Immune modulatory

  • 文章类型: Journal Article
    目的:多种常见癌症受益于免疫治疗;然而,对罕见肿瘤的疗效知之甚少。我们报告了罕见肿瘤中NCI/SWOGS1609双重抗CTLA-4和抗PD-1阻断的肾上腺皮质癌队列的结果。
    方法:前瞻性,ipilimumab联合nivolumab的2期临床试验由SWOG早期治疗学和罕见癌症委员会针对超过1,000个国家临床试验网络站点的多个罕见肿瘤队列进行.
    方法:登记了21例符合条件的患者。中位年龄为53岁(范围26-69);16(76%)为女性。
    方法:伊匹单抗每6周静脉注射1mg/kg,纳武单抗每2周静脉注射240mg,直至疾病进展,症状恶化,任何原因的治疗延迟>56天,不可接受的或与免疫相关的毒性,无法将泼尼松降低至每天<10mg,或根据患者要求。
    方法:主要终点是总缓解率(ORR)(RECISTV.1.1)。次要终点包括临床获益率(CBR)(包括疾病稳定(SD)>6个月),无进展生存期(PFS),总生存期(OS),和毒性。免疫相关结果包括免疫ORR(iORR),免疫CBR(iCBR),免疫PFS(iPFS)。采用两阶段设计,假设:null=5%替代=30%,n=6在第一阶段,16max,单侧α=13%。
    结果:先前治疗线的中位数为2(范围:1-9)。21例患者中有3例获得确认的部分缓解(PR)(ORR=14%)。此外,一名患者有未经证实的公关;一名,稳定疾病(SD)>6个月;一,免疫相关RECIST(IRECIST)PR(iPR);一名患者达到iSD>6个月:临床获益率(反应或SD>6个月)=5/21(24%),iORR=4/21(19%),iCBR=7/21(33%)。6个月PFS为24%;6个月iPFS,33%。iRECIST临床获益患者(N=7)的PFS分别为57、52、18、15、13、7和7个月。6个月OS为76%;OS中位数,是15.8个月。最常见的毒性是疲劳(62%)和皮疹(38%),最常见的3/4级免疫相关不良事件是肝功能障碍(9.5%)和肾上腺功能不全(9.5%).治疗相关的不良事件导致4例患者停止治疗(21%)。没有发生5级不良事件。
    结论:Ipilimumab联合nivolumab在难治性转移性肾上腺皮质癌中具有活性,达到研究的主要终点,iORR为19%,iCBR为33%(包括SD/iSD>6个月),PFS/iPFS最长为52个月和57个月。
    背景:NCT02834013(7月15日注册,2016;https://clinicaltrials.gov/ct2/show/NCT02834013)。
    OBJECTIVE: Multiple common cancers benefit from immunotherapy; however, less is known about efficacy in rare tumors. We report the results of the adrenocortical carcinoma cohort of NCI/SWOG S1609 Dual Anti-CTLA-4 and Anti-PD-1 blockade in Rare Tumors.
    METHODS: A prospective, phase 2 clinical trial of ipilimumab plus nivolumab was conducted by the SWOG Early Therapeutics and Rare Cancers Committee for multiple rare tumor cohorts across >1,000 National Clinical Trial Network sites.
    METHODS: 21 eligible patients were registered. Median age was 53 years (range 26-69); 16 (76%) were women.
    METHODS: Ipilimumab 1 mg/kg intravenously every 6 weeks with nivolumab 240 mg intravenously every 2 weeks was administered until disease progression, symptomatic deterioration, treatment delay for any reason >56 days, unacceptable or immune-related toxicity with inability to decrease prednisone to <10 mg daily, or per patient request.
    METHODS: The primary endpoint was the overall response rate (ORR) (RECIST V.1.1). Secondary endpoints include clinical benefit rate (CBR) (includes stable disease (SD)>6 months), progression-free survival (PFS), overall survival (OS), and toxicity. Immune-related outcomes included immune ORR (iORR), immune CBR (iCBR), and immune PFS (iPFS). A two-stage design was used assuming: null=5% alternative=30%, n=6 in the first stage, 16 max, one-sided alpha=13%.
    RESULTS: The median number of prior therapy lines was 2 (range: 1-9). 3 of 21 patients attained confirmed partial response (PR) (ORR=14%). In addition, one patient had an unconfirmed PR; one, stable disease (SD)>6 months; one, immune-related RECIST (iRECIST) PR (iPR); and one patient attained iSD>6 months: clinical benefit rate (response or SD>6 months)=5/21 (24%), iORR=4/21 (19%), iCBR=7/21 (33%). The 6-month PFS was 24%; 6-month iPFS, 33%. The PFS for patients (N=7) with iRECIST clinical benefit were 57, 52, 18, 15, 13, 7, and 7 months. The 6-month OS was 76%; the median OS, was 15.8 months. The most common toxicities were fatigue (62%) and rash (38%), and the most common grade 3/4 immune-related adverse events were hepatic dysfunction (9.5%) and adrenal insufficiency (9.5%). Treatment-related adverse events leading to discontinuation of therapy in four patients (21%). There were no grade 5 adverse events.
    CONCLUSIONS: Ipilimumab plus nivolumab is active in refractory metastatic adrenocortical cancer meeting the primary endpoint of the study, with a 19% iORR and 33% iCBR (includes SD/iSD>6 months) and with the longest PFS/iPFS of 52 and 57 months.
    BACKGROUND: NCT02834013 (registered 15 July, 2016; https://clinicaltrials.gov/ct2/show/NCT02834013).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:自然杀伤(NK)细胞作为癌症的细胞疗法正在被广泛研究。这些细胞通过识别肿瘤细胞上的配体和抗原而被激活。细胞因子疗法,例如IL-15,也广泛用于刺激癌症患者的内源性和过继转移的NK细胞。这些刺激激活膜蛋白酶ADAM17,其作为负反馈回路裂解NK细胞上的各种细胞表面受体以限制其细胞溶解功能。ADAM17抑制可以在体外和体内增强IL-15介导的NK细胞增殖。在这项研究中,我们研究了这个过程的潜在机制。
    方法:来自人外周血的外周血单核细胞(PBMC)或富集的NK细胞,未标记或用细胞增殖染料标记,在rhIL-15±ADAM17功能阻断抗体存在下培养长达7天。产生了不同的完全人类版本的抗体;Medi-1(IgG1),Medi-4(IgG4),Medi-PGLALA,Medi-F(ab')2和TAB16(抗ADAM17和抗CD16双特异性)调节CD16A结合。流式细胞术用于评估NK细胞增殖和表型标志物,免疫印迹检查CD16A信号,和基于IncuCyte的活细胞成像来测量NK细胞抗肿瘤活性。
    结果:ADAM17功能阻断单克隆抗体(mAb)Medi-1显著增加了IL-15对早期NK细胞的活化。通过使用不同的工程化版本的抗体,我们展示了CD16A的参与,激活Fcγ受体和良好描述的ADAM17底物。因此,当结合到NK细胞上的ADAM17时,Medi-1被CD16A接合并阻断其脱落,诱导和延长其信号传导。此过程并未促进明显的NK细胞自相残杀或功能障碍。在存在PBMC辅助细胞或抗CD137激动性mAb的情况下,Medi-1和IL-15的协同信号传导增强了CD137在CD16ANK细胞上的上调,并增强了它们的增殖。
    结论:我们的数据首次揭示CD16A和CD137支持Medi-1增强IL-15驱动的NK细胞活化和增殖,分别,后者需要PBMC辅助细胞。使用Medi-1代表了增强IL-15驱动的NK细胞增殖的新策略。通过增加癌症患者NK细胞的抗肿瘤活性可能具有治疗意义。
    BACKGROUND: Natural killer (NK) cells are being extensively studied as a cell therapy for cancer. These cells are activated by recognition of ligands and antigens on tumor cells. Cytokine therapies, such as IL-15, are also broadly used to stimulate endogenous and adoptively transferred NK cells in patients with cancer. These stimuli activate the membrane protease ADAM17, which cleaves various cell-surface receptors on NK cells as a negative feedback loop to limit their cytolytic function. ADAM17 inhibition can enhance IL-15-mediated NK cell proliferation in vitro and in vivo. In this study, we investigated the underlying mechanism of this process.
    METHODS: Peripheral blood mononuclear cells (PBMCs) or enriched NK cells from human peripheral blood, either unlabeled or labeled with a cell proliferation dye, were cultured for up to 7 days in the presence of rhIL-15±an ADAM17 function-blocking antibody. Different fully human versions of the antibody were generated; Medi-1 (IgG1), Medi-4 (IgG4), Medi-PGLALA, Medi-F(ab\')2, and TAB16 (anti-ADAM17 and anti-CD16 bispecific) to modulate CD16A binding. Flow cytometry was used to assess NK cell proliferation and phenotypic markers, immunoblotting to examine CD16A signaling, and IncuCyte-based live cell imaging to measure NK cell antitumor activity.
    RESULTS: The ADAM17 function-blocking monoclonal antibody (mAb) Medi-1 markedly increased early NK cell activation by IL-15. By using different engineered versions of the antibody, we demonstrate involvement by CD16A, an activating Fcγ receptor and well-described ADAM17 substrate. Hence, Medi-1 when bound to ADAM17 on NK cells is engaged by CD16A and blocks its shedding, inducing and prolonging its signaling. This process did not promote evident NK cell fratricide or dysfunction. Synergistic signaling by Medi-1 and IL-15 enhanced the upregulation of CD137 on CD16A+ NK cells and augmented their proliferation in the presence of PBMC accessory cells or an anti-CD137 agonistic mAb.
    CONCLUSIONS: Our data reveal for the first time that CD16A and CD137 underpin Medi-1 enhancement of IL-15-driven NK cell activation and proliferation, respectively, with the latter requiring PBMC accessory cells. The use of Medi-1 represents a novel strategy to enhance IL-15-driven NK cell proliferation, and it may be of therapeutic importance by increasing the antitumor activity of NK cells in patients with cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:宫颈癌的淋巴扩散主要从前哨淋巴结(SLN)开始,强调它们在疾病转移中的关键作用。然而,这些节点的免疫基因表达谱和免疫调节机制还有待探索。
    方法:我们的研究旨在使用NanostringRNAseq分析阐明免疫细胞群体及其在阴性SLN与阳性SLN和非SLN的免疫基因表达谱中的作用。我们对nCounterPanCancer免疫分析小组中685个内源性基因的log2标准化表达进行了主成分分析,然后评估基因和免疫细胞类型丰度的差异表达。
    结果:我们发现各组基因表达存在显著差异,阴性SLN显示与肿瘤浸润免疫细胞相关的基因过表达,特别是先天细胞群。他们还证明了参与抗原呈递和T细胞引发的基因的上调。相比之下,积极的SLN在监管网络中得到了丰富,表明它们在逃避免疫中的潜在作用。阴性SLN和非SLN的比较显示先天和适应性免疫细胞类型增加,强调正在进行的T细胞对肿瘤抗原的反应。
    结论:我们的发现强调了阴性SLN中特定的免疫遗传学表型谱,强调它们在初始抗癌反应中的关键作用,免疫监视,以及原发性宫颈肿瘤免疫耐受的传播。这些结果突出了SLN作为免疫治疗策略的新靶标的潜力,并强调了新的成像方法对于准确识别SLN状态而不去除的重要性。需要进行进一步的研究以进一步了解SLN内的免疫相互作用及其对宫颈癌进展的影响。
    BACKGROUND: Cervical cancer\'s lymphatic spread primarily begins from the sentinel lymph nodes (SLNs), underlining their pivotal role in disease metastasis. However, these nodes\' immune gene expression profiles and immunoregulation mechanisms have yet to be explored.
    METHODS: Our study aimed to elucidate the immune cell populations and their roles in the immune gene expression profile of negative SLNs compared with positive SLNs and non-SLNs using Nanostring RNA seq analysis. We performed a principal component analysis on the log2 normalized expression of 685 endogenous genes in the nCounter PanCancer Immune Profiling Panel, followed by an assessment of the differential expression of genes and immune cell type abundance.
    RESULTS: We found significant variations in gene expression among the groups, with negative SLNs displaying overexpression of genes related to tumor-infiltrating immune cells, specifically innate cell populations. They also demonstrated the upregulation of genes involved in antigen presentation and T-cell priming. In contrast, positive SLNs were enriched in regulatory networks, suggesting their potential role in immune evasion. A comparison of negative SLNs and non-SLNs revealed increased innate and adaptive immune cell types, underscoring the ongoing T cell response to tumor antigens.
    CONCLUSIONS: Our findings underscore a specific immunogenetic phenotype profile in negative SLNs, emphasizing their crucial role in the initial anticancer response, immunosurveillance, and the propagation of immune tolerance from the primary cervical tumor. These results highlight the potential of SLNs as a novel target for immunotherapy strategies and underscore the importance of new imaging methods for accurately identifying SLN status without removal. Future investigations are needed to understand further the immunological interplay within SLNs and their influence on cervical cancer progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    疫苗的发展,尤其是基于RNA的,针对患者特异性肿瘤新表位是癌症免疫治疗的一个活跃和富有成效的领域.黑色素瘤和其他实体瘤类型的有希望的临床结果正在出现。与所有癌症治疗方式一样,新表位疫苗的开发和递送也有一些缺点,包括开发患者特定产品的努力程度,预测新表位的算法的准确性,除了黑色素瘤和一些其他类型的肿瘤,实体瘤转移性病变的活检通常不可用.我们假设,在某些情况下,使用合理设计的“现成”药物组合可能证明是使患者能够原位生产自己的“新表位疫苗”的另一种途径。这些联合疗法可能由激活肿瘤相关T细胞反应的药物组成。增强这种反应,减少或消除肿瘤微环境中的免疫抑制实体,和/或改变肿瘤细胞的表型以使它们对免疫介导的裂解更敏感。在临床前和临床研究中都提供了实例,其中“现成”试剂的组合导致产生针对肿瘤衍生的新表位的T细胞,从而具有抗肿瘤活性。
    The development of vaccines, especially RNA-based, directed against patient-specific tumor neoepitopes is an active and productive area of cancer immunotherapy. Promising clinical results in melanoma and other solid tumor types are emerging. As with all cancer therapy modalities, neoepitope vaccine development and delivery also has some drawbacks, including the level of effort to develop a patient-specific product, accuracy of algorithms to predict neoepitopes, and with the exception of melanoma and some other tumor types, biopsies of metastatic lesions of solid tumors are often not available. We hypothesize that in some circumstances the use of rationally designed combinations of \"off-the-shelf\" agents may prove an additional path to enable the patient to produce his/her own \"neoepitope vaccine\" in situ. These combination therapies may consist of agents to activate a tumor-associated T-cell response, potentiate that response, reduce or eliminate immunosuppressive entities in the tumor microenvironment, and/or alter the phenotype of tumor cells to render them more susceptible to immune-mediated lysis. Examples are provided in both preclinical and clinical studies in which combinations of \"off-the-shelf\" agents lead to the generation of T cells directed against tumor-derived neoepitopes with consequent antitumor activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:OX40已被广泛研究为具有激动剂抗体的免疫疗法的靶标,这些抗体已进入癌症的临床试验,但尚未显示出实质性功效。这里,我们研究了抗小鼠(m)OX40和抗人(h)OX40抗体的潜在作用机制,包括临床相关的单克隆抗体(mAb)(GSK3174998),并评估了同种型如何改变这些机制,目的是开发用于癌症合理联合治疗的改进抗体。
    方法:在许多体内模型中评估了抗mOX40和抗hOX40mAb,包括在hOX40敲入(KI)小鼠和同基因肿瘤模型中的OT-I过继转移免疫模型。在缺乏Fcγ受体(FcγR)的hOX40KI小鼠中评估FcγR接合的影响。此外,评估了使用抗小鼠程序性细胞死亡蛋白-1(mPD-1)的联合研究.还进行了使用外周血单核细胞(PBMC)检查可能的抗hOX40mAb作用机制的体外实验。
    结果:临床相关mAbGSK3174998的同种型变体显示出不同机制的免疫调节作用;mIgG1介导直接T细胞激动,而mIgG2a间接起作用,可能通过激活FcγRs消耗调节性T细胞(Tregs)。在OT-I和EG.7-OVA模型中,hIgG1是最有效的人类同种型,能够直接和通过Treg消耗作用。抗hOX40hIgG1与抗mPD-1协同以改善EG.7-OVA模型中的治疗结果。最后,人外周血单核细胞(hPBMC)的体外测定,抗hOX40hIgG1也显示了T细胞刺激和Treg消耗的潜力。
    结论:这些发现强调了理解同种型在治疗性单克隆抗体作用机制中的作用的重要性。作为hIgG1,抗hOX40mAb可以引发多种作用机制,可以帮助或阻碍治疗结果,依赖于微环境。在设计潜在的组合伙伴及其FcγR要求时,应考虑到这一点,以实现最大的益处并改善患者的预后。
    BACKGROUND: OX40 has been widely studied as a target for immunotherapy with agonist antibodies taken forward into clinical trials for cancer where they are yet to show substantial efficacy. Here, we investigated potential mechanisms of action of anti-mouse (m) OX40 and anti-human (h) OX40 antibodies, including a clinically relevant monoclonal antibody (mAb) (GSK3174998) and evaluated how isotype can alter those mechanisms with the aim to develop improved antibodies for use in rational combination treatments for cancer.
    METHODS: Anti-mOX40 and anti-hOX40 mAbs were evaluated in a number of in vivo models, including an OT-I adoptive transfer immunization model in hOX40 knock-in (KI) mice and syngeneic tumor models. The impact of FcγR engagement was evaluated in hOX40 KI mice deficient for Fc gamma receptors (FcγR). Additionally, combination studies using anti-mouse programmed cell death protein-1 (mPD-1) were assessed. In vitro experiments using peripheral blood mononuclear cells (PBMCs) examining possible anti-hOX40 mAb mechanisms of action were also performed.
    RESULTS: Isotype variants of the clinically relevant mAb GSK3174998 showed immunomodulatory effects that differed in mechanism; mIgG1 mediated direct T-cell agonism while mIgG2a acted indirectly, likely through depletion of regulatory T cells (Tregs) via activating FcγRs. In both the OT-I and EG.7-OVA models, hIgG1 was the most effective human isotype, capable of acting both directly and through Treg depletion. The anti-hOX40 hIgG1 synergized with anti-mPD-1 to improve therapeutic outcomes in the EG.7-OVA model. Finally, in vitro assays with human peripheral blood mononuclear cells (hPBMCs), anti-hOX40 hIgG1 also showed the potential for T-cell stimulation and Treg depletion.
    CONCLUSIONS: These findings underline the importance of understanding the role of isotype in the mechanism of action of therapeutic mAbs. As an hIgG1, the anti-hOX40 mAb can elicit multiple mechanisms of action that could aid or hinder therapeutic outcomes, dependent on the microenvironment. This should be considered when designing potential combinatorial partners and their FcγR requirements to achieve maximal benefit and improvement of patient outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:肺转移是乳腺癌相关死亡的主要原因。中性粒细胞胞外诱捕网(NETs)参与乳腺癌的进展。然而,NET形成的机制尚未完全了解。这项研究认为,肿瘤细胞释放的自噬体(TRAPs)在这一过程中起着至关重要的作用。
    方法:从乳腺癌细胞系中分离TRAP,以分析其对人和小鼠中性粒细胞中NET形成的影响。这项研究使用了体外和体内模型,包括Toll样受体4(TLR4-/-)小鼠和工程化乳腺癌细胞系。免疫荧光,ELISA,西方印迹,RNA测序,和流式细胞术被用来解剖导致NET产生的信号通路,并探索其免疫抑制作用,特别关注NETs对T细胞功能的影响。使用DNaseI和αPD-L1抗体评估靶向TRAP诱导的NETs的治疗潜力及其免疫抑制功能。通过将TRAP和NETs的循环水平与乳腺癌患者的肺转移相关联来评估临床相关性。
    结果:这项研究表明,TRAP通过使用高迁移率组框1并激活TLR4-Myd88-ERK/p38信号轴,在人和小鼠中性粒细胞中诱导NETs的形成。更重要的是,TRAP诱导的NETs携带的PD-L1在体外和体内抑制T细胞功能,从而有助于形成肺转移前生态位(PMN)免疫抑制。相比之下,体内循环TRAP减少的Becn1KD-4T1乳腺肿瘤减少了NETs的形成,这反过来减弱了PMN的免疫抑制作用,并导致小鼠模型中乳腺癌肺转移的减少。此外,αPD-L1联合DNaseI降解NETs的治疗可恢复T细胞功能并显着减少肿瘤转移。乳腺癌患者外周血TRAP水平与NET水平和肺转移呈正相关。
    结论:我们的结果证明了TRAP在PD-L1修饰的NET形成中的新作用,这可能为乳腺癌患者肺转移的早期发现和治疗提供新的策略。
    BACKGROUND: Lung metastasis is the primary cause of breast cancer-related mortality. Neutrophil extracellular traps (NETs) are involved in the progression of breast cancer. However, the mechanism of NET formation is not fully understood. This study posits that tumor cell-released autophagosomes (TRAPs) play a crucial role in this process.
    METHODS: TRAPs were isolated from breast cancer cell lines to analyze their impact on NET formation in both human and mouse neutrophils. The study used both in vitro and in vivo models, including Toll-like receptor 4 (TLR4-/-) mice and engineered breast cancer cell lines. Immunofluorescence, ELISA, Western blotting, RNA sequencing, and flow cytometry were employed to dissect the signaling pathways leading to NET production and to explore their immunosuppressive effects, particularly focusing on the impact of NETs on T-cell function. The therapeutic potential of targeting TRAP-induced NETs and their immunosuppressive functions was evaluated using DNase I and αPD-L1 antibodies. Clinical relevance was assessed by correlating circulating levels of TRAPs and NETs with lung metastasis in patients with breast cancer.
    RESULTS: This study showed that TRAPs induced the formation of NETs in both human and mouse neutrophils by using the high mobility group box 1 and activating the TLR4-Myd88-ERK/p38 signaling axis. More importantly, PD-L1 carried by TRAP-induced NETs inhibited T-cell function in vitro and in vivo, thereby contributing to the formation of lung premetastatic niche (PMN) immunosuppression. In contrast, Becn1 KD-4T1 breast tumors with decreased circulating TRAPs in vivo reduced the formation of NETs, which in turn attenuated the immunosuppressive effects in PMN and resulted in a reduction of breast cancer pulmonary metastasis in murine models. Moreover, treatment with αPD-L1 in combination with DNase I that degraded NETs restored T-cell function and significantly reduced tumor metastasis. TRAP levels in the peripheral blood positively correlated with NET levels and lung metastasis in patients with breast cancer.
    CONCLUSIONS: Our results demonstrate a novel role of TRAPs in the formation of PD-L1-decorated NETs, which may provide a new strategy for early detection and treatment of pulmonary metastasis in patients with breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:大多数抗程序性细胞死亡1(PD-1)单克隆抗体(mAb)使用S228P突变IgG4作为结构基础,以避免免疫细胞或补体的激活。然而,IgG4和其他IgGFc片段之间的Fc-Fc相互作用可能导致不良反应的研究很少.Fc-无效IgG1框架是避免用IgG4观察到的不期望的Fc-Fc相互作用和Fc受体结合衍生效应的潜在更安全的替代方案。本研究提供了对这两种框架的抗PD-1mAb的综合评价。
    方法:曲妥珠单抗和利妥昔单抗(均为IgG1),野生型IgG1和IgG4固定在硝酸纤维素膜上,涂在微孔板和生物传感器芯片上,并结合肿瘤细胞作为Fc-Fc相互作用的靶标。野生型IgG1和IgG4,抗PD-1单克隆抗体nivolumab(IgG4S228P),penpullimab(Fc-nullIgG1),评估和tislelizumab(Fc-nullIgG4S228P-R409K)与固定的IgG蛋白的结合反应,并获得定量动力学数据。评估两种抗PD-1单克隆抗体对联合治疗中曲妥珠单抗和利妥昔单抗介导的免疫反应的影响,我们采用经典的抗体依赖性细胞毒性免疫模型,抗体依赖性细胞吞噬作用,和补体依赖性细胞毒性。荷瘤小鼠模型,野生型和人源化,用于体内研究。此外,我们还检查了IgG1和IgG4对不同免疫细胞群体的影响。结果:实验证明野生型IgG4和nivolumab通过Fc-Fc相互作用与固定化IgG结合,减少抗体依赖性细胞介导的细胞毒性和吞噬反应。动力学参数的定量分析表明,纳武单抗和野生型IgG4在非变性和变性状态下都表现出与固定化IgG1相当的结合亲和力。IgG4对各种免疫细胞类型发挥抑制作用。野生型IgG4和nivolumab均促进野生型小鼠模型中的肿瘤生长。相反,野生型IgG1,penpulimab,和tislelizumab未显示类似的不良反应.
    结论:通过避免IgG4的不利的Fc-Fc相互作用和Fc相关的免疫抑制作用,Fc-nullIgG1代表抗PD-1免疫疗法的更安全的选择。Fc-无效IgG4S228P-R409K和Fc-无效IgG1显示相似的结构特性和益处。这项研究有助于了解免疫疗法的耐药性和更安全的癌症免疫疗法的发展。
    BACKGROUND: The majority of anti-programmed cell-death 1 (PD-1) monoclonal antibodies (mAbs) use S228P mutation IgG4 as the structural basis to avoid the activation of immune cells or complement. However, little attention has been paid to the Fc-Fc interactions between IgG4 and other IgG Fc fragments that could result in adverse effects. Fc-null IgG1 framework is a potential safer alternative to avoid the undesirable Fc-Fc interactions and Fc receptor binding derived effects observed with IgG4. This study provides a comprehensive evaluation of anti-PD-1 mAbs of these two frameworks.
    METHODS: Trastuzumab and rituximab (both IgG1), wildtype IgG1 and IgG4 were immobilized on nitrocellulose membranes, coated to microplates and biosensor chips, and bound to tumor cells as targets for Fc-Fc interactions. Wildtype IgG1 and IgG4, anti-PD-1 mAb nivolumab (IgG4 S228P), penpulimab (Fc-null IgG1), and tislelizumab (Fc-null IgG4 S228P-R409K) were assessed for their binding reactions to the immobilized IgG proteins and quantitative kinetic data were obtained. To evaluate the effects of the two anti-PD-1 mAbs on immune responses mediated by trastuzumab and rituximab in the context of combination therapy, we employed classic immune models for antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and complement dependent cytotoxicity. Tumor-bearing mouse models, both wildtype and humanized, were used for in vivo investigation. Furthermore, we also examined the effects of IgG1 and IgG4 on diverse immune cell populations RESULTS: Experiments demonstrated that wildtype IgG4 and nivolumab bound to immobilized IgG through Fc-Fc interactions, diminishing antibody-dependent cell-mediated cytotoxicity and phagocytosis reactions. Quantitative analysis of kinetic parameters suggests that nivolumab and wildtype IgG4 exhibit comparable binding affinities to immobilized IgG1 in both non-denatured and denatured states. IgG4 exerted inhibitory effects on various immune cell types. Wildtype IgG4 and nivolumab both promoted tumor growth in wildtype mouse models. Conversely, wildtype IgG1, penpulimab, and tislelizumab did not show similar adverse effects.
    CONCLUSIONS: Fc-null IgG1 represents a safer choice for anti-PD-1 immunotherapies by avoiding both the adverse Fc-Fc interactions and Fc-related immune inhibitory effects of IgG4. Fc-null IgG4 S228P-R409K and Fc-null IgG1 displayed similar structural properties and benefits. This study contributes to the understanding of immunotherapy resistance and the advancement of safer immune therapies for cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    癌症免疫疗法在过去的10-15年里蓬勃发展,改变肿瘤学的实践,并为一些患者提供长期的临床益处。在此期间,三类不同的免疫检查点抑制剂,对两个靶标具有特异性的嵌合抗原受体-T细胞疗法,和两类不同的双特异性T细胞衔接者,疫苗,和溶瘤病毒已经加入细胞因子作为癌症治疗的标准。同时,科学进步带来了大量新知识。例如,单细胞测序和空间转录组学等技术的进步为肿瘤微环境的免疫生物学提供了深刻的见解。随着这种快速的临床和科学进步,癌症免疫治疗领域目前正处于一个关键的拐点,具有未来十年指数增长的潜力。认识到这一点,癌症免疫治疗学会召集了代表学术界的癌症免疫治疗专家,制药和生物技术行业,病人的倡导,和监管社区确定当前的机遇和挑战,目标是优先考虑具有最高临床影响潜力的领域。共识小组确定了该领域当前机会的七个高度优先领域:抗肿瘤活性和毒性机制;耐药性机制;生物标志物和生物标本;新疗法的独特方面;宿主和环境相互作用;癌前免疫,免疫拦截,和免疫预防措施;和临床试验设计,端点,和行为。此外,讨论了进展的潜在障碍,并确定了几个主题作为优化的交叉工具,每个都有可能影响多个科学优先领域。这些交叉工具包括临床前模型,数据管理和共享,活检和生物标本,资金来源多样化,定义和标准,和病人的参与。最后,确定了三项关键指导原则,这些原则将优化和最大限度地提高该领域的进展。这些包括参与患者社区;培养多样性,股本,inclusion,和可访问性;并利用人工智能的力量加速进步。这里,我们将这些讨论的结果作为激发该领域未来十年癌症免疫疗法呈指数级进展的战略愿景.
    Cancer immunotherapy has flourished over the last 10-15 years, transforming the practice of oncology and providing long-term clinical benefit to some patients. During this time, three distinct classes of immune checkpoint inhibitors, chimeric antigen receptor-T cell therapies specific for two targets, and two distinct classes of bispecific T cell engagers, a vaccine, and an oncolytic virus have joined cytokines as a standard of cancer care. At the same time, scientific progress has delivered vast amounts of new knowledge. For example, advances in technologies such as single-cell sequencing and spatial transcriptomics have provided deep insights into the immunobiology of the tumor microenvironment. With this rapid clinical and scientific progress, the field of cancer immunotherapy is currently at a critical inflection point, with potential for exponential growth over the next decade. Recognizing this, the Society for Immunotherapy of Cancer convened a diverse group of experts in cancer immunotherapy representing academia, the pharmaceutical and biotechnology industries, patient advocacy, and the regulatory community to identify current opportunities and challenges with the goal of prioritizing areas with the highest potential for clinical impact. The consensus group identified seven high-priority areas of current opportunity for the field: mechanisms of antitumor activity and toxicity; mechanisms of drug resistance; biomarkers and biospecimens; unique aspects of novel therapeutics; host and environmental interactions; premalignant immunity, immune interception, and immunoprevention; and clinical trial design, endpoints, and conduct. Additionally, potential roadblocks to progress were discussed, and several topics were identified as cross-cutting tools for optimization, each with potential to impact multiple scientific priority areas. These cross-cutting tools include preclinical models, data curation and sharing, biopsies and biospecimens, diversification of funding sources, definitions and standards, and patient engagement. Finally, three key guiding principles were identified that will both optimize and maximize progress in the field. These include engaging the patient community; cultivating diversity, equity, inclusion, and accessibility; and leveraging the power of artificial intelligence to accelerate progress. Here, we present the outcomes of these discussions as a strategic vision to galvanize the field for the next decade of exponential progress in cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    细胞因子是调节免疫细胞生长和功能活性的小蛋白,和一些已被批准用于癌症治疗。溶瘤病毒是通过直接杀死肿瘤细胞和诱导免疫应答来介导抗肿瘤活性的试剂。Talimogenelaherparepvec是一种溶瘤性单纯疱疹病毒1型(oHSV),被批准用于治疗复发性黑色素瘤,病毒编码人类细胞因子,粒细胞-巨噬细胞集落刺激因子(GM-CSF)。溶瘤病毒的一个显著优点是能够将治疗性有效载荷递送到肿瘤部位,其可以帮助驱动抗肿瘤免疫。虽然细胞因子作为有效载荷特别有趣,溶瘤病毒中使用的最佳细胞因子仍存在争议.在这次审查中,我们强调了几种细胞因子和趋化因子的初步数据,包括GM-CSF,白细胞介素12,FMS样酪氨酸激酶3配体,肿瘤坏死因子α,白介素2,白介素15,白介素18,趋化因子(C-C基序)配体2,趋化因子(C-C基序)配体5,趋化因子(C-X-C基序)配体4或其组合,并显示这些有效载荷如何进一步增强oHSV的抗肿瘤免疫力。更好地了解oHSV的细胞因子递送可以帮助提高癌症患者溶瘤病毒免疫疗法的临床益处。
    Cytokines are small proteins that regulate the growth and functional activity of immune cells, and several have been approved for cancer therapy. Oncolytic viruses are agents that mediate antitumor activity by directly killing tumor cells and inducing immune responses. Talimogene laherparepvec is an oncolytic herpes simplex virus type 1 (oHSV), approved for the treatment of recurrent melanoma, and the virus encodes the human cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). A significant advantage of oncolytic viruses is the ability to deliver therapeutic payloads to the tumor site that can help drive antitumor immunity. While cytokines are especially interesting as payloads, the optimal cytokine(s) used in oncolytic viruses remains controversial. In this review, we highlight preliminary data with several cytokines and chemokines, including GM-CSF, interleukin 12, FMS-like tyrosine kinase 3 ligand, tumor necrosis factor α, interleukin 2, interleukin 15, interleukin 18, chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 5, chemokine (C-X-C motif) ligand 4, or their combinations, and show how these payloads can further enhance the antitumor immunity of oHSV. A better understanding of cytokine delivery by oHSV can help improve clinical benefit from oncolytic virus immunotherapy in patients with cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:肿瘤靶向治疗导致令人印象深刻的肿瘤消退,但耐药性的出现限制了患者的长期生存获益.关于骨髓细胞网络的作用的信息很少,尤其是树突状细胞(DC)在肿瘤靶向治疗期间。
    方法:这里,我们使用高维多色流式细胞术结合多重免疫组织化学,研究了D4M.3A临床前黑色素瘤小鼠模型(携带V-Raf鼠肉瘤病毒癌基因同源物B(BRAF)V600E突变)中肿瘤微环境(TME)和肿瘤引流淋巴结(LN)的治疗介导的免疫学改变.这补充了RNA测序和细胞因子定量以表征肿瘤的免疫状态。通过在荷瘤小鼠中消耗CD4+或CD8+T细胞来研究T细胞在肿瘤靶向治疗期间的重要性。通过进行体内T细胞增殖测定来表征肿瘤抗原特异性T细胞应答,并且使用缺乏cDC1的Batf3-/-小鼠评估在肿瘤靶向治疗期间常规1型DC(cDC1)对T细胞免疫的贡献。
    结果:我们的发现表明BRAF抑制剂治疗增加了肿瘤的免疫原性,反映在与免疫激活相关的基因上调上。T细胞发炎的TME含有较高数量的活化cDC1和cDC2,但也含有炎性CCR2表达单核细胞。同时,肿瘤靶向治疗增加了迁移的频率,肿瘤引流LN中激活的DC亚群。甚至更多,我们鉴定了在肿瘤和LN中表达Fcγ受体I(FcγRI)/CD64的cDC2群体,其显示高水平的CD40和CCR7,表明参与了T细胞介导的肿瘤免疫.cDC2的重要性通过在cDC1缺陷型小鼠模型中仅部分丧失治疗反应来强调。CD4+和CD8+T细胞对于治疗反应都是必需的,因为它们各自的耗竭损害了治疗成功。关于抗性发展,肿瘤恢复到免疫惰性状态,DC和炎性单核细胞的损失以及调节性T细胞的积累。此外,肿瘤抗原特异性CD8+T细胞在增殖和干扰素-γ产生方面受损.
    结论:我们的研究结果为肿瘤靶向治疗的骨髓景观重塑提供了新的见解。我们证明了短暂的免疫原性肿瘤环境含有更多的活化DC。这些知识对未来组合疗法的发展具有重要意义。
    BACKGROUND: Tumor-targeted therapy causes impressive tumor regression, but the emergence of resistance limits long-term survival benefits in patients. Little information is available on the role of the myeloid cell network, especially dendritic cells (DC) during tumor-targeted therapy.
    METHODS: Here, we investigated therapy-mediated immunological alterations in the tumor microenvironment (TME) and tumor-draining lymph nodes (LN) in the D4M.3A preclinical melanoma mouse model (harboring the V-Raf murine sarcoma viral oncogene homolog B (BRAF)V600E mutation) by using high-dimensional multicolor flow cytometry in combination with multiplex immunohistochemistry. This was complemented with RNA sequencing and cytokine quantification to characterize the immune status of the tumors. The importance of T cells during tumor-targeted therapy was investigated by depleting CD4+ or CD8+ T cells in tumor-bearing mice. Tumor antigen-specific T-cell responses were characterized by performing in vivo T-cell proliferation assays and the contribution of conventional type 1 DC (cDC1) to T-cell immunity during tumor-targeted therapy was assessed using Batf3-/- mice lacking cDC1.
    RESULTS: Our findings reveal that BRAF-inhibitor therapy increased tumor immunogenicity, reflected by an upregulation of genes associated with immune activation. The T cell-inflamed TME contained higher numbers of activated cDC1 and cDC2 but also inflammatory CCR2-expressing monocytes. At the same time, tumor-targeted therapy enhanced the frequency of migratory, activated DC subsets in tumor-draining LN. Even more, we identified a cDC2 population expressing the Fc gamma receptor I (FcγRI)/CD64 in tumors and LN that displayed high levels of CD40 and CCR7 indicating involvement in T cell-mediated tumor immunity. The importance of cDC2 is underlined by just a partial loss of therapy response in a cDC1-deficient mouse model. Both CD4+ and CD8+ T cells were essential for therapy response as their respective depletion impaired therapy success. On resistance development, the tumors reverted to an immunologically inert state with a loss of DC and inflammatory monocytes together with the accumulation of regulatory T cells. Moreover, tumor antigen-specific CD8+ T cells were compromised in proliferation and interferon-γ-production.
    CONCLUSIONS: Our results give novel insights into the remodeling of the myeloid landscape by tumor-targeted therapy. We demonstrate that the transient immunogenic tumor milieu contains more activated DC. This knowledge has important implications for the development of future combinatorial therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号