三级淋巴结构在肾细胞癌中的作用:从预测性生物标志物到治疗靶点

IF 1.9
Chongxiang Gao, Du Cai, Jianguang Qiu, Feng Gao
{"title":"三级淋巴结构在肾细胞癌中的作用:从预测性生物标志物到治疗靶点","authors":"Chongxiang Gao,&nbsp;Du Cai,&nbsp;Jianguang Qiu,&nbsp;Feng Gao","doi":"10.1002/ctd2.70085","DOIUrl":null,"url":null,"abstract":"<p>In the evolving landscape of cancer immunotherapy, tertiary lymphoid structures (TLS) have rapidly transitioned from a histological curiosity to a central mediator of anti-tumour immunity.<span><sup>1</sup></span> A growing body of evidence, particularly within renal cell carcinoma (RCC), has established a strong association between the presence of dense, mature TLS in the tumour microenvironment (TME) and significant clinical benefit from immune checkpoint inhibitor (ICI) therapy. A recent meta-analysis provided high-level evidence for this, confirming that high TLS density correlates with superior objective response rates and prolonged progression-free survival across solid tumours, including RCC.<span><sup>2</sup></span> This consensus is not only reshaping our understanding of the TME but also charting new paths for therapeutic innovation.</p><p>The significance of TLS lies in their function as fully equipped, in situ immune factories. Groundbreaking work using spatial transcriptomics has beautifully elucidated that within the TLS of RCC, a complete B cell maturation process occurs — from B cell recruitment and germinal centre reactions to the generation of antibody-producing plasma cells.<span><sup>3</sup></span> These locally produced IgG antibodies can directly bind to and mark tumour cells for apoptosis. This elegant mechanism provides a compelling explanation for why TLS signatures are a key immunogenomic determinant for ‘exceptional responders’ to combination immunotherapy in RCC.<span><sup>4</sup></span></p><p>However, the clinical utility of TLS is not a simple binary question; its value is deeply rooted in its heterogeneity. First, location and maturity are paramount. Studies indicate that it is the intratumoral and mature (i.e. secondary follicle-like) TLS that are most strongly associated with favourable outcomes and ICI efficacy.<span><sup>5</sup></span> Second, the value of TLS is highly context-dependent on the surrounding immune milieu. A pivotal study proposed a more refined paradigm: optimal response to PD-1 blockade in RCC requires the combination of high TLS density with a low abundance of tissue-resident exhausted CD8<sup>+</sup> T cells.<span><sup>6</sup></span> This suggests that even a potent ‘immune factory’ is of limited use if the effector T cells are already terminally dysfunctional. This complex interplay may explain paradoxical reports of TLS correlating with poor prognosis in some RCC cohorts.<span><sup>7</sup></span></p><p>These deepening insights are paving the way for clinical translation and future therapeutic strategies. To overcome the challenges of invasive biopsies, developing non-invasive surrogates is critical. An important clinical study has linked the baseline level of circulating unswitched memory B cells with the presence of intratumoral TLS and improved survival in patients with RCC treated with nivolumab, presenting a promising avenue for a liquid biopsy approach.<span><sup>8</sup></span> This principle is not unique to RCC; similar efforts in other cancers have also successfully identified circulating T cell signatures that correlate with intratumoral TLS, reinforcing the feasibility of this approach.<span><sup>9</sup></span> Furthermore, in the highly aggressive yet paradoxically ICI-sensitive sarcomatoid RCC subtype, TLS are now understood to be ‘pervasive’, providing a key mechanistic explanation for its unique immune sensitivity.<span><sup>10</sup></span></p><p>Looking ahead, the field is poised to shift from passive prediction to active modulation. The ultimate goal is to therapeutically induce the formation and maturation of TLS, thereby converting immunologically ‘cold’ tumours into ‘hot’ ones. Excitingly, forward-looking research in bioengineering has already demonstrated the potential to generate functional TLS-like organoids in vitro that, when transplanted, can recruit immune cells and exert anti-tumour effects in vivo.<span><sup>11</sup></span></p><p>In conclusion, TLS represent not only a powerful predictive biomarker in RCC but also an actionable therapeutic target. First, future efforts must focus on standardizing TLS assessment and validating reliable circulating biomarkers. Furthermore, it is crucial to elucidate the key molecular pathways that govern TLS formation and maturation. Ultimately, this knowledge should guide the development of innovative therapies that can safely and effectively induce mature TLS in situ. By pursuing these avenues, we can translate our biological understanding of TLS into tangible clinical strategies that will profoundly improve outcomes for patients with RCC.</p><p>Chongxiang Gao and Du Cai drafted the initial version of the manuscript. Jianguang Qiu and Feng Gao reviewed the manuscript. All authors approved and agreed to publish the final version of the manuscript.</p><p>Not applicable.</p>","PeriodicalId":72605,"journal":{"name":"Clinical and translational discovery","volume":"5 5","pages":""},"PeriodicalIF":1.9000,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/ctd2.70085","citationCount":"0","resultStr":"{\"title\":\"The role of tertiary lymphoid structures in renal cell carcinoma: From predictive biomarker to therapeutic target\",\"authors\":\"Chongxiang Gao,&nbsp;Du Cai,&nbsp;Jianguang Qiu,&nbsp;Feng Gao\",\"doi\":\"10.1002/ctd2.70085\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>In the evolving landscape of cancer immunotherapy, tertiary lymphoid structures (TLS) have rapidly transitioned from a histological curiosity to a central mediator of anti-tumour immunity.<span><sup>1</sup></span> A growing body of evidence, particularly within renal cell carcinoma (RCC), has established a strong association between the presence of dense, mature TLS in the tumour microenvironment (TME) and significant clinical benefit from immune checkpoint inhibitor (ICI) therapy. A recent meta-analysis provided high-level evidence for this, confirming that high TLS density correlates with superior objective response rates and prolonged progression-free survival across solid tumours, including RCC.<span><sup>2</sup></span> This consensus is not only reshaping our understanding of the TME but also charting new paths for therapeutic innovation.</p><p>The significance of TLS lies in their function as fully equipped, in situ immune factories. Groundbreaking work using spatial transcriptomics has beautifully elucidated that within the TLS of RCC, a complete B cell maturation process occurs — from B cell recruitment and germinal centre reactions to the generation of antibody-producing plasma cells.<span><sup>3</sup></span> These locally produced IgG antibodies can directly bind to and mark tumour cells for apoptosis. This elegant mechanism provides a compelling explanation for why TLS signatures are a key immunogenomic determinant for ‘exceptional responders’ to combination immunotherapy in RCC.<span><sup>4</sup></span></p><p>However, the clinical utility of TLS is not a simple binary question; its value is deeply rooted in its heterogeneity. First, location and maturity are paramount. Studies indicate that it is the intratumoral and mature (i.e. secondary follicle-like) TLS that are most strongly associated with favourable outcomes and ICI efficacy.<span><sup>5</sup></span> Second, the value of TLS is highly context-dependent on the surrounding immune milieu. A pivotal study proposed a more refined paradigm: optimal response to PD-1 blockade in RCC requires the combination of high TLS density with a low abundance of tissue-resident exhausted CD8<sup>+</sup> T cells.<span><sup>6</sup></span> This suggests that even a potent ‘immune factory’ is of limited use if the effector T cells are already terminally dysfunctional. This complex interplay may explain paradoxical reports of TLS correlating with poor prognosis in some RCC cohorts.<span><sup>7</sup></span></p><p>These deepening insights are paving the way for clinical translation and future therapeutic strategies. To overcome the challenges of invasive biopsies, developing non-invasive surrogates is critical. An important clinical study has linked the baseline level of circulating unswitched memory B cells with the presence of intratumoral TLS and improved survival in patients with RCC treated with nivolumab, presenting a promising avenue for a liquid biopsy approach.<span><sup>8</sup></span> This principle is not unique to RCC; similar efforts in other cancers have also successfully identified circulating T cell signatures that correlate with intratumoral TLS, reinforcing the feasibility of this approach.<span><sup>9</sup></span> Furthermore, in the highly aggressive yet paradoxically ICI-sensitive sarcomatoid RCC subtype, TLS are now understood to be ‘pervasive’, providing a key mechanistic explanation for its unique immune sensitivity.<span><sup>10</sup></span></p><p>Looking ahead, the field is poised to shift from passive prediction to active modulation. The ultimate goal is to therapeutically induce the formation and maturation of TLS, thereby converting immunologically ‘cold’ tumours into ‘hot’ ones. Excitingly, forward-looking research in bioengineering has already demonstrated the potential to generate functional TLS-like organoids in vitro that, when transplanted, can recruit immune cells and exert anti-tumour effects in vivo.<span><sup>11</sup></span></p><p>In conclusion, TLS represent not only a powerful predictive biomarker in RCC but also an actionable therapeutic target. First, future efforts must focus on standardizing TLS assessment and validating reliable circulating biomarkers. Furthermore, it is crucial to elucidate the key molecular pathways that govern TLS formation and maturation. Ultimately, this knowledge should guide the development of innovative therapies that can safely and effectively induce mature TLS in situ. By pursuing these avenues, we can translate our biological understanding of TLS into tangible clinical strategies that will profoundly improve outcomes for patients with RCC.</p><p>Chongxiang Gao and Du Cai drafted the initial version of the manuscript. Jianguang Qiu and Feng Gao reviewed the manuscript. All authors approved and agreed to publish the final version of the manuscript.</p><p>Not applicable.</p>\",\"PeriodicalId\":72605,\"journal\":{\"name\":\"Clinical and translational discovery\",\"volume\":\"5 5\",\"pages\":\"\"},\"PeriodicalIF\":1.9000,\"publicationDate\":\"2025-09-09\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/ctd2.70085\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical and translational discovery\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/ctd2.70085\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical and translational discovery","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/ctd2.70085","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

在不断发展的癌症免疫治疗领域,三级淋巴样结构(TLS)已经迅速从组织学上的好奇心转变为抗肿瘤免疫的中心介质越来越多的证据,特别是在肾细胞癌(RCC)中,已经建立了肿瘤微环境(TME)中致密、成熟的TLS的存在与免疫检查点抑制剂(ICI)治疗的显着临床益处之间的密切关联。最近的一项荟萃分析为此提供了高水平的证据,证实高TLS密度与实体肿瘤(包括rcc)中优越的客观缓解率和延长的无进展生存期相关。这一共识不仅重塑了我们对TME的理解,而且为治疗创新开辟了新的途径。TLS的意义在于它们作为装备齐全的原位免疫工厂的功能。利用空间转录组学的开创性工作已经很好地阐明了在RCC的TLS中,发生了一个完整的B细胞成熟过程-从B细胞募集和生发中心反应到产生抗体的浆细胞这些局部产生的IgG抗体可以直接结合并标记肿瘤细胞凋亡。这种优雅的机制为为什么TLS签名是rcc联合免疫治疗“特殊应答者”的关键免疫基因组决定因素提供了令人信服的解释。它的价值深深植根于它的异质性。首先,地点和成熟度是最重要的。研究表明,肿瘤内和成熟的(即继发卵泡样)TLS与良好的预后和ICI疗效最密切相关其次,TLS的值高度依赖于周围的免疫环境。一项关键研究提出了一个更完善的范例:在RCC中,对PD-1阻断的最佳反应需要高TLS密度与低含量的组织驻留耗尽CD8+ T细胞的结合这表明,如果效应T细胞已经最终功能失调,即使是一个强大的“免疫工厂”的作用也是有限的。这种复杂的相互作用可以解释一些RCC队列中TLS与不良预后相关的矛盾报道。这些深入的见解为临床转化和未来的治疗策略铺平了道路。为了克服侵入性活检的挑战,开发非侵入性替代品至关重要。一项重要的临床研究将循环未切换记忆B细胞的基线水平与肿瘤内TLS的存在和nivolumab治疗的RCC患者的生存率提高联系起来,为液体活检方法提供了一个有希望的途径这一原则并非碾压混凝土所独有;在其他癌症中,类似的研究也成功地识别了与肿瘤内TLS相关的循环T细胞特征,加强了这种方法的可行性此外,在高度侵袭性但对ici敏感的肉瘤样RCC亚型中,TLS现在被认为是“普遍存在的”,这为其独特的免疫敏感性提供了关键的机制解释。展望未来,该领域将从被动预测转向主动调制。最终目标是通过治疗诱导TLS的形成和成熟,从而将免疫上的“冷”肿瘤转化为“热”肿瘤。令人兴奋的是,生物工程的前瞻性研究已经证明了在体外产生功能性的类tls器官的潜力,当移植时,可以在体内招募免疫细胞并发挥抗肿瘤作用。综上所述,TLS不仅是一种强大的预测RCC的生物标志物,也是一种可行的治疗靶点。首先,未来的努力必须集中在标准化TLS评估和验证可靠的循环生物标志物上。此外,阐明控制TLS形成和成熟的关键分子途径至关重要。最终,这些知识将指导能够安全有效地原位诱导成熟TLS的创新疗法的发展。通过追求这些途径,我们可以将我们对TLS的生物学理解转化为切实的临床策略,这将极大地改善RCC患者的预后。高崇祥和杜才起草了初稿。邱建光和高峰审阅了稿件。所有作者都批准并同意出版手稿的最终版本。不适用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
The role of tertiary lymphoid structures in renal cell carcinoma: From predictive biomarker to therapeutic target

In the evolving landscape of cancer immunotherapy, tertiary lymphoid structures (TLS) have rapidly transitioned from a histological curiosity to a central mediator of anti-tumour immunity.1 A growing body of evidence, particularly within renal cell carcinoma (RCC), has established a strong association between the presence of dense, mature TLS in the tumour microenvironment (TME) and significant clinical benefit from immune checkpoint inhibitor (ICI) therapy. A recent meta-analysis provided high-level evidence for this, confirming that high TLS density correlates with superior objective response rates and prolonged progression-free survival across solid tumours, including RCC.2 This consensus is not only reshaping our understanding of the TME but also charting new paths for therapeutic innovation.

The significance of TLS lies in their function as fully equipped, in situ immune factories. Groundbreaking work using spatial transcriptomics has beautifully elucidated that within the TLS of RCC, a complete B cell maturation process occurs — from B cell recruitment and germinal centre reactions to the generation of antibody-producing plasma cells.3 These locally produced IgG antibodies can directly bind to and mark tumour cells for apoptosis. This elegant mechanism provides a compelling explanation for why TLS signatures are a key immunogenomic determinant for ‘exceptional responders’ to combination immunotherapy in RCC.4

However, the clinical utility of TLS is not a simple binary question; its value is deeply rooted in its heterogeneity. First, location and maturity are paramount. Studies indicate that it is the intratumoral and mature (i.e. secondary follicle-like) TLS that are most strongly associated with favourable outcomes and ICI efficacy.5 Second, the value of TLS is highly context-dependent on the surrounding immune milieu. A pivotal study proposed a more refined paradigm: optimal response to PD-1 blockade in RCC requires the combination of high TLS density with a low abundance of tissue-resident exhausted CD8+ T cells.6 This suggests that even a potent ‘immune factory’ is of limited use if the effector T cells are already terminally dysfunctional. This complex interplay may explain paradoxical reports of TLS correlating with poor prognosis in some RCC cohorts.7

These deepening insights are paving the way for clinical translation and future therapeutic strategies. To overcome the challenges of invasive biopsies, developing non-invasive surrogates is critical. An important clinical study has linked the baseline level of circulating unswitched memory B cells with the presence of intratumoral TLS and improved survival in patients with RCC treated with nivolumab, presenting a promising avenue for a liquid biopsy approach.8 This principle is not unique to RCC; similar efforts in other cancers have also successfully identified circulating T cell signatures that correlate with intratumoral TLS, reinforcing the feasibility of this approach.9 Furthermore, in the highly aggressive yet paradoxically ICI-sensitive sarcomatoid RCC subtype, TLS are now understood to be ‘pervasive’, providing a key mechanistic explanation for its unique immune sensitivity.10

Looking ahead, the field is poised to shift from passive prediction to active modulation. The ultimate goal is to therapeutically induce the formation and maturation of TLS, thereby converting immunologically ‘cold’ tumours into ‘hot’ ones. Excitingly, forward-looking research in bioengineering has already demonstrated the potential to generate functional TLS-like organoids in vitro that, when transplanted, can recruit immune cells and exert anti-tumour effects in vivo.11

In conclusion, TLS represent not only a powerful predictive biomarker in RCC but also an actionable therapeutic target. First, future efforts must focus on standardizing TLS assessment and validating reliable circulating biomarkers. Furthermore, it is crucial to elucidate the key molecular pathways that govern TLS formation and maturation. Ultimately, this knowledge should guide the development of innovative therapies that can safely and effectively induce mature TLS in situ. By pursuing these avenues, we can translate our biological understanding of TLS into tangible clinical strategies that will profoundly improve outcomes for patients with RCC.

Chongxiang Gao and Du Cai drafted the initial version of the manuscript. Jianguang Qiu and Feng Gao reviewed the manuscript. All authors approved and agreed to publish the final version of the manuscript.

Not applicable.

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
1.00
自引率
0.00%
发文量
0
×
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