anti-PD-1

抗 PD - 1
  • 文章类型: Case Reports
    手术切除的III期黑色素瘤是一种具有高复发风险的疾病。免疫检查点抑制剂(ICIs)以及BRAF和MEK抑制剂的靶向治疗显着改变了转移性黑色素瘤患者的预后,并且一些研究也显示了它们在III期黑色素瘤患者中延迟复发的辅助治疗中的益处。在转移背景下,观察到过度进展疾病可能是对免疫疗法的不良反应,这表明一些患者可能面临ICI进展的额外风险,尽管对于该事件的正确定义尚未达成共识.
    我们在此描述了III期黑素瘤切除患者在辅助免疫疗法期间快速多器官转移的两例病例。尽管将这种综合征定义为过度进展是不准确的,因为在辅助治疗中明显没有初始疾病,我们观察到,在这2例病例中,在首次给予辅助ICIs后,同样快速的进展导致患者在治疗开始后2个月内死亡.两名患者均患有NRAS突变的黑色素瘤。
    迫切需要更好地了解这些致命结果的原因,并识别生物标志物,以便在为患者提供辅助治疗之前选择患者。降低过度进展的风险。从这些案例中,我们建议,在基于分子谱提出ICI辅助治疗时,应特别注意这一点.
    UNASSIGNED: Stage III surgically resected melanoma is a disease at high risk of recurrence. Immune checkpoint inhibitors (ICIs) and the target therapy with BRAF and MEK inhibitors significantly changed the outcome of patients with metastatic melanoma and several studies have also shown their benefit in the adjuvant setting for the delay of recurrence in stage III melanoma patients. Hyperprogression disease was observed as a possible adverse response to immunotherapy in the metastatic setting, suggesting that some patients could face additional risk of progression with ICIs, although no consensus was found for the correct definition of this event.
    UNASSIGNED: We describe here two cases of rapid multiorgan metastatization during adjuvant immunotherapy in patients with stage III resected melanoma. Even though it would be not accurate to define this syndrome as hyperprogression because of apparent absence of the initial disease in the adjuvant setting, we observed in these two cases the same very rapid progression after first administration of adjuvant ICIs that resulted in death of patients within two months from the starting of treatment. Both patients had NRAS mutated melanoma.
    UNASSIGNED: There is an urgent need for a better understanding of the causes of these fatal outcomes and for the identification of biomarkers that would allow to select the patients before offering them an adjuvant treatment, reducing the risk of hyperprogression. From these cases, we suggest that it could be useful a particular attention in proposing ICI adjuvant treatment based on the molecular profile.
    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
    克服免疫介导的对PD-1阻断的抗性仍然是主要的临床挑战。在联合使用nivolumab(抗PD-1)和relatlimab(抗LAG-3)治疗的黑色素瘤患者中,已证明疗效增强。这是同类产品中第一个获得FDA批准的。然而,这两种抑制性受体如何协同作用以阻碍抗肿瘤免疫仍然未知。这里,我们显示,CD8+T细胞缺乏PD-1和LAG-3,与缺乏任一受体的CD8+T细胞相反,在黑色素瘤小鼠模型中介导增强的肿瘤清除和长期存活。PD-1-和LAG-3缺陷型CD8+T细胞在转录上不同,具有广泛的TCR克隆性和效应物样和干扰素反应基因的富集,导致增强的IFN-γ释放指示功能性。LAG-3和PD-1联合驱动T细胞耗尽,在调节TOX表达中起主导作用。机械上,自分泌,PD-1-和LAG-3-缺陷的CD8+T细胞需要细胞固有的IFN-γ信号传导来增强抗肿瘤免疫力,深入了解LAG-3和PD-1的组合靶向如何增强疗效。
    Overcoming immune-mediated resistance to PD-1 blockade remains a major clinical challenge. Enhanced efficacy has been demonstrated in melanoma patients with combined nivolumab (anti-PD-1) and relatlimab (anti-LAG-3) treatment, the first in its class to be FDA approved. However, how these two inhibitory receptors synergize to hinder anti-tumor immunity remains unknown. Here, we show that CD8+ T cells deficient in both PD-1 and LAG-3, in contrast to CD8+ T cells lacking either receptor, mediate enhanced tumor clearance and long-term survival in mouse models of melanoma. PD-1- and LAG-3-deficient CD8+ T cells were transcriptionally distinct, with broad TCR clonality and enrichment of effector-like and interferon-responsive genes, resulting in enhanced IFN-γ release indicative of functionality. LAG-3 and PD-1 combined to drive T cell exhaustion, playing a dominant role in modulating TOX expression. Mechanistically, autocrine, cell-intrinsic IFN-γ signaling was required for PD-1- and LAG-3-deficient CD8+ T cells to enhance anti-tumor immunity, providing insight into how combinatorial targeting of LAG-3 and PD-1 enhances efficacy.
    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
    目的:本文旨在研究抗程序性死亡1(anti-PD-1)治疗引起的脊髓损伤(SCI)的毒性机制。方法:采用骨髓移植Rag1-/-小鼠建立SCI模型。结果:抗PD-1通过CD8+T细胞活化导致SCI,而CD8+T细胞的过度活化进一步加重SCI。抗PD-1和CD8+T细胞活化均诱导凋亡相关穿孔素的表达,GrB和FasL,但抑制了PI-9的水平。在神经血清素对这些因素的影响中观察到相反的结果。CD8+T细胞活化通过上调穿孔素诱导神经毒性,GrB和FasL并抑制PI-9。此外,Neuroserpin通过穿孔素/GrB/PI-9/FasL途径抑制CD8+T细胞活化。结论:本研究结果可为抗PD-1所致SCI的临床治疗提供理论依据。
    这篇文章是关于什么的?在治疗癌症的过程中,免疫检查点抑制剂,如抗程序性死亡1(抗PD-1)治疗,作为一种免疫疗法,发展迅速,并显著改变了癌症的管理方式。然而,一些接受免疫检查点阻断治疗的癌症患者遭受严重的不良反应,包括脊髓损伤(SCI).本文首次构建骨髓移植小鼠模型,探讨抗PD-1治疗SCI的毒性机制。结果是什么?我们发现抗PD-1治疗可以诱导免疫细胞的激活,而免疫细胞活化通过调节细胞死亡途径进一步促进神经细胞的自我破坏。研究结果是什么意思?抗PD-1治疗引起SCI的机制是通过调节细胞死亡途径来激活免疫细胞,从而诱导神经细胞的自我破坏。本研究结果为抗PD-1治疗脊髓损伤的临床治疗提供了理论依据。
    Aims: This paper was to scrutinize the toxicity mechanism of anti-programmed death 1 (anti-PD-1) therapy-caused spinal cord injury (SCI). Methods: Bone marrow transplant Rag1-/- mice were used to establish SCI model. Results: Anti-PD-1 results in SCI via CD8+ T-cells activation, while excessive activation of CD8+ T-cells further aggravated SCI. Both anti-PD-1 and the activation of CD8+ T-cells induced the expression of apoptosis-related perforin, GrB and FasL, but suppressed PI-9 level. The opposite results were observed in the effects of neuroserpin on these factors. CD8+ T-cells activation induced neurotoxicity via upregulation perforin, GrB and FasL and inhibiting PI-9. Additionally, neuroserpin suppressed CD8+ T-cells activation via perforin/GrB/PI-9/FasL pathways. Conclusion: These results may provide theoretical foundation for the clinical treatment of SCI caused by anti-PD-1.
    What is this article about? In the process of treating cancer, immune checkpoint inhibitors such as anti-programmed death 1 (anti-PD-1) therapy, as a form of immunotherapy, have developed rapidly and changed the way to manage cancers significantly. However, some cancer patients who receive immune checkpoint blockade treatment suffer from severe adverse effects including spinal cord injury (SCI). This article for the first time constructed a bone marrow transplant mouse model to explore the toxicity mechanism of anti-PD-1 therapy-caused SCI.What were the results? We found that anti-PD-1 therapy can induce the activation of immune cells, while immune cell activation further promotes self-destruction of nerve cells by regulating cell death pathways.What do the results of the study mean? The mechanism of anti-PD-1 therapy-caused SCI is to activate of immune cells through regulating cell death pathways, thereby inducing self-destruction of nerve cells. These findings provide theoretical foundation for the clinical treatment of SCI caused by anti-PD-1 therapy.
    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: Case Reports
    胸腺癌(TC)是一种罕见的胸腺上皮肿瘤。复发或难治性TC患者预后不良。免疫检查点抑制剂单一疗法可以作为这种情况的二线治疗。这项研究报告了一名TC患者,该患者对常规化疗和放疗无反应,但在使用抗程序性细胞死亡1抑制剂sintilimab进行三线治疗后,获得了持续17个月的长期部分缓解。该患者没有经历任何与sintilimab治疗相关的严重副作用。以上结果表明,辛替利玛可能是难治性TC患者的可行治疗选择。
    Thymic carcinoma (TC) is an uncommon type of thymic epithelial tumors. Patients with relapsed or refractory TCs have a poor prognosis. Immune checkpoint inhibitor monotherapy can be applied as a second-line treatment for such cases. This study reported a TC patient who did not respond to conventional chemotherapy and radiotherapy but achieved prolonged partial remission lasting 17 months following the third-line treatment with anti-programmed cell death-1 inhibitor sintilimab. This patient did not experience any serious side effects associated with sintilimab treatment. The above results demonstrated that sintilimab could be a feasible therapeutic option for refractory TC 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
    胰腺导管腺癌(PDAC)的存活率为12%,并且针对PDAC的抗PD1疗法的多项临床试验均失败,表明需要新的治疗策略。在这项研究中,我们评估了米尔贝霉素肟(MBO)的潜力,一种抗寄生虫化合物,作为PDAC中的免疫调节剂。我们的结果表明,MBO通过诱导凋亡抑制多种PDAC细胞系的生长。体内研究表明,口服5mg/kgMBO可以抑制皮下和原位模型中PDAC肿瘤的生长49%和56%,分别。此外,与对照组相比,MBO治疗显著增加了荷瘤小鼠27天的存活率。有趣的是,MBO治疗小鼠的肿瘤CD8+T细胞浸润增加.值得注意的是,CD8+T细胞的消耗显著降低了MBO在小鼠中的抗肿瘤功效。Further,MBO显著增强抗PD1治疗的疗效,和联合治疗导致TME内更大比例的活性细胞毒性T细胞。在我们所有的临床前毒理学研究中,MBO是安全且耐受性良好的。总的来说,我们的研究为MBO对抗PDAC的应用提供了新的方向,并强调了MBO再利用以增强抗PD1免疫治疗的潜力.
    Pancreatic ductal adenocarcinoma (PDAC) has a survival rate of 12%, and multiple clinical trials testing anti-PD-1 therapies against PDAC have failed, suggesting a need for a novel therapeutic strategy. In this study, we evaluated the potential of milbemycin oxime (MBO), an antiparasitic compound, as an immunomodulatory agent in PDAC. Our results show that MBO inhibited the growth of multiple PDAC cell lines by inducing apoptosis. In vivo studies showed that the oral administration of 5 mg/kg MBO inhibited PDAC tumor growth in both subcutaneous and orthotopic models by 49% and 56%, respectively. Additionally, MBO treatment significantly increased the survival of tumor-bearing mice by 27 days as compared to the control group. Interestingly, tumors from MBO-treated mice had increased infiltration of CD8+ T cells. Notably, depletion of CD8+ T cells significantly reduced the anti-tumor efficacy of MBO in mice. Furthermore, MBO significantly augmented the efficacy of anti-PD-1 therapy, and the combination treatment resulted in a greater proportion of active cytotoxic T cells within the tumor microenvironment. MBO was safe and well tolerated in all our preclinical toxicological studies. Overall, our study provides a new direction for the use of MBO against PDAC and highlights the potential of repurposing MBO for enhancing anti-PD-1 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
    目的:CheckMate227(NCT02477826)评估了一线nivolumab联合ipilimumab与化疗在转移性非小细胞肺癌(NSCLC)患者中的程序性死亡配体1(PD-L1)表达≥1%或<1%且无EGFR/ALK改变。然而,许多随机接受化疗的患者随后接受了免疫治疗.这里,对nivolumab联合ipilimumab的总生存期(OS)和相对OS获益进行了校正,校正了治疗切换带来的潜在偏倚.
    方法:治疗切换调整分析是根据NICE决策支持部门技术支持文件16进行的,对于接受治疗的患者的CheckMate227第1部分OS数据(数据库锁定,2019年7月2日)。在基本情况分析中使用了审查权重的逆概率(IPCW);探索了其他方法作为敏感性分析。
    结果:在1166名随机患者中,391例(PD-L1≥1%)和185例(PD-L1<1%)患者接受了纳武单抗联合伊匹单抗;387例(PD-L1≥1%)和183例(PD-L1<1%)患者接受了化疗,最低随访时间为29.3个月。在接受化疗的患者中,169/387(43.7%;PD-L1≥1%)和66/183(36.1%;PD-L1<1%)在研究后转为免疫治疗。在接受治疗的患者中,nivolumab联合ipilimumab的中位OS为17.4个月,而化疗的中位OS为14.9个月(风险比[HR],0.80;95%置信区间[CI],0.68-0.95)在PD-L1≥1%亚组和17.1对12.4个月(HR,0.62;95%CI,0.49-0.80)在PD-L1<1%亚组中。使用IPCW进行治疗切换调整后,nivolumab+ipilimumab与化疗相比,OS的HR(95%CI)降至0.68(0.56~0.83;PD-L1≥1%)和0.53(0.40~0.69;PD-L1<1%).敏感性分析支持结果的稳健性。
    结论:治疗转换调整导致一线纳武单抗联合伊匹单抗治疗转移性NSCLC患者相对于化疗的相对OS获益更大。
    OBJECTIVE: CheckMate 227 (NCT02477826) evaluated first-line nivolumab-plus-ipilimumab versus chemotherapy in patients with metastatic nonsmall cell lung cancer (NSCLC) with programmed death ligand 1 (PD-L1) expression ≥ 1% or < 1% and no EGFR/ALK alterations. However, many patients randomized to chemotherapy received subsequent immunotherapy. Here, overall survival (OS) and relative OS benefit of nivolumab-plus-ipilimumab were adjusted for potential bias introduced by treatment switching.
    METHODS: Treatment-switching adjustment analyses were conducted following the NICE Decision Support Unit Technical Support Document 16, for CheckMate 227 Part 1 OS data from treated patients (database lock, July 2, 2019). Inverse probability of censoring weighting (IPCW) was used in the base-case analysis; other methods were explored as sensitivity analyses.
    RESULTS: Of 1166 randomized patients, 391 (PD-L1 ≥ 1%) and 185 (PD-L1 < 1%) patients received nivolumab-plus-ipilimumab; 387 (PD-L1 ≥ 1%) and 183 (PD-L1 < 1%) patients received chemotherapy, with 29.3-month minimum follow-up. Among chemotherapy-treated patients, 169/387 (43.7%; PD-L1 ≥ 1%) and 66/183 (36.1%; PD-L1 < 1%) switched to immunotherapy poststudy. Among treated patients, median OS was 17.4 months with nivolumab-plus-ipilimumab versus 14.9 months with chemotherapy (hazard ratio [HR], 0.80; 95% confidence interval [CI], 0.68-0.95) in the PD-L1 ≥ 1% subgroup and 17.1 versus 12.4 months (HR, 0.62; 95% CI, 0.49-0.80) in the PD-L1 < 1% subgroup. After treatment-switching adjustment using IPCW, the HR (95% CI) for OS for nivolumab-plus-ipilimumab versus chemotherapy was reduced to 0.68 (0.56-0.83; PD-L1 ≥ 1%) and 0.53 (0.40-0.69; PD-L1 < 1%). Sensitivity analyses supported the robustness of the results.
    CONCLUSIONS: Treatment-switching adjustments resulted in a greater estimated relative OS benefit with first-line nivolumab-plus-ipilimumab versus chemotherapy in patients with metastatic NSCLC.
    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: Case Reports
    抗PD-1免疫疗法通过阻断PD-1与其配体之间的相互作用来增强针对肿瘤细胞的T细胞应答,PD-L1.虽然这些疗法在治疗各种恶性肿瘤方面具有显著的益处,它们还可能导致几种免疫相关的不良事件(irAE),最明显的表现在皮肤上。苔藓样反应,湿疹,白癜风是皮肤irAE的三种最常见形式。
    这里,我们报道了一例罕见的在pembrolizumab治疗浸润性膀胱癌期间发生的地衣皮及变形杆菌糠疹(PLEVA)病例.一个53岁的男人,接受pembrolizumab治疗浸润性膀胱癌,第11次输液后,他的腿上出现了红斑丘疹。皮肤损伤逐渐扩散到他的整个躯干和四肢。穿刺活检显示几个凋亡的角质形成细胞和海绵体,伴随血管周围和苔藓样淋巴细胞浸润,空泡改变。免疫组织化学显示表皮和真皮中均有CD4+和CD8+T细胞浸润。颗粒酶B-阳性炎症细胞也轻微存在。从这些结果来看,他被诊断出患有PLEVA,这可能被归类为苔藓样喷发,尤其是基于组织学发现。
    我们假设抗PD-1抗体可能通过扩增细胞溶解分子如粒酶B在CD8+T细胞中的表达而导致表皮坏死。
    UNASSIGNED: Anti-PD-1 immunotherapies enhance T-cell responses against tumor cells by blocking the interaction between PD-1 and its ligand, PD-L1. While these therapies offer significant benefits in treating various malignancies, they can also lead to several immune-related adverse events (irAEs), most notably manifesting in the skin. Lichenoid reactions, eczema, and vitiligo are the three most prevalent forms of cutaneous irAE.
    UNASSIGNED: Here, we report a rare case of a pityriasis lichenoides et varioliformis acuta (PLEVA) that developed during pembrolizumab treatment for invasive bladder cancer. A 53-year-old man, receiving pembrolizumab for invasive bladder cancer, developed erythematous papules on his legs after his 11th infusion. The skin lesions gradually spread to his entire trunk and extremities. A punch biopsy revealed several apoptotic keratinocytes and spongiosis, along with perivascular and lichenoid lymphocytic infiltration with vacuolar alteration. Immunohistochemistry showed infiltration of CD4+ and CD8+ T cells in both the epidermis and dermis. Granzyme B-positive inflammatory cells were also slightly present. From these results, he was diagnosed with PLEVA, which might be classified as a lichenoid eruption, especially based on the histological findings.
    UNASSIGNED: We hypothesize that the anti-PD-1 antibody might lead to epidermal necrosis by amplifying the expression of cytolytic molecules such as granzyme B in CD8+ T cells.
    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
    目的:本研究旨在探讨MDSCs在CRC免疫抑制中的关键作用。专注于CSF1R和JAK/STAT3信令轴。此外,它评估了LNCs@CSF1RsiRNA和抗PD-1联合治疗的疗效。
    方法:来自CRC和邻近正常组织的单细胞转录组测序数据鉴定了MDSC相关的差异表达基因。RNA-seq分析全面分析小鼠CRC肿瘤中的MDSC基因表达。LNCs@CSF1RsiRNA纳米载体有效靶向并抑制CSF1R。流式细胞术定量CSF1R抑制后MDSC表面标志物的变化。RNA-seq和途径富集分析揭示了CSF1R对MDSC代谢和信号传导的影响。使用Colivelin和代谢评估来验证CSF1R抑制对JAK/STAT3信号传导轴的影响。通过基于荧光的流式细胞术测量葡萄糖和脂肪酸摄取。LNCs@CSF1RsiRNA和抗PD-1的疗效,单独和联合,在具有广泛肿瘤切片分析的小鼠CRC模型中进行评估。
    结果:CSF1R在MDSC介导的免疫抑制中起重要作用。LNCs@CSF1RsiRNA纳米载体有效靶向MDSCs并抑制CSF1R。CSF1R通过JAK/STAT3信号轴调节MDSC脂肪酸代谢和免疫抑制。抑制CSF1R降低了STAT3的激活和靶基因的表达,是Colivelin救的.用LNC@CSF1RsiRNA和抗PD-1联合治疗显著减缓肿瘤生长并降低CRC肿瘤内的MDSC丰度。
    结论:CSF1R通过JAK/STAT3轴关键调节MDSCs,特别是脂肪酸代谢和免疫抑制。LNCs@CSF1RsiRNA和抗PD-1联合治疗可增强小鼠CRC模型的疗效,为未来的临床应用奠定了坚实的基础。
    OBJECTIVE: This study aimed to investigate the critical role of MDSCs in CRC immune suppression, focusing on the CSF1R and JAK/STAT3 signaling axis. Additionally, it assessed the therapeutic efficacy of LNCs@CSF1R siRNA and anti-PD-1 in combination.
    METHODS: Single-cell transcriptome sequencing data from CRC and adjacent normal tissues identified MDSC-related differentially expressed genes. RNA-seq analysis comprehensively profiled MDSC gene expression in murine CRC tumors. LNCs@CSF1R siRNA nanocarriers effectively targeted and inhibited CSF1R. Flow cytometry quantified changes in MDSC surface markers post-CSF1R inhibition. RNA-seq and pathway enrichment analyses revealed the impact of CSF1R on MDSC metabolism and signaling. The effect of CSF1R inhibition on the JAK/STAT3 signaling axis was validated using Colivelin and metabolic assessments. Glucose and fatty acid uptake were measured via fluorescence-based flow cytometry. The efficacy of LNCs@CSF1R siRNA and anti-PD-1, alone and in combination, was evaluated in a murine CRC model with extensive tumor section analyses.
    RESULTS: CSF1R played a significant role in MDSC-mediated immune suppression. LNCs@CSF1R siRNA nanocarriers effectively targeted MDSCs and inhibited CSF1R. CSF1R regulated MDSC fatty acid metabolism and immune suppression through the JAK/STAT3 signaling axis. Inhibition of CSF1R reduced STAT3 activation and target gene expression, which was rescued by Colivelin. Combined treatment with LNCs@CSF1R siRNA and anti-PD-1 significantly slowed tumor growth and reduced MDSC abundance within CRC tumors.
    CONCLUSIONS: CSF1R via the JAK/STAT3 axis critically regulates MDSCs, particularly in fatty acid metabolism and immune suppression. Combined therapy with LNCs@CSF1R siRNA and anti-PD-1 enhances therapeutic efficacy in a murine CRC model, providing a strong foundation for future clinical applications.
    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:在非小细胞肺癌(NSCLC)中的先前研究表明,具有特定驱动突变的肿瘤可能不太可能对免疫检查点抑制剂(ICI)产生反应。在这个分析中,我们评估了ICI具有持久临床获益(DCB)的患者与没有持久临床获益(NDB)的患者的特征,强调分子改变在EGFR中的作用,ALK,和ROS1和治疗前中性粒细胞与淋巴细胞比率(NLR)。
    方法:我们回顾性收集了2015年4月至2018年5月在斯坦福大学开始ICI单药治疗晚期NSCLC患者的临床特征和结局。如果ICI治疗的时间大于或等于180天,则将患者分类为患有DCB。或NDB,如果少于180天。结果包括ICI时的最佳影像学益处和ICI开始时的生存率。
    结果:在123例ICI治疗非小细胞肺癌的患者中,28例患者有DCB(23%),95人拥有NDB(77%)。在33例EGFR分子改变的患者中,ICI开始后的中位总生存期(n=31),ALK,或ROS1和NLR为5.9或更高为2.0个月,这些基因组改变和NLR小于5.9的患者为8.1个月。EGFR无改变患者的中位总生存期,ALK,或ROS1和NLR为5.9或更高为4.3个月,相比,NLR低于5.9的患者为12.1个月(P=0.023)。
    结论:治疗前NLR升高与ICI开始后总中位生存期显著降低相关,尤其是与EGFR改变的NSCLC联合使用时,ALK,或者ROS1。这一发现可能会影响临床实践,因为NLR很容易通过常规血液检测获得。
    BACKGROUND: Prior research in non-small cell lung cancer (NSCLC) has shown that tumors with specific driver mutations may be less likely to respond to immune checkpoint inhibitors (ICI). In this analysis, we evaluated the characteristics of patients with durable clinical benefit (DCB) to ICI compared to those with no durable clinical benefit (NDB), with emphasis on the role of molecular alterations in EGFR, ALK, and ROS1 and pretreatment neutrophil-to-lymphocyte ratio (NLR).
    METHODS: We retrospectively collected clinical characteristics and outcomes for patients who initiated ICI monotherapy for advanced NSCLC at Stanford University between April 2015 and May 2018. Patients were classified as having DCB if time on ICI therapy was greater than or equal to 180 days, or NDB if less than 180 days. Outcomes included best radiographic benefit while on ICI and survival from time of ICI initiation.
    RESULTS: Of 123 patients treated with ICI for NSCLC, 28 patients had DCB (23%), while 95 had NDB (77%). Median overall survival from initiation of ICI in the 33 patients with molecular alterations in EGFR (n = 31), ALK, or ROS1 and NLR of 5.9 or higher was 2.0 months, compared to 8.1 months in patients with these genomic alterations and NLR less than 5.9. Median overall survival in patients without alterations in EGFR, ALK, or ROS1 and NLR of 5.9 or higher was 4.3 months, compared to 12.1 months in patients with NLR less than 5.9 (P = .023).
    CONCLUSIONS: Elevation in pretreatment NLR was associated with significantly lower overall median survival from initiation of ICI, particularly when in combination with NSCLC with alterations in EGFR, ALK, or ROS1. This finding could influence clinical practice as NLR is readily available through routine blood testing.
    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
    探讨重组人血管内皮抑素注射液(名为Endo)与抗PD-1联合治疗80岁及以上老年非小细胞肺癌(NSCLC)的疗效和安全性。
    巢湖医院呼吸与重症医学科收治的181例80岁及以上非小细胞肺癌患者的回顾性分析,安徽医科大学附属,从2019年6月到2024年1月。基于纳入标准,纳入接受至少一个周期的Endo与抗PD-1联合治疗的患者。收集临床和病理资料,包括全血细胞计数,肝肾功能,心电图,凝血功能,甲状腺功能,心肌酶,和全身成像。在2024年1月25日的最后随访中记录了不良事件。主要终点是无进展生存期(PFS)和总生存期(OS),以安全为次要终点。
    本研究涉及14例80岁以上的老年非小细胞肺癌患者。中位无进展生存期(mPFS)为102天,中位总生存期(mOS)为311天。基于治疗周期的亚组分析显示,长期组的441天mPFS无显著增加(≥6个周期,5名患者)与短期组(<6个周期,9名患者)。然而,长期组的mOS显著超过短期组141天,具有统计学意义(P=0.048)。进一步分类显示,与联合维持组(Endo联合免疫,441天)。单药治疗维持组的mOS(686天)比联合治疗维持组(311天)更长,但无统计学意义(P=0.710、0.920)。在整个治疗过程中,记录77起不良事件,主要为1-2级,没有新的治疗相关反应发生。总的来说,Endo联合抗PD-1的安全性被认为是良好且可控的.
    对于80岁及以上的非小细胞肺癌患者,Endo和抗PD-1的联合治疗可能是一种有效的治疗选择。
    UNASSIGNED: To investigate the efficacy and safety of combining Recombinant Human Endostatin Injection (marketed as Endo) with anti-PD-1 in elderly patients aged 80 and above with non-small cell lung cancer (NSCLC).
    UNASSIGNED: Retrospective analysis of 181 patients with NSCLC aged 80 and above treated in the Department of Respiratory and Critical Care Medicine at Chaohu Hospital, affiliated with Anhui Medical University, from June 2019 to January 2024. Patients who received at least one cycle of combined Endo with anti-PD-1 were included based on inclusion criteria. Clinical and pathological data were collected, including complete blood count, liver and kidney function, electrocardiogram, coagulation function, thyroid function, cardiac enzymes, and whole-body imaging. Adverse events were recorded with a final follow-up on January 25, 2024. The primary endpoints were progression-free survival (PFS) and overall survival (OS), with safety as a secondary endpoint.
    UNASSIGNED: This study involved 14 elderly patients with NSCLC aged over 80. Median progression-free survival (mPFS) was 102 days, and median overall survival (mOS) was 311 days. Subgroup analyses based on treatment cycles showed a non-significant 441-day mPFS increase in the long-term group (≥6 cycles, 5 patients) compared to the short-term group (<6 cycles, 9 patients). However, the mOS in the long-term group significantly exceeded the short-term group by 141 days, with statistical significance (P=0.048). Further categorization revealed a 204-day shorter mPFS in the monotherapy maintenance group (Endo or Immunol) compared to the combination maintenance group (Endo combined with Immunol, 441 days). The mOS of the monotherapy maintenance group was longer (686 days) than the combination maintenance group (311 days), but no statistical significance (P= 0.710, 0.920). Throughout the treatment, 77 adverse events were recorded, mainly grade 1-2, with no new treatment-related reactions occurred. Overall, the safety of Endo combined with anti-PD-1 was considered good and manageable.
    UNASSIGNED: The combination of Endo and anti-PD-1 could be an effective treatment choice for patients with NSCLC aged 80 and above.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号