checkpoint blockade

检查站封锁
  • 文章类型: Journal Article
    免疫检查点抑制剂(CPI)诱导的糖尿病(CPI-DM)是一种罕见的免疫相关不良事件(irAE)。患者和提供者担心持续的CPIs会使患者面临其他irAE的风险,因此可能会停止治疗。目前,几乎没有数据可以告知这一决定。因此,本研究旨在阐明CPI-DM诊断后停止CPIs是否会影响未来irAE的发展以及进展和死亡等癌症结局.对2015年7月1日至2023年7月5日在UCSF治疗期间发生CPI-DM的患者进行癌症结果和irAE发展分析。费希尔的精确检验,学生t检验,Kaplan-Meier方法,适当时使用Cox回归。在43例CPI-DM患者中,20(47%)在IRAE发生后90天内恢复了CPIs,4例(9%)患者在90天后重新开始,和19(44%)患者从未重新启动。在恢复CPIs的24人中有9人(38%)和停止CPIs的19人中有3人(16%)被诊断出随后的irAE(p=0.17)。两组之间的死亡(p=0.74)或癌症进展(p=0.55)没有显着差异。虽然我们的单机构研究在停止CPIs后没有显示更坏的癌症结果,许多变量会影响结果,我们的研究没有足够的能力来评估。需要一种细致入微的方法来决定在CPI-DM等严重的irAE之后是否继续CPI治疗。
    Immune checkpoint inhibitor (CPI)-induced diabetes mellitus (CPI-DM) is a rare immune-related adverse event (irAE). Patients and providers fear that continuing CPIs puts patients at risk for additional irAEs and thus may discontinue therapy. Currently, there are little data to inform this decision. Therefore, this study aims to elucidate whether discontinuing CPIs after diagnosis of CPI-DM impacts the development of future irAEs and cancer outcomes such as progression and death. Patients who developed CPI-DM during cancer treatment at UCSF from 1 July 2015 to 5 July 2023 were analyzed for cancer outcomes and irAE development. Fisher\'s exact tests, Student t-tests, Kaplan-Meier methods, and Cox regression were used as appropriate. Of the 43 patients with CPI-DM, 20 (47%) resumed CPIs within 90 days of the irAE, 4 (9%) patients restarted after 90 days, and 19 (44%) patients never restarted. Subsequent irAEs were diagnosed in 9 of 24 (38%) who resumed CPIs and 3 of 19 (16%) who discontinued CPIs (p = 0.17). There was no significant difference in death (p = 0.74) or cancer progression (p = 0.55) between these two groups. While our single-institution study did not show worse cancer outcomes after discontinuing CPIs, many variables can impact outcomes, which our study was not adequately powered to evaluate. A nuanced approach is needed to decide whether to continue CPI treatment after a severe irAE like CPI-DM.
    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
    临床前研究表明,二甲双胍可以减少肿瘤内缺氧,改善T细胞功能,并增加对PD-1阻断的敏感性,二甲双胍暴露与各种类型癌症的临床结局改善相关。然而,该药物对糖尿病黑色素瘤患者的影响尚未完全阐明.
    我们回顾了1996-2020年间在UPMC-Hillman癌症中心和纪念SloanKettering癌症中心治疗的4,790名I-IV期皮肤黑色素瘤糖尿病患者。主要终点包括复发率,无进展生存期(PFS),以及有和没有二甲双胍暴露的总生存期(OS)。列表变量包括BRAF突变状态,免疫疗法(IMT)类型,和脑转移的发生率。
    二甲双胍暴露后,I/II期患者的五年复发率显着降低(32.3%vs47.7%,p=0.012)。III期患者的五年复发率也显着降低(58.3%vs77.3%,p=0.013)在二甲双胍队列中。在暴露于二甲双胍的几乎所有阶段,OS都在数值上增加,尽管这没有达到统计学意义。在二甲双胍队列中脑转移的发生率显著较低(8.9%vs14.6%,p=0.039)。
    这是第一项研究,证明糖尿病黑色素瘤患者暴露于二甲双胍后的临床结果显著改善。总的来说,这些结果为正在进行的临床试验提供了进一步的理论基础,这些临床试验研究了二甲双胍对晚期黑色素瘤检查点阻断的潜在增强作用.
    UNASSIGNED: Pre-clinical studies have shown that metformin reduces intratumoral hypoxia, improves T-cell function, and increases sensitivity to PD-1 blockade, and metformin exposure has been associated with improved clinical outcomes in various types of cancer. However, the impact of this drug in diabetic melanoma patients has not yet been fully elucidated.
    UNASSIGNED: We reviewed 4,790 diabetic patients with stage I-IV cutaneous melanoma treated at the UPMC-Hillman Cancer Center and Memorial Sloan Kettering Cancer Center between 1996-2020. The primary endpoints included recurrence rates, progression free survival (PFS), and overall survival (OS) with and without metformin exposure. Tabulated variables included BRAF mutational status, immunotherapy (IMT) by type, and incidence of brain metastases.
    UNASSIGNED: The five-year incidence of recurrence in stage I/II patients was significantly reduced with metformin exposure (32.3% vs 47.7%, p=0.012). The five-year recurrence rate for stage III patients was also significantly reduced (58.3% vs 77.3%, p=0.013) in the metformin cohort. OS was numerically increased in nearly all stages exposed to metformin, though this did not reach statistical significance. The incidence of brain metastases was significantly lower in the metformin cohort (8.9% vs 14.6%, p=0.039).
    UNASSIGNED: This is the first study to demonstrate significantly improved clinical outcomes in diabetic melanoma patients exposed to metformin. Overall, these results provide further rationale for ongoing clinical trials studying the potential augmentation of checkpoint blockade with metformin in advanced melanoma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase II
    绝大多数HER2阳性转移性乳腺癌(MBC)患者最终会对抗HER2治疗产生耐药性并死于这种疾病。尽管,相对高水平的间质瘤浸润淋巴细胞(sTIL),PD1封锁仅显示出适度的反应。Monalizumab靶向抑制性免疫检查点NKG2A,从而释放NK-和CD8T细胞。我们假设monalizumab通过促进抗体依赖性细胞介导的细胞毒性与曲妥珠单抗协同作用。在MIMOSA第二阶段试验中,HER2阳性MBC患者每两周接受曲妥珠单抗和750mgmonalizumab治疗。按照西蒙的两阶段设计,11名患者被纳入试验的I期。治疗耐受性良好,无剂量限制性毒性。没有观察到客观反应。因此,MIMOSA试验未达到主要终点.总之,尽管有很强的临床前理由,monalizumab和曲妥珠单抗的新组合在HER2阳性MBC重度治疗前患者中不能诱导客观缓解.
    The large majority of patients with HER2-positive metastatic breast cancer (MBC) will eventually develop resistance to anti-HER2 therapy and die of this disease. Despite, relatively high levels of stromal tumor infiltrating lymphocytes (sTILs), PD1-blockade has only shown modest responses. Monalizumab targets the inhibitory immune checkpoint NKG2A, thereby unleashing NK- and CD8 T cells. We hypothesized that monalizumab synergizes with trastuzumab by promoting antibody-dependent cell-mediated cytotoxicity. In the phase II MIMOSA-trial, HER2-positive MBC patients were treated with trastuzumab and 750 mg monalizumab every two weeks. Following a Simon\'s two-stage design, 11 patients were included in stage I of the trial. Treatment was well tolerated with no dose-limiting toxicities. No objective responses were observed. Therefore, the MIMOSA-trial did not meet its primary endpoint. In summary, despite the strong preclinical rationale, the novel combination of monalizumab and trastuzumab does not induce objective responses in heavily pre-treated HER2-positive MBC 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
    过去的几十年见证了靶向疗法的临床应用,包括但不限于抑制多种促肿瘤发生信号的酪氨酸激酶抑制剂(TKIs)。TKI可分为(i)直接靶向癌细胞的试剂,(ii)使血管生成正常化或(iii)影响血液谱系的细胞。然而,基于该定义的TKIs的明确区分受到以下事实的限制:许多旨在抑制癌细胞的TKIs对正在发现的免疫细胞也有影响.此外,最初设计为靶向血液癌症的TKI对相同血液谱系的健康细胞表现出生物活性。TKIs已被描述为提高免疫识别和癌症免疫监视,为将TKIs与免疫治疗相结合提供科学依据。的确,TKIs与免疫治疗的组合在临床前模型和临床试验中显示出协同作用,一些使血管生成正常化的TKIs与免疫检查点阻断抗体的组合已被FDA批准用于癌症治疗.然而,为了在癌症治疗中取得切实进展,需要改进适当药物组合的确定以及最佳剂量和计划。本试验观察总结了将TKIs与各种免疫治疗策略结合起来治疗癌症患者的积极临床试验。
    The past decades witnessed the clinical employment of targeted therapies including but not limited to tyrosine kinase inhibitors (TKIs) that restrain a broad variety of pro-tumorigenic signals. TKIs can be categorized into (i) agents that directly target cancer cells, (ii) normalize angiogenesis or (iii) affect cells of the hematologic lineage. However, a clear distinction of TKIs based on this definition is limited by the fact that many TKIs designed to inhibit cancer cells have also effects on immune cells that are being discovered. Additionally, TKIs originally designed to target hematological cancers exhibit bioactivities on healthy cells of the same hematological lineage. TKIs have been described to improve immune recognition and cancer immunosurveillance, providing the scientific basis to combine TKIs with immunotherapy. Indeed, combination of TKIs with immunotherapy showed synergistic effects in preclinical models and clinical trials and some combinations of TKIs normalizing angiogenesis with immune checkpoint blocking antibodies have already been approved by the FDA for cancer therapy. However, the identification of appropriate drug combinations as well as optimal dosing and scheduling needs to be improved in order to obtain tangible progress in cancer care. This Trial Watch summarizes active clinical trials combining TKIs with various immunotherapeutic strategies to treat 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
    背景:EORTC62961-ESHO95随机试验显示,在新辅助化疗的基础上增加局部热疗可改善高危软组织肉瘤患者的长期生存率。我们假设接受新辅助治疗的患者的免疫浸润与临床结果相关。
    方法:在四个周期的治疗后,在患者的序贯活检中评估肿瘤浸润淋巴细胞(TIL)和CD8,FOXP3,PD-1和PD-L1。
    结果:在1997年7月至2006年11月期间随机接受新辅助化疗(53例)或局部热疗新辅助化疗(56例)的109例患者亚组,获得137个活检。配对的第二次活检中的TIL增加,与治疗分配无关(p<0.001)。FOXP3调节性T细胞减少(p=0.002),肿瘤的PD-L1表达变得无法检测。在多变量分析中,治疗后高TIL与LPFS(HR:0.34;95%CI0.15-0.75;p=0.008)和DFS(HR:0.38;95%CI0.17-0.82;p=0.015)相关。在比较治疗组之间的治疗后免疫浸润时,肿瘤反应与局部热疗(p=0.013)和高TIL(p=0.064)的新辅助化疗相关.高CD8细胞浸润与改善的LPFS(HR:0.27;95%CI0.09-0.79;Log-rankp=0.011)和DFS(HR:0.25;95%CI0.09-0.73;Log-rankp=0.006)相关。10年生存率的提高与局部热疗新辅助化疗后的免疫浸润有关。
    结论:术前治疗将基线时未发炎的肿瘤重新编程为发炎的肿瘤。治疗后的免疫浸润成为临床结果的预测指标。与局部热疗相结合可启动肿瘤微环境,在高风险软组织肉瘤中能够增强抗肿瘤免疫活性。
    背景:ClinicalTrials.gov,NCT00003052。
    BACKGROUND: The EORTC 62961-ESHO 95 randomised trial showed improved long-term survival of patients with high-risk soft-tissue sarcoma by adding regional hyperthermia to neoadjuvant chemotherapy. We hypothesised that immune infiltrate of patients treated with neoadjuvant therapy associate with clinical outcome.
    METHODS: Tumour infiltrating lymphocytes (TILs) and CD8, FOXP3, PD-1, and PD-L1 were evaluated in sequential biopsies of patients after four cycles of therapy.
    RESULTS: From a subgroup of 109 patients who had been randomised between July 1997 and November 2006 to neoadjuvant chemotherapy (53 patients) or neoadjuvant chemotherapy with regional hyperthermia (56 patients), 137 biopsies were obtained. TILs increased in paired second biopsies independent of treatment allocation (p < 0.001). FOXP3 regulatory T cells decreased (p = 0.002), and PD-L1 expression of tumours became undetectable. In the multivariate analysis, post-treatment high TILs correlated to LPFS (HR: 0.34; 95% CI 0.15-0.75; p = 0.008) and DFS (HR: 0.38; 95% CI 0.17-0.82; p = 0.015). In comparing post-treatment immune infiltrate between treatment arms, tumour response was associated with neoadjuvant chemotherapy with regional hyperthermia (p = 0.013) and high TILs (p = 0.064). High CD8 cell infiltration was associated with improved LPFS (HR: 0.27; 95% CI 0.09-0.79; Log-rank p = 0.011) and DFS (HR: 0.25; 95% CI 0.09-0.73; Log-rank p = 0.006). Improved survival at 10 years was associated with immune infiltrate after neoadjuvant chemotherapy with regional hyperthermia.
    CONCLUSIONS: Preoperative therapy re-programs a non-inflamed tumour at baseline into an inflamed tumour. The post-treatment immune infiltrate became predictive for clinical outcomes. The combination with regional hyperthermia primes the tumour microenvironment, enabling enhanced anti-tumour immune activity in high-risk soft tissue sarcomas.
    BACKGROUND: ClinicalTrials.gov, NCT00003052.
    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
    Pembrolizumab, a PD1 immune checkpoint inhibitor (ICI), was recently reported to be very effective in patients with microsatellite instable/deficient mismatch repair metastatic colorectal cancer (MSI/dMMR mCRC), unlike patients with microsatellite stable/proficient MMR (MSS/pMMR) mCRC, in whom ICIs are generally ineffective. However, about 15% of MSS/pMMR CRCs are highly infiltrated by tumour infiltrating lymphocytes. In addition, both oxaliplatin and bevacizumab have been shown to have immunomodulatory properties that may increase the efficacy of an ICI. We formulated the hypothesis that patients with MSS/pMMR mCRC with a high immune infiltrate can be sensitive to ICI plus oxalipatin and bevacizumab-based chemotherapy. POCHI is a multicenter, open-label, single-arm phase II trial to evaluate efficacy of Pembrolizumab with Capox Bevacizumab as first-line treatment of MSS/pMMR mCRC with a high immune infiltrate for which we plan to enrol 55 patients. Primary endpoint is progression-free survival (PFS) at 10 months, which is expected greater than 50%, but a 70% rate is hoped for. Main secondary objectives are overall survival, secondary resection rate and depth of response. Patients must have been resected of their primary tumour so as to evaluate two different immune scores (Immunoscore® and TuLIS) and are eligible if one score is \"high\". The first patient was included on April 20, 2021.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Clinical Trial, Phase II
    The purpose of this study was to investigate whether combining pembrolizumab with palliative radiation therapy (RT) improves outcomes in patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC).
    Eligible patients had HR+/human epidermal growth factor receptor 2-negative MBC; were candidates for RT to ≥ 1 bone, soft tissue, or lymph node lesion; and had ≥ 1 lesion outside the RT field. Patients received 200 mg pembrolizumab intravenously 2 to 7 days prior to RT and on day 1 of repeating 21-day cycles. RT was delivered to a previously unirradiated area in 5 treatments each of 4 Gy. The primary endpoint was objective response rate. The study used a 2-stage design: 8 women were enrolled into the first stage, and if at least 1 of 8 patients experienced an objective response, 19 more would be enrolled. Secondary endpoints included progression-free survival, overall survival, and safety. Exploratory endpoints included association of overall response rate with programmed death-ligand 1 status and tumor-infiltrating lymphocytes.
    Eight patients were enrolled in stage 1. The median age was 59 years, and the median prior lines of chemotherapy for metastatic disease was 2. There were no objective responses, and the study was closed to further accrual. The median progression-free survival was 1.4 months (95% confidence interval, 0.4-2.1 months), and the median overall survival was 2.9 months (95% confidence interval, 0.9-3.6 months). All-cause adverse events occurred in 87.5% of patients, including just 1 grade 3 event (elevation of aspartate aminotransferase).
    RT combined with pembrolizumab did not produce an objective response in patients with heavily pre-treated HR+ MBC. Future studies should consider alternative radiation dosing and fractionation in patients with less heavily pre-treated HR+ MBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Clinical Trial, Phase II
    本研究旨在评估pembrolizumab介导的程序性细胞死亡蛋白1抑制加放疗(RT)在未选择程序性死亡配体1表达的转移性三阴性乳腺癌患者中的疗效和安全性。
    目前的研究是单臂,西蒙2级,2期临床试验共纳入17名患者,中位年龄为52岁(范围,37-73岁)。以5个每日分数递送3000厘米灰色(cGy)的RT剂量。Pembrolizumab在第一个RT部分的3天内以200mg的剂量静脉内施用,然后每3周±3天直到疾病进展。中位随访时间为34.5周(范围,2.1-108.3周)。本研究的主要终点是使用实体瘤的反应评估标准(RECIST;1.1版)测量的未照射病变患者在第13周的总反应率(ORR)。次要终点包括安全性和无进展生存期。探索性目标是确定预测ORR和无进展生存期的生物标志物。
    整个队列的ORR为17.6%(17例患者中有3例;95%CI,4.7%-44.2%),有3个完整的响应(CR),1例病情稳定,进展性疾病13例。由于疾病进展,八名患者在第13周之前死亡。在第13周使用RECIST1.1版进行评估的9名女性中,第3周(33%)获得了CR,在被照射的门静脉外,肿瘤体积减少了100%。CR持续18周,20周,108周,分别。最常见的1至2级毒性(根据美国国家癌症研究所常见不良事件术语标准进行评估,4.0版)为皮炎(29%)。四个3级不良事件归因于pembrolizumab:疲劳,淋巴细胞减少,和感染。无4级不良事件或治疗相关死亡报告。
    发现pembrolizumab和RT的组合是安全的,并且在预后不良的患者中表现出令人鼓舞的活性。转移性,未选择程序性死亡-配体1表达的三阴性乳腺癌。需要更大规模的检查点阻断加RT与反应的预测生物标志物的临床试验。
    The current study was conducted to evaluate the efficacy and safety of pembrolizumab-mediated programmed cell death protein 1 inhibition plus radiotherapy (RT) in patients with metastatic triple-negative breast cancer who were unselected for programmed death-ligand 1 expression.
    The current study was a single-arm, Simon 2-stage, phase 2 clinical trial that enrolled a total of 17 patients with a median age of 52 years (range, 37-73 years). An RT dose of 3000 centigrays (cGy) was delivered in 5 daily fractions. Pembrolizumab was administered intravenously at a dose of 200 mg within 3 days of the first RT fraction, and then every 3 weeks ± 3 days until disease progression. The median follow-up was 34.5 weeks (range, 2.1-108.3 weeks). The primary endpoint of the current study was the overall response rate (ORR) at week 13 in patients with unirradiated lesions measured using Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1). Secondary endpoints included safety and progression-free survival. Exploratory objectives were to identify biomarkers predictive of ORR and progression-free survival.
    The ORR for the entire cohort was 17.6% (3 of 17 patients; 95% CI, 4.7%-44.2%), with 3 complete responses (CRs), 1 case of stable disease, and 13 cases of progressive disease. Eight patients died prior to week 13 due to disease progression. Among the 9 women assessed using RECIST version 1.1 at week 13, 3 (33%) achieved a CR, with a 100% reduction in tumor volume outside of the irradiated portal. The CRs were durable for 18 weeks, 20 weeks, and 108 weeks, respectively. The most common grade 1 to 2 toxicity (assessed according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0) was dermatitis (29%). Four grade 3 adverse events were attributed to pembrolizumab: fatigue, lymphopenia, and infection. No were no grade 4 adverse events or treatment-related deaths reported.
    The combination of pembrolizumab and RT was found to be safe and demonstrated encouraging activity in patients with poor-prognosis, metastatic, triple-negative breast cancer who were unselected for programmed death-ligand 1 expression. Larger clinical trials of checkpoint blockade plus RT with predictive biomarkers of response are needed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    癌细胞利用多种机制来逃避和抑制抗癌免疫反应,从而产生“冷”免疫抑制肿瘤微环境。溶瘤病毒疗法正在成为一种有希望的方法来恢复肿瘤免疫抑制并增强其他形式的免疫疗法的功效。越来越多的证据表明,溶瘤病毒(OVs)以多模态方式起作用,诱导免疫原性细胞死亡,从而引发强大的抗癌免疫反应。在这次审查中,我们总结了有关OV介导的肿瘤微环境免疫转换的信息。作为案例研究,我们专注于啮齿动物原病毒H-1PV及其作为溶瘤和免疫调节剂的双重作用。还讨论了提高H-1PV抗癌功效的潜在策略。
    Cancer cells utilize multiple mechanisms to evade and suppress anticancer immune responses creating a \"cold\" immunosuppressive tumor microenvironment. Oncolytic virotherapy is emerging as a promising approach to revert tumor immunosuppression and enhance the efficacy of other forms of immunotherapy. Growing evidence indicates that oncolytic viruses (OVs) act in a multimodal fashion, inducing immunogenic cell death and thereby eliciting robust anticancer immune responses. In this review, we summarize information about OV-mediated immune conversion of the tumor microenvironment. As a case study we focus on the rodent protoparvovirus H-1PV and its dual role as an oncolytic and immune modulatory agent. Potential strategies to improve H-1PV anticancer efficacy are also discussed.
    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
    Immunostimulatory monoclonal antibodies (mAbs) exert antineoplastic effects by eliciting a novel or reinstating a pre-existing antitumor immune response. Most often, immunostimulatory mAbs activate T lymphocytes or natural killer (NK) cells by inhibiting immunosuppressive receptors, such as cytotoxic T lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1 (PDCD1, best known as PD-1), or by engaging co-stimulatory receptors, like CD40, tumor necrosis factor receptor superfamily, member 4 (TNFRSF4, best known as OX40) or TNFRSF18 (best known as GITR). The CTLA4-targeting mAb ipilimumab has been approved by the US Food and Drug Administration for use in patients with unresectable or metastatic melanoma in 2011. The therapeutic profile of ipilimumab other CTLA4-blocking mAbs, such as tremelimumab, is currently being assessed in subjects affected by a large panel of solid neoplasms. In the last few years, promising clinical results have also been obtained with nivolumab, a PD-1-targeting mAb formerly known as BMS-936558. Accordingly, the safety and efficacy of nivolumab and other PD-1-blocking molecules are being actively investigated. Finally, various clinical trials are underway to test the therapeutic potential of OX40- and GITR-activating mAbs. Here, we summarize recent findings on the therapeutic profile of immunostimulatory mAbs and discuss clinical trials that have been launched in the last 14 months to assess the therapeutic profile of these immunotherapeutic agents.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号