溶酶体靶向嵌合体(LYTAC):靶向降解致癌膜蛋白的银弹

Qingquan Zheng, Jiawei Guo, Rui Ma, Wenchen Pu
{"title":"溶酶体靶向嵌合体(LYTAC):靶向降解致癌膜蛋白的银弹","authors":"Qingquan Zheng,&nbsp;Jiawei Guo,&nbsp;Rui Ma,&nbsp;Wenchen Pu","doi":"10.1002/mog2.64","DOIUrl":null,"url":null,"abstract":"<p>Recently, the group of Prof. Carolyn Bertozzi, a laureate of the Nobel Prize in chemistry 2022, reported the detailed mechanism of lysosome-targeting chimera (LYTAC) in the journal of <i>Science</i>,<span><sup>1</sup></span> after the publication of their first LYTAC molecule in <i>Nature</i> in 2020.<span><sup>2</sup></span> The establishment of LYTAC, a subtype of targeted protein degradation technology, expands the scope of protein degradation to extracellular and membrane-associated targets, and Bertozzi group's new discovery is expected to accelerate the development of LYTAC in cancer therapy.</p><p>Cell membranes play a critical role in various cellular processes, including signaling transduction, cell adhesion, transport of biomolecules and immunity. Proteins embedded in or associated with the cell membrane are key executants of the function of cell membrane, and their dysregulation contributes to tumorigenesis and development of human cancers.<span><sup>3</sup></span> For example, epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase for epithelial growth factor (EGF) and transforming growth factor α (TGF-α), belonging to the ErbB receptor family. Activation of EGFR signaling promotes cell proliferation, survival, angiogenesis and metastasis of diverse malignancies.<span><sup>3</sup></span> Moreover, hepatocyte growth factor receptor (c-Met, HGFR) is another oncogenic receptor tyrosine kinase in diverse cancers. Upon the binding to hepatocyte growth factor (HGF), c-Met is activated via autophosphorylation, leading to the initiation of oncogenic downstream signaling cascades, such as PI3K/AKT and RAS/ERK pathways.<span><sup>3</sup></span> Given their central role in cancer-promoting processes, EGFR and c-Met has become attractive targets for cancer therapies. Small-molecule tyrosine kinase inhibitors (EGFR: gefitinib, afatinib, osimertinib, etc.; c-Met: capmatinib, tepotinib, savolitinib, etc.) and monoclonal antibodies (EGFR: cetuximab, panitumumab; EGFR/c-Met: amivantamab; c-Met: emibetuzumab), have been developed and approved for the treatment of various cancers, including lung and colorectal cancers (Figure 1A). But severe acquired resistance (e.g., via EGFR mutations) and limited therapeutic efficacy (slightly prolonged overall survival) of these treatments restrict their clinical benefit for patients. Moreover, nonenzymatic function of membrane proteins, such as protein–protein interactions, could not be interfered with by kinase inhibitors or monoclonal antibodies, calling for new strategies to control these oncogenic membrane proteins.</p><p>Targeted protein degradation (TPD) is a therapeutic approach that aims to selectively remove disease-causing or undesirable proteins from cells by inducing their degradation, with multiple therapies entering clinical trials and targeting proteins that are previously considered “undruggable.”<span><sup>4</sup></span> There are two main protein degradation mechanisms within cells, including ubiquitin-proteasome system (UPS) and lysosome pathway. By triggering chemically induced proximity to form ternary complex assembly via heterobifunctional molecules, various TPD techniques have been developed based on proteasome (PROTAC, molecular glue, etc.), lysosome (LYTAC, AUTAC, ATTEC, etc.), or both (PROTAB).<span><sup>4</sup></span> Currently, some membrane proteins could be degraded through TPD technologies. For example, Jang et al. developed an allosteric EGFR proteolysis-targeting chimera (PROTAC), DDC-01-163, which with selective activity against various clinically relevant EGFR mutants (L858R/T790M) as a single agent or combined with an ATP-site inhibitor osimertinib (Figure 1B, left).<span><sup>5</sup></span> Recently, Marei et al. reported proteolysis-targeting antibodies (PROTABs, a class of bispecific antibodies) that tethered cell-surface E3 ubiquitin ligases (RNF43 or ZNRF3) to transmembrane proteins (IGF1R, HER2, and PD-L1) for targeted degradation via both UPS and lysosome pathway, providing a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of membrane proteins.<span><sup>6</sup></span></p><p>In 2020, Bertozzi group prepared bifunctional molecules that consisted of antibodies conjugated to chemically synthesized glycopeptide ligands, the agonists of the cation-independent mannose-6-phosphate receptor (CI-M6PR, a cell-surface lysosome-shuttling receptor). These conjugates recognized both CI-M6PR and the extracellular domain of target proteins, inducing the lysosome-mediated targeted degradation of membrane proteins, such as EGFR, CD71, programmed death-ligand 1 (PD-L1) and apolipoprotein E4. Notably, the data from CRISPR interference screen suggested an involvement of CI-M6PR-mediated cargo internalization in cell lines, and uncovered the participation of exocyst complex in these processes. Thus, these evidence demonstrated the feasibility of degrading membrane proteins through lysosomes by activating chemically induced proximity. Thus, this technique was termed as lysosome-targeting chimaeras (LYTACs).<span><sup>2</sup></span> However, the cellular characteristics that regulate the behavior of LYTACs to hijack lysosome machinery for membrane protein degradation are largely unknown. There is an urgent need for identifying the cellular determinants that modulate the efficacy of LYTACs-induced lysosomal degradation, facilitating the understanding of their molecular and cellular mechanisms.</p><p>To this end, Bertozzi group subsequently performed an unbiased genome-wide CRISPR knockout screening approach complemented by proteomics to map the key regulators of LYTAC-mediated membrane protein degradation in human cells. The results indicated that inhibiting retromer genes (e.g., <i>VPS35</i>, <i>SNX3</i>, <i>VPS29</i>, and <i>VPS26A</i>) to reduce LYTAC recycling enhanced the target degradation. Moreover, genes that involved in cullin3 (CUL3) neddylation, such as <i>CUL3</i>, <i>UBA3</i>, and <i>CAND1</i>, promoted the E3 ligase activity as well as the transport of LYTAC-target protein complexes to lysosomes. Thereby, levels of neddylated CUL3 could act as a predictive marker for LYTAC efficacy. Additionally, membrane CI-M6PR receptors were partially engaged by endogenous mannose 6-phosphate (M6P)-modified lysosomal glycoproteins. Blockage of M6P biosynthesis genes (e.g., <i>ALGO12</i>, <i>GNPTAB</i>) upregulated the ratio of unoccupied receptors, increasing LYTAC-receptor internalization and the degradation of cell surface proteins, including EGFR and c-Met.<span><sup>1</sup></span> Overall, this work discovered a series of critical cellular regulators that modulated LYTAC-mediated degradation of EGFR and c-Met (Figure 1B, right), giving important support for understanding LYTAC mechanism and developing next-generation LYTAC with improved clinical potential.</p><p>With the development and maturity of monoclonal antibody technology based on phage display, hybridoma cell and single B cell techniques, discovery and optimization of specific monoclonal antibodies is highly efficient. Furthermore, the synthesis and characterization of LYTAC could follow the route of antibody-drug conjugates (ADCs), because they share similar chemical compositions. Therefore, LYTAC is highly promising as a general platform for targeting extracellular and membrane proteins, the products of 40% of all protein-encoding genes. However, for targeted therapy of cancers, the following issues still need to be considered: Comprehensive in vivo evidence is needed to support the pharmacodynamics, pharmacokinetics and safety of LYTAC as a treatment option; Cooperative diagnosis is necessary for characterizing target protein, predictive biomarkers and LYTAC-related regulators to confirm suitable cancer patients, and for determining whether combination therapy is needed, such as in combination with the inhibitors of <i>ALGO12</i> or <i>GNPTAB</i>; Mutations of many oncogenic membrane proteins lead to tumor heterogeneity and/or drug resistance, and it is still uncertain whether LYTAC could overcome or bypass the negative effect of protein mutations. Since 2020, companies, such as Lycia Therapeutics (founded by Prof. Bertozzi) and Avilar Therapeutics, have undergone translational research around LYTAC technology. We look forward to these new discoveries driving LYTAC into novel therapies that benefit patients.</p><p>All authors were involved in the writing of the manuscript. Qingquan Zheng and Wenchen Pu initiated the conception and outline. Qingquan Zheng, Jiawei Guo, and Wenchen Pu organized and processed the figure. Jiawei Guo and Rui Ma revised manuscript. Jiawei Guo and Wenchen Pu were involved in study supervision. All authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>The authors declare that human ethics approval was not needed for this highlight.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0000,"publicationDate":"2024-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.64","citationCount":"0","resultStr":"{\"title\":\"Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins\",\"authors\":\"Qingquan Zheng,&nbsp;Jiawei Guo,&nbsp;Rui Ma,&nbsp;Wenchen Pu\",\"doi\":\"10.1002/mog2.64\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Recently, the group of Prof. Carolyn Bertozzi, a laureate of the Nobel Prize in chemistry 2022, reported the detailed mechanism of lysosome-targeting chimera (LYTAC) in the journal of <i>Science</i>,<span><sup>1</sup></span> after the publication of their first LYTAC molecule in <i>Nature</i> in 2020.<span><sup>2</sup></span> The establishment of LYTAC, a subtype of targeted protein degradation technology, expands the scope of protein degradation to extracellular and membrane-associated targets, and Bertozzi group's new discovery is expected to accelerate the development of LYTAC in cancer therapy.</p><p>Cell membranes play a critical role in various cellular processes, including signaling transduction, cell adhesion, transport of biomolecules and immunity. Proteins embedded in or associated with the cell membrane are key executants of the function of cell membrane, and their dysregulation contributes to tumorigenesis and development of human cancers.<span><sup>3</sup></span> For example, epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase for epithelial growth factor (EGF) and transforming growth factor α (TGF-α), belonging to the ErbB receptor family. Activation of EGFR signaling promotes cell proliferation, survival, angiogenesis and metastasis of diverse malignancies.<span><sup>3</sup></span> Moreover, hepatocyte growth factor receptor (c-Met, HGFR) is another oncogenic receptor tyrosine kinase in diverse cancers. Upon the binding to hepatocyte growth factor (HGF), c-Met is activated via autophosphorylation, leading to the initiation of oncogenic downstream signaling cascades, such as PI3K/AKT and RAS/ERK pathways.<span><sup>3</sup></span> Given their central role in cancer-promoting processes, EGFR and c-Met has become attractive targets for cancer therapies. Small-molecule tyrosine kinase inhibitors (EGFR: gefitinib, afatinib, osimertinib, etc.; c-Met: capmatinib, tepotinib, savolitinib, etc.) and monoclonal antibodies (EGFR: cetuximab, panitumumab; EGFR/c-Met: amivantamab; c-Met: emibetuzumab), have been developed and approved for the treatment of various cancers, including lung and colorectal cancers (Figure 1A). But severe acquired resistance (e.g., via EGFR mutations) and limited therapeutic efficacy (slightly prolonged overall survival) of these treatments restrict their clinical benefit for patients. Moreover, nonenzymatic function of membrane proteins, such as protein–protein interactions, could not be interfered with by kinase inhibitors or monoclonal antibodies, calling for new strategies to control these oncogenic membrane proteins.</p><p>Targeted protein degradation (TPD) is a therapeutic approach that aims to selectively remove disease-causing or undesirable proteins from cells by inducing their degradation, with multiple therapies entering clinical trials and targeting proteins that are previously considered “undruggable.”<span><sup>4</sup></span> There are two main protein degradation mechanisms within cells, including ubiquitin-proteasome system (UPS) and lysosome pathway. By triggering chemically induced proximity to form ternary complex assembly via heterobifunctional molecules, various TPD techniques have been developed based on proteasome (PROTAC, molecular glue, etc.), lysosome (LYTAC, AUTAC, ATTEC, etc.), or both (PROTAB).<span><sup>4</sup></span> Currently, some membrane proteins could be degraded through TPD technologies. For example, Jang et al. developed an allosteric EGFR proteolysis-targeting chimera (PROTAC), DDC-01-163, which with selective activity against various clinically relevant EGFR mutants (L858R/T790M) as a single agent or combined with an ATP-site inhibitor osimertinib (Figure 1B, left).<span><sup>5</sup></span> Recently, Marei et al. reported proteolysis-targeting antibodies (PROTABs, a class of bispecific antibodies) that tethered cell-surface E3 ubiquitin ligases (RNF43 or ZNRF3) to transmembrane proteins (IGF1R, HER2, and PD-L1) for targeted degradation via both UPS and lysosome pathway, providing a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of membrane proteins.<span><sup>6</sup></span></p><p>In 2020, Bertozzi group prepared bifunctional molecules that consisted of antibodies conjugated to chemically synthesized glycopeptide ligands, the agonists of the cation-independent mannose-6-phosphate receptor (CI-M6PR, a cell-surface lysosome-shuttling receptor). These conjugates recognized both CI-M6PR and the extracellular domain of target proteins, inducing the lysosome-mediated targeted degradation of membrane proteins, such as EGFR, CD71, programmed death-ligand 1 (PD-L1) and apolipoprotein E4. Notably, the data from CRISPR interference screen suggested an involvement of CI-M6PR-mediated cargo internalization in cell lines, and uncovered the participation of exocyst complex in these processes. Thus, these evidence demonstrated the feasibility of degrading membrane proteins through lysosomes by activating chemically induced proximity. Thus, this technique was termed as lysosome-targeting chimaeras (LYTACs).<span><sup>2</sup></span> However, the cellular characteristics that regulate the behavior of LYTACs to hijack lysosome machinery for membrane protein degradation are largely unknown. There is an urgent need for identifying the cellular determinants that modulate the efficacy of LYTACs-induced lysosomal degradation, facilitating the understanding of their molecular and cellular mechanisms.</p><p>To this end, Bertozzi group subsequently performed an unbiased genome-wide CRISPR knockout screening approach complemented by proteomics to map the key regulators of LYTAC-mediated membrane protein degradation in human cells. The results indicated that inhibiting retromer genes (e.g., <i>VPS35</i>, <i>SNX3</i>, <i>VPS29</i>, and <i>VPS26A</i>) to reduce LYTAC recycling enhanced the target degradation. Moreover, genes that involved in cullin3 (CUL3) neddylation, such as <i>CUL3</i>, <i>UBA3</i>, and <i>CAND1</i>, promoted the E3 ligase activity as well as the transport of LYTAC-target protein complexes to lysosomes. Thereby, levels of neddylated CUL3 could act as a predictive marker for LYTAC efficacy. Additionally, membrane CI-M6PR receptors were partially engaged by endogenous mannose 6-phosphate (M6P)-modified lysosomal glycoproteins. Blockage of M6P biosynthesis genes (e.g., <i>ALGO12</i>, <i>GNPTAB</i>) upregulated the ratio of unoccupied receptors, increasing LYTAC-receptor internalization and the degradation of cell surface proteins, including EGFR and c-Met.<span><sup>1</sup></span> Overall, this work discovered a series of critical cellular regulators that modulated LYTAC-mediated degradation of EGFR and c-Met (Figure 1B, right), giving important support for understanding LYTAC mechanism and developing next-generation LYTAC with improved clinical potential.</p><p>With the development and maturity of monoclonal antibody technology based on phage display, hybridoma cell and single B cell techniques, discovery and optimization of specific monoclonal antibodies is highly efficient. Furthermore, the synthesis and characterization of LYTAC could follow the route of antibody-drug conjugates (ADCs), because they share similar chemical compositions. Therefore, LYTAC is highly promising as a general platform for targeting extracellular and membrane proteins, the products of 40% of all protein-encoding genes. However, for targeted therapy of cancers, the following issues still need to be considered: Comprehensive in vivo evidence is needed to support the pharmacodynamics, pharmacokinetics and safety of LYTAC as a treatment option; Cooperative diagnosis is necessary for characterizing target protein, predictive biomarkers and LYTAC-related regulators to confirm suitable cancer patients, and for determining whether combination therapy is needed, such as in combination with the inhibitors of <i>ALGO12</i> or <i>GNPTAB</i>; Mutations of many oncogenic membrane proteins lead to tumor heterogeneity and/or drug resistance, and it is still uncertain whether LYTAC could overcome or bypass the negative effect of protein mutations. Since 2020, companies, such as Lycia Therapeutics (founded by Prof. Bertozzi) and Avilar Therapeutics, have undergone translational research around LYTAC technology. We look forward to these new discoveries driving LYTAC into novel therapies that benefit patients.</p><p>All authors were involved in the writing of the manuscript. Qingquan Zheng and Wenchen Pu initiated the conception and outline. Qingquan Zheng, Jiawei Guo, and Wenchen Pu organized and processed the figure. Jiawei Guo and Rui Ma revised manuscript. Jiawei Guo and Wenchen Pu were involved in study supervision. All authors have read and approved the final manuscript.</p><p>The authors declare no conflict of interest.</p><p>The authors declare that human ethics approval was not needed for this highlight.</p>\",\"PeriodicalId\":100902,\"journal\":{\"name\":\"MedComm – Oncology\",\"volume\":\"3 1\",\"pages\":\"\"},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-01-14\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.64\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm – Oncology\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mog2.64\",\"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 – Oncology","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mog2.64","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,继 2020 年在《自然》(Nature)杂志上发表首个 LYTAC 分子之后,2022 年诺贝尔化学奖得主卡罗琳-贝托齐(Carolyn Bertozzi)教授的研究小组又在《科学》(Science)杂志1 上报告了溶酶体靶向嵌合体(LYTAC)的详细机制。2 LYTAC 是靶向蛋白质降解技术的一个亚型,它的建立将蛋白质降解的范围扩大到细胞外和与膜相关的靶点,Bertozzi 小组的新发现有望加速 LYTAC 在癌症治疗中的发展。细胞膜在信号转导、细胞粘附、生物大分子运输和免疫等各种细胞过程中发挥着关键作用。3 例如,表皮生长因子受体(EGFR)是上皮生长因子(EGF)和转化生长因子α(TGF-α)的受体酪氨酸激酶,属于 ErbB 受体家族。表皮生长因子受体信号激活可促进多种恶性肿瘤的细胞增殖、存活、血管生成和转移。3 此外,肝细胞生长因子受体(c-Met,HGFR)也是多种癌症中的另一种致癌受体酪氨酸激酶。与肝细胞生长因子(HGF)结合后,c-Met 通过自身磷酸化被激活,导致致癌下游信号级联的启动,如 PI3K/AKT 和 RAS/ERK 通路。小分子酪氨酸激酶抑制剂(表皮生长因子受体:吉非替尼、阿法替尼、奥西莫替尼等;c-Met:卡帕替尼、替泊替尼、沙夫利替尼等)和单克隆抗体(表皮生长因子受体:西妥昔单抗、帕尼妥单抗;表皮生长因子受体/c-Met:阿米万坦单抗;c-Met:埃米特珠单抗)已被开发并批准用于治疗各种癌症,包括肺癌和结直肠癌(图 1A)。但是,这些疗法严重的获得性耐药性(如通过表皮生长因子受体突变)和有限的疗效(略微延长总生存期)限制了它们对患者的临床益处。此外,激酶抑制剂或单克隆抗体无法干扰膜蛋白的非酶功能,如蛋白质与蛋白质之间的相互作用,这就要求采用新策略来控制这些致癌膜蛋白。靶向蛋白降解(TPD)是一种治疗方法,旨在通过诱导细胞内致病蛋白或不良蛋白的降解,有选择性地将其从细胞中清除,目前已有多种疗法进入临床试验阶段,其靶向蛋白都是以前被认为 "不可药用 "的蛋白。通过异功能分子触发化学诱导接近形成三元复合物组装,目前已开发出各种基于蛋白酶体(PROTAC、分子胶等)、溶酶体(LYTAC、AUTAC、ATTEC 等)或两者(PROTAB)的 TPD 技术。例如,Jang 等人开发了一种异位表皮生长因子受体蛋白水解靶向嵌合体(PROTAC)--DDC-01-163,该嵌合体对各种临床相关的表皮生长因子受体突变体(L858R/T790M)具有选择性活性,可作为单药或与 ATP 位点抑制剂奥希替尼联合使用(图 1B 左)。5 最近,Marei 等人报道了蛋白水解靶向抗体(PROTABs,一类双特异性抗体),该抗体将细胞表面 E3 泛素连接酶(RNF43 或 ZNRF3)与跨膜蛋白(IGF1R、HER2 和 PD-L1)拴在一起,通过 UPS 和溶酶体途径进行靶向降解,为快速开发强效、生物可用且具有组织选择性的膜蛋白降解剂提供了一种策略。2020 年,Bertozzi 小组制备了双功能分子,其中包括与化学合成的糖肽配体共轭的抗体,糖肽配体是不依赖阳离子的 6-磷酸甘露糖受体(CI-M6PR,一种细胞表面溶酶体关闭受体)的激动剂。这些共轭物能识别 CI-M6PR 和靶蛋白的胞外结构域,诱导溶酶体介导的膜蛋白靶向降解,如表皮生长因子受体、CD71、程序性死亡配体 1 (PD-L1) 和脂蛋白 E4。值得注意的是,CRISPR 干扰筛选的数据表明,在细胞系中,CI-M6PR 介导的货物内化参与其中,并发现外囊复合体参与了这些过程。因此,这些证据证明了通过激活化学诱导的接近性,通过溶酶体降解膜蛋白的可行性。因此,这种技术被称为溶酶体靶向嵌合体(LYTACs)。 2 然而,调控 LYTACs 劫持溶酶体机制降解膜蛋白行为的细胞特征在很大程度上尚属未知。为此,Bertozzi 小组随后采用无偏见的全基因组 CRISPR 基因敲除筛选方法,并辅以蛋白质组学研究,绘制了人体细胞中 LYTAC 介导的膜蛋白降解的关键调控因子图。结果表明,抑制 retromer 基因(如 VPS35、SNX3、VPS29 和 VPS26A)以减少 LYTAC 的循环,可增强目标降解。此外,参与cullin3 (CUL3)内切酶化的基因,如CUL3、UBA3和CAND1,促进了E3连接酶的活性以及LYTAC-靶蛋白复合物向溶酶体的转运。因此,Neddylated CUL3 的水平可作为 LYTAC 疗效的预测指标。此外,膜CI-M6PR受体部分被内源性6-磷酸甘露糖(M6P)修饰的溶酶体糖蛋白啮合。阻断 M6P 生物合成基因(如 ALGO12、GNPTAB)可提高未被占用受体的比例,增加 LYTAC 受体的内化和细胞表面蛋白(包括表皮生长因子受体和 c-Met)的降解。随着基于噬菌体展示、杂交瘤细胞和单 B 细胞技术的单克隆抗体技术的发展和成熟,特异性单克隆抗体的发现和优化变得非常高效。此外,LYTAC 的合成和表征也可以遵循抗体药物共轭物(ADC)的路线,因为它们具有相似的化学成分。因此,LYTAC 很有希望成为靶向细胞外蛋白和膜蛋白的通用平台,因为所有蛋白编码基因中有 40% 是细胞外蛋白和膜蛋白的产物。然而,对于癌症的靶向治疗,还需要考虑以下问题:需要全面的体内证据来支持 LYTAC 作为一种治疗选择的药效学、药代动力学和安全性;需要合作诊断来确定靶蛋白、预测性生物标志物和 LYTAC 相关调节剂的特征,以确认合适的癌症患者,并确定是否需要联合治疗,例如与 ALGO12 或 GNPTAB 抑制剂联合治疗;许多致癌膜蛋白的突变会导致肿瘤的异质性和/或耐药性,LYTAC 能否克服或绕过蛋白突变的负面影响尚不确定。自 2020 年以来,Lycia Therapeutics(由 Bertozzi 教授创立)和 Avilar Therapeutics 等公司围绕 LYTAC 技术开展了转化研究。我们期待这些新发现能推动LYTAC成为造福患者的新型疗法。郑清泉和蒲文臣提出了构思和大纲。郑清泉、郭嘉伟和蒲文臣组织并处理了图表。郭嘉伟和马锐修改了稿件。郭嘉伟和蒲文臣参与研究督导。所有作者均已阅读并批准最终稿件。作者声明无利益冲突。作者声明本亮点研究无需人类伦理批准。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins

Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins

Recently, the group of Prof. Carolyn Bertozzi, a laureate of the Nobel Prize in chemistry 2022, reported the detailed mechanism of lysosome-targeting chimera (LYTAC) in the journal of Science,1 after the publication of their first LYTAC molecule in Nature in 2020.2 The establishment of LYTAC, a subtype of targeted protein degradation technology, expands the scope of protein degradation to extracellular and membrane-associated targets, and Bertozzi group's new discovery is expected to accelerate the development of LYTAC in cancer therapy.

Cell membranes play a critical role in various cellular processes, including signaling transduction, cell adhesion, transport of biomolecules and immunity. Proteins embedded in or associated with the cell membrane are key executants of the function of cell membrane, and their dysregulation contributes to tumorigenesis and development of human cancers.3 For example, epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase for epithelial growth factor (EGF) and transforming growth factor α (TGF-α), belonging to the ErbB receptor family. Activation of EGFR signaling promotes cell proliferation, survival, angiogenesis and metastasis of diverse malignancies.3 Moreover, hepatocyte growth factor receptor (c-Met, HGFR) is another oncogenic receptor tyrosine kinase in diverse cancers. Upon the binding to hepatocyte growth factor (HGF), c-Met is activated via autophosphorylation, leading to the initiation of oncogenic downstream signaling cascades, such as PI3K/AKT and RAS/ERK pathways.3 Given their central role in cancer-promoting processes, EGFR and c-Met has become attractive targets for cancer therapies. Small-molecule tyrosine kinase inhibitors (EGFR: gefitinib, afatinib, osimertinib, etc.; c-Met: capmatinib, tepotinib, savolitinib, etc.) and monoclonal antibodies (EGFR: cetuximab, panitumumab; EGFR/c-Met: amivantamab; c-Met: emibetuzumab), have been developed and approved for the treatment of various cancers, including lung and colorectal cancers (Figure 1A). But severe acquired resistance (e.g., via EGFR mutations) and limited therapeutic efficacy (slightly prolonged overall survival) of these treatments restrict their clinical benefit for patients. Moreover, nonenzymatic function of membrane proteins, such as protein–protein interactions, could not be interfered with by kinase inhibitors or monoclonal antibodies, calling for new strategies to control these oncogenic membrane proteins.

Targeted protein degradation (TPD) is a therapeutic approach that aims to selectively remove disease-causing or undesirable proteins from cells by inducing their degradation, with multiple therapies entering clinical trials and targeting proteins that are previously considered “undruggable.”4 There are two main protein degradation mechanisms within cells, including ubiquitin-proteasome system (UPS) and lysosome pathway. By triggering chemically induced proximity to form ternary complex assembly via heterobifunctional molecules, various TPD techniques have been developed based on proteasome (PROTAC, molecular glue, etc.), lysosome (LYTAC, AUTAC, ATTEC, etc.), or both (PROTAB).4 Currently, some membrane proteins could be degraded through TPD technologies. For example, Jang et al. developed an allosteric EGFR proteolysis-targeting chimera (PROTAC), DDC-01-163, which with selective activity against various clinically relevant EGFR mutants (L858R/T790M) as a single agent or combined with an ATP-site inhibitor osimertinib (Figure 1B, left).5 Recently, Marei et al. reported proteolysis-targeting antibodies (PROTABs, a class of bispecific antibodies) that tethered cell-surface E3 ubiquitin ligases (RNF43 or ZNRF3) to transmembrane proteins (IGF1R, HER2, and PD-L1) for targeted degradation via both UPS and lysosome pathway, providing a strategy for the rapid development of potent, bioavailable and tissue-selective degraders of membrane proteins.6

In 2020, Bertozzi group prepared bifunctional molecules that consisted of antibodies conjugated to chemically synthesized glycopeptide ligands, the agonists of the cation-independent mannose-6-phosphate receptor (CI-M6PR, a cell-surface lysosome-shuttling receptor). These conjugates recognized both CI-M6PR and the extracellular domain of target proteins, inducing the lysosome-mediated targeted degradation of membrane proteins, such as EGFR, CD71, programmed death-ligand 1 (PD-L1) and apolipoprotein E4. Notably, the data from CRISPR interference screen suggested an involvement of CI-M6PR-mediated cargo internalization in cell lines, and uncovered the participation of exocyst complex in these processes. Thus, these evidence demonstrated the feasibility of degrading membrane proteins through lysosomes by activating chemically induced proximity. Thus, this technique was termed as lysosome-targeting chimaeras (LYTACs).2 However, the cellular characteristics that regulate the behavior of LYTACs to hijack lysosome machinery for membrane protein degradation are largely unknown. There is an urgent need for identifying the cellular determinants that modulate the efficacy of LYTACs-induced lysosomal degradation, facilitating the understanding of their molecular and cellular mechanisms.

To this end, Bertozzi group subsequently performed an unbiased genome-wide CRISPR knockout screening approach complemented by proteomics to map the key regulators of LYTAC-mediated membrane protein degradation in human cells. The results indicated that inhibiting retromer genes (e.g., VPS35, SNX3, VPS29, and VPS26A) to reduce LYTAC recycling enhanced the target degradation. Moreover, genes that involved in cullin3 (CUL3) neddylation, such as CUL3, UBA3, and CAND1, promoted the E3 ligase activity as well as the transport of LYTAC-target protein complexes to lysosomes. Thereby, levels of neddylated CUL3 could act as a predictive marker for LYTAC efficacy. Additionally, membrane CI-M6PR receptors were partially engaged by endogenous mannose 6-phosphate (M6P)-modified lysosomal glycoproteins. Blockage of M6P biosynthesis genes (e.g., ALGO12, GNPTAB) upregulated the ratio of unoccupied receptors, increasing LYTAC-receptor internalization and the degradation of cell surface proteins, including EGFR and c-Met.1 Overall, this work discovered a series of critical cellular regulators that modulated LYTAC-mediated degradation of EGFR and c-Met (Figure 1B, right), giving important support for understanding LYTAC mechanism and developing next-generation LYTAC with improved clinical potential.

With the development and maturity of monoclonal antibody technology based on phage display, hybridoma cell and single B cell techniques, discovery and optimization of specific monoclonal antibodies is highly efficient. Furthermore, the synthesis and characterization of LYTAC could follow the route of antibody-drug conjugates (ADCs), because they share similar chemical compositions. Therefore, LYTAC is highly promising as a general platform for targeting extracellular and membrane proteins, the products of 40% of all protein-encoding genes. However, for targeted therapy of cancers, the following issues still need to be considered: Comprehensive in vivo evidence is needed to support the pharmacodynamics, pharmacokinetics and safety of LYTAC as a treatment option; Cooperative diagnosis is necessary for characterizing target protein, predictive biomarkers and LYTAC-related regulators to confirm suitable cancer patients, and for determining whether combination therapy is needed, such as in combination with the inhibitors of ALGO12 or GNPTAB; Mutations of many oncogenic membrane proteins lead to tumor heterogeneity and/or drug resistance, and it is still uncertain whether LYTAC could overcome or bypass the negative effect of protein mutations. Since 2020, companies, such as Lycia Therapeutics (founded by Prof. Bertozzi) and Avilar Therapeutics, have undergone translational research around LYTAC technology. We look forward to these new discoveries driving LYTAC into novel therapies that benefit patients.

All authors were involved in the writing of the manuscript. Qingquan Zheng and Wenchen Pu initiated the conception and outline. Qingquan Zheng, Jiawei Guo, and Wenchen Pu organized and processed the figure. Jiawei Guo and Rui Ma revised manuscript. Jiawei Guo and Wenchen Pu were involved in study supervision. All authors have read and approved the final manuscript.

The authors declare no conflict of interest.

The authors declare that human ethics approval was not needed for this highlight.

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
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