SMARCAL1:驯服肿瘤免疫逃避的新靶点

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL MedComm Pub Date : 2024-09-07 DOI:10.1002/mco2.730
Ting Xiao, Shiqi Li, Xinghua Long
{"title":"SMARCAL1:驯服肿瘤免疫逃避的新靶点","authors":"Ting Xiao,&nbsp;Shiqi Li,&nbsp;Xinghua Long","doi":"10.1002/mco2.730","DOIUrl":null,"url":null,"abstract":"<p>The viability and development of cancer cells depend on their ability to effectively evade the surveillance of the immune system. To achieve this, cancer cells alter the expression or function of specific molecules to become “invisible”. In a recent elegant study in <i>Cell</i>, Leuzzi et al.<span><sup>1</sup></span> unveiled a novel player in the intricate game of tumor immune evasion: SMARCAL1, linked to DNA repair, emerges as a dual regulator with a surprising ability to modulate both innate immune signaling and PD-L1 expression. This work not only sheds light on the complex interplay between DNA damage response, innate immunity, and immune checkpoint regulation, but also identifies a potential new target for cancer immunotherapy.</p><p>Using a CRISPR–Cas9 screen, the authors identified SMARCAL1 as one of the sgRNA-targeted genes that are distinctly enriched in IRF3<sup>High</sup>/PD-L1<sup>Low</sup> cells (Figure 1A). Interferon regulatory factor 3 (IRF3) is a central signaling mediator of the innate immune signaling pathway, selected as a screening factor. In the innate immunity cGAS–STING pathway, abnormally exposed double-stranded DNA (dsDNA) in the cytoplasm binds to cGAS to produce cGAMP.<span><sup>2</sup></span> cGAMP binding by STING is activated, recruiting TANK-binding kinase to phosphorylate IRF3 leading to its nuclear translocation and the induction of type I interferons (IFNs), which then triggers the expression of interferon stimulated genes (ISGs) expression to enhance cellular autonomous defense mechanisms.<span><sup>3</sup></span> Furthermore, PD-L1 can undergo upregulation in antigen-presenting cells following IFN-γ stimulation, which leads to immune tolerance. This observation provides fresh perspectives on the significant role of SMARCAL1 in modulating IRF3-mediated immune stimulation and the expression levels of the immune checkpoint PD-L1.</p><p>SMARCAL1 is an ATP-dependent, SWI/SNF-related annealing helicase that stabilizes replication forks during DNA damage. Mutations in this gene are responsible for Schimke immune-osseous dysplasia, an autosomal recessive disorder characterized by T-cell immunodeficiency and growth dysfunctions.<span><sup>4</sup></span> In this study, SMARCAL1 operates on dual fronts. First, it dampens the cGAS–STING pathway, which is triggered by abnormal DNA in the cytoplasm. Chromosomal instability is a primary source of cytosolic dsDNA, a hallmark of cancer cells.<span><sup>2</sup></span> Second, it promotes PD-L1 expression, which shields tumors from T cell attack and allows cancer cells to enhance their viability and metastatic potential. SMARCAL1-deficient tumor cells show higher levels of IRF3 phosphorylation and reduced levels of PD-L1. This is consistent with the screening results, indicating that SMARCAL1 deficiency simultaneously induces IRF3 activation to promote the cGAS–STING pathway and downregulates PD-L1. This dual functionality renders SMARCAL1 an exceptional and potentially powerful candidate for immunotherapy.</p><p>Mechanistically, SMARCAL1 can facilitate the repair of collapsed DNA replication forks, alleviating the DNA replication stress caused by oncogenes.<span><sup>5</sup></span> Loss of SMARCAL1 induces genome instability and leads to micronuclei formation. Disruption of the micronuclear envelopes exposes dsDNA to the cytosol, leading to the activation of the cGAS–STING signaling pathway. Furthermore, SMARCAL1 does not directly mediate chromatin remodeling. Notably, SMARCAL1 upholds chromatin accessibility of P3 element located downstream of the PD-L1 transcription start site through its ATPase activity, and cooperates with the AP-1 family member JUN to efficiently promote PD-L1 expression. The research opens a new window to reveal how a single molecule plays a dual role in cancer cells, both hiding itself from the immune system's attack and facilitating the immune system's fight back under certain conditions. This discovery not only provides new insights into the complex mechanisms of cancer, but also opens up the possibility of developing more effective cancer treatment strategies.</p><p>In mouse models, SMARCAL1 deficiency significantly inhibited tumor growth and increased animal survival. Notably, SMARCAL1 is cGAS-dependent for antitumor immunity and enhances T cell-mediated antitumor competence. Due to the lack of accessible P3 regions in B16/F10 mouse cells, SMARCAL1-deficiency-mediated regulation of PD-L1 levels could not be assessed for antitumor effects in the study. Interestingly, PD-L1 downregulation induced by SMARCAL1 deletion in MDA-MB-436 cells made them more sensitive to CD8<sup>+</sup> T cell-mediated killing, which could be partially rescued by PD-L1 overexpression. Moreover, treatment of SMARCAL1-deficient B16/F10 tumors with anti-PD-L1 antibody or anti-CTLA-4 antibody significantly reduced tumor growth and improved animal survival. Combined treatment with both antibodies enhanced the effect. SMARCAL1 deficiency has a synergistic effect with immune checkpoint blockade treatment for antitumor immunity.</p><p>Immune checkpoint inhibitors (ICIs) are the mainstay of tumor immunotherapy, blocking cancer cell evasion mechanisms to enhance the immune system's attack on cancer cells. Nevertheless, inherent or acquired tumor resistance to ICIs and treatment-related toxicity limit their clinical application.<span><sup>6</sup></span> The dual mechanism of action of SMARCAL1 may be one of the key factors in enhancing the efficacy of ICIs. Targeting SMARCAL1 can synergize with ICIs to treat tumors, providing a novel and effective approach to tumor immunotherapy. Active DNA-dependent ATPase A domain inhibitor (ADAADi) targets the ATPase domain of SMARCAL1, which was identified as a potent inhibitor of ADAAD, the bovine homologue of SMARCAL1, but SMARCAL1 does not determine the cellular response to ADAADi.<span><sup>7</sup></span> Although there was no significant correlation between SMARCAL1 expression and cellular responsiveness to ADAADi, ADAADi provides a viable way to target inhibition of SMARCAL1 for tumor treatment.</p><p>Although intriguing, several questions persist. Initially, SMARCAL1-mediated regulation of PD-L1 in cancer patients could not be fully mimicked due to the study's heavy reliance on preclinical models, and the fact that SMARCAL1 does not affect the levels of PD-L1 in mouse cells. Further evaluation of SMARCAL1-mediated antitumor effects in mouse tumor models is needed. And it is required to conduct comprehensive research across various types of cancers. Second, minimizing off-target effects and ensuring safety using known and newly developed SMARCAL1 inhibitors is a demanding task. Finally, further exploration is needed to achieve synergistic treatments combining SMARCAL1 targeting with other immunotherapies to enhance clinical benefits.</p><p>Overall, the work of Leuzzi et al.<span><sup>1</sup></span> sheds light on a previously unknown mechanism of tumor immune evasion and highlights the potential of targeting SMARCAL1 as a novel therapeutic strategy (Figure 1B). Targeted inhibition of SMARCAL1 induces cancer innate immune signaling while downregulating PD-L1 levels, resulting in a double whammy for cancer and better cancer treatment outcomes. Further investigation is required to elucidate the precise mechanisms through which SMARCAL1 controls PD-L1 expression and its interactions with other gene targets. Additionally, exploring the potential of SMARCAL1 inhibitors or SMARCAL1-targeting therapies in combination with existing immunotherapies is crucial. This approach is important for translating these findings into clinical applications and propelling them toward the advancement of efficacious cancer therapeutics.</p><p><b>Ting Xiao</b>: drafted the manuscript. <b>Shiqi Li</b>: drew the figure. and <b>Xinghua Long</b>: drafted and reviewed the manuscript. All the authors read and approved the final manuscript.</p><p>The authors declare that they have no conflict of interest.</p><p>Not applicable.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":null,"pages":null},"PeriodicalIF":10.7000,"publicationDate":"2024-09-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.730","citationCount":"0","resultStr":"{\"title\":\"SMARCAL1: a new target for taming tumor immune evasion\",\"authors\":\"Ting Xiao,&nbsp;Shiqi Li,&nbsp;Xinghua Long\",\"doi\":\"10.1002/mco2.730\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>The viability and development of cancer cells depend on their ability to effectively evade the surveillance of the immune system. To achieve this, cancer cells alter the expression or function of specific molecules to become “invisible”. In a recent elegant study in <i>Cell</i>, Leuzzi et al.<span><sup>1</sup></span> unveiled a novel player in the intricate game of tumor immune evasion: SMARCAL1, linked to DNA repair, emerges as a dual regulator with a surprising ability to modulate both innate immune signaling and PD-L1 expression. This work not only sheds light on the complex interplay between DNA damage response, innate immunity, and immune checkpoint regulation, but also identifies a potential new target for cancer immunotherapy.</p><p>Using a CRISPR–Cas9 screen, the authors identified SMARCAL1 as one of the sgRNA-targeted genes that are distinctly enriched in IRF3<sup>High</sup>/PD-L1<sup>Low</sup> cells (Figure 1A). Interferon regulatory factor 3 (IRF3) is a central signaling mediator of the innate immune signaling pathway, selected as a screening factor. In the innate immunity cGAS–STING pathway, abnormally exposed double-stranded DNA (dsDNA) in the cytoplasm binds to cGAS to produce cGAMP.<span><sup>2</sup></span> cGAMP binding by STING is activated, recruiting TANK-binding kinase to phosphorylate IRF3 leading to its nuclear translocation and the induction of type I interferons (IFNs), which then triggers the expression of interferon stimulated genes (ISGs) expression to enhance cellular autonomous defense mechanisms.<span><sup>3</sup></span> Furthermore, PD-L1 can undergo upregulation in antigen-presenting cells following IFN-γ stimulation, which leads to immune tolerance. This observation provides fresh perspectives on the significant role of SMARCAL1 in modulating IRF3-mediated immune stimulation and the expression levels of the immune checkpoint PD-L1.</p><p>SMARCAL1 is an ATP-dependent, SWI/SNF-related annealing helicase that stabilizes replication forks during DNA damage. Mutations in this gene are responsible for Schimke immune-osseous dysplasia, an autosomal recessive disorder characterized by T-cell immunodeficiency and growth dysfunctions.<span><sup>4</sup></span> In this study, SMARCAL1 operates on dual fronts. First, it dampens the cGAS–STING pathway, which is triggered by abnormal DNA in the cytoplasm. Chromosomal instability is a primary source of cytosolic dsDNA, a hallmark of cancer cells.<span><sup>2</sup></span> Second, it promotes PD-L1 expression, which shields tumors from T cell attack and allows cancer cells to enhance their viability and metastatic potential. SMARCAL1-deficient tumor cells show higher levels of IRF3 phosphorylation and reduced levels of PD-L1. This is consistent with the screening results, indicating that SMARCAL1 deficiency simultaneously induces IRF3 activation to promote the cGAS–STING pathway and downregulates PD-L1. This dual functionality renders SMARCAL1 an exceptional and potentially powerful candidate for immunotherapy.</p><p>Mechanistically, SMARCAL1 can facilitate the repair of collapsed DNA replication forks, alleviating the DNA replication stress caused by oncogenes.<span><sup>5</sup></span> Loss of SMARCAL1 induces genome instability and leads to micronuclei formation. Disruption of the micronuclear envelopes exposes dsDNA to the cytosol, leading to the activation of the cGAS–STING signaling pathway. Furthermore, SMARCAL1 does not directly mediate chromatin remodeling. Notably, SMARCAL1 upholds chromatin accessibility of P3 element located downstream of the PD-L1 transcription start site through its ATPase activity, and cooperates with the AP-1 family member JUN to efficiently promote PD-L1 expression. The research opens a new window to reveal how a single molecule plays a dual role in cancer cells, both hiding itself from the immune system's attack and facilitating the immune system's fight back under certain conditions. This discovery not only provides new insights into the complex mechanisms of cancer, but also opens up the possibility of developing more effective cancer treatment strategies.</p><p>In mouse models, SMARCAL1 deficiency significantly inhibited tumor growth and increased animal survival. Notably, SMARCAL1 is cGAS-dependent for antitumor immunity and enhances T cell-mediated antitumor competence. Due to the lack of accessible P3 regions in B16/F10 mouse cells, SMARCAL1-deficiency-mediated regulation of PD-L1 levels could not be assessed for antitumor effects in the study. Interestingly, PD-L1 downregulation induced by SMARCAL1 deletion in MDA-MB-436 cells made them more sensitive to CD8<sup>+</sup> T cell-mediated killing, which could be partially rescued by PD-L1 overexpression. Moreover, treatment of SMARCAL1-deficient B16/F10 tumors with anti-PD-L1 antibody or anti-CTLA-4 antibody significantly reduced tumor growth and improved animal survival. Combined treatment with both antibodies enhanced the effect. SMARCAL1 deficiency has a synergistic effect with immune checkpoint blockade treatment for antitumor immunity.</p><p>Immune checkpoint inhibitors (ICIs) are the mainstay of tumor immunotherapy, blocking cancer cell evasion mechanisms to enhance the immune system's attack on cancer cells. Nevertheless, inherent or acquired tumor resistance to ICIs and treatment-related toxicity limit their clinical application.<span><sup>6</sup></span> The dual mechanism of action of SMARCAL1 may be one of the key factors in enhancing the efficacy of ICIs. Targeting SMARCAL1 can synergize with ICIs to treat tumors, providing a novel and effective approach to tumor immunotherapy. Active DNA-dependent ATPase A domain inhibitor (ADAADi) targets the ATPase domain of SMARCAL1, which was identified as a potent inhibitor of ADAAD, the bovine homologue of SMARCAL1, but SMARCAL1 does not determine the cellular response to ADAADi.<span><sup>7</sup></span> Although there was no significant correlation between SMARCAL1 expression and cellular responsiveness to ADAADi, ADAADi provides a viable way to target inhibition of SMARCAL1 for tumor treatment.</p><p>Although intriguing, several questions persist. Initially, SMARCAL1-mediated regulation of PD-L1 in cancer patients could not be fully mimicked due to the study's heavy reliance on preclinical models, and the fact that SMARCAL1 does not affect the levels of PD-L1 in mouse cells. Further evaluation of SMARCAL1-mediated antitumor effects in mouse tumor models is needed. And it is required to conduct comprehensive research across various types of cancers. Second, minimizing off-target effects and ensuring safety using known and newly developed SMARCAL1 inhibitors is a demanding task. Finally, further exploration is needed to achieve synergistic treatments combining SMARCAL1 targeting with other immunotherapies to enhance clinical benefits.</p><p>Overall, the work of Leuzzi et al.<span><sup>1</sup></span> sheds light on a previously unknown mechanism of tumor immune evasion and highlights the potential of targeting SMARCAL1 as a novel therapeutic strategy (Figure 1B). Targeted inhibition of SMARCAL1 induces cancer innate immune signaling while downregulating PD-L1 levels, resulting in a double whammy for cancer and better cancer treatment outcomes. Further investigation is required to elucidate the precise mechanisms through which SMARCAL1 controls PD-L1 expression and its interactions with other gene targets. Additionally, exploring the potential of SMARCAL1 inhibitors or SMARCAL1-targeting therapies in combination with existing immunotherapies is crucial. This approach is important for translating these findings into clinical applications and propelling them toward the advancement of efficacious cancer therapeutics.</p><p><b>Ting Xiao</b>: drafted the manuscript. <b>Shiqi Li</b>: drew the figure. and <b>Xinghua Long</b>: drafted and reviewed the manuscript. All the authors read and approved the final manuscript.</p><p>The authors declare that they have no conflict of interest.</p><p>Not applicable.</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2024-09-07\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.730\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.730\",\"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/mco2.730","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

免疫检查点抑制剂(ICIs)是肿瘤免疫疗法的主要药物,可阻断癌细胞的逃避机制,增强免疫系统对癌细胞的攻击。6 SMARCAL1的双重作用机制可能是提高ICIs疗效的关键因素之一。靶向 SMARCAL1 可与 ICIs 协同治疗肿瘤,为肿瘤免疫疗法提供了一种新颖有效的方法。活性 DNA 依赖性 ATPase A 结构域抑制剂(ADAADi)靶向 SMARCAL1 的 ATPase 结构域,被确认为 SMARCAL1 牛同源物 ADAAD 的强效抑制剂,但 SMARCAL1 并不决定细胞对 ADAADi 的反应。虽然 SMARCAL1 的表达与细胞对 ADAADi 的反应之间没有明显的相关性,但 ADAADi 为靶向抑制 SMARCAL1 治疗肿瘤提供了一种可行的方法。最初,由于该研究严重依赖临床前模型,而且SMARCAL1不会影响小鼠细胞中PD-L1的水平,因此无法完全模拟SMARCAL1介导的对癌症患者PD-L1的调控。需要进一步评估 SMARCAL1 在小鼠肿瘤模型中介导的抗肿瘤作用。而且需要对各种类型的癌症进行全面研究。其次,使用已知和新开发的 SMARCAL1 抑制剂最大限度地减少脱靶效应并确保安全性是一项艰巨的任务。总之,Leuzzi 等人1 的研究揭示了一种之前未知的肿瘤免疫逃避机制,并强调了靶向 SMARCAL1 作为一种新型治疗策略的潜力(图 1B)。靶向抑制 SMARCAL1 可诱导癌症先天免疫信号转导,同时下调 PD-L1 水平,从而对癌症产生双重打击,改善癌症治疗效果。要阐明 SMARCAL1 控制 PD-L1 表达及其与其他基因靶点相互作用的确切机制,还需要进一步的研究。此外,探索 SMARCAL1 抑制剂或 SMARCAL1 靶向疗法与现有免疫疗法相结合的潜力也至关重要。这种方法对于将这些发现转化为临床应用并推动其向有效的癌症疗法发展非常重要。李世奇:绘制图表。龙兴华:起草并审阅手稿。所有作者阅读并批准了最终稿件。作者声明他们没有利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
SMARCAL1: a new target for taming tumor immune evasion

The viability and development of cancer cells depend on their ability to effectively evade the surveillance of the immune system. To achieve this, cancer cells alter the expression or function of specific molecules to become “invisible”. In a recent elegant study in Cell, Leuzzi et al.1 unveiled a novel player in the intricate game of tumor immune evasion: SMARCAL1, linked to DNA repair, emerges as a dual regulator with a surprising ability to modulate both innate immune signaling and PD-L1 expression. This work not only sheds light on the complex interplay between DNA damage response, innate immunity, and immune checkpoint regulation, but also identifies a potential new target for cancer immunotherapy.

Using a CRISPR–Cas9 screen, the authors identified SMARCAL1 as one of the sgRNA-targeted genes that are distinctly enriched in IRF3High/PD-L1Low cells (Figure 1A). Interferon regulatory factor 3 (IRF3) is a central signaling mediator of the innate immune signaling pathway, selected as a screening factor. In the innate immunity cGAS–STING pathway, abnormally exposed double-stranded DNA (dsDNA) in the cytoplasm binds to cGAS to produce cGAMP.2 cGAMP binding by STING is activated, recruiting TANK-binding kinase to phosphorylate IRF3 leading to its nuclear translocation and the induction of type I interferons (IFNs), which then triggers the expression of interferon stimulated genes (ISGs) expression to enhance cellular autonomous defense mechanisms.3 Furthermore, PD-L1 can undergo upregulation in antigen-presenting cells following IFN-γ stimulation, which leads to immune tolerance. This observation provides fresh perspectives on the significant role of SMARCAL1 in modulating IRF3-mediated immune stimulation and the expression levels of the immune checkpoint PD-L1.

SMARCAL1 is an ATP-dependent, SWI/SNF-related annealing helicase that stabilizes replication forks during DNA damage. Mutations in this gene are responsible for Schimke immune-osseous dysplasia, an autosomal recessive disorder characterized by T-cell immunodeficiency and growth dysfunctions.4 In this study, SMARCAL1 operates on dual fronts. First, it dampens the cGAS–STING pathway, which is triggered by abnormal DNA in the cytoplasm. Chromosomal instability is a primary source of cytosolic dsDNA, a hallmark of cancer cells.2 Second, it promotes PD-L1 expression, which shields tumors from T cell attack and allows cancer cells to enhance their viability and metastatic potential. SMARCAL1-deficient tumor cells show higher levels of IRF3 phosphorylation and reduced levels of PD-L1. This is consistent with the screening results, indicating that SMARCAL1 deficiency simultaneously induces IRF3 activation to promote the cGAS–STING pathway and downregulates PD-L1. This dual functionality renders SMARCAL1 an exceptional and potentially powerful candidate for immunotherapy.

Mechanistically, SMARCAL1 can facilitate the repair of collapsed DNA replication forks, alleviating the DNA replication stress caused by oncogenes.5 Loss of SMARCAL1 induces genome instability and leads to micronuclei formation. Disruption of the micronuclear envelopes exposes dsDNA to the cytosol, leading to the activation of the cGAS–STING signaling pathway. Furthermore, SMARCAL1 does not directly mediate chromatin remodeling. Notably, SMARCAL1 upholds chromatin accessibility of P3 element located downstream of the PD-L1 transcription start site through its ATPase activity, and cooperates with the AP-1 family member JUN to efficiently promote PD-L1 expression. The research opens a new window to reveal how a single molecule plays a dual role in cancer cells, both hiding itself from the immune system's attack and facilitating the immune system's fight back under certain conditions. This discovery not only provides new insights into the complex mechanisms of cancer, but also opens up the possibility of developing more effective cancer treatment strategies.

In mouse models, SMARCAL1 deficiency significantly inhibited tumor growth and increased animal survival. Notably, SMARCAL1 is cGAS-dependent for antitumor immunity and enhances T cell-mediated antitumor competence. Due to the lack of accessible P3 regions in B16/F10 mouse cells, SMARCAL1-deficiency-mediated regulation of PD-L1 levels could not be assessed for antitumor effects in the study. Interestingly, PD-L1 downregulation induced by SMARCAL1 deletion in MDA-MB-436 cells made them more sensitive to CD8+ T cell-mediated killing, which could be partially rescued by PD-L1 overexpression. Moreover, treatment of SMARCAL1-deficient B16/F10 tumors with anti-PD-L1 antibody or anti-CTLA-4 antibody significantly reduced tumor growth and improved animal survival. Combined treatment with both antibodies enhanced the effect. SMARCAL1 deficiency has a synergistic effect with immune checkpoint blockade treatment for antitumor immunity.

Immune checkpoint inhibitors (ICIs) are the mainstay of tumor immunotherapy, blocking cancer cell evasion mechanisms to enhance the immune system's attack on cancer cells. Nevertheless, inherent or acquired tumor resistance to ICIs and treatment-related toxicity limit their clinical application.6 The dual mechanism of action of SMARCAL1 may be one of the key factors in enhancing the efficacy of ICIs. Targeting SMARCAL1 can synergize with ICIs to treat tumors, providing a novel and effective approach to tumor immunotherapy. Active DNA-dependent ATPase A domain inhibitor (ADAADi) targets the ATPase domain of SMARCAL1, which was identified as a potent inhibitor of ADAAD, the bovine homologue of SMARCAL1, but SMARCAL1 does not determine the cellular response to ADAADi.7 Although there was no significant correlation between SMARCAL1 expression and cellular responsiveness to ADAADi, ADAADi provides a viable way to target inhibition of SMARCAL1 for tumor treatment.

Although intriguing, several questions persist. Initially, SMARCAL1-mediated regulation of PD-L1 in cancer patients could not be fully mimicked due to the study's heavy reliance on preclinical models, and the fact that SMARCAL1 does not affect the levels of PD-L1 in mouse cells. Further evaluation of SMARCAL1-mediated antitumor effects in mouse tumor models is needed. And it is required to conduct comprehensive research across various types of cancers. Second, minimizing off-target effects and ensuring safety using known and newly developed SMARCAL1 inhibitors is a demanding task. Finally, further exploration is needed to achieve synergistic treatments combining SMARCAL1 targeting with other immunotherapies to enhance clinical benefits.

Overall, the work of Leuzzi et al.1 sheds light on a previously unknown mechanism of tumor immune evasion and highlights the potential of targeting SMARCAL1 as a novel therapeutic strategy (Figure 1B). Targeted inhibition of SMARCAL1 induces cancer innate immune signaling while downregulating PD-L1 levels, resulting in a double whammy for cancer and better cancer treatment outcomes. Further investigation is required to elucidate the precise mechanisms through which SMARCAL1 controls PD-L1 expression and its interactions with other gene targets. Additionally, exploring the potential of SMARCAL1 inhibitors or SMARCAL1-targeting therapies in combination with existing immunotherapies is crucial. This approach is important for translating these findings into clinical applications and propelling them toward the advancement of efficacious cancer therapeutics.

Ting Xiao: drafted the manuscript. Shiqi Li: drew the figure. and Xinghua Long: drafted and reviewed the manuscript. All the authors read and approved the final manuscript.

The authors declare that they have no conflict 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
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
期刊最新文献
Copper homeostasis and cuproptosis in health and disease Liver cirrhosis: molecular mechanisms and therapeutic interventions High serum magnesium level is associated with increased mortality in patients with sepsis: an international, multicenter retrospective study N1-methylpseudouridine modification level correlates with protein expression, immunogenicity, and stability of mRNA Issue Information
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1