The mechanism of resistance to CDK4/6 inhibition and novel combination therapy with RNR inhibition for chemo-resistant bladder cancer

IF 20.1 1区 医学 Q1 ONCOLOGY Cancer Communications Pub Date : 2024-03-11 DOI:10.1002/cac2.12532
Zhichao Tong, Yubo Zhao, Shiyu Bai, Benedikt Ebner, Lou Lienhard, Yuling Zhao, Ziqi Wang, Qi Pan, Pengyu Guo, Thilo Bracht, Barbara Sitek, Jürgen E. Gschwend, Wanhai Xu, Roman Nawroth
{"title":"The mechanism of resistance to CDK4/6 inhibition and novel combination therapy with RNR inhibition for chemo-resistant bladder cancer","authors":"Zhichao Tong,&nbsp;Yubo Zhao,&nbsp;Shiyu Bai,&nbsp;Benedikt Ebner,&nbsp;Lou Lienhard,&nbsp;Yuling Zhao,&nbsp;Ziqi Wang,&nbsp;Qi Pan,&nbsp;Pengyu Guo,&nbsp;Thilo Bracht,&nbsp;Barbara Sitek,&nbsp;Jürgen E. Gschwend,&nbsp;Wanhai Xu,&nbsp;Roman Nawroth","doi":"10.1002/cac2.12532","DOIUrl":null,"url":null,"abstract":"<p>Bladder cancer (BCa) is the most prevalent urological cancer worldwide [<span>1</span>]. A significant proportion of BCa (89%) exhibits molecular alterations in the cell cycle pathway, and targeting cyclin-dependent kinases 4 and 6 (CDK4/6) is deemed as a promising therapeutic strategy [<span>2</span>]. Selective inhibitors of CDK4/6 (CDK4/6is) have been approved by the US Food and Drug Administration (FDA) [<span>3</span>]. They could induce cell cycle arrest in BCa immediately, and after this “sensitive stage”, unknown compensatory mechanism may cause acquired resistance [<span>4, 5</span>]. To address this issue, our study employed multi-omics and identified ribonucleotide reductase regulatory subunit M2 (RRM2), a crucial component of the ribonucleotide reductase (RNR) complex [<span>6</span>], as a key mediator in conferring acquired resistance. We further investigated whether Palbociclib activates proteolysis of RRM2 by the ubiquitin-proteasome system (UPS) and the ubiquitin-like proteins (UBLs) during the sensitive stage. Additionally, we explored whether RRM2 is controlled by E2F transcription factor 3 (E2F3) when acquired resistance is established. Interestingly, upregulation of RRM2 may also cause chemotherapy resistance [<span>7</span>]. Thus, we verified if concurrent inhibition of RNR and CDK4/6 holds promise as a novel therapeutic strategy for BCa patients, especially those exhibit resistance to chemotherapy. All the study designs and methods are described in the Supplementary file.</p><p>Retinoblastoma (RB)-positive BCa elicits a sequential progression from sensitivity to resistance to Palbociclib [<span>8</span>]. We utilized multi-omics to identify key regulators of this process (Figure 1A, Supplementary Tables S1-S5). The only candidate matching all three high-throughput screening approaches was RRM2, and pathway analysis further demonstrated related mechanisms (Supplementary Figures S1-S2). To validate this finding, we examined the cell cycle distribution and expression levels of the other RNR subunit RRM1 and RRM2 in a time kinetic (Figure 1B-C, Supplementary Figure S3A-D). Transcript levels were initially downregulated, followed by a partial recovery, while the decline and recovery pattern of proteins mirrored this. We then transduced single-guide RNAs of RRM1 and RRM2 into T24 synergistic activation mediator (SAM) cells and confirmed partial resistance (Figure 1D-E, Supplementary Figure S3E). However, degradation of RRM2 was still observed at early time points (Supplementary Figure S3F), indicating that proteolysis might be essential for therapy response. We then applied the proteasome inhibitors Epoxomicin/MG-132 in combination with Palbociclib. As shown, protein degradation of RRM2 was effectively blocked, but only partially for RRM1 (Figure 1F, Supplementary Figure S4A-B). We next tested the combination of Palbociclib with ubiquitin-like proteins (UBLs) inhibitor MLN4924 and proved that the initial degradation of RRM1/2 was UPS-dependent but UBL-independent (Figure 1G, Supplementary Figure S4C-E).</p><p>To further investigate the regulation network of RRM2, we interrogated RRM2 in The Cancer Genome Atlas (TCGA) bladder carcinoma study for mRNA co-expression (Supplementary Table S6) and performed network analysis using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) (Supplementary Figure S5A). Additionally, by analyzing spatial transcriptomics data on Spatial Omics DataBase (SODB) and ConsensusPathDB (CPDB), we found that E2F1 was spatially co-expressed with RRM2 (Supplementary Figure S5B-C). We confirmed that Palbociclib mainly affected the expression levels of E2F1 and E2F3, and notably, E2F3 was consistently upregulated (Supplementary Figure S5D-E). By knocking down E2F1, we observed a significant re-expression of RRM2, whereas blocking E2F3 maintained its downregulation (Figure 1H-I). Surprisingly, after long-term suppression of RRM2 expression by siE2F3, we observed an upregulation of E2F1 and blocking of cell cycle re-entry (Figure 1I, Supplementary Figure S5F). Double knockdown of E2F1 and E2F3 slightly prevented the recovery of RRM2 expression levels (Supplementary Figure S5G). These findings collectively suggest that E2F1 might have a suppressive function, while E2F3 drives the RRM2 re-expression.</p><p>We conducted comprehensive tests with the novel validated RNR inhibitor COH29 [<span>9</span>] in combination with Palbociclib to overcome acquired resistance. The combination successfully blocked long-term colony formation (Figure 1J, Supplementary Figure S6A). Additionally, cell viability assays confirmed synergism (Figure 1K, Supplementary Figure S6B). It is worth noting that the combination partially blocked the recovery of RRM2 in RT112 and UM-UC-3 cells, but not in T24 and 253J cells (Figure 1L, Supplementary Figure S6C). These suggested that the synergism is independent of RRM2 expression level, as COH29 blocked the function of RNR (Supplementary Figure S6D). Furthermore, synergism and senescent morphology were also observed using a living cell monitoring system in the combination groups (Supplementary Figure S7A). However, apoptosis through Caspase3/7 could not be observed (Supplementary Figure S7B). Subsequently, the combination was tested in two models: the chicken chorioallantoic membrane (CAM) and the murine subcutaneous xenograft model. Both monotherapies suppressed tumor growth, while the combination induced more potent effect (Figure 1M, Supplementary Figure S8A-E). This effect was attributed to proliferation as indicated by a significant decrease in Ki-67 expression (Supplementary Figure S8F).</p><p>By utilizing Gene Expression Profiling Interactive Analysis (GEPIA), we found that RRM2 was significantly upregulated in tumor tissues, while RRM1 and RB showed no difference (Figure 1N, Supplementary Figure S9A). Higher expression of them was related with significantly worse disease-free survival (DFS) (Supplementary Figure S9B). We also identified 4 patients from the Cornell/Trento cohort [<span>10</span>] who had primary and paired advanced tumors and were treated with chemotherapy. Among them, 2 patients (WCM088 and WCM117) showed amplified RRM2 only in the metastasis (Supplementary Figure S9C), suggesting that amplification of RRM2 may contribute to chemo-resistance. In addition, we retrospectively collected 20 muscle-invasive BCa specimens from patients who underwent radical cystectomy (Supplementary Table S7). These specimens were divided into three subgroups according to their response to chemotherapy. We observed lower RRM2 levels in the chemo-responded group compared to chemotherapy-naïve patients, but an increase in the chemo-resistant group (Supplementary Figure S9D-E). We tested the combination therapy in chemo-resistant BCa cell lines and found that they responded synergistically (Figure 1O, Supplementary Figure S9F-L).</p><p>In conclusion, we identified RRM2 as key mediator in conferring acquired resistance to Palbociclib in BCa (Figure 1P). Dual inhibition of RNR and CDK4/6 could effectively overcome this acquired resistance and holds promise as a potential therapeutic strategy for chemo-resistant BCa.</p><p>Zhichao Tong, Wanhai Xu, Roman Nawroth and Jürgen E. Gschwend designed the study and wrote the manuscript, Zhichao Tong, Yubo Zhao, Lou Lienhard, Shiyu Bai, Ziqi Wang, Yuling Zhao, Thilo Bracht, Barbara Sitek performed the experiments, Qi Pan, Pengyu Guo, Benedikt Ebner analyzed the data.</p><p>The authors declare no competing interests.</p><p>This work was supported by the National Natural Science Foundation Fund of China (82002688; U20A20385), China Postdoctoral Science Foundation (2021M693828), Postdoctoral Scientific Research Development Fund of Heilongjiang Province (LBH-Z22030), National Key Research and Development Program of China (2021YFB3801000).</p><p>The research procedures were approved by the Committees for Ethical Review of Harbin Medical University (2022-DWSYLLCZ-38; KY2023-62).</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1000,"publicationDate":"2024-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11194448/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12532","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Bladder cancer (BCa) is the most prevalent urological cancer worldwide [1]. A significant proportion of BCa (89%) exhibits molecular alterations in the cell cycle pathway, and targeting cyclin-dependent kinases 4 and 6 (CDK4/6) is deemed as a promising therapeutic strategy [2]. Selective inhibitors of CDK4/6 (CDK4/6is) have been approved by the US Food and Drug Administration (FDA) [3]. They could induce cell cycle arrest in BCa immediately, and after this “sensitive stage”, unknown compensatory mechanism may cause acquired resistance [4, 5]. To address this issue, our study employed multi-omics and identified ribonucleotide reductase regulatory subunit M2 (RRM2), a crucial component of the ribonucleotide reductase (RNR) complex [6], as a key mediator in conferring acquired resistance. We further investigated whether Palbociclib activates proteolysis of RRM2 by the ubiquitin-proteasome system (UPS) and the ubiquitin-like proteins (UBLs) during the sensitive stage. Additionally, we explored whether RRM2 is controlled by E2F transcription factor 3 (E2F3) when acquired resistance is established. Interestingly, upregulation of RRM2 may also cause chemotherapy resistance [7]. Thus, we verified if concurrent inhibition of RNR and CDK4/6 holds promise as a novel therapeutic strategy for BCa patients, especially those exhibit resistance to chemotherapy. All the study designs and methods are described in the Supplementary file.

Retinoblastoma (RB)-positive BCa elicits a sequential progression from sensitivity to resistance to Palbociclib [8]. We utilized multi-omics to identify key regulators of this process (Figure 1A, Supplementary Tables S1-S5). The only candidate matching all three high-throughput screening approaches was RRM2, and pathway analysis further demonstrated related mechanisms (Supplementary Figures S1-S2). To validate this finding, we examined the cell cycle distribution and expression levels of the other RNR subunit RRM1 and RRM2 in a time kinetic (Figure 1B-C, Supplementary Figure S3A-D). Transcript levels were initially downregulated, followed by a partial recovery, while the decline and recovery pattern of proteins mirrored this. We then transduced single-guide RNAs of RRM1 and RRM2 into T24 synergistic activation mediator (SAM) cells and confirmed partial resistance (Figure 1D-E, Supplementary Figure S3E). However, degradation of RRM2 was still observed at early time points (Supplementary Figure S3F), indicating that proteolysis might be essential for therapy response. We then applied the proteasome inhibitors Epoxomicin/MG-132 in combination with Palbociclib. As shown, protein degradation of RRM2 was effectively blocked, but only partially for RRM1 (Figure 1F, Supplementary Figure S4A-B). We next tested the combination of Palbociclib with ubiquitin-like proteins (UBLs) inhibitor MLN4924 and proved that the initial degradation of RRM1/2 was UPS-dependent but UBL-independent (Figure 1G, Supplementary Figure S4C-E).

To further investigate the regulation network of RRM2, we interrogated RRM2 in The Cancer Genome Atlas (TCGA) bladder carcinoma study for mRNA co-expression (Supplementary Table S6) and performed network analysis using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) (Supplementary Figure S5A). Additionally, by analyzing spatial transcriptomics data on Spatial Omics DataBase (SODB) and ConsensusPathDB (CPDB), we found that E2F1 was spatially co-expressed with RRM2 (Supplementary Figure S5B-C). We confirmed that Palbociclib mainly affected the expression levels of E2F1 and E2F3, and notably, E2F3 was consistently upregulated (Supplementary Figure S5D-E). By knocking down E2F1, we observed a significant re-expression of RRM2, whereas blocking E2F3 maintained its downregulation (Figure 1H-I). Surprisingly, after long-term suppression of RRM2 expression by siE2F3, we observed an upregulation of E2F1 and blocking of cell cycle re-entry (Figure 1I, Supplementary Figure S5F). Double knockdown of E2F1 and E2F3 slightly prevented the recovery of RRM2 expression levels (Supplementary Figure S5G). These findings collectively suggest that E2F1 might have a suppressive function, while E2F3 drives the RRM2 re-expression.

We conducted comprehensive tests with the novel validated RNR inhibitor COH29 [9] in combination with Palbociclib to overcome acquired resistance. The combination successfully blocked long-term colony formation (Figure 1J, Supplementary Figure S6A). Additionally, cell viability assays confirmed synergism (Figure 1K, Supplementary Figure S6B). It is worth noting that the combination partially blocked the recovery of RRM2 in RT112 and UM-UC-3 cells, but not in T24 and 253J cells (Figure 1L, Supplementary Figure S6C). These suggested that the synergism is independent of RRM2 expression level, as COH29 blocked the function of RNR (Supplementary Figure S6D). Furthermore, synergism and senescent morphology were also observed using a living cell monitoring system in the combination groups (Supplementary Figure S7A). However, apoptosis through Caspase3/7 could not be observed (Supplementary Figure S7B). Subsequently, the combination was tested in two models: the chicken chorioallantoic membrane (CAM) and the murine subcutaneous xenograft model. Both monotherapies suppressed tumor growth, while the combination induced more potent effect (Figure 1M, Supplementary Figure S8A-E). This effect was attributed to proliferation as indicated by a significant decrease in Ki-67 expression (Supplementary Figure S8F).

By utilizing Gene Expression Profiling Interactive Analysis (GEPIA), we found that RRM2 was significantly upregulated in tumor tissues, while RRM1 and RB showed no difference (Figure 1N, Supplementary Figure S9A). Higher expression of them was related with significantly worse disease-free survival (DFS) (Supplementary Figure S9B). We also identified 4 patients from the Cornell/Trento cohort [10] who had primary and paired advanced tumors and were treated with chemotherapy. Among them, 2 patients (WCM088 and WCM117) showed amplified RRM2 only in the metastasis (Supplementary Figure S9C), suggesting that amplification of RRM2 may contribute to chemo-resistance. In addition, we retrospectively collected 20 muscle-invasive BCa specimens from patients who underwent radical cystectomy (Supplementary Table S7). These specimens were divided into three subgroups according to their response to chemotherapy. We observed lower RRM2 levels in the chemo-responded group compared to chemotherapy-naïve patients, but an increase in the chemo-resistant group (Supplementary Figure S9D-E). We tested the combination therapy in chemo-resistant BCa cell lines and found that they responded synergistically (Figure 1O, Supplementary Figure S9F-L).

In conclusion, we identified RRM2 as key mediator in conferring acquired resistance to Palbociclib in BCa (Figure 1P). Dual inhibition of RNR and CDK4/6 could effectively overcome this acquired resistance and holds promise as a potential therapeutic strategy for chemo-resistant BCa.

Zhichao Tong, Wanhai Xu, Roman Nawroth and Jürgen E. Gschwend designed the study and wrote the manuscript, Zhichao Tong, Yubo Zhao, Lou Lienhard, Shiyu Bai, Ziqi Wang, Yuling Zhao, Thilo Bracht, Barbara Sitek performed the experiments, Qi Pan, Pengyu Guo, Benedikt Ebner analyzed the data.

The authors declare no competing interests.

This work was supported by the National Natural Science Foundation Fund of China (82002688; U20A20385), China Postdoctoral Science Foundation (2021M693828), Postdoctoral Scientific Research Development Fund of Heilongjiang Province (LBH-Z22030), National Key Research and Development Program of China (2021YFB3801000).

The research procedures were approved by the Committees for Ethical Review of Harbin Medical University (2022-DWSYLLCZ-38; KY2023-62).

Abstract Image

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
CDK4/6 抑制剂的耐药机制以及与 RNR 抑制剂联合治疗化疗耐药膀胱癌的新方法。
此外,使用活细胞监测系统也观察到了组合组的协同作用和衰老形态(补充图 S7A)。然而,无法观察到通过 Caspase3/7 发生的细胞凋亡(补充图 S7B)。随后,在鸡绒毛膜(CAM)和小鼠皮下异种移植模型这两种模型中对联合疗法进行了测试。两种单一疗法都能抑制肿瘤生长,而联合疗法的效果更强(图 1M,补充图 S8A-E)。通过基因表达谱交互分析(GEPIA),我们发现 RRM2 在肿瘤组织中显著上调,而 RRM1 和 RB 则没有差异(图 1N,补充图 S9A)。它们的高表达与较差的无病生存期(DFS)有关(补充图 S9B)。我们还从康奈尔/特伦托队列[10]中发现了 4 例原发性和配对晚期肿瘤并接受化疗的患者。其中,2 例患者(WCM088 和 WCM117)仅在转移瘤中出现 RRM2 扩增(补充图 S9C),提示 RRM2 扩增可能导致化疗耐药。此外,我们回顾性地从接受根治性膀胱切除术的患者中收集了20份肌肉浸润性BCa标本(补充表S7)。根据化疗反应将这些标本分为三个亚组。我们观察到化疗反应组的 RRM2 水平低于化疗未反应组,但化疗耐受组的 RRM2 水平有所升高(补充图 S9D-E)。我们在化疗耐药的BCa细胞系中测试了联合疗法,发现它们具有协同反应(图1O,补充图S9F-L)。总之,我们发现RRM2是BCa对Palbociclib产生获得性耐药性的关键介质(图1P)。RNR和CDK4/6的双重抑制可有效克服这种获得性耐药,有望成为化疗耐药BCa的潜在治疗策略。童志超、赵玉波、Lou Lienhard、白诗雨、王梓琪、赵玉玲、Thilo Bracht、Barbara Sitek设计研究并撰写手稿,潘琦、郭鹏宇、Benedikt Ebner分析数据。本研究得到了国家自然科学基金(82002688;U20A20385)、中国博士后科学基金(2021M693828)、黑龙江省博士后科研发展基金(LBH-Z22030)、国家重点研发计划(2021YFB3801000)的资助,研究程序得到了哈尔滨医科大学伦理审查委员会(2022-DWSYLLCZ-38;KY2023-62)的批准。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
期刊最新文献
Perioperative toripalimab plus neoadjuvant chemotherapy might improve outcomes in resectable esophageal cancer: an interim analysis of a phase III randomized clinical trial. Tryptophan 2,3-dioxygenase-positive matrix fibroblasts fuel breast cancer lung metastasis via kynurenine-mediated ferroptosis resistance of metastatic cells and T cell dysfunction. Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Unraveling the routes to distant metastases in colorectal cancer: Tumor deposits and lymph node metastases as the gateway. 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