阻断 TIM-1 对 B 细胞的抗肿瘤作用

Wenwen Liu, Jian Huang
{"title":"阻断 TIM-1 对 B 细胞的抗肿瘤作用","authors":"Wenwen Liu,&nbsp;Jian Huang","doi":"10.1002/mef2.71","DOIUrl":null,"url":null,"abstract":"<p>A research by Bod et al.<span><sup>1</sup></span> has been published recently in <i>Nature</i> entitled “B-cell-specific checkpoint molecules that regulate anti-tumor immunity‘.” It revealed that T-cell immunoglobulin and mucin domain protein 1 (TIM-1) blockade can strikingly reduce the melanoma size in mouse models.</p><p>In the field of antitumor immunotherapy, researchers have been extensively studying the role of T cells in fighting against tumors. However, the function of B cells in antitumor immunity has remained unclear or controversial. To address this gap, this collaboration study among Harvard, MIT, and many others has shed light on a crucial molecule called TIM-1, which for the first time, has been claimed to act as a specific checkpoint on B cells.<span><sup>1</sup></span></p><p>TIM-1 is also known as hepatitis A virus cellular receptor 1 (HAVCR1) and kidney injury molecule 1 as well. It is a membrane glycoprotein encoded by the <i>Havcr1</i> gene. Expressed on various cells as a receptor for many viruses and ligands, TIM-1 has been reported to involve in kidney diseases, atopic diseases, T-cell activation and cancers.<span><sup>2-4</sup></span> Antibody–drug targeting TIM-1 has also been developed for treating cancers with high TIM-1 expression.<span><sup>4</sup></span> In Phase I clinical trial, CDX-014, an antibody–drug conjugate against TIM-1, exhibited a manageable toxicity profile and early signs of activity in 16 patients with advanced refractory renal cell cancer.<span><sup>4</sup></span> However, no scholar considers the role of TIM-1 on B cells in these studies, not to speak of as a B-cell-specific checkpoint molecule.</p><p>Immune checkpoints are host molecules that have the ability to suppress immune responses.<span><sup>5</sup></span> Their expression will suppress antitumor immune responses in cancer. In past studies, researchers have primarily focused on enhancing the immune response of T cells, particularly cytotoxic T lymphocytes, in the fight against cancer. The discovery of immune checkpoints, such as the PD-1/PD-L1 and the CTLA4/B7 pathways sparked a wave of interest in T-cell-specific immune checkpoint inhibitors for cancer treatment. However, the authors of this study sought to investigate B cells, as they are one of the most abundant cell types that infiltrate tumors, especially in melanoma.</p><p>To understand the role of B cell subsets in melanoma, the researchers employed the well-established B16F10 melanoma mouse model and collected immune cells at 7, 10, and 16 days after tumor cell injection. Subsequently, bulk messenger RNA-sequencing was performed on the tumor group along with draining lymph nodes (dLNs), nondraining lymph nodes (ndLNs), and spleen groups at Day 16 postinjection to examine the expression profiles of B cells across different groups. To further comprehend the heterogeneity of B cells within tumor infiltration, single-cell RNA sequencing (scRNA-seq) combined with B-cell receptor sequencing analysis were utilized to identify expanding subsets of B cells within these three groups over time. Notably, a distinct cluster of proliferative germinal center-like B cells (cluster 3) was observed in dLNs that exhibited a corresponding increase in tumor growth. The researchers identified a key gene, <i>Havcr1</i>, encoding TIM-1 molecule, specifically expressed by cluster 3. Based on bulk RNA-seq and flow cytometry analysis, TIM-1<sup>+</sup> B cells from dLNs and ndLNs showed increased proliferation and tended to differentiate into plasma cells compared with TIM-1<sup>−</sup> B cells. Furthermore, TIM-1<sup>+</sup> B cells expressed higher levels of various coinhibitory and immunoregulatory molecules, including PD-1, TIGIT, LAG3, TIM-3, CD39, CD73, and IL-10.</p><p>Subsequently, the researchers found that conditional deletion of <i>Havcr1</i> gene in B cells significantly inhibited tumor growth in multiple melanoma models as well as other cancer models. However, genetic deletion of known checkpoints TIM-3, TIGIT, PD1, or LAG3 in B cells had little or no effect on tumor growth. In addition, deletion of <i>Havcr1</i> in all T cells had no effect on tumor growth, demonstrating the vital role of TIM-1 specifically expressed on B cells. Therefore antitumor effect of anti-TIM-1 therapy was completely dependent on TIM-1 expression on B cells. Furthermore, they found that the TIM-1 blockade of B cells can also enhance the antitumor immune effect of PD-1 blocking therapy, providing a possible new choice for checkpoint combination therapy. Moreover, after analyzing public data of tumor-infiltrating leukocytes (TILs) in human tumors, Bod et al.<span><sup>1</sup></span> also found that higher expression of <i>Havcr1</i> corresponded to poorer overall survival in lung, pancreatic and stomach adenocarcinoma patients. They identified a cluster of TIM-1<sup>+</sup> B cells simultaneously expressing several know coinhibitory molecules. The signature of this subset of B cells is somehow similar to human exhausted T cells.</p><p>As summarized in Figure 1, the experimental results and analysis of scRNA-seq profile revealed several impacts of <i>Havcr1</i> deficiency in B cells, which might explain why it can suppress tumor growth. First, T-cell receptor sequencing analysis revealed that deficiency of <i>Havcr1</i> in B cells led to an increased immune cell infiltration and an increased antigen-specific CD8<sup>+</sup> TILs to enhance effector T-cell responses, including stronger cytotoxicity of CD8<sup>+</sup> T cells, higher CD107a expression, and more coexpression of granzyme B as well as perforin. Second, <i>Havcr1</i> deficiency in B cells has minimal effect on the humoral immunity against tumors and lymphatic organs. However, it enhanced B-cell antigen presentation to CD4<sup>+</sup> T cells, expanded CD4<sup>+</sup> T helper cells, and reduced FOXP3<sup>+</sup> T<sub>reg</sub> cell expansion. Third, TIM-1-deficient B cells showed higher expression and sensitivity to type I and type II interferon.</p><p>In summary, this study challenges the previously controversial role of B cells in antitumor immunotherapy and paves the way for innovative therapeutic approaches. TIM-1 blockade therapy has shown early signs of effect in cancers with high TIM-1 expression in clinical trials.<span><sup>5</sup></span> The significance of TIM-1 as a checkpoint molecule seems to lie in its expression on B cells rather than on T cells or cancer cells. By understanding this, researchers can expand TIM-1 blockade therapy to cancers with low or no TIM-1 expression. This breakthrough has the potential to revolutionize the design of immunotherapeutic strategies, opening up new avenues for treating cancer patients. However, further studies are warranted to explore which receptor makes TIM-1 the immune checkpoint on B cells. Additionally, further investigating the therapeutic potential TIM-1 blockade of B cells in preclinical and clinical settings could lead to the development of more novel treatments that harness the power of immune checkpoint on B cells in combating tumors.</p><p><b>Wenwen Liu</b>: Writing—original draft (lead); writing—review and editing (equal). <b>Jian Huang</b>: Conceptualization (lead); funding acquisition (lead); supervision (lead); writing—review and editing (equal). Both authors have written and approved the article.</p><p>The authors declare no conflicts of interest.</p><p>Not applicable.</p>","PeriodicalId":74135,"journal":{"name":"MedComm - Future medicine","volume":"3 1","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.71","citationCount":"0","resultStr":"{\"title\":\"Antitumor effects of TIM-1 blockade in B cells\",\"authors\":\"Wenwen Liu,&nbsp;Jian Huang\",\"doi\":\"10.1002/mef2.71\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>A research by Bod et al.<span><sup>1</sup></span> has been published recently in <i>Nature</i> entitled “B-cell-specific checkpoint molecules that regulate anti-tumor immunity‘.” It revealed that T-cell immunoglobulin and mucin domain protein 1 (TIM-1) blockade can strikingly reduce the melanoma size in mouse models.</p><p>In the field of antitumor immunotherapy, researchers have been extensively studying the role of T cells in fighting against tumors. However, the function of B cells in antitumor immunity has remained unclear or controversial. To address this gap, this collaboration study among Harvard, MIT, and many others has shed light on a crucial molecule called TIM-1, which for the first time, has been claimed to act as a specific checkpoint on B cells.<span><sup>1</sup></span></p><p>TIM-1 is also known as hepatitis A virus cellular receptor 1 (HAVCR1) and kidney injury molecule 1 as well. It is a membrane glycoprotein encoded by the <i>Havcr1</i> gene. Expressed on various cells as a receptor for many viruses and ligands, TIM-1 has been reported to involve in kidney diseases, atopic diseases, T-cell activation and cancers.<span><sup>2-4</sup></span> Antibody–drug targeting TIM-1 has also been developed for treating cancers with high TIM-1 expression.<span><sup>4</sup></span> In Phase I clinical trial, CDX-014, an antibody–drug conjugate against TIM-1, exhibited a manageable toxicity profile and early signs of activity in 16 patients with advanced refractory renal cell cancer.<span><sup>4</sup></span> However, no scholar considers the role of TIM-1 on B cells in these studies, not to speak of as a B-cell-specific checkpoint molecule.</p><p>Immune checkpoints are host molecules that have the ability to suppress immune responses.<span><sup>5</sup></span> Their expression will suppress antitumor immune responses in cancer. In past studies, researchers have primarily focused on enhancing the immune response of T cells, particularly cytotoxic T lymphocytes, in the fight against cancer. The discovery of immune checkpoints, such as the PD-1/PD-L1 and the CTLA4/B7 pathways sparked a wave of interest in T-cell-specific immune checkpoint inhibitors for cancer treatment. However, the authors of this study sought to investigate B cells, as they are one of the most abundant cell types that infiltrate tumors, especially in melanoma.</p><p>To understand the role of B cell subsets in melanoma, the researchers employed the well-established B16F10 melanoma mouse model and collected immune cells at 7, 10, and 16 days after tumor cell injection. Subsequently, bulk messenger RNA-sequencing was performed on the tumor group along with draining lymph nodes (dLNs), nondraining lymph nodes (ndLNs), and spleen groups at Day 16 postinjection to examine the expression profiles of B cells across different groups. To further comprehend the heterogeneity of B cells within tumor infiltration, single-cell RNA sequencing (scRNA-seq) combined with B-cell receptor sequencing analysis were utilized to identify expanding subsets of B cells within these three groups over time. Notably, a distinct cluster of proliferative germinal center-like B cells (cluster 3) was observed in dLNs that exhibited a corresponding increase in tumor growth. The researchers identified a key gene, <i>Havcr1</i>, encoding TIM-1 molecule, specifically expressed by cluster 3. Based on bulk RNA-seq and flow cytometry analysis, TIM-1<sup>+</sup> B cells from dLNs and ndLNs showed increased proliferation and tended to differentiate into plasma cells compared with TIM-1<sup>−</sup> B cells. Furthermore, TIM-1<sup>+</sup> B cells expressed higher levels of various coinhibitory and immunoregulatory molecules, including PD-1, TIGIT, LAG3, TIM-3, CD39, CD73, and IL-10.</p><p>Subsequently, the researchers found that conditional deletion of <i>Havcr1</i> gene in B cells significantly inhibited tumor growth in multiple melanoma models as well as other cancer models. However, genetic deletion of known checkpoints TIM-3, TIGIT, PD1, or LAG3 in B cells had little or no effect on tumor growth. In addition, deletion of <i>Havcr1</i> in all T cells had no effect on tumor growth, demonstrating the vital role of TIM-1 specifically expressed on B cells. Therefore antitumor effect of anti-TIM-1 therapy was completely dependent on TIM-1 expression on B cells. Furthermore, they found that the TIM-1 blockade of B cells can also enhance the antitumor immune effect of PD-1 blocking therapy, providing a possible new choice for checkpoint combination therapy. Moreover, after analyzing public data of tumor-infiltrating leukocytes (TILs) in human tumors, Bod et al.<span><sup>1</sup></span> also found that higher expression of <i>Havcr1</i> corresponded to poorer overall survival in lung, pancreatic and stomach adenocarcinoma patients. They identified a cluster of TIM-1<sup>+</sup> B cells simultaneously expressing several know coinhibitory molecules. The signature of this subset of B cells is somehow similar to human exhausted T cells.</p><p>As summarized in Figure 1, the experimental results and analysis of scRNA-seq profile revealed several impacts of <i>Havcr1</i> deficiency in B cells, which might explain why it can suppress tumor growth. First, T-cell receptor sequencing analysis revealed that deficiency of <i>Havcr1</i> in B cells led to an increased immune cell infiltration and an increased antigen-specific CD8<sup>+</sup> TILs to enhance effector T-cell responses, including stronger cytotoxicity of CD8<sup>+</sup> T cells, higher CD107a expression, and more coexpression of granzyme B as well as perforin. Second, <i>Havcr1</i> deficiency in B cells has minimal effect on the humoral immunity against tumors and lymphatic organs. However, it enhanced B-cell antigen presentation to CD4<sup>+</sup> T cells, expanded CD4<sup>+</sup> T helper cells, and reduced FOXP3<sup>+</sup> T<sub>reg</sub> cell expansion. Third, TIM-1-deficient B cells showed higher expression and sensitivity to type I and type II interferon.</p><p>In summary, this study challenges the previously controversial role of B cells in antitumor immunotherapy and paves the way for innovative therapeutic approaches. TIM-1 blockade therapy has shown early signs of effect in cancers with high TIM-1 expression in clinical trials.<span><sup>5</sup></span> The significance of TIM-1 as a checkpoint molecule seems to lie in its expression on B cells rather than on T cells or cancer cells. By understanding this, researchers can expand TIM-1 blockade therapy to cancers with low or no TIM-1 expression. This breakthrough has the potential to revolutionize the design of immunotherapeutic strategies, opening up new avenues for treating cancer patients. However, further studies are warranted to explore which receptor makes TIM-1 the immune checkpoint on B cells. Additionally, further investigating the therapeutic potential TIM-1 blockade of B cells in preclinical and clinical settings could lead to the development of more novel treatments that harness the power of immune checkpoint on B cells in combating tumors.</p><p><b>Wenwen Liu</b>: Writing—original draft (lead); writing—review and editing (equal). <b>Jian Huang</b>: Conceptualization (lead); funding acquisition (lead); supervision (lead); writing—review and editing (equal). Both authors have written and approved the article.</p><p>The authors declare no conflicts of interest.</p><p>Not applicable.</p>\",\"PeriodicalId\":74135,\"journal\":{\"name\":\"MedComm - Future medicine\",\"volume\":\"3 1\",\"pages\":\"\"},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-01-29\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mef2.71\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm - Future medicine\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mef2.71\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm - Future medicine","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mef2.71","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

最近,《自然》杂志发表了 Bod 等人1 的一项研究,题为 "调节抗肿瘤免疫的 B 细胞特异性检查点分子"。在抗肿瘤免疫疗法领域,研究人员一直在广泛研究 T 细胞在抗肿瘤中的作用。在抗肿瘤免疫疗法领域,研究人员已经广泛研究了T细胞在抗肿瘤中的作用,但B细胞在抗肿瘤免疫中的功能仍不明确或存在争议。为了填补这一空白,哈佛大学、麻省理工学院和许多其他机构合作开展的这项研究揭示了一种名为 TIM-1 的关键分子,并首次声称它是 B 细胞的特异性检查点。它是由 Havcr1 基因编码的一种膜糖蛋白。4 在 I 期临床试验中,针对 TIM-1 的抗体药物共轭物 CDX-014 在 16 例晚期难治性肾细胞癌患者中表现出了可控的毒性特征和早期活性迹象。免疫检查点是一种具有抑制免疫反应能力的宿主分子5 。在过去的研究中,研究人员主要关注增强 T 细胞,尤其是细胞毒性 T 淋巴细胞在抗癌中的免疫反应。免疫检查点(如 PD-1/PD-L1 和 CTLA4/B7 通路)的发现引发了人们对 T 细胞特异性免疫检查点抑制剂治疗癌症的兴趣。为了了解B细胞亚群在黑色素瘤中的作用,研究人员采用了成熟的B16F10黑色素瘤小鼠模型,并在肿瘤细胞注射后7天、10天和16天收集免疫细胞。随后,研究人员在注射后第16天对肿瘤组、引流淋巴结(dLNs)组、非引流淋巴结(ndLNs)组和脾脏组进行了大量信使RNA测序,以检测不同组别B细胞的表达谱。为了进一步了解肿瘤浸润内 B 细胞的异质性,研究人员利用单细胞 RNA 测序(scRNA-seq)结合 B 细胞受体测序分析来确定这三组中随着时间推移不断扩大的 B 细胞亚群。值得注意的是,在dLNs中观察到了一个独特的增殖生殖中心样B细胞群(群3),该群的肿瘤生长也出现了相应的增长。研究人员发现了一个关键基因,即编码TIM-1分子的Havcr1,该基因在群集3中特异性表达。根据大量RNA-seq和流式细胞术分析,与TIM-1- B细胞相比,来自dLNs和ndLNs的TIM-1+ B细胞增殖速度加快,并倾向于分化成浆细胞。此外,TIM-1+ B 细胞表达更高水平的各种协同抑制和免疫调节分子,包括 PD-1、TIGIT、LAG3、TIM-3、CD39、CD73 和 IL-10。然而,在 B 细胞中基因缺失已知的检查点 TIM-3、TIGIT、PD1 或 LAG3 对肿瘤生长几乎没有影响。此外,在所有 T 细胞中删除 Havcr1 对肿瘤生长也没有影响,这证明了特异性表达于 B 细胞的 TIM-1 的重要作用。因此,抗 TIM-1 疗法的抗肿瘤效果完全依赖于 B 细胞中 TIM-1 的表达。此外,他们还发现阻断B细胞的TIM-1还能增强PD-1阻断疗法的抗肿瘤免疫效果,为检查点联合疗法提供了可能的新选择。此外,在分析了人类肿瘤中肿瘤浸润白细胞(TILs)的公开数据后,Bod 等人1 还发现,在肺癌、胰腺癌和胃腺癌患者中,Havcr1 的高表达与较差的总生存率相对应。他们发现了一组同时表达多种已知共抑制分子的 TIM-1+ B 细胞。如图 1 所示,实验结果和 scRNA-seq 图谱分析揭示了 Havcr1 缺乏对 B 细胞的多种影响,这或许能解释为什么它能抑制肿瘤生长。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Antitumor effects of TIM-1 blockade in B cells

Antitumor effects of TIM-1 blockade in B cells

A research by Bod et al.1 has been published recently in Nature entitled “B-cell-specific checkpoint molecules that regulate anti-tumor immunity‘.” It revealed that T-cell immunoglobulin and mucin domain protein 1 (TIM-1) blockade can strikingly reduce the melanoma size in mouse models.

In the field of antitumor immunotherapy, researchers have been extensively studying the role of T cells in fighting against tumors. However, the function of B cells in antitumor immunity has remained unclear or controversial. To address this gap, this collaboration study among Harvard, MIT, and many others has shed light on a crucial molecule called TIM-1, which for the first time, has been claimed to act as a specific checkpoint on B cells.1

TIM-1 is also known as hepatitis A virus cellular receptor 1 (HAVCR1) and kidney injury molecule 1 as well. It is a membrane glycoprotein encoded by the Havcr1 gene. Expressed on various cells as a receptor for many viruses and ligands, TIM-1 has been reported to involve in kidney diseases, atopic diseases, T-cell activation and cancers.2-4 Antibody–drug targeting TIM-1 has also been developed for treating cancers with high TIM-1 expression.4 In Phase I clinical trial, CDX-014, an antibody–drug conjugate against TIM-1, exhibited a manageable toxicity profile and early signs of activity in 16 patients with advanced refractory renal cell cancer.4 However, no scholar considers the role of TIM-1 on B cells in these studies, not to speak of as a B-cell-specific checkpoint molecule.

Immune checkpoints are host molecules that have the ability to suppress immune responses.5 Their expression will suppress antitumor immune responses in cancer. In past studies, researchers have primarily focused on enhancing the immune response of T cells, particularly cytotoxic T lymphocytes, in the fight against cancer. The discovery of immune checkpoints, such as the PD-1/PD-L1 and the CTLA4/B7 pathways sparked a wave of interest in T-cell-specific immune checkpoint inhibitors for cancer treatment. However, the authors of this study sought to investigate B cells, as they are one of the most abundant cell types that infiltrate tumors, especially in melanoma.

To understand the role of B cell subsets in melanoma, the researchers employed the well-established B16F10 melanoma mouse model and collected immune cells at 7, 10, and 16 days after tumor cell injection. Subsequently, bulk messenger RNA-sequencing was performed on the tumor group along with draining lymph nodes (dLNs), nondraining lymph nodes (ndLNs), and spleen groups at Day 16 postinjection to examine the expression profiles of B cells across different groups. To further comprehend the heterogeneity of B cells within tumor infiltration, single-cell RNA sequencing (scRNA-seq) combined with B-cell receptor sequencing analysis were utilized to identify expanding subsets of B cells within these three groups over time. Notably, a distinct cluster of proliferative germinal center-like B cells (cluster 3) was observed in dLNs that exhibited a corresponding increase in tumor growth. The researchers identified a key gene, Havcr1, encoding TIM-1 molecule, specifically expressed by cluster 3. Based on bulk RNA-seq and flow cytometry analysis, TIM-1+ B cells from dLNs and ndLNs showed increased proliferation and tended to differentiate into plasma cells compared with TIM-1 B cells. Furthermore, TIM-1+ B cells expressed higher levels of various coinhibitory and immunoregulatory molecules, including PD-1, TIGIT, LAG3, TIM-3, CD39, CD73, and IL-10.

Subsequently, the researchers found that conditional deletion of Havcr1 gene in B cells significantly inhibited tumor growth in multiple melanoma models as well as other cancer models. However, genetic deletion of known checkpoints TIM-3, TIGIT, PD1, or LAG3 in B cells had little or no effect on tumor growth. In addition, deletion of Havcr1 in all T cells had no effect on tumor growth, demonstrating the vital role of TIM-1 specifically expressed on B cells. Therefore antitumor effect of anti-TIM-1 therapy was completely dependent on TIM-1 expression on B cells. Furthermore, they found that the TIM-1 blockade of B cells can also enhance the antitumor immune effect of PD-1 blocking therapy, providing a possible new choice for checkpoint combination therapy. Moreover, after analyzing public data of tumor-infiltrating leukocytes (TILs) in human tumors, Bod et al.1 also found that higher expression of Havcr1 corresponded to poorer overall survival in lung, pancreatic and stomach adenocarcinoma patients. They identified a cluster of TIM-1+ B cells simultaneously expressing several know coinhibitory molecules. The signature of this subset of B cells is somehow similar to human exhausted T cells.

As summarized in Figure 1, the experimental results and analysis of scRNA-seq profile revealed several impacts of Havcr1 deficiency in B cells, which might explain why it can suppress tumor growth. First, T-cell receptor sequencing analysis revealed that deficiency of Havcr1 in B cells led to an increased immune cell infiltration and an increased antigen-specific CD8+ TILs to enhance effector T-cell responses, including stronger cytotoxicity of CD8+ T cells, higher CD107a expression, and more coexpression of granzyme B as well as perforin. Second, Havcr1 deficiency in B cells has minimal effect on the humoral immunity against tumors and lymphatic organs. However, it enhanced B-cell antigen presentation to CD4+ T cells, expanded CD4+ T helper cells, and reduced FOXP3+ Treg cell expansion. Third, TIM-1-deficient B cells showed higher expression and sensitivity to type I and type II interferon.

In summary, this study challenges the previously controversial role of B cells in antitumor immunotherapy and paves the way for innovative therapeutic approaches. TIM-1 blockade therapy has shown early signs of effect in cancers with high TIM-1 expression in clinical trials.5 The significance of TIM-1 as a checkpoint molecule seems to lie in its expression on B cells rather than on T cells or cancer cells. By understanding this, researchers can expand TIM-1 blockade therapy to cancers with low or no TIM-1 expression. This breakthrough has the potential to revolutionize the design of immunotherapeutic strategies, opening up new avenues for treating cancer patients. However, further studies are warranted to explore which receptor makes TIM-1 the immune checkpoint on B cells. Additionally, further investigating the therapeutic potential TIM-1 blockade of B cells in preclinical and clinical settings could lead to the development of more novel treatments that harness the power of immune checkpoint on B cells in combating tumors.

Wenwen Liu: Writing—original draft (lead); writing—review and editing (equal). Jian Huang: Conceptualization (lead); funding acquisition (lead); supervision (lead); writing—review and editing (equal). Both authors have written and approved the article.

The authors declare no conflicts of interest.

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:481959085
Book学术官方微信