Avelumab通过自然杀伤细胞介导对单核细胞来源的树突状细胞的抗体依赖性细胞毒性

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
MedComm Pub Date : 2025-02-18 DOI:10.1002/mco2.70111
Tatjana Sauerer, Franziska Bremm, Amke C. Beenen, Lukas Heger, Diana Dudziak, Naomi C. Bosch, Michael Erdmann, Carola Berking, Niels Schaft, Jan Dörrie
{"title":"Avelumab通过自然杀伤细胞介导对单核细胞来源的树突状细胞的抗体依赖性细胞毒性","authors":"Tatjana Sauerer,&nbsp;Franziska Bremm,&nbsp;Amke C. Beenen,&nbsp;Lukas Heger,&nbsp;Diana Dudziak,&nbsp;Naomi C. Bosch,&nbsp;Michael Erdmann,&nbsp;Carola Berking,&nbsp;Niels Schaft,&nbsp;Jan Dörrie","doi":"10.1002/mco2.70111","DOIUrl":null,"url":null,"abstract":"<p>Dear Editor,</p><p>In recent years, treatment with immune checkpoint inhibitors (ICI) has revolutionized cancer therapy. Monoclonal antibodies that block immune checkpoint receptors such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) or its ligand PD-L1 prevent the tumor from suppressing adaptive immune responses.<span><sup>1</sup></span> Especially PD-L1 is expressed on tumor cells, but also on various healthy cell types.<span><sup>2</sup></span> When we evaluated a combination therapy of active immunization via dendritic cells (DCs) and ICIs in the context of Merkel cell carcinoma (MCC) in order to improve the efficacy of the treatment, we observed an unexpected inhibitory effect of the anti-PD-L1 ICI Avelumab. In cell culture experiments with the most commonly used type of DCs in clinical trials on therapeutic cancer vaccination,<span><sup>3</sup></span> the priming capacity of the tumor antigen-loaded human monocyte-derived DCs was reduced in the presence of Avelumab but not Pembrolizumab (anti-PD-1; Figure S1A). Experiments with GFP-expressing DCs showed a disappearance of the DCs in co-cultures with autologous lymphocytes and Avelumab over time (data not shown). However, no direct toxic effect of the antibody against pure DCs was observed (Figure S1B). Moreover, the DCs’ ability to stimulate T cell receptor-transfected pure CD8<sup>+</sup> T cells was not influenced by the ICI antibodies (Figure S1C).</p><p>From these unanticipated findings, we concluded that Avelumab, in the presence of autologous lymphocytes, had a detrimental effect on the DCs. In contrast to other approved anti-PD-L1 antibodies like Atezolizumab and Durvalumab, Avelumab contains a constant region (Fc-part) of the IgG1 isotype that is capable of inducing antibody-dependent cellular cytotoxicity (ADCC) against PD-L1 expressing tumor cells, which has been shown to be beneficial in preclinical studies.<span><sup>4</sup></span> However, healthy cells including DCs also express PD-L1. Therefore, Avelumab could induce an unwanted ADCC reaction against the DCs, as any antibody with a suitable Fc-part that binds efficiently to these cells would do.</p><p>Flow cytometry experiments showed that monocyte-derived DCs expressed high levels of PD-L1 and that Avelumab, Atezolizumab, and Durvalumab but not Pembrolizumab efficiently bound to the DCs (see Figure 1A). Also, the human primary DC subpopulations cDC1, DC2, and DC3, which are responsible for antigen presentation,<span><sup>5</sup></span> expressed PD-L1 upon Toll-like receptor 7/8 stimulation with R848 (see Figure 1A). Primarily, two types of cells exert ADCC: macrophages and natural killer (NK) cells. Since the latter are present in substantial numbers in the lymphocyte fractions we used, we examined whether this was the specific cell type that carried out an Avelumab-dependent ADCC against DCs. In a classical chromium-release cytotoxicity assay, we were able to show that in the presence of Avelumab, DCs were efficiently lysed by purified NK cells whereas intermediate lysis was seen when complete peripheral blood mononuclear cells (PBMCs) were used as effector cells (Figure 1B). Consequently, NK-depleted PBMCs showed no lysis. Likewise, there was also no cytotoxicity observed in the control conditions containing no antibody or the anti-PD-1 antibody Pembrolizumab (Figure 1B). The Fc-part-mutated antibodies Durvalumab and Atezolizumab were also not able to induce lysis of the DCs (Figure 1C), although they bound to the DCs to a similar extent (Figure 1A). This clearly shows that Avelumab induced an efficient ADCC reaction by NK cells against autologous DCs. In the experiments described above, monocyte-derived cytokine-matured DCs were used, as these cells are the most commonly used in clinical trials for DC-based therapeutic tumor vaccination. Hence, we examined whether the maturation type and state influenced the susceptibility of DCs to Avelumab-mediated ADCC. We observed that DCs matured with several different stimuli (cytokine cocktail, lipopolysaccharide (LPS), R848, or polyinosinic:polycytidylic acid [poly I:C]) all expressed high levels of PD-L1. Even immature monocyte-derived DCs expressed PD-L1, however to a lower extent. Therefore, the cytotoxicity assay was repeated with purified NK cells and immature DCs, as well as DCs treated with the various maturation stimuli. In the presence of Avelumab all differently matured DCs and even immature DCs were lysed with similarly high efficiency (Figure 1D). This implies that the observed phenomenon may be of general relevance as it seems to apply to all cells expressing a sufficient level of PD-L1. Next to classical ADCC, additional antibody-mediated cytotoxic effects may exist in vivo, like complement-dependent cytotoxicity and antibody-dependent phagocytosis, which may aggravate the effect.</p><p>In summary, we demonstrated that Avelumab-mediated ADCC via NK-cell activation can lead to the killing of DCs. These findings are of high clinical relevance for combination therapies with Avelumab. Therefore we recommend that patients receiving active immunotherapy like therapeutic vaccination should not simultaneously receive Avelumab treatment because this could result in an inhibitory effect on healthy PD-L1-expressing immune cells—especially when using <i>ex vivo-</i>generated DCs as vaccine. Either a sequential approach with initial vaccination and subsequent Avelumab treatment, or the use of Atezolizumab, Durvalumab, or Pembrolizumab instead of Avelumab is recommended.</p><p>Avelumab is currently approved for MCC, urothelial carcinoma, and renal cell carcinoma. According to clinicaltrials.gov, Avelumab has been tested in hundreds of clinical trials from phase 1 to phase 3. Most of these combined the antibody with other treatment regimens such as chemotherapy, small molecule inhibitors, other therapeutic antibodies, oncolytic viruses, adoptive cell transfer, and therapeutic vaccination. Two trials used DCs together with Avelumab (NCT03707808 and NCT03152565) and both applied the DCs and the antibody simultaneously. Unfortunately, both did not address the question of whether the antibody had any effect on the DCs. Other trials used other types of vaccines, including peptides, adenoviral vectors, yeast formulations, and again, Avelumab was given at the same time as the vaccine. In a series of discontinued trials (QUILT-3 series) Avelumab was given together with allogenic NK-cells and a recombinant IL-15 superagonist. In one trial with highly progressed MCC patients (NCT03853317), this led to serious adverse events in more than half of the patients, and in another similar trial with pancreatic cancer patients (NCT03136406), one patient reported lymph node pain as an adverse event. In a breast cancer trial (NCT04215146) with paclitaxel and an oncolytic reovirus, the additional application of Avelumab resulted in fewer clinical responses but increased serious adverse events. However, in none of these trials, the effects of the antibody on DCs were examined. Hence, we think that awareness that Avelumab can probably kill important antigen-presenting cells will enable researchers to design more efficient treatment protocols for combination therapies with Avelumab.</p><p>T.S., F.B., A.C.B., and L.H. performed experiments; T.S., F.B., and J.D. wrote the manuscript; N.C.B. and M.E. provided essential materials; T.S., J.D., N.S., and D.D. supervised experiments; A.C.B., L.H., D.D., N.B., M.E., C.B., and N.S. corrected the manuscript. All authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) via the Research Training Group GRK2504/1 (project number 401821119), research project B2 to Diana Dudziak and B4 to Jan Dörrie.</p><p>The blood of healthy donors was obtained following informed consent and approval of the institutional review board (Ethics Committee of the Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany: Ref. no. 4158).</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 3","pages":""},"PeriodicalIF":10.7000,"publicationDate":"2025-02-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70111","citationCount":"0","resultStr":"{\"title\":\"Avelumab mediates antibody-dependent cellular cytotoxicity against monocyte-derived dendritic cells through natural killer cells\",\"authors\":\"Tatjana Sauerer,&nbsp;Franziska Bremm,&nbsp;Amke C. Beenen,&nbsp;Lukas Heger,&nbsp;Diana Dudziak,&nbsp;Naomi C. Bosch,&nbsp;Michael Erdmann,&nbsp;Carola Berking,&nbsp;Niels Schaft,&nbsp;Jan Dörrie\",\"doi\":\"10.1002/mco2.70111\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Dear Editor,</p><p>In recent years, treatment with immune checkpoint inhibitors (ICI) has revolutionized cancer therapy. Monoclonal antibodies that block immune checkpoint receptors such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) or its ligand PD-L1 prevent the tumor from suppressing adaptive immune responses.<span><sup>1</sup></span> Especially PD-L1 is expressed on tumor cells, but also on various healthy cell types.<span><sup>2</sup></span> When we evaluated a combination therapy of active immunization via dendritic cells (DCs) and ICIs in the context of Merkel cell carcinoma (MCC) in order to improve the efficacy of the treatment, we observed an unexpected inhibitory effect of the anti-PD-L1 ICI Avelumab. In cell culture experiments with the most commonly used type of DCs in clinical trials on therapeutic cancer vaccination,<span><sup>3</sup></span> the priming capacity of the tumor antigen-loaded human monocyte-derived DCs was reduced in the presence of Avelumab but not Pembrolizumab (anti-PD-1; Figure S1A). Experiments with GFP-expressing DCs showed a disappearance of the DCs in co-cultures with autologous lymphocytes and Avelumab over time (data not shown). However, no direct toxic effect of the antibody against pure DCs was observed (Figure S1B). Moreover, the DCs’ ability to stimulate T cell receptor-transfected pure CD8<sup>+</sup> T cells was not influenced by the ICI antibodies (Figure S1C).</p><p>From these unanticipated findings, we concluded that Avelumab, in the presence of autologous lymphocytes, had a detrimental effect on the DCs. In contrast to other approved anti-PD-L1 antibodies like Atezolizumab and Durvalumab, Avelumab contains a constant region (Fc-part) of the IgG1 isotype that is capable of inducing antibody-dependent cellular cytotoxicity (ADCC) against PD-L1 expressing tumor cells, which has been shown to be beneficial in preclinical studies.<span><sup>4</sup></span> However, healthy cells including DCs also express PD-L1. Therefore, Avelumab could induce an unwanted ADCC reaction against the DCs, as any antibody with a suitable Fc-part that binds efficiently to these cells would do.</p><p>Flow cytometry experiments showed that monocyte-derived DCs expressed high levels of PD-L1 and that Avelumab, Atezolizumab, and Durvalumab but not Pembrolizumab efficiently bound to the DCs (see Figure 1A). Also, the human primary DC subpopulations cDC1, DC2, and DC3, which are responsible for antigen presentation,<span><sup>5</sup></span> expressed PD-L1 upon Toll-like receptor 7/8 stimulation with R848 (see Figure 1A). Primarily, two types of cells exert ADCC: macrophages and natural killer (NK) cells. Since the latter are present in substantial numbers in the lymphocyte fractions we used, we examined whether this was the specific cell type that carried out an Avelumab-dependent ADCC against DCs. In a classical chromium-release cytotoxicity assay, we were able to show that in the presence of Avelumab, DCs were efficiently lysed by purified NK cells whereas intermediate lysis was seen when complete peripheral blood mononuclear cells (PBMCs) were used as effector cells (Figure 1B). Consequently, NK-depleted PBMCs showed no lysis. Likewise, there was also no cytotoxicity observed in the control conditions containing no antibody or the anti-PD-1 antibody Pembrolizumab (Figure 1B). The Fc-part-mutated antibodies Durvalumab and Atezolizumab were also not able to induce lysis of the DCs (Figure 1C), although they bound to the DCs to a similar extent (Figure 1A). This clearly shows that Avelumab induced an efficient ADCC reaction by NK cells against autologous DCs. In the experiments described above, monocyte-derived cytokine-matured DCs were used, as these cells are the most commonly used in clinical trials for DC-based therapeutic tumor vaccination. Hence, we examined whether the maturation type and state influenced the susceptibility of DCs to Avelumab-mediated ADCC. We observed that DCs matured with several different stimuli (cytokine cocktail, lipopolysaccharide (LPS), R848, or polyinosinic:polycytidylic acid [poly I:C]) all expressed high levels of PD-L1. Even immature monocyte-derived DCs expressed PD-L1, however to a lower extent. Therefore, the cytotoxicity assay was repeated with purified NK cells and immature DCs, as well as DCs treated with the various maturation stimuli. In the presence of Avelumab all differently matured DCs and even immature DCs were lysed with similarly high efficiency (Figure 1D). This implies that the observed phenomenon may be of general relevance as it seems to apply to all cells expressing a sufficient level of PD-L1. Next to classical ADCC, additional antibody-mediated cytotoxic effects may exist in vivo, like complement-dependent cytotoxicity and antibody-dependent phagocytosis, which may aggravate the effect.</p><p>In summary, we demonstrated that Avelumab-mediated ADCC via NK-cell activation can lead to the killing of DCs. These findings are of high clinical relevance for combination therapies with Avelumab. Therefore we recommend that patients receiving active immunotherapy like therapeutic vaccination should not simultaneously receive Avelumab treatment because this could result in an inhibitory effect on healthy PD-L1-expressing immune cells—especially when using <i>ex vivo-</i>generated DCs as vaccine. Either a sequential approach with initial vaccination and subsequent Avelumab treatment, or the use of Atezolizumab, Durvalumab, or Pembrolizumab instead of Avelumab is recommended.</p><p>Avelumab is currently approved for MCC, urothelial carcinoma, and renal cell carcinoma. According to clinicaltrials.gov, Avelumab has been tested in hundreds of clinical trials from phase 1 to phase 3. Most of these combined the antibody with other treatment regimens such as chemotherapy, small molecule inhibitors, other therapeutic antibodies, oncolytic viruses, adoptive cell transfer, and therapeutic vaccination. Two trials used DCs together with Avelumab (NCT03707808 and NCT03152565) and both applied the DCs and the antibody simultaneously. Unfortunately, both did not address the question of whether the antibody had any effect on the DCs. Other trials used other types of vaccines, including peptides, adenoviral vectors, yeast formulations, and again, Avelumab was given at the same time as the vaccine. In a series of discontinued trials (QUILT-3 series) Avelumab was given together with allogenic NK-cells and a recombinant IL-15 superagonist. In one trial with highly progressed MCC patients (NCT03853317), this led to serious adverse events in more than half of the patients, and in another similar trial with pancreatic cancer patients (NCT03136406), one patient reported lymph node pain as an adverse event. In a breast cancer trial (NCT04215146) with paclitaxel and an oncolytic reovirus, the additional application of Avelumab resulted in fewer clinical responses but increased serious adverse events. However, in none of these trials, the effects of the antibody on DCs were examined. Hence, we think that awareness that Avelumab can probably kill important antigen-presenting cells will enable researchers to design more efficient treatment protocols for combination therapies with Avelumab.</p><p>T.S., F.B., A.C.B., and L.H. performed experiments; T.S., F.B., and J.D. wrote the manuscript; N.C.B. and M.E. provided essential materials; T.S., J.D., N.S., and D.D. supervised experiments; A.C.B., L.H., D.D., N.B., M.E., C.B., and N.S. corrected the manuscript. All authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) via the Research Training Group GRK2504/1 (project number 401821119), research project B2 to Diana Dudziak and B4 to Jan Dörrie.</p><p>The blood of healthy donors was obtained following informed consent and approval of the institutional review board (Ethics Committee of the Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany: Ref. no. 4158).</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":\"6 3\",\"pages\":\"\"},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2025-02-18\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70111\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.70111\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, RESEARCH & EXPERIMENTAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.70111","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

摘要

近年来,免疫检查点抑制剂(ICI)的治疗已经彻底改变了癌症治疗。单克隆抗体阻断免疫检查点受体,如细胞毒性t淋巴细胞相关蛋白4 (CTLA-4)和程序性细胞死亡蛋白1 (PD-1)或其配体PD-L1,可防止肿瘤抑制适应性免疫反应特别是PD-L1在肿瘤细胞上表达,但也在各种健康细胞上表达当我们在默克尔细胞癌(MCC)的背景下评估通过树突状细胞(dc)和ICI的主动免疫联合治疗以提高治疗效果时,我们观察到抗pd - l1 ICI Avelumab的意想不到的抑制作用。在治疗性癌症疫苗临床试验中最常用的dc类型的细胞培养实验中,3在Avelumab的存在下,负载肿瘤抗原的人单核细胞来源dc的启动能力降低,但Pembrolizumab(抗pd -1;图S1A)。表达gfp的dc实验显示,随着时间的推移,与自体淋巴细胞和Avelumab共培养的dc消失(数据未显示)。然而,没有观察到抗体对纯dc的直接毒性作用(图S1B)。此外,DCs刺激T细胞受体转染的纯CD8+ T细胞的能力不受ICI抗体的影响(图S1C)。从这些意想不到的发现中,我们得出结论,在自体淋巴细胞存在的情况下,Avelumab对dc有不利影响。与其他批准的抗PD-L1抗体如Atezolizumab和Durvalumab相比,Avelumab含有IgG1同型的恒定区域(Fc-part),能够诱导抗体依赖性细胞毒性(ADCC)对抗表达PD-L1的肿瘤细胞,这在临床前研究中已被证明是有益的然而,包括dc在内的健康细胞也表达PD-L1。因此,Avelumab可以诱导针对dc的不必要的ADCC反应,因为任何具有合适fc部分的抗体都可以有效地与这些细胞结合。流式细胞术实验显示,单核细胞来源的dc表达高水平的PD-L1,并且Avelumab, Atezolizumab和Durvalumab(而不是Pembrolizumab)有效地结合到dc上(见图1A)。此外,负责抗原呈递的人原发性DC亚群cDC1、DC2和DC3在R848刺激toll样受体7/8后表达PD-L1(见图1A)。主要有两种细胞发挥ADCC作用:巨噬细胞和自然杀伤细胞(NK)。由于后者在我们使用的淋巴细胞分数中大量存在,因此我们检查了这是否是对dc进行avelumab依赖性ADCC的特定细胞类型。在经典的铬释放细胞毒性试验中,我们能够证明,在Avelumab存在的情况下,纯化的NK细胞可以有效地裂解dc,而当完整的外周血单核细胞(PBMCs)用作效应细胞时,可以看到中间裂解(图1B)。因此,nk -贫PBMCs没有裂解。同样,在不含抗体或抗pd -1抗体Pembrolizumab的对照条件下,也没有观察到细胞毒性(图1B)。fc部分突变的抗体Durvalumab和Atezolizumab也不能诱导dc的裂解(图1C),尽管它们与dc的结合程度相似(图1A)。这清楚地表明,Avelumab诱导NK细胞对自体dc产生有效的ADCC反应。在上述实验中,使用了单核细胞衍生的细胞因子成熟dc,因为这些细胞是临床试验中最常用的基于dc的治疗性肿瘤疫苗。因此,我们研究了成熟类型和状态是否影响dc对avelumab介导的ADCC的敏感性。我们观察到,在几种不同的刺激(细胞因子混合物、脂多糖(LPS)、R848或多肌苷:多胞酸[poly I:C])下成熟的树突状细胞都表达了高水平的PD-L1。即使是未成熟的单核细胞来源的dc也表达PD-L1,但表达程度较低。因此,用纯化的NK细胞和未成熟的树突状细胞以及用各种成熟刺激处理的树突状细胞重复细胞毒性试验。在Avelumab存在下,所有不同成熟的dc甚至未成熟的dc都以同样高的效率被裂解(图1D)。这意味着观察到的现象可能具有普遍相关性,因为它似乎适用于所有表达足够水平PD-L1的细胞。除了经典的ADCC外,体内还可能存在其他抗体介导的细胞毒性作用,如补体依赖性细胞毒性和抗体依赖性吞噬作用,这些作用可能会加重ADCC的作用。总之,我们证明了avelumab通过nk细胞激活介导的ADCC可以导致dc的杀伤。这些发现对于与Avelumab联合治疗具有很高的临床相关性。 因此,我们建议接受主动免疫治疗(如治疗性疫苗接种)的患者不应同时接受Avelumab治疗,因为这可能导致对健康的pd - l1表达免疫细胞的抑制作用,特别是当使用体外生成的dc作为疫苗时。建议采用初始疫苗接种和随后的Avelumab治疗的顺序方法,或使用Atezolizumab, Durvalumab或Pembrolizumab代替Avelumab。Avelumab目前被批准用于MCC、尿路上皮癌和肾细胞癌。根据clinicaltrials.gov的数据,Avelumab已经在从1期到3期的数百项临床试验中进行了测试。其中大多数将抗体与其他治疗方案联合使用,如化疗、小分子抑制剂、其他治疗性抗体、溶瘤病毒、过继细胞转移和治疗性疫苗接种。两项试验将DCs与Avelumab (NCT03707808和NCT03152565)联合使用,并同时应用DCs和抗体。不幸的是,两者都没有解决抗体是否对dc有任何影响的问题。其他试验使用其他类型的疫苗,包括多肽、腺病毒载体、酵母制剂,同样,Avelumab与疫苗同时给予。在一系列中断的试验(QUILT-3系列)中,Avelumab与同种异体nk细胞和重组IL-15超级激动剂一起给予。在一项高度进展的MCC患者(NCT03853317)的试验中,超过一半的患者出现了严重的不良事件,而在另一项胰腺癌患者(NCT03136406)的类似试验中,一名患者报告了淋巴结疼痛作为不良事件。在一项使用紫杉醇和溶瘤呼出气病毒的乳腺癌试验(NCT04215146)中,额外应用Avelumab导致临床反应减少,但增加了严重不良事件。然而,在这些试验中都没有检测抗体对dc的影响。因此,我们认为,认识到Avelumab可能会杀死重要的抗原呈递细胞,将使研究人员能够设计出更有效的治疗方案,与Avelumab. ts联合治疗。, f.b., a.c.b.和L.H.进行实验;t.s., f.b.和J.D.写了手稿;N.C.B.和M.E.提供了必要的材料;t.s., j.d., n.s.和D.D.监督实验;A.C.B L.H。公司代码,注意,当,C.B。n。纠正了手稿。所有作者都阅读并批准了最终稿件。作者声明无利益冲突。这项工作由德国研究基金会(DFG)通过研究培训小组GRK2504/1(项目编号401821119)支持,研究项目B2给Diana Dudziak, B4给Jan Dörrie。健康献血者的血液是在机构审查委员会(Friedrich-Alexander-Universität Erlangen- n<e:1>伦理委员会,Erlangen, Germany: Ref. no. 5)的知情同意和批准下获得的。4158)。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Avelumab mediates antibody-dependent cellular cytotoxicity against monocyte-derived dendritic cells through natural killer cells

Avelumab mediates antibody-dependent cellular cytotoxicity against monocyte-derived dendritic cells through natural killer cells

Dear Editor,

In recent years, treatment with immune checkpoint inhibitors (ICI) has revolutionized cancer therapy. Monoclonal antibodies that block immune checkpoint receptors such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) or its ligand PD-L1 prevent the tumor from suppressing adaptive immune responses.1 Especially PD-L1 is expressed on tumor cells, but also on various healthy cell types.2 When we evaluated a combination therapy of active immunization via dendritic cells (DCs) and ICIs in the context of Merkel cell carcinoma (MCC) in order to improve the efficacy of the treatment, we observed an unexpected inhibitory effect of the anti-PD-L1 ICI Avelumab. In cell culture experiments with the most commonly used type of DCs in clinical trials on therapeutic cancer vaccination,3 the priming capacity of the tumor antigen-loaded human monocyte-derived DCs was reduced in the presence of Avelumab but not Pembrolizumab (anti-PD-1; Figure S1A). Experiments with GFP-expressing DCs showed a disappearance of the DCs in co-cultures with autologous lymphocytes and Avelumab over time (data not shown). However, no direct toxic effect of the antibody against pure DCs was observed (Figure S1B). Moreover, the DCs’ ability to stimulate T cell receptor-transfected pure CD8+ T cells was not influenced by the ICI antibodies (Figure S1C).

From these unanticipated findings, we concluded that Avelumab, in the presence of autologous lymphocytes, had a detrimental effect on the DCs. In contrast to other approved anti-PD-L1 antibodies like Atezolizumab and Durvalumab, Avelumab contains a constant region (Fc-part) of the IgG1 isotype that is capable of inducing antibody-dependent cellular cytotoxicity (ADCC) against PD-L1 expressing tumor cells, which has been shown to be beneficial in preclinical studies.4 However, healthy cells including DCs also express PD-L1. Therefore, Avelumab could induce an unwanted ADCC reaction against the DCs, as any antibody with a suitable Fc-part that binds efficiently to these cells would do.

Flow cytometry experiments showed that monocyte-derived DCs expressed high levels of PD-L1 and that Avelumab, Atezolizumab, and Durvalumab but not Pembrolizumab efficiently bound to the DCs (see Figure 1A). Also, the human primary DC subpopulations cDC1, DC2, and DC3, which are responsible for antigen presentation,5 expressed PD-L1 upon Toll-like receptor 7/8 stimulation with R848 (see Figure 1A). Primarily, two types of cells exert ADCC: macrophages and natural killer (NK) cells. Since the latter are present in substantial numbers in the lymphocyte fractions we used, we examined whether this was the specific cell type that carried out an Avelumab-dependent ADCC against DCs. In a classical chromium-release cytotoxicity assay, we were able to show that in the presence of Avelumab, DCs were efficiently lysed by purified NK cells whereas intermediate lysis was seen when complete peripheral blood mononuclear cells (PBMCs) were used as effector cells (Figure 1B). Consequently, NK-depleted PBMCs showed no lysis. Likewise, there was also no cytotoxicity observed in the control conditions containing no antibody or the anti-PD-1 antibody Pembrolizumab (Figure 1B). The Fc-part-mutated antibodies Durvalumab and Atezolizumab were also not able to induce lysis of the DCs (Figure 1C), although they bound to the DCs to a similar extent (Figure 1A). This clearly shows that Avelumab induced an efficient ADCC reaction by NK cells against autologous DCs. In the experiments described above, monocyte-derived cytokine-matured DCs were used, as these cells are the most commonly used in clinical trials for DC-based therapeutic tumor vaccination. Hence, we examined whether the maturation type and state influenced the susceptibility of DCs to Avelumab-mediated ADCC. We observed that DCs matured with several different stimuli (cytokine cocktail, lipopolysaccharide (LPS), R848, or polyinosinic:polycytidylic acid [poly I:C]) all expressed high levels of PD-L1. Even immature monocyte-derived DCs expressed PD-L1, however to a lower extent. Therefore, the cytotoxicity assay was repeated with purified NK cells and immature DCs, as well as DCs treated with the various maturation stimuli. In the presence of Avelumab all differently matured DCs and even immature DCs were lysed with similarly high efficiency (Figure 1D). This implies that the observed phenomenon may be of general relevance as it seems to apply to all cells expressing a sufficient level of PD-L1. Next to classical ADCC, additional antibody-mediated cytotoxic effects may exist in vivo, like complement-dependent cytotoxicity and antibody-dependent phagocytosis, which may aggravate the effect.

In summary, we demonstrated that Avelumab-mediated ADCC via NK-cell activation can lead to the killing of DCs. These findings are of high clinical relevance for combination therapies with Avelumab. Therefore we recommend that patients receiving active immunotherapy like therapeutic vaccination should not simultaneously receive Avelumab treatment because this could result in an inhibitory effect on healthy PD-L1-expressing immune cells—especially when using ex vivo-generated DCs as vaccine. Either a sequential approach with initial vaccination and subsequent Avelumab treatment, or the use of Atezolizumab, Durvalumab, or Pembrolizumab instead of Avelumab is recommended.

Avelumab is currently approved for MCC, urothelial carcinoma, and renal cell carcinoma. According to clinicaltrials.gov, Avelumab has been tested in hundreds of clinical trials from phase 1 to phase 3. Most of these combined the antibody with other treatment regimens such as chemotherapy, small molecule inhibitors, other therapeutic antibodies, oncolytic viruses, adoptive cell transfer, and therapeutic vaccination. Two trials used DCs together with Avelumab (NCT03707808 and NCT03152565) and both applied the DCs and the antibody simultaneously. Unfortunately, both did not address the question of whether the antibody had any effect on the DCs. Other trials used other types of vaccines, including peptides, adenoviral vectors, yeast formulations, and again, Avelumab was given at the same time as the vaccine. In a series of discontinued trials (QUILT-3 series) Avelumab was given together with allogenic NK-cells and a recombinant IL-15 superagonist. In one trial with highly progressed MCC patients (NCT03853317), this led to serious adverse events in more than half of the patients, and in another similar trial with pancreatic cancer patients (NCT03136406), one patient reported lymph node pain as an adverse event. In a breast cancer trial (NCT04215146) with paclitaxel and an oncolytic reovirus, the additional application of Avelumab resulted in fewer clinical responses but increased serious adverse events. However, in none of these trials, the effects of the antibody on DCs were examined. Hence, we think that awareness that Avelumab can probably kill important antigen-presenting cells will enable researchers to design more efficient treatment protocols for combination therapies with Avelumab.

T.S., F.B., A.C.B., and L.H. performed experiments; T.S., F.B., and J.D. wrote the manuscript; N.C.B. and M.E. provided essential materials; T.S., J.D., N.S., and D.D. supervised experiments; A.C.B., L.H., D.D., N.B., M.E., C.B., and N.S. corrected the manuscript. All authors have read and approved the final manuscript.

The authors declare no conflict of interest.

This work was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) via the Research Training Group GRK2504/1 (project number 401821119), research project B2 to Diana Dudziak and B4 to Jan Dörrie.

The blood of healthy donors was obtained following informed consent and approval of the institutional review board (Ethics Committee of the Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany: Ref. no. 4158).

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信