1 regulatory T (T<sub>R</sub>1) CD4 T cells can subvert cancer immunotherapy by killing type 1 conventional dendritic cells (cDC1s) that present tumor antigens bound to MHC class II. This underlines the importance of cDC1s for eliciting anticancer immunity but poses a novel clinical challenge.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2412874"},"PeriodicalIF":6.5,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11457612/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394377","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
Peptide-guided adaptor-CAR T-Cell therapy for the treatment of SSTR2-expressing neuroendocrine tumors. 肽引导的适配体-CAR T 细胞疗法用于治疗表达 SSTR2 的神经内分泌肿瘤。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-03 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2412371
Christian Pellegrino, Nicholas Favalli, Laura Volta, Ramon Benz, Sara Puglioli, Gabriele Bassi, Kathrin Zitzmann, Christoph Josef Auernhammer, Svenja Nölting, Chiara F Magnani, Dario Neri, Felix Beuschlein, Markus G Manz

Somatostatin receptor type 2 (SSTR2) is one of the five subtypes of somatostatin receptors and is overexpressed on the surface of most gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs), pituitary tumors, paraganglioma, and meningioma, as well as hepatocellular carcinoma and breast cancer. Chimeric antigen receptor (CAR) T-cells are genetically engineered to express an artificial, T-cell activating binder, leading upon ligation to biocidal activity against target-antigen expressing cells. Adaptor-CAR T-cells recognize, via the CAR, a tag on an antigen-binding molecule, building an activating bridge between the CAR and the target cell. We hypothesized that a novel fluorescent-peptide antagonist of SSTR2, called Octo-Fluo, in combination with anti-FITC adaptor CAR (AdFITC(E2)-CAR) T-cells, may function as an on-off tunable activating bridge between the CAR and SSTR2 expressing target cells. In vitro studies confirmed the binding of Octo-Fluo to Bon1-SSTR2 mCherry-Luc cells without evidence of internalization. AdFITC(E2)-CAR T-cells were activated and efficiently induced Bon1-SSTR2 cell death in vitro, in an Octo-Fluo concentration-dependent manner. Similarly, AdFITC(E2)-CAR T-cells in combination with Octo-Fluo efficiently infiltrated the tumor and eliminated Bon1-SSTR2 tumors in immunodeficient mice in therapeutic settings. Both, AdFITC(E2)-CAR T-cell tumor infiltration and biocidal activity were Octo-Fluo concentration-dependent, with high doses of Octo-Fluo, saturating both the CAR and the SSTR2 antigen independently, leading to the loss of tumor infiltration and biocidal activity due to the loss of bridge formation. Our findings demonstrate the potential of using AdFITC(E2)-CAR T-cells with Octo-Fluo as a versatile, on-off tunable bispecific adaptor for targeted CAR T-cell immunotherapy against SSTR2-positive NETs.

体生长抑素受体 2 型(SSTR2)是体生长抑素受体的五种亚型之一,在大多数胃肠胰神经内分泌肿瘤(GEP-NET)、垂体瘤、副神经节瘤、脑膜瘤以及肝细胞癌和乳腺癌的表面过度表达。嵌合抗原受体(CAR)T 细胞经过基因工程改造,表达一种人工的 T 细胞活化粘合剂,在连接后对表达靶抗原的细胞产生生物杀伤活性。适配器-CAR T 细胞通过 CAR 识别抗原结合分子上的标签,在 CAR 和靶细胞之间架起一座激活桥梁。我们假设,一种名为 Octo-Fluo 的新型 SSTR2 荧光肽拮抗剂与抗 FITC 适配 CAR(AdFITC(E2)-CAR)T 细胞结合,可以在 CAR 和表达 SSTR2 的靶细胞之间起到开关可调的激活桥梁作用。体外研究证实,Octo-Fluo 可与 Bon1-SSTR2 mCherry-Luc 细胞结合,但无内化迹象。AdFITC(E2)-CAR T 细胞在体外被激活并有效诱导 Bon1-SSTR2 细胞死亡,其方式与 Octo-Fluo 浓度有关。同样,在治疗过程中,AdFITC(E2)-CAR T 细胞与 Octo-Fluo 结合使用可有效渗透肿瘤,并消除免疫缺陷小鼠的 Bon1-SSTR2 肿瘤。AdFITC(E2)-CAR T 细胞的肿瘤浸润和生物杀伤活性都与 Octo-Fluo 的浓度有关,高剂量的 Octo-Fluo 会使 CAR 和 SSTR2 抗原独立达到饱和,从而导致肿瘤浸润和生物杀伤活性因桥的形成而丧失。我们的研究结果表明,AdFITC(E2)-CAR T细胞与Octo-Fluo可作为一种多功能、开关可调的双特异性适配体,用于针对SSTR2阳性NET的靶向CAR T细胞免疫疗法。
{"title":"Peptide-guided adaptor-CAR T-Cell therapy for the treatment of SSTR2-expressing neuroendocrine tumors.","authors":"Christian Pellegrino, Nicholas Favalli, Laura Volta, Ramon Benz, Sara Puglioli, Gabriele Bassi, Kathrin Zitzmann, Christoph Josef Auernhammer, Svenja Nölting, Chiara F Magnani, Dario Neri, Felix Beuschlein, Markus G Manz","doi":"10.1080/2162402X.2024.2412371","DOIUrl":"https://doi.org/10.1080/2162402X.2024.2412371","url":null,"abstract":"<p><p>Somatostatin receptor type 2 (SSTR2) is one of the five subtypes of somatostatin receptors and is overexpressed on the surface of most gastro-entero-pancreatic neuroendocrine tumors (GEP-NETs), pituitary tumors, paraganglioma, and meningioma, as well as hepatocellular carcinoma and breast cancer. Chimeric antigen receptor (CAR) T-cells are genetically engineered to express an artificial, T-cell activating binder, leading upon ligation to biocidal activity against target-antigen expressing cells. Adaptor-CAR T-cells recognize, via the CAR, a tag on an antigen-binding molecule, building an activating bridge between the CAR and the target cell. We hypothesized that a novel fluorescent-peptide antagonist of SSTR2, called Octo-Fluo, in combination with anti-FITC adaptor CAR (AdFITC(E2)-CAR) T-cells, may function as an on-off tunable activating bridge between the CAR and SSTR2 expressing target cells. In vitro studies confirmed the binding of Octo-Fluo to Bon1-SSTR2 mCherry-Luc cells without evidence of internalization. AdFITC(E2)-CAR T-cells were activated and efficiently induced Bon1-SSTR2 cell death in vitro, in an Octo-Fluo concentration-dependent manner. Similarly, AdFITC(E2)-CAR T-cells in combination with Octo-Fluo efficiently infiltrated the tumor and eliminated Bon1-SSTR2 tumors in immunodeficient mice in therapeutic settings. Both, AdFITC(E2)-CAR T-cell tumor infiltration and biocidal activity were Octo-Fluo concentration-dependent, with high doses of Octo-Fluo, saturating both the CAR and the SSTR2 antigen independently, leading to the loss of tumor infiltration and biocidal activity due to the loss of bridge formation. Our findings demonstrate the potential of using AdFITC(E2)-CAR T-cells with Octo-Fluo as a versatile, on-off tunable bispecific adaptor for targeted CAR T-cell immunotherapy against SSTR2-positive NETs.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2412371"},"PeriodicalIF":6.5,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11457607/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142394378","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
High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1α activity in neutrophils. 中性粒细胞中的IRE1α活性驱动了高级别浆液性卵巢癌的发展和抗PD-1的耐药性。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-02 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2411070
Alexander Emmanuelli, Camilla Salvagno, Sung-Min Hwang, Deepika Awasthi, Tito A Sandoval, Chang-Suk Chae, Jin-Gyu Cheong, Chen Tan, Takao Iwawaki, Juan R Cubillos-Ruiz

High-grade serious ovarian cancer (HGSOC) is an aggressive malignancy that remains refractory to current immunotherapies. While advanced stage disease has been extensively studied, the cellular and molecular mechanisms that promote early immune escape in HGSOC remain largely unexplored. Here, we report that primary HGSO tumors program neutrophils to inhibit T cell anti-tumor function by activating the endoplasmic reticulum (ER) stress sensor IRE1α. We found that intratumoral neutrophils exhibited overactivation of ER stress response markers compared with their counterparts at non-tumor sites. Selective deletion of IRE1α in neutrophils delayed primary ovarian tumor growth and extended the survival of mice with HGSOC by enabling early T cell-mediated tumor control. Notably, loss of IRE1α in neutrophils sensitized tumor-bearing mice to PD-1 blockade, inducing HGSOC regression and long-term survival in ~ 50% of the treated hosts. Hence, neutrophil-intrinsic IRE1α facilitates early adaptive immune escape in HGSOC and targeting this ER stress sensor might be used to unleash endogenous and immunotherapy-elicited immunity that controls metastatic disease.

高级别重度卵巢癌(HGSOC)是一种侵袭性恶性肿瘤,对目前的免疫疗法仍有耐药性。虽然晚期疾病已被广泛研究,但促进 HGSOC 早期免疫逃逸的细胞和分子机制在很大程度上仍未被探索。在这里,我们报告了原发性HGSO肿瘤通过激活内质网(ER)应激传感器IRE1α,使中性粒细胞抑制T细胞的抗肿瘤功能。我们发现,与非肿瘤部位的中性粒细胞相比,瘤内中性粒细胞表现出过度激活ER应激反应标记物。选择性地删除中性粒细胞中的IRE1α可延缓原发性卵巢肿瘤的生长,并通过早期T细胞介导的肿瘤控制延长HGSOC小鼠的生存期。值得注意的是,中性粒细胞中 IRE1α 的缺失会使肿瘤小鼠对 PD-1 阻滞剂敏感,从而诱导 HGSOC 消退,并使约 50% 的受试宿主长期存活。因此,中性粒细胞内在的IRE1α有助于HGSOC早期适应性免疫逃逸,靶向这种ER应激传感器可用于释放内源性免疫和免疫疗法诱导的免疫,从而控制转移性疾病。
{"title":"High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1α activity in neutrophils.","authors":"Alexander Emmanuelli, Camilla Salvagno, Sung-Min Hwang, Deepika Awasthi, Tito A Sandoval, Chang-Suk Chae, Jin-Gyu Cheong, Chen Tan, Takao Iwawaki, Juan R Cubillos-Ruiz","doi":"10.1080/2162402X.2024.2411070","DOIUrl":"10.1080/2162402X.2024.2411070","url":null,"abstract":"<p><p>High-grade serious ovarian cancer (HGSOC) is an aggressive malignancy that remains refractory to current immunotherapies. While advanced stage disease has been extensively studied, the cellular and molecular mechanisms that promote early immune escape in HGSOC remain largely unexplored. Here, we report that primary HGSO tumors program neutrophils to inhibit T cell anti-tumor function by activating the endoplasmic reticulum (ER) stress sensor IRE1α. We found that intratumoral neutrophils exhibited overactivation of ER stress response markers compared with their counterparts at non-tumor sites. Selective deletion of IRE1α in neutrophils delayed primary ovarian tumor growth and extended the survival of mice with HGSOC by enabling early T cell-mediated tumor control. Notably, loss of IRE1α in neutrophils sensitized tumor-bearing mice to PD-1 blockade, inducing HGSOC regression and long-term survival in ~ 50% of the treated hosts. Hence, neutrophil-intrinsic IRE1α facilitates early adaptive immune escape in HGSOC and targeting this ER stress sensor might be used to unleash endogenous and immunotherapy-elicited immunity that controls metastatic disease.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2411070"},"PeriodicalIF":6.5,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448341/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373288","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
Clinical prognosticators and targets in the immune microenvironment of intrahepatic cholangiocarcinoma. 肝内胆管癌免疫微环境中的临床预后指标和靶点。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2406052
Isis Lozzi, Alexander Arnold, Matthias Barone, Juliette Claire Johnson, Bruno V Sinn, Johannes Eschrich, Pimrapat Gebert, Ruonan Wang, Mengwen Hu, Linda Feldbrügge, Anja Schirmeier, Anja Reutzel-Selke, Thomas Malinka, Felix Krenzien, Wenzel Schöning, Dominik P Modest, Johann Pratschke, Igor M Sauer, Matthäus Felsenstein

Background: Intrahepatic cholangiocarcinoma (ICC) is a disease with poor prognosis and limited therapeutic options. We investigated the tumor immune microenvironment (TIME) to identify predictors of disease outcome and to explore targets for therapeutic modulation.

Methods: Liver tissue samples were collected during 2008-2019 from patients (n = 139) diagnosed with ICC who underwent curative intent surgery without neoadjuvant chemotherapy. Samples from the discovery cohort (n = 86) were immunohistochemically analyzed on tissue microarrays (TMAs) for the expression of CD68, CD3, CD4, CD8, Foxp3, PD-L1, STAT1, and p-STAT1 in tumor core and stroma areas. Results were digitally analyzed using QuPath software and correlated with clinicopathological characteristics. For validation of TIME-related biomarkers, we performed multiplex imaging mass cytometry (IMC) in a validation cohort (n = 53).

Results: CD68+ cells were the predominant immune cell type in the TIME of ICC. CD4+high T cell density correlated with better overall survival (OS). Prediction modeling together with validation cohort confirmed relevance of CD4+ cells, PD-L1 expression by immune cells in the stroma and N-stage on overall disease outcome. In turn, IMC analyses revealed that silent CD3+CD4+ clusters inversely impacted survival. Among annotated immune cell clusters, PD-L1 was most relevantly expressed by CD4+FoxP3+ cells. A subset of tumors with high density of immune cells ("hot" cluster) correlated with PD-L1 expression and could identify a group of candidates for immune checkpoint inhibition (ICI). Ultimately, higher levels of STAT1 expression were associated with higher lymphocyte infiltration and PD-L1 expression.

Conclusions: These results highlight the importance of CD4+ T cells in immune response against ICC. Secondly, a subset of tumors with "hot" TIME represents potential candidates for ICI, while stimulation of STAT1 pathway could be a potential target to turn "cold" into "hot" TIME in ICC.

背景:肝内胆管癌(ICC肝内胆管癌(ICC)是一种预后不良且治疗方案有限的疾病。我们研究了肿瘤免疫微环境(TIME),以确定疾病预后的预测因素,并探索治疗调节的靶点:2008-2019年期间,我们收集了被诊断为ICC的患者(139人)的肝脏组织样本,这些患者接受了根治性手术,但未接受新辅助化疗。对发现队列中的样本(n = 86)进行组织芯片(TMA)免疫组化分析,检测肿瘤核心区和基质区的 CD68、CD3、CD4、CD8、Foxp3、PD-L1、STAT1 和 p-STAT1 的表达。使用 QuPath 软件对结果进行数字分析,并将其与临床病理特征相关联。为了验证与 TIME 相关的生物标记物,我们在一个验证队列(n = 53)中进行了多重成像质谱(IMC)分析:结果:CD68+细胞是ICC TIME中最主要的免疫细胞类型。CD4+高T细胞密度与较好的总生存期(OS)相关。预测模型和验证队列证实了CD4+细胞、基质中免疫细胞的PD-L1表达和N期对总体疾病预后的相关性。反过来,IMC分析显示,沉默的CD3+CD4+细胞群对生存率有反向影响。在注释的免疫细胞群中,CD4+FoxP3+细胞表达的PD-L1最为相关。免疫细胞密度高的肿瘤亚群("热 "群)与PD-L1的表达相关,可以确定一组免疫检查点抑制剂(ICI)的候选者。最终,较高水平的STAT1表达与较高的淋巴细胞浸润和PD-L1表达相关:这些结果凸显了 CD4+ T 细胞在针对 ICC 的免疫反应中的重要性。其次,具有 "热 "TIME的肿瘤亚群是ICI的潜在候选者,而刺激STAT1通路可能是将ICC中的 "冷 "TIME变为 "热 "TIME的潜在靶点。
{"title":"Clinical prognosticators and targets in the immune microenvironment of intrahepatic cholangiocarcinoma.","authors":"Isis Lozzi, Alexander Arnold, Matthias Barone, Juliette Claire Johnson, Bruno V Sinn, Johannes Eschrich, Pimrapat Gebert, Ruonan Wang, Mengwen Hu, Linda Feldbrügge, Anja Schirmeier, Anja Reutzel-Selke, Thomas Malinka, Felix Krenzien, Wenzel Schöning, Dominik P Modest, Johann Pratschke, Igor M Sauer, Matthäus Felsenstein","doi":"10.1080/2162402X.2024.2406052","DOIUrl":"10.1080/2162402X.2024.2406052","url":null,"abstract":"<p><strong>Background: </strong>Intrahepatic cholangiocarcinoma (ICC) is a disease with poor prognosis and limited therapeutic options. We investigated the tumor immune microenvironment (TIME) to identify predictors of disease outcome and to explore targets for therapeutic modulation.</p><p><strong>Methods: </strong>Liver tissue samples were collected during 2008-2019 from patients (<i>n</i> = 139) diagnosed with ICC who underwent curative intent surgery without neoadjuvant chemotherapy. Samples from the discovery cohort (<i>n</i> = 86) were immunohistochemically analyzed on tissue microarrays (TMAs) for the expression of CD68, CD3, CD4, CD8, Foxp3, PD-L1, STAT1, and p-STAT1 in tumor core and stroma areas. Results were digitally analyzed using QuPath software and correlated with clinicopathological characteristics. For validation of TIME-related biomarkers, we performed multiplex imaging mass cytometry (IMC) in a validation cohort (<i>n</i> = 53).</p><p><strong>Results: </strong>CD68+ cells were the predominant immune cell type in the TIME of ICC. CD4+<sup>high</sup> T cell density correlated with better overall survival (OS). Prediction modeling together with validation cohort confirmed relevance of CD4+ cells, PD-L1 expression by immune cells in the stroma and N-stage on overall disease outcome. In turn, IMC analyses revealed that silent CD3+CD4+ clusters inversely impacted survival. Among annotated immune cell clusters, PD-L1 was most relevantly expressed by CD4+FoxP3+ cells. A subset of tumors with high density of immune cells (\"hot\" cluster) correlated with PD-L1 expression and could identify a group of candidates for immune checkpoint inhibition (ICI). Ultimately, higher levels of STAT1 expression were associated with higher lymphocyte infiltration and PD-L1 expression.</p><p><strong>Conclusions: </strong>These results highlight the importance of CD4+ T cells in immune response against ICC. Secondly, a subset of tumors with \"hot\" TIME represents potential candidates for ICI, while stimulation of STAT1 pathway could be a potential target to turn \"cold\" into \"hot\" TIME in ICC.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2406052"},"PeriodicalIF":6.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11445892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142367086","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
Enhancing T-cell recruitment in renal cell carcinoma with cytokine-armed adenoviruses. 用细胞因子武装的腺病毒增强肾细胞癌中的 T 细胞募集。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-25 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2407532
Michaela Feodoroff, Firas Hamdan, Gabriella Antignani, Sara Feola, Manlio Fusciello, Salvatore Russo, Jacopo Chiaro, Katja Välimäki, Teijo Pellinen, Rui M Branca, Janne Lehtiö, Federica D Alessio, Paolo Bottega, Virpi Stigzelius, Janita Sandberg, Jonna Clancy, Jukka Partanen, Minna Malmstedt, Antti Rannikko, Vilja Pietiäinen, Mikaela Grönholm, Vincenzo Cerullo

Immunotherapy has emerged as a promising approach for cancer treatment, with oncolytic adenoviruses showing power as immunotherapeutic agents. In this study, we investigated the immunotherapeutic potential of an adenovirus construct expressing CXCL9, CXCL10, or IL-15 in clear cell renal cell carcinoma (ccRCC) tumor models. Our results demonstrated robust cytokine secretion upon viral treatment, suggesting effective transgene expression. Subsequent analysis using resistance-based transwell migration and microfluidic chip assays demonstrated increased T-cell migration in response to chemokine secretion by infected cells in both 2D and 3D cell models. Flow cytometry analysis revealed CXCR3 receptor expression across T-cell subsets, with the highest percentage found on CD8+ T-cells, underscoring their key role in immune cell migration. Alongside T-cells, we also detected NK-cells in the tumors of immunocompromised mice treated with cytokine-encoding adenoviruses. Furthermore, we identified potential immunogenic antigens that may enhance the efficacy and specificity of our armed oncolytic adenoviruses in ccRCC. Overall, our findings using ccRCC cell line, in vivo humanized mice, physiologically relevant PDCs in 2D and patient-derived organoids (PDOs) in 3D suggest that chemokine-armed adenoviruses hold promise for enhancing T-cell migration and improving immunotherapy outcomes in ccRCC. Our study contributes to the development of more effective ccRCC treatment strategies by elucidating immune cell infiltration and activation mechanisms within the tumor microenvironment (TME) and highlights the usefulness of PDOs for predicting clinical relevance and validating novel immunotherapeutic approaches. Overall, our research offers insights into the rational design and optimization of viral-based immunotherapies for ccRCC.

免疫疗法已成为一种前景广阔的癌症治疗方法,溶瘤腺病毒作为免疫治疗药物显示出强大的威力。在本研究中,我们研究了表达 CXCL9、CXCL10 或 IL-15 的腺病毒构建体在透明细胞肾细胞癌(ccRCC)肿瘤模型中的免疫治疗潜力。我们的结果表明,病毒处理后细胞因子分泌旺盛,表明转基因表达有效。随后使用基于抗性的透孔迁移和微流控芯片测定法进行的分析表明,在二维和三维细胞模型中,T细胞迁移增加是对感染细胞分泌趋化因子的反应。流式细胞术分析表明,CXCR3受体在各T细胞亚群中均有表达,其中CD8+ T细胞的表达量最高,这表明它们在免疫细胞迁移中发挥着关键作用。除了 T 细胞,我们还在接受细胞因子编码腺病毒治疗的免疫缺陷小鼠肿瘤中检测到了 NK 细胞。此外,我们还发现了潜在的免疫原性抗原,这些抗原可能会提高我们的武装溶瘤腺病毒在ccRCC中的疗效和特异性。总之,我们使用ccRCC细胞系、体内人源化小鼠、二维生理学相关PDCs和三维患者衍生器官组织(PDOs)的研究结果表明,趋化因子武装腺病毒有望增强T细胞迁移,改善ccRCC的免疫治疗效果。我们的研究通过阐明肿瘤微环境(TME)中的免疫细胞浸润和激活机制,为开发更有效的ccRCC治疗策略做出了贡献,并强调了PDOs在预测临床相关性和验证新型免疫治疗方法方面的有用性。总之,我们的研究为合理设计和优化基于病毒的 ccRCC 免疫疗法提供了启示。
{"title":"Enhancing T-cell recruitment in renal cell carcinoma with cytokine-armed adenoviruses.","authors":"Michaela Feodoroff, Firas Hamdan, Gabriella Antignani, Sara Feola, Manlio Fusciello, Salvatore Russo, Jacopo Chiaro, Katja Välimäki, Teijo Pellinen, Rui M Branca, Janne Lehtiö, Federica D Alessio, Paolo Bottega, Virpi Stigzelius, Janita Sandberg, Jonna Clancy, Jukka Partanen, Minna Malmstedt, Antti Rannikko, Vilja Pietiäinen, Mikaela Grönholm, Vincenzo Cerullo","doi":"10.1080/2162402X.2024.2407532","DOIUrl":"10.1080/2162402X.2024.2407532","url":null,"abstract":"<p><p>Immunotherapy has emerged as a promising approach for cancer treatment, with oncolytic adenoviruses showing power as immunotherapeutic agents. In this study, we investigated the immunotherapeutic potential of an adenovirus construct expressing CXCL9, CXCL10, or IL-15 in clear cell renal cell carcinoma (ccRCC) tumor models. Our results demonstrated robust cytokine secretion upon viral treatment, suggesting effective transgene expression. Subsequent analysis using resistance-based transwell migration and microfluidic chip assays demonstrated increased T-cell migration in response to chemokine secretion by infected cells in both 2D and 3D cell models. Flow cytometry analysis revealed CXCR3 receptor expression across T-cell subsets, with the highest percentage found on CD8+ T-cells, underscoring their key role in immune cell migration. Alongside T-cells, we also detected NK-cells in the tumors of immunocompromised mice treated with cytokine-encoding adenoviruses. Furthermore, we identified potential immunogenic antigens that may enhance the efficacy and specificity of our armed oncolytic adenoviruses in ccRCC. Overall, our findings using ccRCC cell line, <i>in vivo</i> humanized mice, physiologically relevant PDCs in 2D and patient-derived organoids (PDOs) in 3D suggest that chemokine-armed adenoviruses hold promise for enhancing T-cell migration and improving immunotherapy outcomes in ccRCC. Our study contributes to the development of more effective ccRCC treatment strategies by elucidating immune cell infiltration and activation mechanisms within the tumor microenvironment (TME) and highlights the usefulness of PDOs for predicting clinical relevance and validating novel immunotherapeutic approaches. Overall, our research offers insights into the rational design and optimization of viral-based immunotherapies for ccRCC.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":"13 1","pages":"2407532"},"PeriodicalIF":6.5,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441019/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142356459","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
期刊
Oncoimmunology
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