immune checkpoint blockade

免疫检查点阻断
  • 文章类型: Journal Article
    免疫疗法正在彻底改变多种癌症类型的管理。然而,只有一部分患者对免疫治疗有反应.抗性的一种机制是肿瘤内不存在免疫浸润。具有可诱导免疫原性细胞死亡的局部肿瘤破坏手段的原位疫苗已显示增强肿瘤T细胞浸润并增加免疫检查点阻断的功效。
    这里,我们比较了三种不同形式的局部肿瘤破坏疗法:放射治疗(RT),血管靶向光动力疗法(VTP)和冷冻消融(Cryo),已知会诱导免疫原性细胞死亡,在小鼠4T1乳腺癌模型中具有诱导局部和全身免疫反应的能力。结合RT的效果,VTP,还评估了抗PD1的冷冻。
    我们观察到RT,VTP和Cryo可显着延迟肿瘤生长并延长总生存期。此外,在提示全身免疫反应的双侧模型中,他们还诱导了未治疗的远处肿瘤消退.流式细胞术显示,VTP和Cryo与CD11b髓样细胞的减少有关(粒细胞,单核细胞,和巨噬细胞)在肿瘤和外周。仅在RT组中观察到CD8+T细胞浸润到肿瘤中的增加。VTP和Cryo与外周CD4+和CD8+细胞的增加相关。
    这些数据表明由VTP和Cryo诱导的细胞死亡引起与局部RT不同的类似免疫应答。
    UNASSIGNED: Immunotherapy is revolutionizing the management of multiple cancer types. However, only a subset of patients responds to immunotherapy. One mechanism of resistance is the absence of immune infiltrates within the tumor. In situ vaccine with local means of tumor destruction that can induce immunogenic cell death have been shown to enhance tumor T cell infiltration and increase efficacy of immune checkpoint blockade.
    UNASSIGNED: Here, we compare three different forms of localize tumor destruction therapies: radiation therapy (RT), vascular targeted photodynamic therapy (VTP) and cryoablation (Cryo), which are known to induce immunogenic cell death, with their ability to induce local and systemic immune responses in a mouse 4T1 breast cancer model. The effects of combining RT, VTP, Cryo with anti-PD1 was also assessed.
    UNASSIGNED: We observed that RT, VTP and Cryo significantly delayed tumor growth and extended overall survival. In addition, they also induced regression of non-treated distant tumors in a bilateral model suggesting a systemic immune response. Flow cytometry showed that VTP and Cryo are associated with a reduction in CD11b+ myeloid cells (granulocytes, monocytes, and macrophages) in tumor and periphery. An increase in CD8+ T cell infiltration into tumors was observed only in the RT group. VTP and Cryo were associated with an increase in CD4+ and CD8+ cells in the periphery.
    UNASSIGNED: These data suggest that cell death induced by VTP and Cryo elicit similar immune responses that differ from local RT.
    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
    冷冻消融(Cryo)是一种针对肿瘤的微创治疗方法。低温可以激活人体的免疫反应,尽管它通常很弱。对Cryo在肝细胞癌(HCC)中诱导的免疫应答知之甚少。PD-1和CTLA-4单克隆抗体是用于肿瘤免疫治疗的免疫检查点抑制剂。这些抗体与Cryo的组合使用可以增强免疫效果。
    建立了Balb/c小鼠肝癌模型并用Cryo治疗,免疫检查点阻断(ICB),或冷冻+ICB(联合治疗)。确定了未治疗的肿瘤的生长趋势和小鼠的存活时间。Westernblot(WB)法检测细胞凋亡相关蛋白的表达。通过流式细胞术分析免疫细胞和免疫抑制细胞的百分比。通过免疫组织化学检查浸润T淋巴细胞的数量,通过定量实时聚合酶链反应(qRT-PCR)测定和酶联免疫吸附测定(ELISA)测定检测T细胞相关细胞因子的水平。
    冷冻+ICB抑制右侧未治疗肿瘤的生长,促进肿瘤细胞凋亡,延长了小鼠的存活时间。联合治疗后右侧肿瘤组织局部T细胞浸润增加,而免疫抑制细胞的数量显著减少。此外,联合治疗可诱导多种Th1型细胞因子的产生,但减少Th2型细胞因子的产生。
    冷冻可以激活CD8+和CD4+T细胞免疫应答。冷冻+ICB可以缓解免疫抑制肿瘤微环境,使Th1/Th2平衡向Th1优势转移,进一步增强了低温诱导的T细胞免疫应答,导致更强的抗肿瘤免疫应答。
    UNASSIGNED: Cryoablation (Cryo) is a minimally invasive treatment for tumors. Cryo can activate the body\'s immune response, although it is typically weak. The immune response induced by Cryo in hepatocellular carcinoma (HCC) is poorly understood. PD-1 and CTLA-4 monoclonal antibodies are immune checkpoint inhibitors used in immunotherapy for tumors. The combined use of these antibodies with Cryo may enhance the immune effect.
    UNASSIGNED: A Balb/c mouse model of HCC was established and treated with Cryo, immune checkpoint blockade (ICB), or Cryo + ICB (combination therapy). The growth trend of right untreated tumors and survival time of mice were determined. The expression of apoptosis-related proteins was detected by Western blot (WB) assay. The percentages of immune cells and immunosuppressive cells were analyzed by flow cytometry. The numbers of infiltrating T lymphocytes were checked by immunohistochemistry, and the levels of T-cell-associated cytokines were detected by Quantitative real-time Polymerase Chain Reaction (qRT-PCR) assays and Enzyme-Linked Immunosorbent Assays (ELISA) assays.
    UNASSIGNED: Cryo + ICB inhibited the growth of right untreated tumors, promoted tumor cell apoptosis, and prolonged the survival time of mice. Local T-cell infiltration in right tumor tissues increased after the combination therapy, while the number of immunosuppressive cells was significantly reduced. In addition, the combination therapy may induce the production of multiple Th1-type cytokines but reduce the production of Th2-type cytokines.
    UNASSIGNED: Cryo can activate CD8+ and CD4+ T-cell immune responses. Cryo + ICB can relieve the immunosuppressive tumor microenvironment and shift the Th1/Th2 balance toward Th1 dominance, further enhancing the Cryo-induced T-cell immune response and resulting in a stronger antitumor immune response.
    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
    颅内肿瘤由于其生理位置而提出了重大的治疗挑战。由于相对低的毒性和肿瘤特异性,免疫疗法提供了靶向这些颅内肿瘤的有吸引力的方法。在这里,我们显示SCIB1,一种TRP-2和gp100定向免疫抗体®DNA疫苗,产生强烈的TRP-2特异性免疫反应,如通过产生大量的TRP2特异性IFNγ斑点和在接种疫苗的小鼠的脾脏中检测到大量的五聚体阳性T细胞所证明的。此外,疫苗诱导的T细胞能够识别并杀死B16HHDII/DR1细胞。发现多形性胶质母细胞瘤(GBM)表达PD-L1和IDO1的显着水平,PD-L1与GBM间质亚型患者的生存率较差相关,我们决定将SCIB1ImmunoBody®与PD-1免疫检查点阻断联合治疗具有表达TRP-2和gp100的颅内肿瘤的小鼠.死亡时间明显延长,这与组织微环境(TME)中CD4和CD8T细胞浸润增加有关。然而,除了PD-L1和IDO,发现GBMTME含有大量免疫调节T(Treg)细胞相关转录本,这种细胞的存在可能会显著影响临床结果,除非也解决。
    Intracranial tumors present a significant therapeutic challenge due to their physiological location. Immunotherapy presents an attractive method for targeting these intracranial tumors due to relatively low toxicity and tumor specificity. Here we show that SCIB1, a TRP-2 and gp100 directed ImmunoBody® DNA vaccine, generates a strong TRP-2 specific immune response, as demonstrated by the high number of TRP2-specific IFNγ spots produced and the detection of a significant number of pentamer positive T cells in the spleen of vaccinated mice. Furthermore, vaccine-induced T cells were able to recognize and kill B16HHDII/DR1 cells after a short in vitro culture. Having found that glioblastoma multiforme (GBM) expresses significant levels of PD-L1 and IDO1, with PD-L1 correlating with poorer survival in patients with the mesenchymal subtype of GBM, we decided to combine SCIB1 ImmunoBody® with PD-1 immune checkpoint blockade to treat mice harboring intracranial tumors expressing TRP-2 and gp100. Time-to-death was significantly prolonged, and this correlated with increased CD4+ and CD8+ T cell infiltration in the tissue microenvironment (TME). However, in addition to PD-L1 and IDO, the GBM TME was found to contain a significant number of immunoregulatory T (Treg) cell-associated transcripts, and the presence of such cells is likely to significantly affect clinical outcome unless also tackled.
    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
    METIMMOX随机试验(NCT03388190)检查了先前未治疗的患者,来自微卫星稳定(MSS)结直肠癌(CRC)的不可切除的腹部转移可能受益于潜在的免疫原性,以奥沙利铂为基础的短期化疗与免疫检查点阻断(ICB)交替进行。38例接受该实验治疗的患者中有3例发生了BRAF突变型MSS-CRC的转移,一般来说,这里探讨了预后不良的亚组。≥70岁的女性患有升结肠腺癌,具有中等肿瘤突变负担(每兆碱基6.2-11.8个突变)。所有患者都经历了原发肿瘤的早期消失,然后是所有明显的转移性疾病的完全反应,导致无进展生存期长达20-35个月。然而,他们在以前未受影响的地点和最终的避难所器官中复发,或作为肝内肿瘤的演变反映在肝转移中最初诱导的T细胞克隆性的最终丧失。然而,以奥沙利铂为基础的短程化疗与ICB交替治疗的一线反应显著,这可能为尤其难以治疗的MSS-CRC亚组提供了一种新的治疗选择.
    The randomized METIMMOX trial (NCT03388190) examined if patients with previously untreated, unresectable abdominal metastases from microsatellite-stable (MSS) colorectal cancer (CRC) might benefit from potentially immunogenic, short-course oxaliplatin-based chemotherapy alternating with immune checkpoint blockade (ICB). Three of 38 patients assigned to this experimental treatment had metastases from BRAF-mutant MSS-CRC, in general a poor-prognostic subgroup explored here. The ≥70-year-old females presented with ascending colon adenocarcinomas with intermediate tumor mutational burden (6.2-11.8 mutations per megabase). All experienced early disappearance of the primary tumor followed by complete response of all overt metastatic disease, resulting in progression-free survival as long as 20-35 months. However, they encountered recurrence at previously unaffected sites and ultimately sanctuary organs, or as intrahepatic tumor evolution reflected in the terminal loss of initially induced T-cell clonality in liver metastases. Yet, the remarkable first-line responses to short-course oxaliplatin-based chemotherapy alternating with ICB may offer a novel therapeutic option to a particularly hard-to-treat MSS-CRC subgroup.
    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
    EMT转录因子ZEB1在结直肠癌(CRC)的肿瘤细胞和癌症相关成纤维细胞(CAF)中异质表达。虽然肿瘤细胞中的ZEB1调节转移和治疗抵抗,其在CAF中的作用在很大程度上是未知的。将成纤维细胞特异性Zeb1缺失与CRC的免疫活性小鼠模型相结合,我们观察到炎症驱动的肿瘤发生加速,而散发性癌症的侵袭和转移减少。单细胞转录组学,组织学特征,体外建模揭示了ZEB1在CAF极化中的关键作用,通过限制炎症激活来促进肌纤维母细胞的特征。Zeb1缺乏损害胶原沉积和CAF屏障功能,但增加NFκB介导的细胞因子产生,共同促进淋巴细胞募集和免疫检查点激活。引人注目的是,缺乏Zeb1的CAF库对免疫检查点抑制敏感,在CAF中提供靶向ZEB1的治疗机会及其作为预后生物标志物的用途。总的来说,我们证明,CAFs的ZEB1依赖性可塑性抑制抗肿瘤免疫并促进转移.
    The EMT-transcription factor ZEB1 is heterogeneously expressed in tumor cells and in cancer-associated fibroblasts (CAFs) in colorectal cancer (CRC). While ZEB1 in tumor cells regulates metastasis and therapy resistance, its role in CAFs is largely unknown. Combining fibroblast-specific Zeb1 deletion with immunocompetent mouse models of CRC, we observe that inflammation-driven tumorigenesis is accelerated, whereas invasion and metastasis in sporadic cancers are reduced. Single-cell transcriptomics, histological characterization, and in vitro modeling reveal a crucial role of ZEB1 in CAF polarization, promoting myofibroblastic features by restricting inflammatory activation. Zeb1 deficiency impairs collagen deposition and CAF barrier function but increases NFκB-mediated cytokine production, jointly promoting lymphocyte recruitment and immune checkpoint activation. Strikingly, the Zeb1-deficient CAF repertoire sensitizes to immune checkpoint inhibition, offering a therapeutic opportunity of targeting ZEB1 in CAFs and its usage as a prognostic biomarker. Collectively, we demonstrate that ZEB1-dependent plasticity of CAFs suppresses anti-tumor immunity and promotes metastasis.
    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:人乳头瘤病毒(HPV)阳性的头颈部鳞状细胞癌(HNSCC)在临床和免疫学上与HPV阴性的HNSCC不同。在这里,我们调查了肿瘤抗原HPVE6/E7和野生型p53特异性T细胞反应的存在,以及免疫检查点阻断对HPV阳性HNSCC患者的影响。
    方法:用HPVE6/E7或野生型p53衍生肽混合物刺激HPV阳性HNSCC患者的外周血单核细胞(PBMC),并使用干扰素-γ酶联免疫吸附斑点测定法进行评估。进行流式细胞术以分析T细胞亚群和表达免疫检查点分子的T细胞的比例。
    结果:23例患者中有22例(95.7%)检测到HPVE6/E7特异性T细胞,而20例患者中有3例(15.0%)检测到野生型p53特异性T细胞。16例患者中有7例(43.8%)表现出野生型p53特异性T细胞反应,使用完整的蛋白质而不是肽确定。免疫检查点阻断增强了20例患者中9例(45.0%)的野生型p53特异性T细胞应答。PBMC的流式细胞术分析显示,免疫检查点阻断后表现出增强的野生型p53特异性T细胞应答的应答者具有显著较高的Ki-67+CD4+T细胞比例,Ki-67+CD8+T细胞,调节性T细胞,PD-1+CD4+T细胞,和TIM-3+CD4+T细胞比非应答者。
    结论:我们的研究结果表明,HPV阳性HNSCC患者的外周血中存在肿瘤抗原特异性T细胞。阻断检查点通路可以增强某些患者的T细胞反应,可能是通过激活的T细胞,Tregs,和/或耗尽的CD4+T细胞。
    OBJECTIVE: Human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC) is clinically and immunologically distinct from HPV-negative HNSCC. Herein, we investigated the presence of tumor antigens HPV E6/E7 and wild-type p53-specific T-cell responses, and the impact of immune checkpoint blockade in patients with HPV-positive HNSCC.
    METHODS: Peripheral blood mononuclear cells (PBMCs) from patients with HPV-positive HNSCC were stimulated with HPV E6/E7 or wild-type p53-derived peptide mixture and evaluated using the interferon-γ enzyme-linked immunosorbent spot assay. Flow cytometry was performed to analyze the proportion of T-cell subsets and T cells expressing immune checkpoint molecules.
    RESULTS: HPV E6/E7-specific T cells were detected in 22 (95.7%) of 23 patients, whereas wild-type p53-specific T cells were detected in 3 (15.0%) of 20 patients. Seven (43.8%) of 16 patients exhibited wild-type p53-specific T-cell responses, as determined using whole proteins instead of peptides. Immune checkpoint blockade enhanced wild-type p53-specific T-cell responses in 9 (45.0%) of 20 patients. Flow cytometric analysis of PBMCs revealed that responders exhibiting enhanced wild-type p53-specific T-cell responses following immune checkpoint blockade had a significantly higher proportion of Ki-67+CD4+ T cells, Ki-67+CD8+ T cells, regulatory T cells, PD-1+CD4+ T cells, and TIM-3+CD4+ T cells than non-responders.
    CONCLUSIONS: Our findings indicate that tumor antigen-specific T cells are present in the peripheral blood of patients with HPV-positive HNSCC. Blockade of checkpoint pathways can enhance T-cell responses in certain patients, probably via activated T cells, Tregs, and/or exhausted CD4+ T cells.
    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
    结直肠癌(CRC)是全球癌症相关死亡的主要原因。虽然治疗进展提高了生存率,主要通过基于KRAS的靶向治疗,NRAS,和BRAF突变,CRC的个性化治疗策略仍然有限.免疫疗法,主要是免疫检查点封锁,已在各种癌症中显示出疗效,但仅在一小部分具有缺陷错配修复(dMMR)蛋白或高微卫星不稳定性(MSI)的CRC患者中有效。最近的研究挑战了CRC免疫惰性的概念,揭示具有高免疫原性和多样化淋巴细胞浸润的亚群。识别dMMR和MSI以外的精确生物标志物对于扩大免疫治疗益处至关重要。因此,探索已经扩展到各种生物标志物来源,比如肿瘤微环境,基因组标记,和肠道微生物群。最近的研究引入了一种新的分类系统,共有分子亚型,这有助于鉴定具有免疫原性的CRC患者。这些发现强调了超越单一生物标志物和走向全面了解CRC免疫学景观的必要性。促进更有效的发展,个性化治疗。
    Colorectal cancer (CRC) is a major cause of cancer-related deaths globally. While treatment advancements have improved survival rates, primarily through targeted therapies based on KRAS, NRAS, and BRAF mutations, personalized treatment strategies for CRC remain limited. Immunotherapy, mainly immune checkpoint blockade, has shown efficacy in various cancers but is effective in only a small subset of patients with CRC with deficient mismatch repair (dMMR) proteins or high microsatellite instability (MSI). Recent research has challenged the notion that CRC is immunologically inert, revealing subsets with high immunogenicity and diverse lymphocytic infiltration. Identifying precise biomarkers beyond dMMR and MSI is crucial to expanding immunotherapy benefits. Hence, exploration has extended to various biomarker sources, such as the tumor microenvironment, genomic markers, and gut microbiota. Recent studies have introduced a novel classification system, consensus molecular subtypes, that aids in identifying patients with CRC with an immunogenic profile. These findings underscore the necessity of moving beyond single biomarkers and toward a comprehensive understanding of the immunological landscape in CRC, facilitating the development of more effective, personalized therapies.
    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
    提高癌症免疫疗法的疗效取决于确定对肿瘤控制和免疫检查点封锁(ICB)反应至关重要的关键T细胞群。我们最近报道,虽然PD-1和CD28的共表达与外周血功能受损有关,它显著增强了非小细胞肺癌(NSCLC)患者肿瘤部位的T细胞适应性.为了揭示潜在的机制,我们探索了CD26的作用,CD26是通过与腺苷脱氨酶(ADA)相互作用而激活T细胞的关键因素,一种能够中和局部腺苷(ADO)的关键细胞内/胞外酶。我们发现自分泌ADA/CD26轴增强CD8+PD-1+CD28+T细胞功能,特别是在以CD39表达为标志的免疫抑制环境中。然后,我们查询了TCGA和OAK数据集,以深入了解我们研究结果的预后/预测潜力.我们确定了预测LUAD患者总生存期(OS)和晚期LUAD患者对阿特珠单抗反应的特征。这些发现提示了靶向ADA/CD26轴的治疗干预的有希望的途径。
    Improving cancer immunotherapy efficacy hinges on identifying key T-cell populations critical for tumor control and response to Immune Checkpoint Blockade (ICB). We have recently reported that while the co-expression of PD-1 and CD28 is associated with impaired functionality in peripheral blood, it significantly enhances T-cell fitness in the tumor site of non-small cell lung cancer (NSCLC) patients. To uncover the underlying mechanisms, we explored the role of CD26, a key player in T-cell activation through its interaction with adenosine deaminase (ADA), a crucial intra/extracellular enzyme able to neutralize local adenosine (ADO). We found that an autocrine ADA/CD26 axis enhances CD8+PD-1+CD28+ T-cell function, particularly within an immunosuppressive environment marked by CD39 expression. Then, we interrogated the TCGA and OAK datasets to gain insight into the prognostic/predictive potential of our findings. We identified a signature predicting overall survival (OS) in LUAD patients and response to atezolizumab in advanced LUAD cases. These findings suggest promising avenues for therapeutic intervention targeting the ADA/CD26 axis.
    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
    尽管转移性葡萄膜黑色素瘤(UM)的治疗有了最新进展,在许多病例中,可获得的进一步治疗方案仍然有限,预后仍然较差.除了Tebentafusp,免疫检查点阻断(ICB,PD-1(+/-)CTLA-4抗体)通常用于转移性UM,特别是在HLA-A02:01阴性患者中。然而,ICB是以潜在的严重免疫相关不良事件(irAE)为代价的。因此,选择更有可能受益于ICB的患者组是可取的.
    在此分析中,包括194例接受ICB的转移性UM患者。患者从德国皮肤癌地点和ADOReg注册表招募。为了研究irAE发生与治疗反应的关系,无进展生存期(PFS),和总生存期(OS)两个队列进行了比较:无irAE或1/2级irAE的患者(n=137)和3/4级irAE的患者(n=57)。
    在整个人口中,中位OS为16.4个月,中位PFS为2.8个月.患有3/4级irAE的患者比没有或没有1/2级irAE的患者表现出更有利的生存率(p=0.0071)。IrAE发生率为44.7%(87/194),29.4%(57/194)的患者出现严重的irAE。有趣的是,结肠炎和肝炎与较长的OS显著相关(分别为p=0.0031和p=0.011)。
    该数据可能表明irAE与接受ICB治疗的转移性UM患者的有利生存结果之间存在关联,并表明对肿瘤抗原的耐受性降低可能与对自身抗原的耐受性降低有关。
    UNASSIGNED: Despite recent advancements in the treatment of metastatic uveal melanoma (UM), the availability of further treatment options remains limited and the prognosis continues to be poor in many cases. In addition to tebentafusp, immune checkpoint blockade (ICB, PD-1 (+/-) CTLA-4 antibodies) is commonly used for metastatic UM, in particular in HLA-A 02:01-negative patients. However, ICB comes at the cost of potentially severe immune-related adverse events (irAE). Thus, the selection of patient groups that are more likely to benefit from ICB is desirable.
    UNASSIGNED: In this analysis, 194 patients with metastatic UM undergoing ICB were included. Patients were recruited from German skin cancer sites and the ADOReg registry. To investigate the association of irAE occurrence with treatment response, progression-free survival (PFS), and overall survival (OS) two cohorts were compared: patients without irAE or grade 1/2 irAE (n=137) and patients with grade 3/4 irAE (n=57).
    UNASSIGNED: In the entire population, the median OS was 16.4 months, and the median PFS was 2.8 months. Patients with grade 3/4 irAE showed more favorable survival than patients without or grade 1/2 irAE (p=0.0071). IrAE occurred in 44.7% (87/194), and severe irAE in 29.4% (57/194) of patients. Interestingly, irColitis and irHepatitis were significantly associated with longer OS (p=0.0031 and p=0.011, respectively).
    UNASSIGNED: This data may indicate an association between irAE and favorable survival outcomes in patients with metastatic UM undergoing ICB treatment and suggests that a reduced tolerance to tumor antigens could be linked to reduced tolerance to self-antigens.
    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
    光热疗法(PTT)是一种有前途的癌症治疗方法,因为它能够诱导肿瘤特异性T细胞反应并增强治疗效果。然而,不完全的PTT可留下残留的肿瘤,在临床情况下往往导致新的转移和降低患者生存率.这主要是由于ATP的释放,一种损伤相关的分子模式,通过CD39迅速转化为免疫抑制代谢物腺苷,在肿瘤微环境中普遍存在,从而促进肿瘤免疫逃避。这项研究提出了一种通过Fe掺杂的聚二氨基吡啶(Fe-PDAP)之间的静电吸附制备的光热纳米药物,吲哚菁绿(ICG),和CD39抑制剂聚氧钨酸钠(POM-1)。当暴露于近红外激光时,构建的Fe-PDAP@ICG@POM-1(FIP)可以诱导肿瘤PTT和免疫原性细胞死亡。重要的是,它可以通过双向免疫代谢调节抑制ATP-腺苷途径,导致ATP水平增加和腺苷合成减少,最终逆转免疫抑制微环境并增加免疫检查点阻断(aPD-1)治疗的敏感性。在aPD-1的帮助下,由FIP介导的双向免疫代谢调控策略可以有效地抑制/根除原发性和远端肿瘤,并唤起长期坚实的免疫记忆。这项研究提出了一种免疫代谢控制策略,为治疗不完全PTT后的残留肿瘤提供了挽救选择。
    Photothermal therapy (PTT) is a promising cancer treatment method due to its ability to induce tumor-specific T cell responses and enhance therapeutic outcomes. However, incomplete PTT can leave residual tumors that often lead to new metastases and decreased patient survival in clinical scenarios. This is primarily due to the release of ATP, a damage-associated molecular pattern that quickly transforms into the immunosuppressive metabolite adenosine by CD39, prevalent in the tumor microenvironment, thus promoting tumor immune evasion. This study presents a photothermal nanomedicine fabricated by electrostatic adsorption among the Fe-doped polydiaminopyridine (Fe-PDAP), indocyanine green (ICG), and CD39 inhibitor sodium polyoxotungstate (POM-1). The constructed Fe-PDAP@ICG@POM-1 (FIP) can induce tumor PTT and immunogenic cell death when exposed to a near-infrared laser. Significantly, it can inhibit the ATP-adenosine pathway by dual-directional immunometabolic regulation, resulting in increased ATP levels and decreased adenosine synthesis, which ultimately reverses the immunosuppressive microenvironment and increases the susceptibility of immune checkpoint blockade (aPD-1) therapy. With the aid of aPD-1, the dual-directional immunometabolic regulation strategy mediated by FIP can effectively suppress/eradicate primary and distant tumors and evoke long-term solid immunological memory. This study presents an immunometabolic control strategy to offer a salvage option for treating residual tumors following incomplete PTT.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号