协同抑制PD-1/LAG-3:增强肿瘤免疫治疗的机制和意义

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
MedComm Pub Date : 2025-05-20 DOI:10.1002/mco2.70216
Jianqiao Shentu, Hening Xu, Shiwei Duan
{"title":"协同抑制PD-1/LAG-3:增强肿瘤免疫治疗的机制和意义","authors":"Jianqiao Shentu,&nbsp;Hening Xu,&nbsp;Shiwei Duan","doi":"10.1002/mco2.70216","DOIUrl":null,"url":null,"abstract":"<p>In the August issue of <i>Cell</i> 2024, three pivotal studies published in <i>Cell</i> by the Vignali and Wherry research teams advanced our understanding of programmed cell death protein 1 (PD-1)/lymphocyte activation gene 3 (LAG-3) synergy in T cell exhaustion [<span>1-3</span>]. This provides new therapeutic strategies for clinical guidance in anti-infection and antitumor treatments.</p><p>PD-1 and LAG-3 are both key inhibitory receptors (IRs). In the context of tumors and chronic viral infections, their expression on CD8<sup>+</sup> T cells increase, hampering the immune system's ability to eliminate these threats. Research indicates that PD-1 and LAG-3 jointly contribute to T cell exhaustion, diminishing effector functions and weakening antitumor immunity. Clinically, the combination of PD-1 and LAG-3 inhibitors has shown remarkable success—particularly in the RELATIVITY-047 melanoma trial—improving progression-free survival (PFS) and demonstrating superior efficacy in neoadjuvant settings compared to PD-1 monotherapy, while maintaining a favorable toxicity profile. However, mechanistic uncertainties persist [<span>4</span>].</p><p>In the first study, the Vignali team utilized mouse models and clinical data to explore the synergistic effects of PD-1 and LAG-3 on T cell function and antitumor immunity [<span>2</span>]. In a melanoma mouse model, CD8<sup>+</sup> T cells deficient in both PD-1 and LAG-3 exhibited enhanced tumor clearance and improved survival rates. Transcriptomic analysis revealed that the absence of PD-1 and LAG-3 led to increased T-cell receptor (TCR) clonality and upregulation of effector-like and interferon-responsive genes. These transcriptional changes boosted IFN-γ secretion and enhanced T cell antitumor efficacy. The study discovered that the increase in IFN-γ does not directly drive antitumor immunity by regulating tumor cells or other cells within the tumor microenvironment. Instead, it acts on CD8<sup>+</sup> T cells themselves in an intracellular and autocrine manner to enhance their function. Particularly, the upregulation of CCL5 may play a crucial role in this process, although CCL5 might have diverse functions in different environments. Further studies have demonstrated that PD-1 and LAG-3 jointly promote the exhaustion of CD8<sup>+</sup> T cells and restrict their proliferation and effector functions. This process is closely associated with the regulation of the transcription factor TOX. Specifically, the loss of PD-1 and LAG-3 significantly reduces the expression level of TOX, indicating that these two receptors have a synergistic effect in regulating the development of T cell exhaustion. Especially in regulating TOX expression, LAG-3 exhibits a more significant dominant role. Additionally, the study also found that in the context of PD-1 and LAG-3 loss, the expression pattern of natural killer group 2 member A (NKG2A) shows interesting inconsistencies in monoclonal and polyclonal systems. This difference suggests that the expression of NKG2A may affect TCR clonal expansion and antitumor immune response. NKG2A may play a key regulatory role in the synergistic effect of PD-1 and LAG-3, and blocking NKG2A could be a beneficial supplement to the combined treatment of PD-1 and LAG-3.</p><p>The second study (initiated by Vignali et al.), focused on the clinical combination of Relatlimab and Nivolumab in advanced melanoma, demonstrated that dual blockade enhanced receptor signaling in CD8<sup>+</sup> T cells, altering their differentiation to improve cytotoxicity while retaining certain exhaustion characteristics [<span>3</span>]. The study revealed that the combination of Relatlimab and Nivolumab led to a unique state of CD8<sup>+</sup> T cells, where enhanced cytotoxicity and exhaustion gene modules coexisted. Despite the presence of exhausted transcriptional signatures, the treatment significantly enhanced the cytotoxicity of CD8<sup>+</sup> T cells, which might suggest that the inhibition of PD-1 and LAG-3 enhances short-term immune responses at the expense of long-term persistence. Further studies indicated that the coexpression of cytotoxic and exhaustion markers of CD8<sup>+</sup> T cells was driven by PRDM1, BATF, ETV7, and TOX. The effector function of clonally expanded CD8<sup>+</sup> T cells after treatment was significantly upregulated, and the characteristics of intratumoral CD8<sup>+</sup> T cells were associated with a favorable prognosis. Notably, in peripheral blood, this marker of combined treatment with Relatlimab and Nivolumab was confirmed as an increased frequency of CD38<sup>+</sup> TIM3<sup>+</sup> CD8<sup>+</sup> T cells. This finding may provide a precise method for identifying pharmacodynamic responses in patients in the clinic and assist in screening out patients who may be resistant to combined treatment with Relatlimab and Nivolumab.</p><p>In the third study, Wherry's team investigated the regulation of Tex cells by PD-1 and LAG-3 in chronic viral infections and cancer [<span>1</span>]. They found that LAG-3 sustains TOX expression and modulates the CD94/NKG2-Qa-1b axis, influencing NK receptor expression and Tex cytotoxicity. LAG-3 drives the formation of a CD94/NKG2<sup>+</sup> Tex subset by recognizing the stress ligand Qa-1b, with similar observations in human studies. This research highlights the distinct roles of PD-1 and LAG-3 in regulating T cell exhaustion, pointing to LAG-3's role in maintaining Tex persistence and enhancing cytotoxicity.</p><p>Together, these studies uncover the mechanisms through which PD-1 and LAG-3 modulate CD8<sup>+</sup> T cell function (Figure 1). Vignali's work emphasizes the balance between effector function and exhaustion, suggesting that LAG-3 inhibition may enhance the immune response at the expense of long-term T cell persistence. Wherry's findings complement this by highlighting LAG-3's role in sustaining T cell cytotoxicity and persistence, providing a comprehensive view of the dual checkpoint blockade.</p><p>While these findings represent significant progress, there are limitations. In the clinical trial led by Vignali et al. [<span>3</span>], Relatlimab monotherapy lasted for only 4 weeks (all patient groups received dual antibody treatment after 4 weeks). As a result, it remains unclear whether the results observed after anti-LAG-3 treatment will persist. Second, the study's reliance on single-cell sequencing presents a problem [<span>3</span>]. In some cases, single-cell sequencing may not be able to capture cell-to-cell interactions and dynamic changes. Moreover, it mainly focuses on gene expression levels while ignoring many other important biological characteristics such as cell metabolic state, protein expression, and signaling pathway activity. Therefore, in the future, it might be necessary to combine multiple technical means, such as multiomics data integration, flow cytometry, and imaging analysis, to further verify its key findings. Additionally, in two other mouse studies, an important finding was made: the combined loss of PD-1 and LAG-3 led to the emergence of NK-like effector CD8<sup>+</sup> T cells, which are characterized by the simultaneous expression of activating and inhibitory NK receptors on the surface [<span>1, 2</span>]. It should be noted that given the differences between the mouse model and the human immune system in some key aspects, the verification of this finding depends on further human research in the future to provide a definitive answer.</p><p>Moreover, current clinical trials must expand to include broader patient populations and diverse tumor types to ensure the safety and efficacy of PD-1 and LAG-3 inhibitors across different contexts. Personalized treatment approaches, guided by biomarkers that predict patient responses, are essential for refining therapeutic regimens. A recent study demonstrated that LAG-3 and PD-1 inhibitors enhance outcomes in advanced nasopharyngeal carcinoma (NPC). Immunotherapy-naïve patients showed 33.3% objective response rate (ORR), 75% disease control rate (DCR), and 10.8-month median PFS, outperforming PD-1 monotherapy. Responses persisted in resistant cases (11.8% ORR, 64.7% DCR). This dual blockade holds clinical promise, particularly as frontline therapy for advanced NPC [<span>5</span>].</p><p>In conclusion, these studies offer new insights into the regulation of immune checkpoints, providing a foundation for future research and clinical applications. They underscore the need for further experiments and trials to better understand the mechanisms of PD-1 and LAG-3, optimize immunotherapy strategies, and ultimately improve patient outcomes.</p><p>Jianqiao Shentu analyzed the literature, wrote the manuscript, Jianqiao Shentu and Hening Xu drafted Figure 1. Jianqiao Shentu and Shiwei Duan conceived the idea. Shiwei Duan reviewed and revised the manuscript. All the authors gave the final approval of the submitted version.</p><p>Not applicable.</p><p>The authors declare no conflicts of interest.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 6","pages":""},"PeriodicalIF":10.7000,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70216","citationCount":"0","resultStr":"{\"title\":\"Synergistic PD-1/LAG-3 Inhibition: Mechanistic Insights and Implications for Enhanced Tumor Immunotherapy\",\"authors\":\"Jianqiao Shentu,&nbsp;Hening Xu,&nbsp;Shiwei Duan\",\"doi\":\"10.1002/mco2.70216\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>In the August issue of <i>Cell</i> 2024, three pivotal studies published in <i>Cell</i> by the Vignali and Wherry research teams advanced our understanding of programmed cell death protein 1 (PD-1)/lymphocyte activation gene 3 (LAG-3) synergy in T cell exhaustion [<span>1-3</span>]. This provides new therapeutic strategies for clinical guidance in anti-infection and antitumor treatments.</p><p>PD-1 and LAG-3 are both key inhibitory receptors (IRs). In the context of tumors and chronic viral infections, their expression on CD8<sup>+</sup> T cells increase, hampering the immune system's ability to eliminate these threats. Research indicates that PD-1 and LAG-3 jointly contribute to T cell exhaustion, diminishing effector functions and weakening antitumor immunity. Clinically, the combination of PD-1 and LAG-3 inhibitors has shown remarkable success—particularly in the RELATIVITY-047 melanoma trial—improving progression-free survival (PFS) and demonstrating superior efficacy in neoadjuvant settings compared to PD-1 monotherapy, while maintaining a favorable toxicity profile. However, mechanistic uncertainties persist [<span>4</span>].</p><p>In the first study, the Vignali team utilized mouse models and clinical data to explore the synergistic effects of PD-1 and LAG-3 on T cell function and antitumor immunity [<span>2</span>]. In a melanoma mouse model, CD8<sup>+</sup> T cells deficient in both PD-1 and LAG-3 exhibited enhanced tumor clearance and improved survival rates. Transcriptomic analysis revealed that the absence of PD-1 and LAG-3 led to increased T-cell receptor (TCR) clonality and upregulation of effector-like and interferon-responsive genes. These transcriptional changes boosted IFN-γ secretion and enhanced T cell antitumor efficacy. The study discovered that the increase in IFN-γ does not directly drive antitumor immunity by regulating tumor cells or other cells within the tumor microenvironment. Instead, it acts on CD8<sup>+</sup> T cells themselves in an intracellular and autocrine manner to enhance their function. Particularly, the upregulation of CCL5 may play a crucial role in this process, although CCL5 might have diverse functions in different environments. Further studies have demonstrated that PD-1 and LAG-3 jointly promote the exhaustion of CD8<sup>+</sup> T cells and restrict their proliferation and effector functions. This process is closely associated with the regulation of the transcription factor TOX. Specifically, the loss of PD-1 and LAG-3 significantly reduces the expression level of TOX, indicating that these two receptors have a synergistic effect in regulating the development of T cell exhaustion. Especially in regulating TOX expression, LAG-3 exhibits a more significant dominant role. Additionally, the study also found that in the context of PD-1 and LAG-3 loss, the expression pattern of natural killer group 2 member A (NKG2A) shows interesting inconsistencies in monoclonal and polyclonal systems. This difference suggests that the expression of NKG2A may affect TCR clonal expansion and antitumor immune response. NKG2A may play a key regulatory role in the synergistic effect of PD-1 and LAG-3, and blocking NKG2A could be a beneficial supplement to the combined treatment of PD-1 and LAG-3.</p><p>The second study (initiated by Vignali et al.), focused on the clinical combination of Relatlimab and Nivolumab in advanced melanoma, demonstrated that dual blockade enhanced receptor signaling in CD8<sup>+</sup> T cells, altering their differentiation to improve cytotoxicity while retaining certain exhaustion characteristics [<span>3</span>]. The study revealed that the combination of Relatlimab and Nivolumab led to a unique state of CD8<sup>+</sup> T cells, where enhanced cytotoxicity and exhaustion gene modules coexisted. Despite the presence of exhausted transcriptional signatures, the treatment significantly enhanced the cytotoxicity of CD8<sup>+</sup> T cells, which might suggest that the inhibition of PD-1 and LAG-3 enhances short-term immune responses at the expense of long-term persistence. Further studies indicated that the coexpression of cytotoxic and exhaustion markers of CD8<sup>+</sup> T cells was driven by PRDM1, BATF, ETV7, and TOX. The effector function of clonally expanded CD8<sup>+</sup> T cells after treatment was significantly upregulated, and the characteristics of intratumoral CD8<sup>+</sup> T cells were associated with a favorable prognosis. Notably, in peripheral blood, this marker of combined treatment with Relatlimab and Nivolumab was confirmed as an increased frequency of CD38<sup>+</sup> TIM3<sup>+</sup> CD8<sup>+</sup> T cells. This finding may provide a precise method for identifying pharmacodynamic responses in patients in the clinic and assist in screening out patients who may be resistant to combined treatment with Relatlimab and Nivolumab.</p><p>In the third study, Wherry's team investigated the regulation of Tex cells by PD-1 and LAG-3 in chronic viral infections and cancer [<span>1</span>]. They found that LAG-3 sustains TOX expression and modulates the CD94/NKG2-Qa-1b axis, influencing NK receptor expression and Tex cytotoxicity. LAG-3 drives the formation of a CD94/NKG2<sup>+</sup> Tex subset by recognizing the stress ligand Qa-1b, with similar observations in human studies. This research highlights the distinct roles of PD-1 and LAG-3 in regulating T cell exhaustion, pointing to LAG-3's role in maintaining Tex persistence and enhancing cytotoxicity.</p><p>Together, these studies uncover the mechanisms through which PD-1 and LAG-3 modulate CD8<sup>+</sup> T cell function (Figure 1). Vignali's work emphasizes the balance between effector function and exhaustion, suggesting that LAG-3 inhibition may enhance the immune response at the expense of long-term T cell persistence. Wherry's findings complement this by highlighting LAG-3's role in sustaining T cell cytotoxicity and persistence, providing a comprehensive view of the dual checkpoint blockade.</p><p>While these findings represent significant progress, there are limitations. In the clinical trial led by Vignali et al. [<span>3</span>], Relatlimab monotherapy lasted for only 4 weeks (all patient groups received dual antibody treatment after 4 weeks). As a result, it remains unclear whether the results observed after anti-LAG-3 treatment will persist. Second, the study's reliance on single-cell sequencing presents a problem [<span>3</span>]. In some cases, single-cell sequencing may not be able to capture cell-to-cell interactions and dynamic changes. Moreover, it mainly focuses on gene expression levels while ignoring many other important biological characteristics such as cell metabolic state, protein expression, and signaling pathway activity. Therefore, in the future, it might be necessary to combine multiple technical means, such as multiomics data integration, flow cytometry, and imaging analysis, to further verify its key findings. Additionally, in two other mouse studies, an important finding was made: the combined loss of PD-1 and LAG-3 led to the emergence of NK-like effector CD8<sup>+</sup> T cells, which are characterized by the simultaneous expression of activating and inhibitory NK receptors on the surface [<span>1, 2</span>]. It should be noted that given the differences between the mouse model and the human immune system in some key aspects, the verification of this finding depends on further human research in the future to provide a definitive answer.</p><p>Moreover, current clinical trials must expand to include broader patient populations and diverse tumor types to ensure the safety and efficacy of PD-1 and LAG-3 inhibitors across different contexts. Personalized treatment approaches, guided by biomarkers that predict patient responses, are essential for refining therapeutic regimens. A recent study demonstrated that LAG-3 and PD-1 inhibitors enhance outcomes in advanced nasopharyngeal carcinoma (NPC). Immunotherapy-naïve patients showed 33.3% objective response rate (ORR), 75% disease control rate (DCR), and 10.8-month median PFS, outperforming PD-1 monotherapy. Responses persisted in resistant cases (11.8% ORR, 64.7% DCR). This dual blockade holds clinical promise, particularly as frontline therapy for advanced NPC [<span>5</span>].</p><p>In conclusion, these studies offer new insights into the regulation of immune checkpoints, providing a foundation for future research and clinical applications. They underscore the need for further experiments and trials to better understand the mechanisms of PD-1 and LAG-3, optimize immunotherapy strategies, and ultimately improve patient outcomes.</p><p>Jianqiao Shentu analyzed the literature, wrote the manuscript, Jianqiao Shentu and Hening Xu drafted Figure 1. Jianqiao Shentu and Shiwei Duan conceived the idea. Shiwei Duan reviewed and revised the manuscript. All the authors gave the final approval of the submitted version.</p><p>Not applicable.</p><p>The authors declare no conflicts of interest.</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":\"6 6\",\"pages\":\"\"},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2025-05-20\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70216\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.70216\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, RESEARCH & EXPERIMENTAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.70216","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

摘要

在《细胞2024》8月刊上,Vignali和Wherry研究小组发表在《细胞》上的三项关键研究提高了我们对程序性细胞死亡蛋白1 (PD-1)/淋巴细胞活化基因3 (LAG-3)在T细胞衰竭中的协同作用的理解[1-3]。这为临床指导抗感染和抗肿瘤治疗提供了新的治疗策略。PD-1和LAG-3都是关键抑制受体(IRs)。在肿瘤和慢性病毒感染的情况下,它们在CD8+ T细胞上的表达增加,阻碍了免疫系统消除这些威胁的能力。研究表明,PD-1和LAG-3共同导致T细胞衰竭、效应功能减弱和抗肿瘤免疫减弱。临床上,PD-1和LAG-3抑制剂的联合治疗取得了显著的成功,特别是在RELATIVITY-047黑色素瘤试验中,与PD-1单药治疗相比,PD-1和LAG-3抑制剂的联合治疗改善了无进展生存期(PFS),并在新辅助治疗中显示出优越的疗效,同时保持了良好的毒性。然而,机制上的不确定性仍然存在。在第一项研究中,Vignali团队利用小鼠模型和临床数据探讨了PD-1和LAG-3对T细胞功能和抗肿瘤免疫[2]的协同作用。在黑色素瘤小鼠模型中,缺乏PD-1和LAG-3的CD8+ T细胞表现出增强的肿瘤清除率和提高的生存率。转录组学分析显示,PD-1和LAG-3的缺失导致t细胞受体(TCR)克隆增加,效应样基因和干扰素应答基因上调。这些转录变化促进了IFN-γ分泌,增强了T细胞抗肿瘤效果。研究发现,IFN-γ的增加并不通过调节肿瘤细胞或肿瘤微环境内的其他细胞直接驱动抗肿瘤免疫。相反,它以细胞内和自分泌的方式作用于CD8+ T细胞本身,以增强其功能。特别是CCL5的上调可能在这一过程中起着至关重要的作用,尽管CCL5在不同的环境中可能具有不同的功能。进一步的研究表明,PD-1和LAG-3共同促进CD8+ T细胞的衰竭,限制其增殖和效应功能。这一过程与转录因子TOX的调控密切相关。具体来说,PD-1和LAG-3的缺失显著降低了TOX的表达水平,表明这两种受体在调节T细胞衰竭的发生中具有协同作用。特别是在调节TOX表达中,LAG-3表现出更显著的主导作用。此外,本研究还发现,在PD-1和LAG-3缺失的情况下,自然杀伤组2成员A (NKG2A)的表达模式在单克隆和多克隆系统中表现出有趣的不一致性。这一差异提示NKG2A的表达可能影响TCR克隆扩增和抗肿瘤免疫应答。NKG2A可能在PD-1和LAG-3的协同作用中发挥关键调节作用,阻断NKG2A可能是PD-1和LAG-3联合治疗的有益补充。第二项研究(由Vignali等人发起)关注的是Relatlimab和Nivolumab在晚期黑色素瘤中的临床联合应用,该研究表明双重阻断增强了CD8+ T细胞中的受体信号,改变了它们的分化以改善细胞毒性,同时保留了一定的衰竭特征[3]。该研究表明,Relatlimab和Nivolumab联合使用可导致CD8+ T细胞的独特状态,其中增强的细胞毒性和耗尽基因模块共存。尽管存在耗尽的转录特征,但治疗显著增强了CD8+ T细胞的细胞毒性,这可能表明抑制PD-1和LAG-3以牺牲长期持续性为代价增强了短期免疫反应。进一步的研究表明,CD8+ T细胞的细胞毒性和衰竭标志物的共表达是由PRDM1、BATF、ETV7和TOX驱动的。治疗后克隆扩增的CD8+ T细胞的效应功能显著上调,肿瘤内CD8+ T细胞的特性与良好的预后相关。值得注意的是,在外周血中,relatlimumab和Nivolumab联合治疗的这一标志物被证实为CD38+ TIM3+ CD8+ T细胞的频率增加。这一发现可能提供一种精确的方法来识别临床患者的药效学反应,并有助于筛选出可能对Relatlimab和Nivolumab联合治疗耐药的患者。在第三项研究中,Wherry的团队研究了PD-1和LAG-3在慢性病毒感染和癌症中对Tex细胞的调节。他们发现LAG-3维持TOX表达并调节CD94/NKG2-Qa-1b轴,影响NK受体表达和Tex细胞毒性。 LAG-3通过识别应激配体Qa-1b来驱动CD94/NKG2+ Tex亚群的形成,在人类研究中也有类似的观察结果。本研究强调了PD-1和LAG-3在调节T细胞衰竭中的不同作用,指出LAG-3在维持Tex持久性和增强细胞毒性方面的作用。总之,这些研究揭示了PD-1和LAG-3调节CD8+ T细胞功能的机制(图1)。Vignali的工作强调了效应功能和衰竭之间的平衡,表明LAG-3抑制可能以牺牲T细胞的长期持久性为代价来增强免疫反应。Wherry的研究结果通过强调LAG-3在维持T细胞毒性和持久性中的作用来补充这一点,提供了双检查点封锁的全面观点。虽然这些发现代表了重大进展,但也存在局限性。在Vignali等人的临床试验中,Relatlimab单药治疗仅持续4周(4周后所有患者组均接受双抗体治疗)。因此,目前尚不清楚抗lag -3治疗后观察到的结果是否会持续。其次,这项研究对单细胞测序的依赖带来了一个问题。在某些情况下,单细胞测序可能无法捕获细胞间的相互作用和动态变化。此外,它主要关注基因表达水平,而忽略了许多其他重要的生物学特性,如细胞代谢状态、蛋白质表达、信号通路活性等。因此,未来可能需要结合多组学数据整合、流式细胞术、影像分析等多种技术手段,进一步验证其关键发现。此外,在另外两项小鼠研究中,有一个重要的发现:PD-1和LAG-3的联合缺失导致了NK样效应CD8+ T细胞的出现,其特征是在表面同时表达激活和抑制NK受体[1,2]。值得注意的是,鉴于小鼠模型与人类免疫系统在某些关键方面存在差异,这一发现的验证取决于未来进一步的人体研究,以提供明确的答案。此外,目前的临床试验必须扩大到包括更广泛的患者群体和不同的肿瘤类型,以确保PD-1和LAG-3抑制剂在不同情况下的安全性和有效性。以预测患者反应的生物标志物为指导的个性化治疗方法,对于完善治疗方案至关重要。最近的一项研究表明,LAG-3和PD-1抑制剂可改善晚期鼻咽癌(NPC)的预后。Immunotherapy-naïve患者的客观缓解率(ORR)为33.3%,疾病控制率(DCR)为75%,中位PFS为10.8个月,优于PD-1单药治疗。耐药病例的反应持续(ORR为11.8%,DCR为64.7%)。这种双重阻断具有临床前景,特别是作为晚期鼻咽癌bbb的一线治疗。总之,这些研究为免疫检查点的调控提供了新的见解,为未来的研究和临床应用提供了基础。他们强调需要进一步的实验和试验来更好地了解PD-1和LAG-3的机制,优化免疫治疗策略,并最终改善患者的预后。申图简桥分析文献,撰写稿件,申图简桥与徐和宁起草图1。申图简桥和段世伟构思了这个想法。段世伟审阅并修改了稿件。所有作者都对提交的版本给予了最终的认可。不适用。作者声明无利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Synergistic PD-1/LAG-3 Inhibition: Mechanistic Insights and Implications for Enhanced Tumor Immunotherapy

In the August issue of Cell 2024, three pivotal studies published in Cell by the Vignali and Wherry research teams advanced our understanding of programmed cell death protein 1 (PD-1)/lymphocyte activation gene 3 (LAG-3) synergy in T cell exhaustion [1-3]. This provides new therapeutic strategies for clinical guidance in anti-infection and antitumor treatments.

PD-1 and LAG-3 are both key inhibitory receptors (IRs). In the context of tumors and chronic viral infections, their expression on CD8+ T cells increase, hampering the immune system's ability to eliminate these threats. Research indicates that PD-1 and LAG-3 jointly contribute to T cell exhaustion, diminishing effector functions and weakening antitumor immunity. Clinically, the combination of PD-1 and LAG-3 inhibitors has shown remarkable success—particularly in the RELATIVITY-047 melanoma trial—improving progression-free survival (PFS) and demonstrating superior efficacy in neoadjuvant settings compared to PD-1 monotherapy, while maintaining a favorable toxicity profile. However, mechanistic uncertainties persist [4].

In the first study, the Vignali team utilized mouse models and clinical data to explore the synergistic effects of PD-1 and LAG-3 on T cell function and antitumor immunity [2]. In a melanoma mouse model, CD8+ T cells deficient in both PD-1 and LAG-3 exhibited enhanced tumor clearance and improved survival rates. Transcriptomic analysis revealed that the absence of PD-1 and LAG-3 led to increased T-cell receptor (TCR) clonality and upregulation of effector-like and interferon-responsive genes. These transcriptional changes boosted IFN-γ secretion and enhanced T cell antitumor efficacy. The study discovered that the increase in IFN-γ does not directly drive antitumor immunity by regulating tumor cells or other cells within the tumor microenvironment. Instead, it acts on CD8+ T cells themselves in an intracellular and autocrine manner to enhance their function. Particularly, the upregulation of CCL5 may play a crucial role in this process, although CCL5 might have diverse functions in different environments. Further studies have demonstrated that PD-1 and LAG-3 jointly promote the exhaustion of CD8+ T cells and restrict their proliferation and effector functions. This process is closely associated with the regulation of the transcription factor TOX. Specifically, the loss of PD-1 and LAG-3 significantly reduces the expression level of TOX, indicating that these two receptors have a synergistic effect in regulating the development of T cell exhaustion. Especially in regulating TOX expression, LAG-3 exhibits a more significant dominant role. Additionally, the study also found that in the context of PD-1 and LAG-3 loss, the expression pattern of natural killer group 2 member A (NKG2A) shows interesting inconsistencies in monoclonal and polyclonal systems. This difference suggests that the expression of NKG2A may affect TCR clonal expansion and antitumor immune response. NKG2A may play a key regulatory role in the synergistic effect of PD-1 and LAG-3, and blocking NKG2A could be a beneficial supplement to the combined treatment of PD-1 and LAG-3.

The second study (initiated by Vignali et al.), focused on the clinical combination of Relatlimab and Nivolumab in advanced melanoma, demonstrated that dual blockade enhanced receptor signaling in CD8+ T cells, altering their differentiation to improve cytotoxicity while retaining certain exhaustion characteristics [3]. The study revealed that the combination of Relatlimab and Nivolumab led to a unique state of CD8+ T cells, where enhanced cytotoxicity and exhaustion gene modules coexisted. Despite the presence of exhausted transcriptional signatures, the treatment significantly enhanced the cytotoxicity of CD8+ T cells, which might suggest that the inhibition of PD-1 and LAG-3 enhances short-term immune responses at the expense of long-term persistence. Further studies indicated that the coexpression of cytotoxic and exhaustion markers of CD8+ T cells was driven by PRDM1, BATF, ETV7, and TOX. The effector function of clonally expanded CD8+ T cells after treatment was significantly upregulated, and the characteristics of intratumoral CD8+ T cells were associated with a favorable prognosis. Notably, in peripheral blood, this marker of combined treatment with Relatlimab and Nivolumab was confirmed as an increased frequency of CD38+ TIM3+ CD8+ T cells. This finding may provide a precise method for identifying pharmacodynamic responses in patients in the clinic and assist in screening out patients who may be resistant to combined treatment with Relatlimab and Nivolumab.

In the third study, Wherry's team investigated the regulation of Tex cells by PD-1 and LAG-3 in chronic viral infections and cancer [1]. They found that LAG-3 sustains TOX expression and modulates the CD94/NKG2-Qa-1b axis, influencing NK receptor expression and Tex cytotoxicity. LAG-3 drives the formation of a CD94/NKG2+ Tex subset by recognizing the stress ligand Qa-1b, with similar observations in human studies. This research highlights the distinct roles of PD-1 and LAG-3 in regulating T cell exhaustion, pointing to LAG-3's role in maintaining Tex persistence and enhancing cytotoxicity.

Together, these studies uncover the mechanisms through which PD-1 and LAG-3 modulate CD8+ T cell function (Figure 1). Vignali's work emphasizes the balance between effector function and exhaustion, suggesting that LAG-3 inhibition may enhance the immune response at the expense of long-term T cell persistence. Wherry's findings complement this by highlighting LAG-3's role in sustaining T cell cytotoxicity and persistence, providing a comprehensive view of the dual checkpoint blockade.

While these findings represent significant progress, there are limitations. In the clinical trial led by Vignali et al. [3], Relatlimab monotherapy lasted for only 4 weeks (all patient groups received dual antibody treatment after 4 weeks). As a result, it remains unclear whether the results observed after anti-LAG-3 treatment will persist. Second, the study's reliance on single-cell sequencing presents a problem [3]. In some cases, single-cell sequencing may not be able to capture cell-to-cell interactions and dynamic changes. Moreover, it mainly focuses on gene expression levels while ignoring many other important biological characteristics such as cell metabolic state, protein expression, and signaling pathway activity. Therefore, in the future, it might be necessary to combine multiple technical means, such as multiomics data integration, flow cytometry, and imaging analysis, to further verify its key findings. Additionally, in two other mouse studies, an important finding was made: the combined loss of PD-1 and LAG-3 led to the emergence of NK-like effector CD8+ T cells, which are characterized by the simultaneous expression of activating and inhibitory NK receptors on the surface [1, 2]. It should be noted that given the differences between the mouse model and the human immune system in some key aspects, the verification of this finding depends on further human research in the future to provide a definitive answer.

Moreover, current clinical trials must expand to include broader patient populations and diverse tumor types to ensure the safety and efficacy of PD-1 and LAG-3 inhibitors across different contexts. Personalized treatment approaches, guided by biomarkers that predict patient responses, are essential for refining therapeutic regimens. A recent study demonstrated that LAG-3 and PD-1 inhibitors enhance outcomes in advanced nasopharyngeal carcinoma (NPC). Immunotherapy-naïve patients showed 33.3% objective response rate (ORR), 75% disease control rate (DCR), and 10.8-month median PFS, outperforming PD-1 monotherapy. Responses persisted in resistant cases (11.8% ORR, 64.7% DCR). This dual blockade holds clinical promise, particularly as frontline therapy for advanced NPC [5].

In conclusion, these studies offer new insights into the regulation of immune checkpoints, providing a foundation for future research and clinical applications. They underscore the need for further experiments and trials to better understand the mechanisms of PD-1 and LAG-3, optimize immunotherapy strategies, and ultimately improve patient outcomes.

Jianqiao Shentu analyzed the literature, wrote the manuscript, Jianqiao Shentu and Hening Xu drafted Figure 1. Jianqiao Shentu and Shiwei Duan conceived the idea. Shiwei Duan reviewed and revised the manuscript. All the authors gave the final approval of the submitted version.

Not applicable.

The authors declare no conflicts of interest.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信