PD-1/PD-L1 axis

  • 文章类型: News
    暂无摘要。
    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
    近年来,几种靶向PD-L1的单克隆抗体(mAb)已被FDA批准用于治疗癌症,证明了阻断免疫检查点的有效性,特别是PD-1/PD-L1途径。尽管基于mAb的疗法取得了长足的进步,他们仍然有自己的局限性,迫切需要能够阻断PD-1/PD-L1轴的新的小分子或PROTAC分子抑制剂。因此,在创建PD-L1阻断疗法时,将最初的体外发现转化为合适的体内动物模型至关重要.由于其广泛的可用性和较低的实验费用,经典的免疫活性小鼠对研究目的很有吸引力。然而,目前尚不清楚小鼠(m)PD-L1与人(h)PD-1的体内相互作用是否会产生功能性免疫检查点.在这次审查中,我们总结了小分子和PROTAC分子的体外和体内实验研究,特别是mPD-L1作为靶标和hPD-L1作为靶标之间的区别。
    In recent years, several monoclonal antibodies (mAbs) targeting PD-L1 have been licensed by the FDA for use in the treatment of cancer, demonstrating the effectiveness of blocking immune checkpoints, particularly the PD-1/PD-L1 pathway. Although mAb-based therapies have made great strides, they still have their limitations, and new small-molecule or PROTAC-molecule inhibitors that can block the PD-1/PD-L1 axis are desperately needed. Therefore, it is crucial to translate initial in vitro discoveries into appropriate in vivo animal models when creating PD-L1-blocking therapies. Due to their widespread availability and low experimental expenses, classical immunocompetent mice are appealing for research purposes. However, it is yet unclear whether the mouse (m) PD-L1 interaction with human (h) PD-1 in vivo would produce a functional immunological checkpoint. In this review, we summarize the in vitro and in vivo experimental studies of small molecules and PROTAC molecules, particularly the distinctions between mPD-L1 as a target and hPD-L1 as a target.
    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
    PD-1/PD-L1的上调允许癌细胞通过功能性失活T细胞免疫监视而从宿主免疫系统逃脱。临床阻断策略导致晚期癌症患者的患病率增加。然而,许多癌症患者对目前的免疫治疗策略反应有限或无反应.因此,如何提高免疫治疗的敏感性成为众多学者关注的焦点。放射治疗在肿瘤微环境中招募T细胞中起作用,增加CD4+和CD8+T细胞,并增加PD-L1表达,导致辐射和PD-L1阻断的协同增强的抗肿瘤作用。放疗可以引起肿瘤代谢的改变,尤其是葡萄糖代谢.肿瘤糖酵解和肿瘤免疫逃避是相互依存的,糖酵解活性增强肿瘤细胞上的PD-L1表达,从而促进抗PD-L1免疫疗法反应。因此,放疗影响肿瘤免疫的机制可能部分是通过对肿瘤糖代谢的干预。此外,一些作者发现2'-脱氧-2'-[18F]氟-D-葡萄糖(18F-FDG)的摄取与PD-1/PD-L1表达相关。正电子发射断层扫描/计算机断层扫描(PET/CT)是一种非侵入性的PD-1/PD-L1表达检测方法,与免疫组织化学(IHC)相比具有若干潜在优势,PET/CT可动态反映肿瘤内PD-1/PD-L1的表达情况,进一步指导临床治疗。
    Upregulation of PD-1/PD-L1 allows cancer cells to escape from host immune systems by functionally inactivating T-cell immune surveillance. Clinical blockade strategies have resulted in an increased prevalence of patients with late-stage cancers. However, many cancer patients had limited or no response to current immunotherapeutic strategies. Therefore, how to improve the sensitivity of immunotherapy has become the focus of attention of many scholars. Radiotherapy plays a role in the recruitment of T cells in the tumor microenvironment, increases CD4 + and CD8 + T cells, and increases PD-L1 expression, resulting in the synergistically enhanced antitumor effect of irradiation and PD-L1 blockade. Radiotherapy can cause changes in tumor metabolism, especially glucose metabolism. Tumor glycolysis and tumor immune evasion are interdependent, glycolytic activity enhances PD-L1 expression on tumor cells and thus promotes anti-PD-L1 immunotherapy response. Therefore, the mechanism of radiotherapy affecting tumor immunity may be partly through intervention of tumor glucose metabolism. Furthermore, some authors had found that the uptake of 2\'-deoxy-2\'-[18F]fluoro-D-glucose(18F-FDG) was correlated with PD-1/PD-L1 expression. Positron emission tomography/computed tomography (PET/CT) is a non-invasive detection method for PD-1/PD-L1 expression and has several potential advantages over immunohistochemical (IHC), PET/CT can dynamically reflect the expression of PD-1/PD-L1 inside the tumor and further guide clinical treatment.
    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
    通过阻断表面程序性死亡-配体1(PD-L1)和程序性细胞死亡蛋白1(PD-1)的相互作用的检查点抑制剂抗体疗法在癌症免疫治疗中具有有希望的优势。然而,许多患者的反应仍然不令人满意,怀疑与位于其他细胞区室的PD-L1相关,抗体无法进入细胞内区室。在这里,我们确定了具有双重作用的PD-L1靶向DNA适体(PA9-1),包括拮抗剂和依赖于PD-L1内化的递送剂。并设计了PD-L1靶向拮抗适体-ASO传递系统(PA9-1-ASO),具有协同抑制性PD-L1活性,涉及阻断和沉默机制的组合。该嵌合体不仅阻断PD-L1/PD-1,而且实现缀合的ASO的靶向递送以减少表面PD-L1和总PD-L1表达两者。与单一封锁相比,这种具有双重抑制功能的嵌合体协同抑制PD-L1增强免疫治疗功效,为免疫治疗提供了一种有前途的协同策略。
    Checkpoint inhibitor antibody therapy by blocking the interaction of surface programmed death-ligand 1(PD-L1) and programmed cell death protein 1(PD-1) has promising advantages in cancer immunotherapy. However, the response of many patients remains unsatisfactorily, suspected to be relevant to PD-L1 located in other cellular compartments and antibodies do not have access to the intracellular compartments. Herein, we identify a PD-L1-targeting DNA aptamer (PA9-1) with dual roles, including an antagonist and a delivery agent dependent on PD-L1 internalization. And we design the PD-L1-targeting antagonistic aptamer-ASO delivery system (PA9-1-ASO), with synergistic inhibitory PD-L1 activity involving the combination of blockade and silencing mechanisms. This chimera not only blocks PD-L1/PD-1 but also achieves targeted delivery of the conjugated ASO to reduce both surface PD-L1 and total PD-L1 expression. Compared with the single blockade, this chimera with the dual inhibitory function synergistically inhibits PD-L1 to amplify immunotherapeutic efficacy, providing a promising synergistic strategy for immunotherapy.
    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
    程序性死亡-1(PD-1)及其程序性死亡配体1(PD-L1)包含PD-1/PD-L1轴并维持肿瘤免疫逃避。基于抗PD-1/PD-L1抗体的癌症免疫疗法是最有前途的抗肿瘤治疗方法,但目前面临着结果不令人满意的棘手问题。中医(TCM),凭借其丰富的中药单体遗产,草药配方,以及针灸等物理疗法,艾灸,和肠线植入,是一种多组分和多目标的药物系统,以增强免疫力和防止疾病传播而闻名。中医在临床实践中经常被用作癌症的辅助治疗,最近的研究已经证明了中药与癌症免疫治疗的协同作用。在这次审查中,我们研究了PD-1/PD-L1轴及其在肿瘤免疫逃逸中的作用,同时探讨了中医药疗法如何调节PD-1/PD-L1轴以提高癌症免疫治疗的疗效.我们的发现表明,中医治疗可以通过降低PD-1和PD-L1的表达,调节T细胞功能来增强癌症的免疫治疗。改善肿瘤免疫微环境,和调节肠道菌群。我们希望这篇综述可以作为未来研究免疫检查点抑制剂(ICIs)治疗敏化的宝贵资源。
    The Programmed death-1 (PD-1) and its programmed death-ligand 1 (PD-L1) comprise the PD-1/PD-L1 axis and maintain tumor immune evasion. Cancer immunotherapy based on anti-PD-1/PD-L1 antibodies is the most promising anti-tumor treatment available but is currently facing the thorny problem of unsatisfactory outcomes. Traditional Chinese Medicine (TCM), with its rich heritage of Chinese medicine monomers, herbal formulas, and physical therapies like acupuncture, moxibustion, and catgut implantation, is a multi-component and multi-target system of medicine known for enhancing immunity and preventing the spread of disease. TCM is often used as an adjuvant therapy for cancer in clinical practices, and recent studies have demonstrated the synergistic effects of combining TCM with cancer immunotherapy. In this review, we examined the PD-1/PD-L1 axis and its role in tumor immune escape while exploring how TCM therapies can modulate the PD-1/PD-L1 axis to improve the efficacy of cancer immunotherapy. Our findings suggest that TCM therapy can enhance cancer immunotherapy by reducing the expression of PD-1 and PD-L1, regulating T-cell function, improving the tumor immune microenvironment, and regulating intestinal flora. We hope this review may serve as a valuable resource for future studies on the sensitization of immune checkpoint inhibitors (ICIs) 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
    整合素β4(ITGB4)是整合素家族的成员,在介导细胞与细胞外基质的粘附中起着至关重要的作用。最近的研究表明,ITGB4参与了肿瘤发生和转移过程。然而,ITGB4在口腔鳞状细胞癌(OSCC)中的作用尚不清楚.多重免疫组织化学(OPAL™,mIHC)测定用于染色ITGB4,ALDH1,PD-L1,细胞角蛋白(CK),人类OSCC组织微阵列中的CD8和PD-1,含有26个正常口腔上皮样本,21个口腔上皮发育不良样本和76个OSCC样本。分析ITGB4的表达模式和临床病理特征,并与PD-1,PD-L1,ALDH1和CD8进行比较。肿瘤细胞亚群之间的相关性,包括ITGB4+PD-L1+和ITGB4+ALDH1+,和T细胞亚群,包括CD8+和CD8+PD-1+,使用双尾皮尔森的统计数据进行评估。建立了卡普兰-迈耶曲线,并进行对数秩检验以分析不同亚组的生存率。mIHC染色结果显示,ITGB4大部分在肿瘤细胞中表达,与正常口腔上皮和口腔上皮发育不良相比,OSCC标本显着增加。配对分析,在OSCC肿瘤组织和正常癌旁粘膜之间进行,证实了结果。该研究进一步揭示了ITGB4+PD-L1+癌细胞,但不是ITGB4+ALDH1+癌细胞,与CD8+T细胞浸润显著相关(阳性p=0.005,阳性数量p=0.03)。此外,ITGB4+PD-L1+肿瘤细胞与CD8+PD-1+T细胞呈正相关(阳性p=0.02,阳性数量p=0.03)。最有趣的是,与其他考虑的亚组相比,CD8/PD-1high的ITGB4/PD-L1亚组的预后最佳.结果表明,与正常口腔粘膜相比,OSCC中ITGB4的表达增加。此外,发现ITGB4/PD-L1和CD8/PD-1高表达的特定亚组与其他亚组相比具有相对更好的预后.最终,本研究揭示了ITGB4在OSCC中的潜在作用,为进一步研究提供了依据。
    Integrin β4 (ITGB4) is a member of the integrin family, which plays a crucial role in mediating cell adhesion to the extracellular matrix. Recent studies have demonstrated that ITGB4 is involved in tumorigenesis and metastasis during the development of cancer. However, the role of ITGB4 in oral squamous cell carcinoma (OSCC) remains unclear. A Multiplex immunohistochemistry (OPAL™, mIHC) assay was employed to stain ITGB4, ALDH1, PD-L1, cytokeratin (CK), CD8 and PD-1 in a human OSCC tissue microarray, containing 26 normal oral epithelium samples, 21 oral epithelium dysplasia samples and 76 OSCC samples. The expression pattern and clinicopathological characteristics of ITGB4 were analyzed and compared with those of PD-1, PD-L1, ALDH1 and CD8. The correlation between subgroups of tumor cells, including ITGB4+PD-L1+ and ITGB4+ALDH1+, and subgroups of T cells, including CD8+ and CD8+PD-1+, was evaluated using two-tailed Pearson\'s statistics. A Kaplan-Meier curve was built, and a log-rank test was performed to analyze the survival rate of different subgroups. The mIHC staining results show that ITGB4 was mostly expressed in the tumor cells, with a significant increase in the OSCC specimens compared with normal oral epithelium and oral epithelium dysplasia. The paired analysis, conducted between the OSCC tumor tissue and normal paracancer mucosa, confirmed the results. The study further revealed that ITGB4+PD-L1+ cancer cells, but not ITGB4+ALDH1+ cancer cells, were significantly associated with the infiltration of CD8+ T cells (positivity p = 0.005, positive number p = 0.03). Additionally, ITGB4+PD-L1+ tumor cells were positively correlated with CD8+PD-1+ T cells (positivity p = 0.02, positive number p = 0.03). Most intriguingly, the subgroup of ITGB4/PD-L1high with CD8/PD-1high displayed the best prognosis compared with the other considered subgroups. The results show that the expression of ITGB4 was increased in OSCC compared with normal oral mucosa. Furthermore, a specific subgroup with high levels of expression of ITGB4/PD-L1 and CD8/PD-1 was found to have a relatively better prognosis compared with the other subgroups. Ultimately, this study sheds light on the potential role of ITGB4 in OSCC and provides a basis for further investigation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    肺母细胞瘤(PB)是一种罕见的侵袭性肺部恶性肿瘤,预后不良。虽然PB的主要治疗方法是手术,放疗和化疗已经有报道,对于晚期不能手术的患者,没有标准的治疗方法。此外,对驱动突变状态和免疫疗法疗效知之甚少。本文介绍了使用CT引导的肺活检病理和免疫组织化学诊断为经典双相PB的男性患者。病人的症状包括咳嗽,胸痛,呼吸急促,咯血,和缺乏活力。本文主要讨论抗PD-1免疫治疗对PB的影响。在sintilimab二线抗PD-1治疗后,患者经历了超过27个月的无进展生存期(PFS)。该患者目前存活了近40个月,生活质量令人满意。
    Pulmonary blastoma (PB) is a rare and invasive malignancy of the lungs with a poor prognosis. Although the mainstay treatment of PB is surgery, and radiotherapy and chemotherapy have been reported, no standard therapy exists for patients inoperable in advanced stages. Moreover, little is known about driver mutation status and immunotherapy efficacy. This paper presents a male patient diagnosed with classic biphasic PB using CT-guided lung biopsy pathology and immunohistochemistry. The patient\'s symptoms included cough, chest pain, shortness of breath, hemoptysis, and hypodynamia. The primary focus of this paper is to discuss the impact of anti-PD-1 immunotherapy on PB. The patient experienced progression-free survival (PFS) of over 27 months following sintilimab second-line anti-PD-1 therapy. The patient has currently survived for nearly 40 months with a satisfactory quality of life.
    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
    转移是宫颈癌(CC)治疗的主要障碍,和SPOP介导的调节作用参与转移。然而,机制尚未完全阐明。
    对CC患者的盆腔淋巴结(pLN)阳性和非pLN组进行蛋白质组测序和SPOP免疫组织化学(IHC)。通过SPOP表达水平对相应患者进行分层,以进行总生存期(OS)和无复发生存期(RFS)分析。进行了体外和体内测试以验证SPOP表达与CC转移之间的因果关系。多重免疫荧光(m-IF)和HALO系统用于分析机制,通过体外实验进一步验证。
    SPOP在pLN转移的CC中上调,与患者预后呈负相关。在体外和体内,SPOP促进CC增殖和转移。根据m-IF和HALO分析,SPOP可能通过促进PD-1与PD-L1的分离来促进CC转移。最后,进一步验证了SPOP可以通过促进PD-1在空间位置和功能上远离PD-L1的运动来实现免疫耐受。
    这项研究表明,SPOP可以通过促进PD-1远离PD-L1的运动来抑制免疫微环境,从而促进CC的pLN转移并导致OS和RFS恶化。
    Metastasis is a major obstacle in the treatment of cervical cancer (CC), and SPOP-mediated regulatory effects are involved in metastasis. However, the mechanisms have not been fully elucidated.
    Proteomic sequencing and SPOP immunohistochemistry (IHC) were performed for the pelvic lymph node (pLN)-positive and non-pLN groups of CC patients. The corresponding patients were stratified by SPOP expression level for overall survival (OS) and relapse-free survival (RFS) analysis. In vitro and in vivo tests were conducted to verify the causal relationship between SPOP expression and CC metastasis. Multiplex immunofluorescence (m-IF) and the HALO system were used to analyse the mechanism, which was further verified by in vitro experiments.
    SPOP is upregulated in CC with pLN metastasis and negatively associated with patient outcome. In vitro and in vivo, SPOP promotes CC proliferation and metastasis. According to m-IF and HALO analysis, SPOP may promote CC metastasis by promoting the separation of PD-1 from PD-L1. Finally, it was further verified that SPOP can achieve immune tolerance by promoting the movement of PD-1 away from PD-L1 in spatial location and function.
    This study shows that SPOP can inhibit the immune microenvironment by promoting the movement of PD-1 away from PD-L1, thereby promoting pLN metastasis of CC and resulting in worse OS and RFS.
    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
    抗程序性细胞死亡1(PD-1)或抗PD-配体(L)1药物,作为经典的免疫检查点抑制剂,被认为是有希望的肿瘤治疗策略。在临床实践中,一些癌症患者在抗PD-1/PD-L1免疫治疗过程中出现耐药和疾病进展。肿瘤相关巨噬细胞(TAMs)通过细胞因子抑制T细胞的募集和功能,在调节PD-1/PD-L1免疫抑制中起关键作用,表面免疫检查点配体,和外泌体。有几种疗法可通过靶向TAM来恢复PD-1/PD-L1抑制剂的抗癌功效,包括TAM分化的抑制和TAM激活的再教育。在这次审查中,我们将总结TAMs在PD-1/PD-L1阻滞剂耐药中的作用和机制。此外,我们将讨论旨在消耗TAM的疗法,再教育TAM,并干预TAMs分泌的趋化因子和M1巨噬细胞的外泌体,提供更多潜在的选择来提高PD-1/PD-L1抑制剂的疗效。
    Anti-programmed cell death 1 (PD-1) or anti-PD-ligand (L) 1 drugs, as classic immune checkpoint inhibitors, are considered promising treatment strategies for tumors. In clinical practice, some cancer patients experience drug resistance and disease progression in the process of anti-PD-1/PD-L1 immunotherapy. Tumor-associated macrophages (TAMs) play key roles in regulating PD-1/PD-L1 immunosuppression by inhibiting the recruitment and function of T cells through cytokines, superficial immune checkpoint ligands, and exosomes. There are several therapies available to recover the anticancer efficacy of PD-1/PD-L1 inhibitors by targeting TAMs, including the inhibition of TAM differentiation and re-education of TAM activation. In this review, we will summarize the roles and mechanisms of TAMs in PD-1/PD-L1 blocker resistance. Furthermore, we will discuss the therapies that were designed to deplete TAMs, re-educate TAMs, and intervene with chemokines secreted by TAMs and exosomes from M1 macrophages, providing more potential options to improve the efficacy of PD-1/PD-L1 inhibitors.
    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
    免疫疗法,特别是针对程序性细胞死亡-1(PD-1)/程序性细胞死亡配体-1(PD-L1)轴的免疫检查点阻断(ICB)治疗,在临床上极大地彻底改变了癌症治疗。抗PD-1/PD-L1治疗旨在通过阻断肿瘤细胞上的PD-L1与CTL上的PD-1之间的相互作用来恢复细胞毒性T细胞(CTL)的抗肿瘤反应。然而,由于肿瘤特异性不足,目前的抗PD-1/PD-L1治疗在多种实体瘤中治疗效果不佳,严重的细胞毒性作用,以及免疫抗性的发生。近年来,纳米药物递送系统(NDDS),具有高效的肿瘤靶向性和联合治疗的多功能性,为癌症免疫疗法开辟了一条新途径。在这篇评论文章中,我们总结了NDDSs用于抗PD-1/PD-L1治疗的最新进展.然后,我们讨论了挑战,并进一步提供了观点,以促进基于NDDS的抗PD-1/PD-L1治疗的临床应用。
    Immunotherapy, in particular immune checkpoint blockade (ICB) therapy targeting the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis, has remarkably revolutionized cancer treatment in the clinic. Anti-PD-1/PD-L1 therapy is designed to restore the antitumor response of cytotoxic T cells (CTLs) by blocking the interaction between PD-L1 on tumour cells and PD-1 on CTLs. Nevertheless, current anti-PD-1/PD-L1 therapy suffers from poor therapeutic outcomes in a large variety of solid tumours due to insufficient tumour specificity, severe cytotoxic effects, and the occurrence of immune resistance. In recent years, nanosized drug delivery systems (NDDSs), endowed with highly efficient tumour targeting and versatility for combination therapy, have paved a new avenue for cancer immunotherapy. In this review article, we summarized the recent advances in NDDSs for anti-PD-1/PD-L1 therapy. We then discussed the challenges and further provided perspectives to promote the clinical application of NDDS-based anti-PD-1/PD-L1 therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号