CD47 Antigen

CD47 抗原
  • 文章类型: Journal Article
    神经母细胞瘤是一种具有显著临床异质性的儿科癌症。尽管付出了广泛的努力,高危神经母细胞瘤患儿仍难以治愈。免疫疗法是治疗患有这种毁灭性疾病的儿童的有希望的方法。我们以前曾报道,巨噬细胞是高危神经母细胞瘤中重要的效应细胞。在这篇透视文章中,我们讨论了巨噬细胞抑制性受体SIRPA在高危神经母细胞瘤肿瘤相关巨噬细胞稳态中的潜在功能.SIRPA的配体是CD47,被称为“不要吃我”信号,与正常细胞相比,在癌细胞上高表达。CD47在肿瘤和基质细胞上表达,而SIRPA的表达仅限于高危神经母细胞瘤组织中的巨噬细胞。值得注意的是,高SIRPA表达与更好的疾病预后相关。根据目前的范式,肿瘤细胞上的CD47与巨噬细胞上的SIRPA之间的相互作用导致肿瘤吞噬作用的抑制。然而,最近的临床试验数据对使用抗CD47抗体治疗成人和儿童癌症提出了质疑.SIRPA在许多组织中的巨噬细胞上的限制性表达表明在CD47/SIRPA阻断疗法中在巨噬细胞上靶向SIRPA而不是CD47。根据迄今为止可用的数据,我们提出,抗CD47抗体破坏CD47-SIRPA相互作用会使巨噬细胞极化状态从M1转变为M2,这是由Timms等人1998年的研究推断的.相比之下,缺乏Fc的抗SIRPAF(ab')2与巨噬细胞上的SIRPA结合,模拟CD47-SIRPA相互作用,从而保持M1极化。抗SIRPAF(ab')2也阻止CD47与SIRPA的结合,从而阻止了“不要吃我”的信号。肿瘤调理和巨噬细胞激活抗体的添加有望增强活跃的肿瘤吞噬作用。
    Neuroblastoma is a pediatric cancer with significant clinical heterogeneity. Despite extensive efforts, it is still difficult to cure children with high-risk neuroblastoma. Immunotherapy is a promising approach to treat children with this devastating disease. We have previously reported that macrophages are important effector cells in high-risk neuroblastoma. In this perspective article, we discuss the potential function of the macrophage inhibitory receptor SIRPA in the homeostasis of tumor-associated macrophages in high-risk neuroblastoma. The ligand of SIRPA is CD47, known as a \"don\'t eat me\" signal, which is highly expressed on cancer cells compared to normal cells. CD47 is expressed on both tumor and stroma cells, whereas SIRPA expression is restricted to macrophages in high-risk neuroblastoma tissues. Notably, high SIRPA expression is associated with better disease outcome. According to the current paradigm, the interaction between CD47 on tumor cells and SIRPA on macrophages leads to the inhibition of tumor phagocytosis. However, data from recent clinical trials have called into question the use of anti-CD47 antibodies for the treatment of adult and pediatric cancers. The restricted expression of SIRPA on macrophages in many tissues argues for targeting SIRPA on macrophages rather than CD47 in CD47/SIRPA blockade therapy. Based on the data available to date, we propose that disruption of the CD47-SIRPA interaction by anti-CD47 antibody would shift the macrophage polarization status from M1 to M2, which is inferred from the 1998 study by Timms et al. In contrast, the anti-SIRPA F(ab\')2 lacking Fc binds to SIRPA on the macrophage, mimics the CD47-SIRPA interaction, and thus maintains M1 polarization. Anti-SIRPA F(ab\')2 also prevents the binding of CD47 to SIRPA, thereby blocking the \"don\'t eat me\" signal. The addition of tumor-opsonizing and macrophage-activating antibodies is expected to enhance active tumor phagocytosis.
    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
    肿瘤相关巨噬细胞(TAM)在免疫抑制肿瘤微环境(TME)中至关重要,最近,已经引起了人们对癌症治疗的高度关注。然而,同时促进TAMs复极化和吞噬癌细胞仍然具有挑战性。这里,构建了基于TAM靶向白蛋白纳米颗粒的递送系统(M@SINP),用于共同递送光敏剂IR820和SHP2抑制剂SHP099,以增强巨噬细胞介导的癌症免疫疗法.激光照射下的M@SINP可以产生细胞内活性氧(ROS)并促进M2-TAM成为M1表型。同时,抑制SHP2可以阻断CD47-SIRPa途径恢复M1巨噬细胞吞噬活性。M@SINP介导的TAM重塑通过将TAM重新极化为M1表型而导致免疫刺激TME,恢复其吞噬功能,促进肿瘤内CTL浸润,显著抑制肿瘤生长。此外,M@SINP与抗PD-1抗体组合还可以改善PD-1阻断的治疗结果并发挥协同抗癌作用。因此,通过M@SINP进行的巨噬细胞复极化/吞噬恢复组合有望作为同时重塑TME中TAM的策略,以提高免疫检查点阻断和常规治疗的抗肿瘤效率.
    Tumor-associated macrophages (TAMs) are pivotal within the immunosuppressive tumor microenvironment (TME), and recently, have attracted intensive attention for cancer treatment. However, concurrently to promote TAMs repolarization and phagocytosis of cancer cells remains challenging. Here, a TAMs-targeted albumin nanoparticles-based delivery system (M@SINPs) was constructed for the co-delivery of photosensitizer IR820 and SHP2 inhibitor SHP099 to potentiate macrophage-mediated cancer immunotherapy. M@SINPs under laser irradiation can generate the intracellular reactive oxygen species (ROS) and facilitate M2-TAMs to an M1 phenotype. Meanwhile, inhibition of SHP2 could block the CD47-SIRPa pathway to restore M1 macrophage phagocytic activity. M@SINPs-mediated TAMs remodeling resulted in the immunostimulatory TME by repolarizing TAMs to an M1 phenotype, restoring its phagocytic function and facilitating intratumoral CTLs infiltration, which significantly inhibited tumor growth. Furthermore, M@SINPs in combination with anti-PD-1 antibody could also improve the treatment outcomes of PD-1 blockade and exert the synergistic anticancer effects. Thus, the macrophage repolarization/phagocytosis restoration combination through M@SINPs holds promise as a strategy to concurrently remodel TAMs in TME for improving the antitumor efficiency of immune checkpoint block and conventional therapy.
    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
    使用免疫检查点抑制剂,激活T细胞,是非小细胞肺癌治疗的范式转变。然而,总体反应仍然很低。为了解决这个限制,在这里我们描述一个新颖的平台,称为抗体缀合的载药纳米治疗剂(ADN),结合了免疫治疗和分子靶向治疗。设计了一种ADN,在纳米颗粒表面上具有抗CD47和抗程序性死亡配体1(PDL1)抗体对和PI3K(磷脂酰肌醇3-激酶)/AKT/mTOR(哺乳动物雷帕霉素靶标)途径的分子靶向抑制剂,PI103,包埋在纳米颗粒中。在侵袭性肺癌免疫活性小鼠模型中,抗CD47-PDL1-ADN表现出比使用PDL1抑制剂的当前治疗选择更大的抗肿瘤功效。双重抗体-药物负载的纳米治疗剂可以成为改善癌症免疫治疗结果的有吸引力的平台。
    The use of immune checkpoint inhibitors, which activate T cells, is a paradigm shift in the treatment of non-small cell lung cancer. However, the overall response remains low. To address this limitation, here we describe a novel platform, termed antibody-conjugated drug-loaded nanotherapeutics (ADN), which combines immunotherapy and molecularly targeted therapy. An ADN was designed with an anti-CD47 and anti-programmed death ligand 1 (PDL1) antibody pair on the surface of the nanoparticle and a molecularly targeted inhibitor of the PI3K (phosphatidylinositol 3-kinase)/AKT/mTOR (mammalian target of rapamycin) pathway, PI103, entrapped in the nanoparticle. The anti-CD47-PDL1-ADN exhibited greater antitumor efficacy than current treatment options with a PDL1 inhibitor in vivo in an aggressive lung cancer immunocompetent mouse model. Dual antibody-drug-loaded nanotherapeutics can emerge as an attractive platform to improve outcomes with 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
    Evorpacept是目前正在开发用于治疗各种癌症的CD47阻断剂。据报道,在有限的血浆浓度下,evorpacept对输血前相容性测试的干扰。尽管已经提出了各种缓解策略,没有一个是实际的。这项体外研究评估了在扩展浓度下evorpacept诱导的干扰,并研究了新型缓解剂的能力,Evo-NR.
    使用手动凝胶卡和自动分析仪,在evorpacept浓度高达2,000μg/mL的含(抗E和抗Jka)或不含同种抗体的血浆中进行抗体筛选测试。Evorpacept包被的红细胞(RBC)(rr[ce/ce],Fy[a+b-],在间接抗球蛋白测试(IAT)阶段,通过直接抗球蛋白测试(DAT)和使用抗Fyb和抗S试剂的抗原分型来测试S-s)。Evo-NR用于解决血浆和RBC样品中的干扰。流式细胞术用于评估缓解效果。
    在使用手动凝胶卡(2+至3+)和自动分析仪(4+)的抗体筛选测试中,Evorpacept-spined血浆显示全反应性干扰。在自动分析仪中也观察到残留效应。使用3至6倍摩尔过量的Evo-NR有效地解决了血浆中的干扰,并能够进行准确的同种抗体鉴定。尽管通过流式细胞术鉴定了evorpacept与红细胞结合的减少,Evo-NR无法解决在DAT和IAT期抗原分型中观察到的血清学干扰。
    Evorpacept在高浓度下显示出恒定的泛反应性和残留效应。Evo-NR成功地解决了血浆样品中的干扰,并且可以被认为是一种实用且有效的缓解解决方案。实施Evo-NR有可能支持接受evorpacept治疗的患者的红细胞输注。
    UNASSIGNED: Evorpacept is a CD47-blocking agent currently being developed for the treatment of various cancers. Interference by evorpacept in pretransfusion compatibility testing has been reported at limited plasma concentrations. Although various mitigation strategies have been proposed, none are practical. This in vitro study assessed evorpacept-induced interference at extended concentrations and investigated the capability of a novel mitigation agent, Evo-NR.
    UNASSIGNED: Antibody screening tests were performed on evorpacept-spiked plasma with (anti-E and anti-Jka) or without alloantibodies at evorpacept concentrations up to 2,000 μg/mL using manual gel cards and automated analyzers. Evorpacept-coated red blood cells (RBCs) (rr [ce/ce], Fy[a+b-], S-s+) were tested by direct antiglobulin testing (DAT) and antigen typing using anti-Fyb and anti-S reagents at indirect antiglobulin testing (IAT) phase. Evo-NR was used to resolve the interference in plasma and RBC samples. Flow cytometry was used to assess the mitigation effects.
    UNASSIGNED: Evorpacept-spiked plasma showed panreactive interference in antibody screening tests using manual gel cards (2+ to 3+) and automated analyzers (4+). A carryover effect was also observed in the automated analyzers. The use of a 3- to 6-fold molar excess of Evo-NR effectively resolved the interference in the plasma and enabled accurate alloantibody identification. Although the reduction in evorpacept binding to RBCs was identified via flow cytometry, Evo-NR was incapable of resolving the serologic interference observed in DAT and antigen typing at IAT phase.
    UNASSIGNED: Evorpacept showed constant panreactivity and a carryover effect at high concentrations. Evo-NR successfully resolved the interference in the plasma samples and could be considered a practical and efficient mitigation solution. Implementation of Evo-NR has the potential to support RBC transfusion for patients undergoing evorpacept treatment.
    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
    细胞膜伪装的纳米粒子具有固有的优势,来源于其膜结构和表面抗原,包括血液中的延长循环,特定的细胞识别和靶向能力,和免疫疗法的潜力。在这里,我们引入了一个细胞膜仿生纳米药物平台,称为MPB-3BP@CMNP。包括微孔普鲁士蓝纳米颗粒(MPBNP),用作3-溴丙酮酸(3BP)的光热敏化剂和载体,这些纳米颗粒隐藏在基因可编程的细胞膜上,显示信号调节蛋白α(SIRPα)的变体,对CD47的亲和力增强。因此,MPB-3BP@CMNP继承了原始细胞膜的特征,在血液中表现出延长的循环时间,并有效地靶向结直肠癌(CRC)细胞的细胞膜上的CD47。值得注意的是,用MPB-3BP@CMNP阻断CD47可增强巨噬细胞对CRC细胞的吞噬作用。此外,3BP,己糖激酶II(HK2)的抑制剂,抑制糖酵解,导致三磷酸腺苷(ATP)水平和乳酸产生的减少。此外,它促进肿瘤相关巨噬细胞(TAMs)向抗肿瘤M1表型的极化。此外,与MPBNP介导的光热疗法(PTT)的整合可增强抗肿瘤的疗效。这些优势使MPB-3BP@CMNP成为未来发展CRC创新治疗方法的有吸引力的平台。同时,它介绍了一种用于肿瘤治疗的疾病定制细胞膜工程的通用方法。
    Cell membrane-camouflaged nanoparticles possess inherent advantages derived from their membrane structure and surface antigens, including prolonged circulation in the bloodstream, specific cell recognition and targeting capabilities, and potential for immunotherapy. Herein, we introduce a cell membrane biomimetic nanodrug platform termed MPB-3BP@CM NPs. Comprising microporous Prussian blue nanoparticles (MPB NPs) serving as both a photothermal sensitizer and carrier for 3-bromopyruvate (3BP), these nanoparticles are cloaked in a genetically programmable cell membrane displaying variants of signal regulatory protein α (SIRPα) with enhanced affinity to CD47. As a result, MPB-3BP@CM NPs inherit the characteristics of the original cell membrane, exhibiting an extended circulation time in the bloodstream and effectively targeting CD47 on the cytomembrane of colorectal cancer (CRC) cells. Notably, blocking CD47 with MPB-3BP@CM NPs enhances the phagocytosis of CRC cells by macrophages. Additionally, 3BP, an inhibitor of hexokinase II (HK2), suppresses glycolysis, leading to a reduction in adenosine triphosphate (ATP) levels and lactate production. Besides, it promotes the polarization of tumor-associated macrophages (TAMs) towards an anti-tumor M1 phenotype. Furthermore, integration with MPB NPs-mediated photothermal therapy (PTT) enhances the therapeutic efficacy against tumors. These advantages make MPB-3BP@CM NPs an attractive platform for the future development of innovative therapeutic approaches for CRC. Concurrently, it introduces a universal approach for engineering disease-tailored cell membranes for tumor therapy.
    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
    与主要依靠重链与其同源抗原建立接触的天然抗体相反,我们开发了一种双特异性抗体形式,其中轻链(LC)驱动抗原结合和特异性.为了更好地理解这种情况下的表位-互补位相互作用,我们测定了与人CD47复合的抗原结合片段(Fab)和与人PD-L1复合的另一个Fab的X-射线晶体结构.这些Fab包含κ-LC和λ-LC,分别,其与相同的重链(HC)配对。这些复合物的结构分析揭示了LC对抗原结合的主要贡献,而且常见的HC在CD47和PD-L1Fab复合物中提供了一些接触。通过使LC的互补决定区多样化,然后进行噬菌体展示选择来优化抗CD47Fab的亲和力。使用同源性建模,分析了氨基酸修饰对亲和力增加的贡献。我们的研究结果表明,尽管在天然抗体中的作用不那么突出,LC可以介导与不同抗原的高亲和力结合并中和它们的生物学功能。重要的是,含有共同可变重(VH)结构域的Fab能够产生保留真正天然结构的双特异性抗体。最大化他们的治疗潜力。
    In contrast to natural antibodies that rely mainly on the heavy chain to establish contacts with their cognate antigen, we have developed a bispecific antibody format in which the light chain (LC) drives antigen binding and specificity. To better understand epitope-paratope interactions in this context, we determined the X-ray crystallographic structures of an antigen binding fragment (Fab) in complex with human CD47 and another Fab in complex with human PD-L1. These Fabs contain a κ-LC and a λ-LC, respectively, which are paired with an identical heavy chain (HC). The structural analysis of these complexes revealed the dominant contribution of the LCs to antigen binding, but also that the common HC provides some contacts in both CD47 and PD-L1 Fab complexes. The anti-CD47 Fab was affinity optimized by diversifying complementary-determining regions of the LC followed by phage display selections. Using homology modeling, the contributions of the amino acid modification to the affinity increase were analyzed. Our results demonstrate that, despite a less prominent role in natural antibodies, the LC can mediate high affinity binding to different antigens and neutralize their biological function. Importantly, Fabs containing a common variable heavy (VH) domain enable the generation of bispecific antibodies retaining a truly native structure, maximizing their therapeutic potential.
    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
    信号调节蛋白α(SIRPα)是包括巨噬细胞和树突状细胞在内的骨髓细胞的免疫抑制受体,与CD47结合,CD47是一种普遍存在的自我结合分子。SIRPα-CD47相互作用被癌细胞利用来抑制骨髓细胞的抗肿瘤活性,因此成为癌症免疫治疗的新型免疫检查点.在血癌中,几种SIRPα-CD47阻断剂显示出令人鼓舞的单一疗法活性。然而,SIRPα-CD47阻断剂在实体瘤中的抗肿瘤活性似乎有限,提示需要联合治疗以充分利用实体瘤中的骨髓免疫检查点。在这里,我们测试了SIRPα-CD47阻断剂与带有拓扑异构酶I抑制剂DXd(DXd-ADC)的抗体-药物缀合物的组合是否会增强实体瘤中的抗肿瘤活性。为此,DS-1103a,一种新开发的抗人SIRPα抗体(Ab),评估了datopotamabderuxtecan(Dato-DXd)和曲妥珠单抗deruxtecan(T-DXd)的潜在组合益处,DXd-ADC分别靶向人滋养层细胞表面抗原2和人表皮生长因子受体2。DS-1103a抑制SIRPα-CD47相互作用,并增强Dato-DXd和T-DXd对人癌细胞的抗体依赖性细胞吞噬作用。在全癌细胞疫苗接种模型中,当与抗小鼠SIRPα(抗mSIRPα)Ab组合时,用DXd处理的癌细胞接种疫苗会导致肿瘤特异性T细胞的激活,暗示将DXd-ADC与抗SIRPαAb组合对抗肿瘤免疫的益处。此外,在同基因小鼠模型中,与单一疗法相比,Dato-DXd和T-DXd与抗mSIRPαAb的组合均显示出更强的抗肿瘤活性。一起来看,这项研究提供了联合SIRPα-CD47阻断剂和DXd-ADC治疗实体肿瘤的新疗法的临床前理论基础.
    Signal regulatory protein alpha (SIRPα) is an immune inhibitory receptor on myeloid cells including macrophages and dendritic cells, which binds to CD47, a ubiquitous self-associated molecule. SIRPα-CD47 interaction is exploited by cancer cells to suppress anti-tumor activity of myeloid cells, therefore emerging as a novel immune checkpoint for cancer immunotherapy. In blood cancer, several SIRPα-CD47 blockers have shown encouraging monotherapy activity. However, the anti-tumor activity of SIRPα-CD47 blockers in solid tumors seems limited, suggesting the need for combination therapies to fully exploit the myeloid immune checkpoint in solid tumors. Here we tested whether combination of SIRPα-CD47 blocker with antibody-drug conjugate bearing a topoisomerase I inhibitor DXd (DXd-ADC) would enhance anti-tumor activity in solid tumors. To this end, DS-1103a, a newly developed anti-human SIRPα antibody (Ab), was assessed for the potential combination benefit with datopotamab deruxtecan (Dato-DXd) and trastuzumab deruxtecan (T-DXd), DXd-ADCs targeting human trophoblast cell-surface antigen 2 and human epidermal growth factor receptor 2, respectively. DS-1103a inhibited SIRPα-CD47 interaction and enhanced antibody-dependent cellular phagocytosis of Dato-DXd and T-DXd against human cancer cells. In a whole cancer cell vaccination model, vaccination with DXd-treated cancer cells led to activation of tumor-specific T cells when combined with an anti-mouse SIRPα (anti-mSIRPα) Ab, implying the benefit of combining DXd-ADCs with anti-SIRPα Ab on anti-tumor immunity. Furthermore, in syngeneic mouse models, both Dato-DXd and T-DXd combination with anti-mSIRPα Ab showed stronger anti-tumor activity over the monotherapies. Taken together, this study provides a preclinical rationale of novel therapies for solid tumors combining SIRPα-CD47 blockers with DXd-ADCs.
    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
    肝细胞癌(HCC)获得免疫抑制微环境,导致不利的治疗结果。透明质酸介导的运动受体(HMMR)在肿瘤进展中起着至关重要的作用。这里,我们发现HMMR的异常表达可能是HCC免疫抑制微环境的预测生物标志物,但机制尚不清楚。我们建立了HMMR-/-肝癌小鼠模型,以阐明HMMR介导的“不要吃我”信号失调的机制。HMMR敲除抑制肝癌生长并诱导吞噬作用。HMMRhigh肝癌细胞通过维持CD47信号从吞噬作用中逃脱。HMMRhighCD47高表达患者的预后比HMMRlowCD47低表达患者差。HMMR在细胞质中与FAK/SRC形成复合物以激活NF-κB信号,这可能与膜与CD44的相互作用无关。值得注意的是,靶向HMMR可以通过募集CD8+T细胞来提高抗PD-1治疗效率。总的来说,我们的数据揭示了“不要吃我”信号和HMMR敲低增强抗PD-1治疗的调节机制。
    Hepatocellular carcinoma (HCC) acquires an immunosuppressive microenvironment, leading to unbeneficial therapeutic outcomes. Hyaluronan-mediated motility receptor (HMMR) plays a crucial role in tumor progression. Here, we found that aberrant expression of HMMR could be a predictive biomarker for the immune suppressive microenvironment of HCC, but the mechanism remains unclear. We established an HMMR-/- liver cancer mouse model to elucidate the HMMR-mediated mechanism of the dysregulated \"don\'t eat me\" signal. HMMR knockout inhibited liver cancer growth and induced phagocytosis. HMMRhigh liver cancer cells escaped from phagocytosis via sustaining CD47 signaling. Patients with HMMRhighCD47high expression showed a worse prognosis than those with HMMRlowCD47low expression. HMMR formed a complex with FAK/SRC in the cytoplasm to activate NF-κB signaling, which could be independent of membrane interaction with CD44. Notably, targeting HMMR could enhance anti-PD-1 treatment efficiency by recruiting CD8+ T cells. Overall, our data revealed a regulatory mechanism of the \"don\'t eat me\" signal and knockdown of HMMR for enhancing anti-PD-1 treatment.
    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
    卵巢癌是最致命的恶性肿瘤之一,具有发病率高、预后差等特点。患者在初始治疗后65-80%复发。迄今为止,对于这些患者,尚未建立有效的治疗方法。最近,CD47被认为是一种有前途的免疫治疗靶点。在本文中,本文就CD47在卵巢癌中的生物学作用及相关机制进行综述。对于大多数类型的癌症,CD47/Sirpα免疫检查点在免疫治疗中最受关注。值得注意的是,CD47单克隆抗体和相关分子在卵巢癌的免疫治疗中很有前景,需要进一步的研究。在未来,针对CD47的新型免疫治疗方案可应用于卵巢癌患者的临床治疗。
    Ovarian cancer is one of the most lethal malignant tumors, characterized by high incidence and poor prognosis. Patients relapse occurred in 65-80% after initial treatment. To date, no effective treatment has been established for these patients. Recently, CD47 has been considered as a promising immunotherapy target. In this paper, we reviewed the biological roles of CD47 in ovarian cancer and summarized the related mechanisms. For most types of cancers, the CD47/Sirpα immune checkpoint has attracted the most attention in immunotherapy. Notably, CD47 monoclonal antibodies and related molecules are promising in the immunotherapy of ovarian cancer, and further research is needed. In the future, new immunotherapy regimens targeting CD47 can be applied to the clinical treatment of ovarian cancer patients.
    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
    靶向HLA-A2限制性癌症睾丸表位NY-ESO-1157-165(A2/NY)的T细胞受体工程化(TCR工程化)T细胞(ACT)的过继转移已经产生了针对几种癌症的有利的临床应答。改善ACT的两种方法是TCR亲和力优化和T细胞共工程以表达可以利用内源性免疫的免疫调节分子。通过计算设计,我们先前开发了一组结合增强的A2/NY-TCR,包括A97L,与WT相比,这增强了基因修饰的T细胞的体外功能。这里,我们证明A97L-T细胞具有更高的持久性和改善的肿瘤控制。为了控制肿瘤中的巨噬细胞,我们进一步共同改造A97L-T细胞以分泌高亲和力信号调节蛋白α(SiRPα)诱饵(CV1),阻断CD47。虽然CV1-Fc-共工程改造的A97L-T细胞在Winn测定中显著更好地控制肿瘤生长和存活,在皮下异种移植模型中,T细胞,用CV1-Fc包被,耗尽了。重要的是,人类巨噬细胞对CV1单体共工程T细胞没有吞噬作用。此外,在CV1存在下,阿维鲁单抗和西妥昔单抗在体外增强了巨噬细胞介导的肿瘤细胞吞噬作用,并在与A97L-T细胞共同给药时改善了肿瘤控制.一起来看,我们的研究表明,通过将CV1-协同工程改造的TCR-T细胞与靶向抗体结合以直接吞噬肿瘤细胞,可以利用巨噬细胞发挥重要的临床作用.
    The adoptive transfer of T cell receptor-engineered (TCR-engineered) T cells (ACT) targeting the HLA-A2-restricted cancer-testis epitope NY-ESO-1157-165 (A2/NY) has yielded favorable clinical responses against several cancers. Two approaches to improve ACT are TCR affinity optimization and T cell coengineering to express immunomodulatory molecules that can exploit endogenous immunity. By computational design we previously developed a panel of binding-enhanced A2/NY-TCRs including A97L, which augmented the in vitro function of gene-modified T cells as compared with WT. Here, we demonstrated higher persistence and improved tumor control by A97L-T cells. In order to harness macrophages in tumors, we further coengineered A97L-T cells to secrete a high-affinity signal regulatory protein α (SiRPα) decoy (CV1) that blocks CD47. While CV1-Fc-coengineered A97L-T cells mediated significantly better control of tumor outgrowth and survival in Winn assays, in subcutaneous xenograft models the T cells, coated by CV1-Fc, were depleted. Importantly, there was no phagocytosis of CV1 monomer-coengineered T cells by human macrophages. Moreover, avelumab and cetuximab enhanced macrophage-mediated phagocytosis of tumor cells in vitro in the presence of CV1 and improved tumor control upon coadministration with A97L-T cells. Taken together, our study indicates important clinical promise for harnessing macrophages by combining CV1-coengineered TCR-T cells with targeted antibodies to direct phagocytosis against tumor cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号