anti-CD40 antibody

  • 文章类型: Journal Article
    近年来,肿瘤免疫治疗在临床上取得了显著进展。尽管年龄是癌症发展的最大主要风险因素之一,老年人代表了大多数癌症患者,只有少数新的癌症免疫治疗干预措施在老年动物中进行了临床前测试.因此,缺乏关注癌症免疫治疗过程中年龄依赖性效应的临床前研究,可能会导致幼年和老年动物的治疗结果不同,并且未来会对人体临床试验进行修改.这里,我们比较了以前开发和测试的肿瘤内免疫疗法的疗效,基于多糖甘露聚糖的组合,toll样受体配体,和抗CD40抗体(MBTA免疫治疗),在患有实验性嗜铬细胞瘤(PHEO)的年轻(6周)和老年(71周)小鼠中。所提出的结果指出,尽管PHEO在老年小鼠中生长更快,但MBTA肿瘤内免疫疗法是没有年龄依赖性的有效方法,并且可能是增强老年和年轻宿主对嗜铬细胞瘤和其他肿瘤类型的免疫反应的可能治疗干预措施之一。
    Cancer immunotherapy has shown remarkable clinical progress in recent years. Although age is one of the biggest leading risk factors for cancer development and older adults represent a majority of cancer patients, only a few new cancer immunotherapeutic interventions have been preclinically tested in aged animals. Thus, the lack of preclinical studies focused on age-dependent effect during cancer immunotherapy could lead to different therapeutic outcomes in young and aged animals and future modifications of human clinical trials. Here, we compare the efficacy of previously developed and tested intratumoral immunotherapy, based on the combination of polysaccharide mannan, toll-like receptor ligands, and anti-CD40 antibody (MBTA immunotherapy), in young (6 weeks) and aged (71 weeks) mice bearing experimental pheochromocytoma (PHEO). The presented results point out that despite faster growth of PHEO in aged mice MBTA intratumoral immunotherapy is effective approach without age dependence and could be one of the possible therapeutic interventions to enhance immune response to pheochromocytoma and perhaps other tumor types in aged and young hosts.
    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
    免疫刺激信号在引发抗肿瘤免疫和提高亚单位癌症疫苗的功效中起重要作用。然而,许多免疫刺激剂的临床使用通常受到其无效的体内递送的阻碍,这可能降低对疫苗接种的免疫应答。为了提高疫苗的效力,我们开发了整合三个关键要素的疫苗制剂:(1)通过将激动性抗CD40单克隆抗体(αCD40)缀合到负载聚IC的脂质纳米颗粒的表面而配制的纳米佐剂,(2)含有源自黑素瘤抗原gp100的优化的CD8+T细胞表位的肽两亲物,(3)增强疫苗接种功效的激动性抗4-1BB单克隆抗体(α4-1BB)。在黑色素瘤的同基因小鼠模型中,疫苗制剂增强了先天免疫,并激活了引流淋巴结内的多种先天免疫信号通路,以及促进抗原特异性免疫反应和减少肿瘤微环境中的免疫抑制,导致严重的肿瘤生长抑制和延长生存期。因此,我们的疫苗制剂代表了刺激抗肿瘤免疫和控制肿瘤进展的有吸引力的策略.重要声明:许多免疫刺激剂的临床使用通常受到其无效的体内递送的阻碍,这可能降低对疫苗接种的免疫应答。提高亚单位疫苗的抗肿瘤免疫力,我们开发了新型疫苗制剂,整合了多功能模式,包括(1)含有抗CD40单克隆抗体(αCD40)和TLR3激动剂的纳米佐剂,通过不同的信号通路激活先天免疫,(2)含有来自肿瘤抗原的优化的CD8+T细胞表位的肽两亲物,(3)增强疫苗接种效力的抗4-1BB单克隆抗体(α4-1BB)。在这项研究中,我们的疫苗配方可刺激优异的抗肿瘤免疫力并控制肿瘤进展.上述纳米工程平台和免疫原性生物大分子可进一步应用于其他T细胞诱导疫苗。
    Immunostimulatory cues play an important role in priming antitumor immunity and promoting the efficacy of subunit cancer vaccines. However, the clinical use of many immunostimulatory agents is often hampered by their inefficient in vivo delivery which may decrease immune response to the vaccination. To promote vaccine efficacy, we develop vaccine formulations which integrate three key elements: (1) a nano-adjuvant formulated by conjugating an agonistic anti-CD40 monoclonal antibody (αCD40) to the surface of a polyIC-loaded lipid nanoparticle, (2) a peptide amphiphile containing an optimized CD8+ T-cell epitope that derived from a melanoma antigen gp100, (3) an agonistic anti-4-1BB monoclonal antibody (α4-1BB) that boosts the efficacy of vaccinations. In a syngeneic mouse model of melanoma, the vaccine formulations enhanced innate immunity and activated multiple innate immune signaling pathways within draining lymph nodes, as well as promoted antigen-specific immune responses and reduced immunosuppression in the tumor microenvironment, leading to profound tumor growth inhibition and prolonged survival. Thus, our vaccine formulations represent an attractive strategy to stimulate antitumor immunity and control tumor progression. STATEMENT OF SIGNIFICANCE: The clinical use of many immunostimulatory agents is often hampered by their inefficient in vivo delivery which may decrease immune response to the vaccination. To promote the antitumor immunity of subunit vaccines, we develop novel vaccine formulations that integrate multifunctional modalities including (1) a nano-adjuvant containing anti-CD40 monoclonal antibody (αCD40) and TLR3 agonist which activate innate immunity through diverse signaling pathways, (2) a peptide amphiphile containing an optimized CD8+ T-cell epitope from tumor antigen, (3) an anti-4-1BB monoclonal antibody (α4-1BB) that boosts the efficacy of vaccinations. In this study, our vaccine formulations stimulate superior antitumor immunity and control tumor progression. The above nano-engineered platform and immunogenic biomacromolecules can be further applied to other T-cell-inducing vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    人和犬肉瘤难以治疗软组织恶性肿瘤,迫切需要新的改进的治疗选择。人类的局部复发率在10%-30%之间,和30%-40%发展转移。患有肉瘤的狗的结果随等级而变化,但可能相似。宠物狗共享人类环境,代表人类癌症,在宿主和肿瘤中具有遗传变异。我们问我们使用基因相同的小鼠和克隆的肿瘤细胞进行的小鼠研究是否可以转化为更大的,基因多样的家犬自然发生肿瘤,(i)开发一种犬癌治疗方法,和(ii)用作人类的翻译途径。我们的小鼠研究表明,肿瘤内递送白细胞介素-2(IL-2)加上激动剂抗CD40抗体(Ab)可诱导30%至100%的长期治愈反应,取决于肿瘤类型。由于适合瘤内注射和直接监测,我们开发了一种激动剂抗犬CD40Ab,并在患有软组织肉瘤的狗(n=27)中进行了I期剂量发现/毒理学33临床试验。用IL-2加抗CD40抗体治疗狗2周。三种剂量水平诱导肿瘤消退,副作用最小,通过监测测量,血液学和生化分析。重要的是,我们的小鼠和犬类研究提供了令人鼓舞的基本概念验证数据,我们可以根据这些数据制定兽医和人类免疫治疗策略.
    Human and canine sarcomas are difficult to treat soft tissue malignancies with an urgent need for new improved therapeutic options. Local recurrence rates for humans are between 10%-30%, and 30%-40% develop metastases. Outcomes for dogs with sarcoma vary with grade but can be similar. Pet dogs share the human environment and represent human cancer with genetic variation in hosts and tumours. We asked if our murine studies using genetically identical mice and cloned tumour cells were translatable to larger, genetically diverse domestic dogs with naturally occurring tumours, to (i) develop a canine cancer therapeutic, and (ii) to use as a translational pathway to humans. Our murine studies showed that intra-tumoral delivery of interleukin-2 (IL-2) plus an agonist anti-CD40 antibody (Ab) induces long-term curative responses ranging from 30% to 100%, depending on tumour type. We developed an agonist anti-canine-CD40 Ab and conducted a phase I dose finding/toxicology 3 + 3 clinical trial in dogs (n = 27) with soft tissue sarcomas on account of suitability for intratumoral injection and straightforward monitoring. Dogs were treated with IL-2 plus anti-CD40 antibody for 2 weeks. Three dose levels induced tumour regression with minimal side effects, measured by monitoring, haematological and biochemical assays. Importantly, our mouse and canine studies provide encouraging fundamental proof-of-concept data upon which we can develop veterinary and human immunotherapeutic strategies.
    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
    Although chimeric antigen receptor (CAR)-T cell therapy has been remarkably successful for haematological malignancies, its efficacy against solid tumors is limited. The combination of CAR-T cell therapy with immune checkpoint inhibitors (CPIs), such as PD-1, PD-L1, and CTLA-4 antibodies, is a promising strategy for enhancing the antitumor efficacy of CAR-T cells. However, because most patients acquire resistance to CPIs, investigating other strategies is necessary to further improve the antitumor efficacy of CAR-T cell therapy for solid tumors. Recently, CD40 agonist antibodies showed potential antitumor efficacy by activating the CD40 pathway.
    Based on the piggyBac transposon system, rather than the widely used viral vectors, we constructed a meso3-CD40 CAR-T targeting region III of mesothelin (MSLN) that possessed the ability to secrete anti-CD40 antibodies. Compared with meso3 CAR-T cells, which did not secrete the anti-CD40 antibody, meso3-CD40 CAR-T cells secreted more cytokines and had a relatively higher proportion of central memory T (TCM) cells after stimulation by the target antigen. In addition, compared with meso3 CAR-T cells, meso3-CD40 CAR-T cells had a more powerful cytotoxic effect on target cells at a relatively low effector-to-target ratio. More importantly, we demonstrated that the antitumor activity of meso3-CD40 CAR-T cells was enhanced in a human ovarian cancer xenograft model in vivo.
    In conclusion, these results highlight anti-CD40-secreting CAR-T cells generated by nonviral vectors as a potential clinical strategy for improving the efficacy of CAR-T cell therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    角膜异种移植是解决人类角膜供体短缺的有效方法,并且猪角膜可以是供体角膜的合适候选物,因为其与人类的光学相似性。然而,有必要施用额外的免疫抑制剂以克服抗原差异。我们旨在研究在临床适用的猪到非人灵长类动物角膜异种移植模型中使用抗CD40抗体介导的共刺激阻断的猪角膜的可行性。
    五只中国恒河猴使用临床上可接受的大小(直径7.5mm)的猪角膜移植物进行了深层状角膜移植。抗CD40抗体以程序化的时间表静脉内施用。移植物存活,中央角膜厚度,并评估眼压。在血液中研究了效应和记忆T和B细胞亚群以及抗αGal和供体特异性抗体的变化,并评估了房水和血液中补体水平的变化。将抗CD40抗体治疗组的记忆细胞谱与抗CD154抗体治疗组的记忆细胞谱或我们先前报告中提出的拒绝对照的记忆细胞谱进行比较。抗αGal的变化,非αGal,6个月后的供体特异性抗体与基线值进行比较.
    抗CD40抗体介导的共刺激阻断导致异种角膜移植物成功存活(>389、>382、>236、>201和>61天),80%的人可存活6个月。抗CD40抗体的注射显著减少了炎性细胞向移植物中的浸润,并且显著阻断了房水中的补体应答(P=.0159,Mann-WhitneyU检验)。与排斥对照中的那些(P<.05,Mann-WhitneyU检验)或在抗CD154抗体处理的灵长类动物中的那些(P>.05,Mann-WhitneyU检验)相比,在抗CD40抗体处理的灵长类动物中消除了中枢或效应记忆T细胞的系统扩增。抗αGal的水平,非αGal,与基线水平相比,6个月时的供体特异性抗体没有显着增加(P>.05,Wilcoxon符号秩检验)。
    抗CD40抗体介导的阻断似乎是灵长类动物猪角膜深层异种移植的有效免疫抑制方法。
    Corneal xenotransplantation is an effective solution for the shortage of human donor corneas, and the porcine cornea may be a suitable candidate for the donor cornea because of its optical similarity with humans. However, it is necessary to administer additional immunosuppressants to overcome antigenic differences. We aimed to investigate the feasibility of porcine corneas with anti-CD40 antibody-mediated costimulation blockade in a clinically applicable pig-to-non-human primate corneal xenotransplantation model.
    Five Chinese rhesus macaques underwent deep-lamellar corneal transplantation using clinically acceptable sized (7.5 mm diameter) porcine corneal grafts. The anti-CD40 antibody was intravenously administered on a programmed schedule. Graft survival, central corneal thickness, and intraocular pressure were evaluated. Changes in effector and memory T and B cell subsets and anti-αGal and donor-specific antibodies were investigated in the blood, and the changes in complement levels in the aqueous humor and blood were evaluated. Memory cell profiles in the anti-CD40 antibody-treated group were compared with those from the anti-CD154 antibody-treated group or rejected controls presented in our previous report. The changes in anti-αGal, non-αGal, and donor-specific antibodies after 6 months were compared with baseline values.
    Anti-CD40 antibody-mediated costimulation blockade resulted in the successful survival of xenocorneal grafts (>389, >382, >236, >201, and >61 days), with 80% reaching 6 months of survival. Injection of anti-CD40 antibody considerably reduced the infiltration of inflammatory cells into the grafts and significantly blocked the complement response in the aqueous humor (P=.0159, Mann-Whitney U test). Systemic expansion of central or effector memory T cells was abrogated in the anti-CD40 antibody-treated primates compared with those in the rejected controls (P<.05, Mann-Whitney U test) or those in the anti-CD154 antibody-treated primates (P>.05, Mann-Whitney U test). The levels of anti-αGal, non-αGal, and donor-specific antibodies at 6 months were not significantly increased compared with baseline levels (P>.05, Wilcoxon signed rank test).
    An anti-CD40 antibody-mediated blockade appears to be effective immunosuppressive approach for porcine corneal deep-lamellar xenotransplantation in primates.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:最近,我们已经证明,包括通过抗CD154抗体的共刺激阻断在内的免疫抑制方案可显著延长GTKO中的心脏异种移植物存活时间.hCD46Tg猪与狒狒异位异种移植模型。不幸的是,使用抗CD154抗体观察到许多凝血障碍,受者生存率因这些并发症而显著降低。
    方法:在本实验中,我们用临床上更可接受的抗CD40抗体替代了抗CD154抗体,同时保持其余免疫抑制方案和供体猪的遗传学相同.进行此评估是为了评估抗体在异种移植物存活和预防凝血病中的作用。测试了抗CD40抗体的两个可用克隆。一种小鼠抗人CD40抗体,(克隆3A8),体外激活的B淋巴细胞,仅在体内适度抑制抗体产生。而针对猕猴CD40的重组小鼠非人灵长类动物嵌合(克隆2C10R4),在体外阻断B细胞活化并在体内完全阻断抗体产生。
    结果:抗CD154抗体可有效避免血栓并发症,但移植物存活,虽然延伸,没有抗CD154抗体治疗所观察到的延长。3A8抗体组的最长存活时间为27天,2C10R4抗体组的最长移植物存活时间为146天。除两个被拒绝并被移植外,所有移植物。由于无关的并发症,只有两个受体狒狒必须被安乐死,其余的狒狒在整个移植物存活期间或移植物外植体后保持健康。与我们的抗CD154抗体治疗的狒狒相反,移植后8周左右B细胞出现后,非Gal抗体水平开始升高.
    结论:当前剂量的抗CD40抗体不会诱导任何凝血功能障碍,但虽然有效,诱导与抗CD154抗体相似的长期移植物存活的功效降低,这可能是由于在当前剂量下对B细胞功能和抗体产生的控制无效。需要更多的实验来确定诱导长期心脏异种移植物存活的抗体亲和力和有效剂量。
    BACKGROUND: Recently, we have shown that an immunosuppression regimen including costimulation blockade via anti-CD154 antibody significantly prolongs the cardiac xenograft survival in a GTKO.hCD46Tg pig-to-baboon heterotopic xenotransplantation model. Unfortunately, many coagulation disorders were observed with the use of anti-CD154 antibody, and recipient survival was markedly reduced by these complications.
    METHODS: In this experiment, we replaced anti-CD154 antibody with a more clinically acceptable anti-CD40 antibody while keeping the rest of the immunosuppressive regimen and the donor pig genetics the same. This was carried out to evaluate the antibody\'s role in xenograft survival and prevention of coagulopathies. Two available clones of anti-CD40 antibody were tested. One mouse anti-human CD40 antibody, (clone 3A8), activated B lymphocytes in vitro and only modestly suppressed antibody production in vivo. Whereas a recombinant mouse non-human primate chimeric raised against macaque CD40, (clone 2C10R4), blocked B-cell activation in vitro and completely blocked antibody production in vivo.
    RESULTS: The thrombotic complications seen with anti-CD154 antibody were effectively avoided but the graft survival, although extended, was not as prolonged as observed with anti-CD154 antibody treatment. The longest survival for the 3A8 antibody group was 27 days, and the longest graft survival in the 2C10R4 antibody group was 146 days. All of the grafts except two rejected and were explanted. Only two recipient baboons had to be euthanized due to unrelated complications, and the rest of the baboons remained healthy throughout the graft survival period or after graft explantation. In contrast to our anti-CD 154 antibody-treated baboons, the non-Gal antibody levels started to rise after B cells made their appearance around 8 weeks post-transplantation.
    CONCLUSIONS: Anti-CD40 antibody at the current dose does not induce any coagulopathies but while effective, had reduced efficacy to induce similar long-term graft survival as with anti-CD154 antibody perhaps due to ineffective control of B-cell function and antibody production at the present dose. More experiments are required to determine antibody affinity and effective dose for inducing long-term cardiac xenograft survival.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号