Proteasome inhibitor

蛋白酶体抑制剂
  • 文章类型: Journal Article
    重新使用FDA批准的药物是从头药物开发的快速且具有成本效益的替代方案。这里,我们确定了与硼替佐米敏感性有关的基因,预测可能受益于硼替佐米治疗的癌症类型,并评估硼替佐米在乳腺癌中的作用机制(BT-474和ZR-75-30),黑色素瘤(A-375),和体外成胶质细胞瘤(A-172)细胞。来自血液癌症的癌细胞系,肾,神经系统,发现皮肤对硼替佐米的敏感性明显高于其他器官系统。体外研究证实,尽管硼替佐米有效抑制了所有四种细胞系中的β5催化位点,细胞周期阻滞仅在G2/M期诱导,24h后A-375和A-172细胞凋亡。基因组和转录组学分析鉴定了与硼替佐米抗性相关的33个基因(例如ALDH18A1、ATAD2)。一起来看,我们确定了预测硼替佐米敏感性的生物标志物和可能受益于硼替佐米治疗的癌症类型.
    Repurposing of FDA-approved drugs is a quick and cost-effective alternative to de novo drug development. Here, we identify genes involved in bortezomib sensitivity, predict cancer types that may benefit from treatment with bortezomib, and evaluate the mechanism-of-action of bortezomib in breast cancer (BT-474 and ZR-75-30), melanoma (A-375), and glioblastoma (A-172) cells in vitro. Cancer cell lines derived from cancers of the blood, kidney, nervous system, and skin were found to be significantly more sensitive to bortezomib than other organ systems. The in vitro studies confirmed that although bortezomib effectively inhibited the β5 catalytic site in all four cell lines, cell cycle arrest was only induced in G2/M phase and apoptosis in A-375 and A-172 after 24h. The genomic and transcriptomic analyses identified 33 genes (e.g. ALDH18A1, ATAD2) associated with bortezomib resistance. Taken together, we identified biomarkers predictive of bortezomib sensitivity and cancer types that might benefit from treatment with bortezomib.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Kirsten大鼠肉瘤(KRAS)是大肠癌(CRC)中最常见的突变癌基因。我们以前报道过微卫星不稳定性(MSI)之间的相互作用,DNA启动子甲基化,和基因表达。在这项研究中,我们寻找KRAS突变之间的关联,基因表达,和甲基化可能有助于精准医学。在配对的CRC肿瘤和周围健康组织中进行全基因组基因表达和DNA甲基化。结果提示(a)在具有KRAS突变的患者中,CRC中许多主要基因通路的失调程度显著更大,(b)这些失调的基因通路的上调和下调可能与相应的低甲基化和高甲基化相关,和(c)CDKN2A的上调在具有KRAS突变的肿瘤中更为明显。最近的细胞系研究表明,在5-FU抗性CRC细胞中存在较高的CDKN2A水平,并且这些可以被Villosol下调。我们的发现表明,在KRAS突变型CRC中,Villosol对抗CDKN2A治疗有更好的反应。此外,CRC组织中蛋白酶体途径的基因上调更明显,尤其是KRAS突变和MSI,可能表明蛋白酶体抑制剂(硼替佐米,Carfilzomib,或艾沙佐米)在必要时用于选定的CRC患者。
    Kirsten Rat Sarcoma (KRAS) is the most commonly mutated oncogene in colorectal carcinoma (CRC). We have previously reported the interactions between microsatellite instability (MSI), DNA promoter methylation, and gene expression. In this study, we looked for associations between KRAS mutation, gene expression, and methylation that may help with precision medicine. Genome-wide gene expression and DNA methylation were done in paired CRC tumor and surrounding healthy tissues. The results suggested that (a) the magnitude of dysregulation of many major gene pathways in CRC was significantly greater in patients with the KRAS mutation, (b) the up- and down-regulation of these dysregulated gene pathways could be correlated with the corresponding hypo- and hyper-methylation, and (c) the up-regulation of CDKN2A was more pronounced in tumors with the KRAS mutation. A recent cell line study showed that there were higher CDKN2A levels in 5-FU-resistant CRC cells and that these could be down-regulated by Villosol. Our findings suggest the possibility of a better response to anti-CDKN2A therapy with Villosol in KRAS-mutant CRC. Also, the more marked up-regulation of genes in the proteasome pathway in CRC tissue, especially with the KRAS mutation and MSI, may suggest a potential role of a proteasome inhibitor (bortezomib, carfilzomib, or ixazomib) in selected CRC patients if necessary.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在过去的几十年里,皮肤黑色素瘤的全球发病率,由黑素细胞引起的恶性肿瘤,明显增加,导致皮肤癌相关死亡率最高。虽然局部肿瘤很容易通过切除手术切除,晚期转移性黑色素瘤难以治疗且预后不良.因此,揭示黑素瘤肿瘤发生和转移的分子机制对于开发新型靶向治疗至关重要。我们发现,在黑色素瘤的临床前异种移植模型中,转化生长因子β(TGFβ)信号通路需要多种内分泌瘤1型(MEN1)基因产物Menin在体外诱导细胞生长停滞和凋亡并防止体内肿瘤发生。我们进一步鉴定了受黑色素瘤影响的两个MEN1家族成员中的点突变,这些点突变导致MEN1基因产物的蛋白酶体降解和TGFβ信号传导的丧失。有趣的是,使用FDA批准的药物和RNAi靶向阻断蛋白酶体降解途径可以有效恢复MEN1表达和TGFβ转录反应。一起,这些结果为皮肤黑色素瘤的治疗提供了新的潜在治疗策略和患者分层.
    Over the past few decades, the worldwide incidence of cutaneous melanoma, a malignant neoplasm arising from melanocytes, has been increasing markedly, leading to the highest rate of skin cancer-related deaths. While localized tumors are easily removed by excision surgery, late-stage metastatic melanomas are refractory to treatment and exhibit a poor prognosis. Consequently, unraveling the molecular mechanisms underlying melanoma tumorigenesis and metastasis is crucial for developing novel targeted therapies. We found that the multiple endocrine neoplasia type 1 (MEN1) gene product Menin is required for the transforming growth factor beta (TGFβ) signaling pathway to induce cell growth arrest and apoptosis in vitro and prevent tumorigenesis in vivo in preclinical xenograft models of melanoma. We further identified point mutations in two MEN1 family members affected by melanoma that led to proteasomal degradation of the MEN1 gene product and to a loss of TGFβ signaling. Interestingly, blocking the proteasome degradation pathway using an FDA-approved drug and RNAi targeting could efficiently restore MEN1 expression and TGFβ transcriptional responses. Together, these results provide new potential therapeutic strategies and patient stratification for the treatment of cutaneous melanoma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    众所周知,非洲侏儒刺猬(Atelerixalbiventris)的肿瘤发病率很高。然而,研究该物种的肿瘤一直受到有限的研究材料,如细胞系和基因组信息的限制。在这项研究中,我们成功地从非洲侏儒刺猬的组织细胞肉瘤(HS)中建立了一个新的细胞系,允许我们进行药物筛选.我们使用FDA批准的药物库筛选进行调查,以确定该肿瘤细胞系对哪种抗癌药物敏感,作为细胞凋亡实验的结果,发现三种蛋白酶体抑制剂中的硼替佐米通过显着增加caspase-3的裂解而诱导癌细胞死亡(P<0.01)。因此,我们阐明了蛋白酶体抑制剂,尤其是硼替佐米,通过与人类肿瘤中描述的机制相当的机制,对非洲侏儒刺猬中HS衍生的细胞系表现出抗肿瘤作用。这项研究报告了来自非洲侏儒刺猬的第一个特征细胞系,并强调了硼替佐米作为该物种HS抗肿瘤治疗的潜在用途。
    The African pygmy hedgehog (Atelerix albiventris) is known to have a high incidence of tumor. However, investigating the tumors of this species has been constrained by the limited availability of research materials such as cell lines and genome information. In this study, we successfully established a novel cell line from a histiocytic sarcoma (HS) of an African pygmy hedgehog, allowing us to conduct a drug screening. We investigated using FDA-approved drug library screening to determine which anticancer drug this tumor cell line is sensitive to, and as a result of apoptosis experiments, bortezomib among the three proteasome inhibitors was found to induce cell death of cancer cells by significantly increasing caspase-3 cleavage (P<0.01). Thus, we elucidated that the proteasome inhibitors, particularly bortezomib, exhibit anti-tumor effects on a cell line derived from an HS in an African pygmy hedgehog through a mechanism comparable to that described in human tumors. This study reports the first characterized cell line from the African pygmy hedgehog and also highlights the potential utility of bortezomib as an anti-tumor treatment for HS in this species.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:为了评估有效性,安全,以及从基于硼替佐米的静脉诱导到基于艾沙佐米的口服方案的课堂过渡(iCT)的便利性。
    方法:这项回顾性研究于2017年10月至2023年4月在16家中国医院进行,分析了新诊断(NDMM)和一线复发多发性骨髓瘤(FRMM)患者,这些患者至少从硼替佐米诱导治疗中获得了部分反应。然后是以ixazomib为基础的口服方案2年或直到疾病进展或无法耐受的毒性。
    结果:该研究招募了199名患者,中位年龄:63岁,男性55.4%,53%为高风险(HR),47%为标准风险。通过中期荧光原位杂交(M-FISH)进行细胞遗传学风险分层,基于梅奥诊所风险分层系统。总PI治疗的中位持续时间为11个月,以艾沙佐米为基础的治疗持续6个月。在20个月的中位随访中,53%的患者仍在接受治疗。从基于硼替佐米的诱导开始,24个月的PFS率为84.3%,从基于艾沙唑米的治疗开始为83.4%。硼替佐米诱导后的总反应率(ORR)为100%,基于伊沙佐米的方案6个周期后为90%。根据桑基图,89.51%的患者在2个周期的iCT后维持或改善了他们的疾病反应,6个周期(90.14%),和12个周期(80%)。发现Mayo的HR水平是缓解不良的重要独立因素(风险比(HR)2.55;p=0.033)。Ixazomib的安全性与以前的临床试验数据一致,49%的患者经历至少一个任何级别的AE。最常见的不良事件包括周围神经病变,恶心和呕吐,腹泻,血小板减少症,和粒细胞减少症.
    结论:在现实世界的中国MM人群中,NDMM和FRMM患者对基于PI的连续治疗反应良好,显示出相当大的反应率。基于ixazomib的iCT允许持续的基于PI的治疗,提供有希望的疗效和可耐受的不良事件。
    OBJECTIVE: To evaluate the effectiveness, safety, and convenience of in-class transition (iCT) from intravenous bortezomib-based induction to ixazomib-based oral regimens.
    METHODS: This retrospective real-world study was conducted in 16 Chinese hospitals between October 2017 and April 2023 and analyzed newly diagnosed (NDMM) and first-line relapsed multiple myeloma (FRMM) patients who attained at least a partial response from bortezomib-based induction therapy, followed by an ixazomib-based oral regimen for 2 year or until disease progression or intolerable toxicity.
    RESULTS: The study enrolled 199 patients, median age: 63 years old, male 55.4%, 53% as high risk (HR), and 47% as standard risk. Cytogenetic risk stratification by metaphase fluorescence in situ hybridization (M-FISH), based on the Mayo Clinic risk stratification system. The median duration of total PI therapy was 11 months, with ixazomib-based treatment spanning 6 months. At the 20-month median follow-up, 53% of patients remained on therapy. The 24-month PFS rate was 84.3% from the initiation of bortezomib-based induction and 83.4% from the start of ixazomib-based treatment. Overall response rate (ORR) was 100% post-bortezomib induction and 90% following 6 cycles of the ixazomib-based regimen. Based on the Sankey diagrams, 89.51% of patients maintained or improved their disease response after 2 cycles of iCT, 6 cycles (90.14%), and 12 cycles (80%). The HR level of Mayo was found to be a significant independent factor in a worse remission (hazard ratio (HR) 2.55; p = 0.033). Ixazomib\'s safety profile aligned with previous clinical trial data, with 49% of patients experiencing at least one AE of any grade. The most common AEs included peripheral neuropathy, nausea and vomiting, diarrhea, thrombocytopenia, and granulocytopenia.
    CONCLUSIONS: In the real-world Chinese MM population, NDMM and FRMM patients responded favorably to PI-based continuous therapy, demonstrating substantial response rates. The ixazomib-based iCT allows for sustained PI-based treatment, offering promising efficacy and tolerable AEs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肺癌是癌症死亡的主要原因。尽管近年来的治疗进展,我们需要新的治疗策略来改善肺癌患者的预后.突变型p53在肺癌中普遍存在,并通过功能获得致癌程序驱动癌症的几个标志,并经常预测预后较差。突变型p53的致癌性与其通过逃避蛋白酶体降解而在细胞中的稳定性和积累有关。因此,已寻求突变p53的去稳定作为一种治疗策略,但到目前为止还没有临床成功。在这项研究中,我们报道,蛋白酶体抑制导致携带R273H突变蛋白的非小细胞肺癌(NSCLC)细胞系中突变p53的降解,并证明这是由hsp70介导的.具有突变R273H等位基因的NSCLC细胞系表现出对蛋白酶体抑制剂的敏感性和凋亡增加。这些数据表明蛋白酶体抑制剂可能在TP53突变的NSCLC的一些亚群中具有治疗意义。
    Lung cancer is the leading cause of cancer mortality. Despite therapeutic advances in recent years, new treatment strategies are needed to improve outcomes of lung cancer patients. Mutant p53 is prevalent in lung cancers and drives several hallmarks of cancer through a gain-of-function oncogenic program, and often predicts a poorer prognosis. The oncogenicity of mutant p53 is related to its stability and accumulation in cells by evading degradation by the proteasome. Therefore, destabilization of mutant p53 has been sought as a therapeutic strategy, but so far without clinical success. In this study, we report that proteasome inhibition results in degradation of mutant p53 in non-small cell lung cancer (NSCLC) cell lines bearing the R273H mutant protein and show evidence that this was mediated by hsp70. NSCLC cell lines with the mutant R273H allele demonstrated increased susceptibility and apoptosis to proteasome inhibitors. These data suggest that proteasome inhibitors could have therapeutic implications in some subsets of TP53 mutated NSCLC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肝细胞癌(HCC)是世界上最常见和最致命的癌症之一,经常在晚期被诊断出来。由于缺乏有效的治疗策略,HCC患者的预后较差。批准的药物再利用是加速药物发现的一种方式,可以显着降低药物开发成本。卡非佐米(CFZ)是第二代蛋白酶体抑制剂,它对多发性骨髓瘤非常有效,并且据报道具有针对多种癌症的潜在抗肿瘤活性。然而,CFZ对HCC的潜在机制尚不清楚。这里,我们表明CFZ通过细胞周期阻滞在G2/M期抑制HCC细胞的增殖,并通过抑制上皮-间质转化抑制HCC细胞的迁移和侵袭。我们还发现,CFZ促进活性氧的产生,以诱导内质网(ER)应激和激活JNK/p38MAPK信号在肝癌细胞,从而诱导HCC细胞死亡。此外,CFZ在异种移植小鼠模型中显著抑制HCC细胞生长。总的来说,我们的研究阐明,CFZ损害线粒体功能,激活内质网应激和JNK/p38MAPK信号,从而抑制HCC细胞和肿瘤生长。这表明CFZ具有作为HCC治疗药物的潜力。
    Hepatocellular carcinoma (HCC) is one of the most prevalent and deadly cancers in the world, which is frequently diagnosed at a late stage. HCC patients have a poor prognosis due to the lack of an efficacious therapeutic strategy. Approved drug repurposing is a way for accelerating drug discovery and can significantly reduce the cost of drug development. Carfilzomib (CFZ) is a second-generation proteasome inhibitor, which is highly efficacious against multiple myeloma and has been reported to possess potential antitumor activities against multiple cancers. However, the underlying mechanism of CFZ on HCC is still unclear. Here, we show that CFZ inhibits the proliferation of HCC cells through cell cycle arrest at the G2/M phase and suppresses the migration and invasion of HCC cells by inhibiting epithelial-mesenchymal transition. We also find that CFZ promotes reactive oxygen species production to induce endoplasmic reticulum (ER) stress and activate JNK/p38 MAPK signaling in HCC cells, thus inducing cell death in HCC cells. Moreover, CFZ significantly inhibits HCC cell growth in a xenograft mouse model. Collectively, our study elucidates that CFZ impairs mitochondrial function and activates ER stress and JNK/p38 MAPK signaling, thus inhibiting HCC cell and tumor growth. This indicates that CFZ has the potential as a therapeutic drug for HCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    蛋白质稳态是正常生命活动的基础,而蛋白酶体家族在这一过程中起着极其重要的作用。蛋白酶体20S是具有两个α环和两个β环重叠的同心圆结构。蛋白酶体20S可以通过与各种亚基(例如19S,11S,和200PA),其通过其活性亚基β1、β2和β5进行。蛋白酶体可以降解错误折叠的,过量的蛋白质来维持体内平衡。同时,它可以被肿瘤用来降解过度增殖的蛋白质和不需要的蛋白质来支持它们的生长。蛋白酶体可以从NF-κB和p53等肿瘤信号通路、细胞周期、免疫调节,和抗药性。已发现蛋白酶体编码基因在多种肿瘤中过表达,为癌症治疗提供了一个潜在的新靶点。此外,蛋白酶体抑制剂,如硼替佐米,Carfilzomib,和艾沙佐米已作为多发性骨髓瘤的一线治疗方法投入临床应用。越来越多的研究表明,它在肝细胞癌等其他肿瘤中也有不同的治疗效果,非小细胞肺癌,胶质母细胞瘤,和神经母细胞瘤.然而,蛋白酶体抑制剂由于它们在其他肿瘤中的耐受性和单一性,效果不大。因此,需要进一步研究它们的作用机制和药物相互作用,以研究它们的治疗潜力.
    Protein homeostasis is the basis of normal life activities, and the proteasome family plays an extremely important function in this process. The proteasome 20S is a concentric circle structure with two α rings and two β rings overlapped. The proteasome 20S can perform both ATP-dependent and non-ATP-dependent ubiquitination proteasome degradation by binding to various subunits (such as 19S, 11S, and 200 PA), which is performed by its active subunit β1, β2, and β5. The proteasome can degrade misfolded, excess proteins to maintain homeostasis. At the same time, it can be utilized by tumors to degrade over-proliferate and unwanted proteins to support their growth. Proteasomes can affect the development of tumors from several aspects including tumor signaling pathways such as NF-κB and p53, cell cycle, immune regulation, and drug resistance. Proteasome-encoding genes have been found to be overexpressed in a variety of tumors, providing a potential novel target for cancer therapy. In addition, proteasome inhibitors such as bortezomib, carfilzomib, and ixazomib have been put into clinical application as the first-line treatment of multiple myeloma. More and more studies have shown that it also has different therapeutic effects in other tumors such as hepatocellular carcinoma, non-small cell lung cancer, glioblastoma, and neuroblastoma. However, proteasome inhibitors are not much effective due to their tolerance and singleness in other tumors. Therefore, further studies on their mechanisms of action and drug interactions are needed to investigate their therapeutic potential.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    包含蛋白酶体抑制剂的三联疗法,免疫调节药物,对于符合移植条件的多发性骨髓瘤(MM)患者,建议采用地塞米松(DEX)诱导/巩固治疗.在2017年至2019年进行的这项日本II期研究中,20-65岁的新诊断MM患者接受了四个硼替佐米(Bor)诱导周期,来那度胺(Len),和DEX(VRD),其次是Bor和大剂量美法仑与自体干细胞抢救。随后,他们用卡非佐米经历了四个巩固周期,Len,和DEX(KRD),然后进行Len维持直至疾病进展。共分析141例患者。在意向治疗人群中,诱导后完全或更好的反应为19.9%,上升至39.7%,58.9%,移植后为62.4%,合并,1年保养,分别。中位随访时间为38个月,3年无进展生存率(PFS)为83.5%,3年总生存率为92.5%.严重不良事件(≥3级)发生在约30%的患者中;然而,无治疗相关死亡.这些发现清楚地表明了该方案的耐受性和有效性。然而,具有高风险细胞遗传学的患者比没有的患者显示出更低的3年PFS趋势(77.8%vs.89.4%,p=0.051),和超高危细胞遗传学(≥2个高危细胞遗传学)的预后更差,3年PFS为61.2%。为了克服这种情况,在未来的研究中,应评估结合CD38抗体等新型药物的更有效的治疗策略.
    Triplet regimen comprising proteasome inhibitors, immunomodulatory drugs, and dexamethasone (DEX) is a recommended induction/consolidation therapy for multiple myeloma (MM) patients eligible for transplant. In this Japanese phase II study conducted from 2017 to 2019, newly diagnosed MM patients aged 20-65 received four induction cycles with bortezomib (Bor), lenalidomide (Len), and DEX (VRD), followed by Bor and high-dose melphalan with autologous stem cell rescue. Subsequently, they underwent four consolidation cycles with carfilzomib, Len, and DEX (KRD), followed by Len maintenance until disease progression. A total of 141 patients were analyzed. In an intent-to-treat population, the complete or better response post induction was 19.9%, rising to 39.7%, 58.9%, and 62.4% after transplant, consolidation, and 1-year maintenance, respectively. With a median follow-up of 38 months, the 3-year progression-free survival (PFS) rate was 83.5% and the 3-year overall survival rate was 92.5%. Severe adverse events (≥grade 3) occurred in ~30% of patients; however, there was no treatment-related mortality. These findings clearly showed the tolerability and effectiveness of this protocol. Nevertheless, patients with high-risk cytogenetics showed a trend toward lower 3-year PFS than those without (77.8% vs. 89.4%, p = 0.051), and ultra-high-risk cytogenetics (≥2 high-risk cytogenetics) had an even worse prognosis, with 61.2% 3-year PFS. To overcome this situation, a more potent treatment strategy incorporating novel agents such as the CD38-antibody should be assessed in future studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    GZ17-6.02,一种含有异香兰素的合成化合物,Harmine和姜黄素,已对实体肿瘤患者(NCT03775525)进行了I期评估,推荐的2期剂量(RP2D)为375mgPOBID。GZ17-6.02作为单一药剂在杀死多发性骨髓瘤细胞方面比先前在实体瘤细胞类型中观察到的更有效。GZ17-6.02以大于累加的方式与蛋白酶体抑制剂相互作用以杀死骨髓瘤细胞,而单独杀死抑制剂抗性细胞的程度相似。GZ17-6.02与硼替佐米联合激活ATM,AMPK和PERK以及灭活的ULK1,mTORC1,eIF2α,NFκB和Hippo途径。该组合增加了ATG13S318的磷酸化和Beclin1,ATG5,BAK和BIM的表达,并降低BCL-XL和MCL1的水平。GZ17-6.02与硼替佐米相互作用以增强自噬体形成和自噬通量,并摧毁自动取款机,AMPKα,ULK1,Beclin1或ATG5显着降低自噬和肿瘤细胞杀伤。BAK和BIM的敲除显著降低了肿瘤细胞的杀伤。HDACs1/2/3的表达显着降低,超出了先前在实体瘤细胞中观察到的水平,并且需要自噬。这与组蛋白H3的乙酰化和甲基化增加有关。HDACs1/2/3的联合敲低导致ATM和AMPK的激活,并导致ULK1,mTORC1,NFκB和Hippo途径的失活。HDAC敲低也增强了ATG13磷酸化,BAK水平升高,BCL-XL水平降低。总的来说,我们目前的研究支持对多发性骨髓瘤细胞进行其他体内研究.
    GZ17-6.02, a synthetically manufactured compound containing isovanillin, harmine and curcumin, has undergone phase I evaluation in patients with solid tumors (NCT03775525) with a recommended phase 2 dose (RP2D) of 375 mg PO BID. GZ17-6.02 was more efficacious as a single agent at killing multiple myeloma cells than had previously been observed in solid tumor cell types. GZ17-6.02 interacted with proteasome inhibitors in a greater than additive fashion to kill myeloma cells and alone it killed inhibitor-resistant cells to a similar extent. The drug combination of GZ17-6.02 and bortezomib activated ATM, the AMPK and PERK and inactivated ULK1, mTORC1, eIF2α, NFκB and the Hippo pathway. The combination increased ATG13 S318 phosphorylation and the expression of Beclin1, ATG5, BAK and BIM, and reduced the levels of BCL-XL and MCL1. GZ17-6.02 interacted with bortezomib to enhance autophagosome formation and autophagic flux, and knock down of ATM, AMPKα, ULK1, Beclin1 or ATG5 significantly reduced both autophagy and tumor cell killing. Knock down of BAK and BIM significantly reduced tumor cell killing. The expression of HDACs1/2/3 was significantly reduced beyond that previously observed in solid tumor cells and required autophagy. This was associated with increased acetylation and methylation of histone H3. Combined knock down of HDACs1/2/3 caused activation of ATM and the AMPK and caused inactivation of ULK1, mTORC1, NFκB and the Hippo pathway. HDAC knock down also enhanced ATG13 phosphorylation, increased BAK levels and reduced those of BCL-XL. Collectively, our present studies support performing additional in vivo studies with multiple myeloma cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号