Proteasome inhibitor

蛋白酶体抑制剂
  • 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
    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
    背景:蛋白酶体抑制剂(PI)是治疗多发性骨髓瘤(MM)的最重要药物之一。然而,几乎所有MM患者都出现PI抵抗,导致治疗失败。因此,MM中PI抵抗的潜在机制需要进一步研究。
    方法:我们使用了几种MM细胞系来建立抗PI的MM细胞系。我们在MM细胞系中进行了RNA微阵列和EccDNA-seq,并收集了人类原代MM样品以探索基因谱。我们使用免疫共沉淀(Co-IP)评估了MUC20对PI抗性MM细胞凋亡的影响,海马生物能谱分析和体内测定。
    结果:这项研究表明,粘蛋白20(MUC20)的下调可以预测MM患者的PI敏感性和预后。此外,MUC20通过抑制细胞周期蛋白依赖性激酶抑制剂2A表达(CDKN2A)诱导细胞凋亡来减弱MM细胞的PI抗性,这是通过阻碍MET原癌基因来实现的,受体酪氨酸激酶(MET)激活。此外,MUC20通过抑制PI抗性MM细胞中的胰岛素样生长因子受体-1(IGF-1R)的乳酸化抑制MET激活。本研究首次对MM进行染色体外环状DNA(eccDNA)测序,结果表明,eccDNA通过扩增驱动蛋白家族成员3C(KIF3C)来降低MM中MUC20的表达,从而诱导PI抗性。
    结论:我们的发现表明,eccDNA调节的MUC20通过调节细胞凋亡来减轻MM的PI抵抗,这将为PI耐药MM的治疗提供新的策略。
    BACKGROUND: Proteasome inhibitors (PIs) are one of the most important classes of drugs for the treatment of multiple myeloma (MM). However, almost all patients with MM develop PI resistance, resulting in therapeutic failure. Therefore, the mechanisms underlying PI resistance in MM require further investigation.
    METHODS: We used several MM cell lines to establish PI-resistant MM cell lines. We performed RNA microarray and EccDNA-seq in MM cell lines and collected human primary MM samples to explore gene profiles. We evaluated the effect of MUC20 on cuproptosis of PI-resistant MM cells using Co-immunoprecipitation (Co-IP), Seahorse bioenergetic profiling and in vivo assay.
    RESULTS: This study revealed that the downregulation of Mucin 20 (MUC20) could predict PI sensitivity and outcomes in MM patients. Besides, MUC20 attenuated PI resistance in MM cells by inducing cuproptosis via the inhibition of cyclin-dependent kinase inhibitor 2 A expression (CDKN2A), which was achieved by hindering MET proto-oncogene, receptor tyrosine kinase (MET) activation. Moreover, MUC20 suppressed MET activation by repressing insulin-like growth factor receptor-1 (IGF-1R) lactylation in PI-resistant MM cells. This study is the first to perform extrachromosomal circular DNA (eccDNA) sequencing for MM, and it revealed that eccDNA induced PI resistance by amplifying kinesin family member 3 C (KIF3C) to reduce MUC20 expression in MM.
    CONCLUSIONS: Our findings indicated that MUC20 regulated by eccDNA alleviates PI resistance of MM by modulating cuproptosis, which would provide novel strategies for the treatment of PI-resistant MM.
    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
    由于多发性神经病的罕见性,器官肿大,内分泌病,单克隆丙种球蛋白病,和皮肤变化(POEMS)综合征,最佳的一线治疗尚未建立,尽管指南中有几种选择。优选的治疗根据医生的偏好和轶事而变化。
    首先,分析以四个周期治疗为诱导方案的POEMS综合征治疗新模式的疗效,随后序贯移植作为符合移植条件的患者的巩固方案,或接受另一个两个周期的治疗移植不合格的患者。第二,比较蛋白酶体抑制剂(硼替佐米-环磷酰胺-地塞米松,BCD)或不含蛋白酶体抑制剂(环磷酰胺-地塞米松±沙利度胺,CD±T)。
    我们使用首都医科大学的真实数据进行了回顾性研究,宣武医院。
    共有34名新诊断的POEMS综合征患者符合Dispenzieri的诊断标准,以及从2013年7月至2021年3月完成至少4个周期治疗的患者纳入.
    这种新治疗模式的总体血管内皮生长因子(VEGF)反应率为100%。累计VEGF完全缓解(CRV)率为67.9%,累积完全血液学应答(CRH)率为55.6%。在中位49个月的随访期间,5年总生存率(OS)为90.7%,3年无进展生存率(PFS)为78.4%,5年PFS率为73.8%。BCD方案实现了75%的CRV率(从诊断到CRV的中位时间=130天)和66.7%的CRH率(从诊断到CRH的中位时间=218天)。此外,4个周期诱导治疗后,VEGF反应小于部分缓解(PRV),which,合并治疗后1年,总体神经限制量表减少少于3分,是一个独立的不良预后因素。
    POEMS综合征患者对硼替佐米的耐受性良好。与CD±T方案相比,BCD作为诱导方案实现了更好的VEGF反应和更早的血液学缓解。自体干细胞移植作为巩固治疗,进一步提高了神经和血液学的缓解率,导致更好的OS和PFS。
    UNASSIGNED: Due to the rarity of polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes (POEMS) syndrome, the best first-line treatment has not been established, although there are several options in guidelines. The preferred treatments vary according to the preference of the physician and anecdote.
    UNASSIGNED: First, to analyze the efficacy of a new treatment mode in POEMS syndrome that uses the four-cycle treatment as the induction regimen, followed by sequential transplantation as the consolidation regimen for transplantation-eligible patients, or received another two-cycle treatment for transplantation-ineligible patients. Second, to compare the efficacy and safety of regimens with a proteasome inhibitor (bortezomib-cyclophosphamide-dexamethasone, BCD) or without a proteasome inhibitor (cyclophosphamide-dexamethasone ± thalidomide, CD ± T).
    UNASSIGNED: We conducted a retrospective study using real-world data from Capital Medical University, Xuanwu Hospital.
    UNASSIGNED: A total of 34 newly diagnosed POEMS syndrome patients met Dispenzieri\'s diagnostic criteria, and those who completed at least four cycles of treatment from July 2013 to March 2021 were included.
    UNASSIGNED: The overall vascular endothelial growth factor (VEGF) response rate of this new treatment mode was 100%. The cumulative VEGF complete remission (CRV) rate was 67.9%, and the cumulative complete hematological response (CRH) rate was 55.6%. During the median 49-month follow-up, the 5-year-overall survival (OS) rate was 90.7%, the 3-year-progression-free survival (PFS) rate was 78.4%, and the 5-year-PFS rate was 73.8%. The BCD regimen achieved a 75% CRV rate (median time from diagnosis to CRV = 130 days) and 66.7% CRH rate (median time from diagnosis to CRH = 218 days). In addition, the VEGF response was less than the partial remission (PRV) after four-cycle induction treatment, which, together with a decrease on the Overall Neurological Limitation Scale of less than three points 1 year after consolidation treatment, was an independent poor prognostic factor.
    UNASSIGNED: Bortezomib was well-tolerated by patients with POEMS syndrome. Compared with CD ± T regimen, BCD as the induction regimen achieved better VEGF response and earlier hematological remission. Autologous stem cell transplantation used as consolidation therapy further improved the neurological and hematological remission rates, resulting in better OS and PFS.
    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
    FMS样酪氨酸激酶3(FLT3-ITDs)的内部串联重复突变发生在25%-30%的急性髓性白血病(AML)患者中,并与预后不良有关。尽管FLT3抑制剂已经证明了最初的临床疗效,FLT3-ITDAML患者的总体结局仍然很差,强调迫切需要制定更有效的治疗策略。在这项研究中,我们发现FLT3抑制剂降低了抗癌蛋白p53的蛋白质稳定性,导致耐药性。用蛋白酶体抑制剂阻断p53降解可恢复细胞内p53蛋白水平,与FLT3-ITD抑制剂联合使用,在细胞中显示针对FLT3-ITDAML的优异治疗效果,小鼠模型,和病人。这些数据表明,这种组合治疗方法可能代表了靶向FLT3-ITDAML的有希望的策略。
    Internal tandem duplication mutations of the FMS-like tyrosine kinase-3 (FLT3-ITDs) occur in 25%-30% of patients with acute myeloid leukemia (AML) and are associated with dismal prognosis. Although FLT3 inhibitors have demonstrated initial clinical efficacy, the overall outcome of patients with FLT3-ITD AML remains poor, highlighting the urgency to develop more effective treatment strategies. In this study, we reveal that FLT3 inhibitors reduced protein stability of the anti-cancer protein p53, resulting in drug resistance. Blocking p53 degradation with proteasome inhibitors restores intracellular p53 protein levels and, in combination with FLT3-ITD inhibitors, shows superior therapeutic effects against FLT3-ITD AML in cells, mouse models, and patients. These data suggest that this combinatorial therapeutic approach may represent a promising strategy to target FLT3-ITD AML.
    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
    药物再利用是扩大肿瘤治疗范围的一个有吸引力的策略。托哌酮是一种用于治疗慢性疼痛病症的古老的中枢作用肌肉松弛剂。在这项研究中,我们研究了托哌酮对人类癌症的治疗效果和机制,并探索了蛋白酶体抑制剂和免疫治疗的联合策略。
    通过测量半数最大抑制浓度来评估托哌酮的抗肿瘤作用,细胞死亡和细胞生长。RNA测序,西方印迹,分子对接,进行酶活性测定和ChIP-qPCR以揭示其潜在机制。使用异种移植模型来评估托哌酮单独或与蛋白酶体抑制剂或免疫疗法组合的功效。
    托哌酮在人癌细胞系中抑制细胞生长并诱导细胞死亡。未折叠蛋白反应(UPR)途径在托哌酮处理的细胞中过度激活。我们进一步确定组蛋白赖氨酸特异性脱甲基酶1(LSD1)作为托哌酮的潜在靶标,直接去甲基化H3K4me2中的UPR相关基因。通过将M2巨噬细胞重编程为M1表型,托哌酮通过增强UPR和对免疫疗法的敏化肿瘤协同地改善MG132的功效。
    托哌酮通过靶向LSD1抑制人类癌症。通过组合策略在癌症治疗中重新使用托哌酮意味着临床潜力。
    UNASSIGNED: Drug repurposing is an attractive strategy for extending the arsenal of oncology therapies. Tolperisone is an old centrally acting muscle relaxant used for treatment of chronic pain conditions. In this study, we investigated the therapeutic effect and mechanism of tolperisone in human cancers and explored the combination strategy with proteasome inhibitor and immunotherapy.
    UNASSIGNED: The antitumor effect of tolperisone was evaluated by measuring half maximal inhibitory concentration, cell death, and cell growth. RNA sequencing, western blotting, molecular docking, enzyme activity assay, and ChIP-qPCR were performed to reveal the underlying mechanism. Xenograft models were used to evaluate the efficacy of tolperisone alone or in combination with proteasome inhibitor or immunotherapy.
    UNASSIGNED: Tolperisone inhibited cell growth and induced cell death in human cancer cell lines. Unfolded protein responses (UPR) pathway was hyperactivated in tolperisone-treated cells. We further identified histone lysine-specific demethylase 1 (LSD1) as a potential target of tolperisone, which directly demethylates UPR-related genes in H3K4me2. Tolperisone synergistically improved the efficacy of MG132 by enhancing UPR and sensitized tumors to immunotherapy by reprogramming M2 macrophages into M1 phenotype.
    UNASSIGNED:
    Tolperisone inhibits human cancer by targeting LSD1. Repurposing tolperisone in cancer therapy by a combination strategy implies clinical potential.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    CD13抑制剂ubenimex由于其作为免疫增强剂的功能而被用作化疗的辅助药物,用于治疗癌症。但它在细胞毒性功效方面有局限性。蛋白酶体抑制剂ixazomib是治疗多发性骨髓瘤的具有高抗癌活性的标志性药物。在这里,我们将乌苯美司的药效团和艾沙唑米的硼酸偶联,获得CD13和蛋白酶体双重抑制剂7(BC-05)。BC-05对人CD13(IC50=0.13μM)和20S蛋白酶体(IC50=1.39μM)均表现出有效的抑制活性。尽管BC-05在体外显示出比Ixazomib更低的抗增殖活性,在体内抗癌功效和延长生存时间方面建立了优势,这可能是由于其抗转移和免疫刺激活性。药代动力学研究表明,BC-05是一种潜在的口服活性剂,F%值为24.9%。此外,在初步毒性研究中,BC-05比艾沙唑米表现出更有利的安全性。总的来说,结果表明,BC-05是治疗多发性骨髓瘤的有希望的候选药物.
    The CD13 inhibitor ubenimex is used as an adjuvant drug with chemotherapy for the treatment of cancer due to its function as an immunoenhancer, but it has limitations in its cytotoxic efficacy. The proteasome inhibitor ixazomib is a landmark drug in the treatment of multiple myeloma with a high anti-cancer activity. Herein, we conjugated the pharmacophore of ubenimex and the boric acid of ixazomib to obtain a dual CD13 and proteasome inhibitor 7 (BC-05). BC-05 exhibited potent inhibitory activity on both human CD13 (IC50 = 0.13 μM) and the 20S proteasome (IC50 = 1.39 μM). Although BC-05 displayed lower anti-proliferative activity than that of ixazomib in vitro, an advantage was established in the in vivo anti-cancer efficacy and prolongation of survival time, which may be due to its anti-metastatic and immune-stimulating activity. A pharmacokinetic study revealed that BC-05 is a potentially orally active agent with an F% value of 24.9%. Moreover, BC-05 showed more favorable safety profiles than those of ixazomib in preliminary toxicity studies. Overall, the results indicate that BC-05 is a promising drug candidate for the treatment of multiple myeloma.
    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
    免疫蛋白酶体是蛋白酶体的变体,在20S亚基中具有结构差异,可优化它们以产生与主要组织相容性复合物(MHC)-I分子具有更高结合亲和力的抗原肽。除了抗原呈递的主要功能外,免疫蛋白酶体也负责蛋白质的降解,用于维持蛋白质稳态的未折叠蛋白质和有助于肿瘤进展的肿瘤抑制蛋白质。在癌症中经常观察到免疫蛋白酶体的表达改变;然而,其表达水平和作用在不同癌症类型之间有所不同,在肿瘤发展中具有拮抗作用。本文综述了免疫蛋白酶体在不同癌症类型中的二分法作用。并总结了目前免疫蛋白酶体激活剂和抑制剂的进展。特异性靶向免疫蛋白酶体可能是癌症治疗中的有益治疗干预,了解免疫蛋白酶体在癌症中的作用将为预防和治疗癌症提供重要的治疗见解。
    Immunoproteasome is a variant of proteasome with structural differences in 20S subunits optimizing them for the production of antigenic peptides with higher binding affinity to major histocompatibility complex (MHC)-I molecules. Apart from this primary function in antigen presentation, immunoproteasome is also responsible for the degradation of proteins, both unfolded proteins for the maintenance of protein homeostasis and tumor suppressor proteins contributing to tumor progression. The altered expression of immunoproteasome is frequently observed in cancers; however, its expression levels and effects vary among different cancer types exhibiting antagonistic roles in tumor development. This review focuses on the dichotomous role of immunoproteasome in different cancer types, as well as summarizes the current progression in immunoproteasome activators and inhibitors. Specifically targeting immunoproteasome may be a beneficial therapeutic intervention in cancer treatment and understanding the role of immunoproteasome in cancers will provide a significant therapeutic insight for the prevention and treatment of cancers.
    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
    背景:综述多发性骨髓瘤(MM)的发病机制和治疗方法。MM是骨髓中浆细胞增殖异常的血液系统恶性肿瘤。由于耐药性的出现,MM仍然是无法治愈的恶性肿瘤,这就需要进一步探索发病机制和有效的治疗靶点。
    方法:在本文中,采用文献综述的方法获取有关MM的信息。根据文献,进行了全面系统的审查。
    结果:MM是一个复杂的病理生理过程,具有很大的异质性,主要表现在基因组不稳定和骨髓微环境。目前,MM的治疗有了很大的进步,蛋白酶体抑制剂和免疫调节药物在临床上应用广泛。异基因干细胞移植可能是治疗MM的唯一有希望的方法。其移植相关的高死亡率限制了其临床应用。
    结论:MM治疗的未来在于开发更有针对性的治疗方法,新的免疫疗法,和更好地了解疾病的分子和遗传基础。
    To review the pathogenesis and treatment of multiple myeloma (MM). MM is a hematological malignancy with abnormal plasma cell proliferation in bone marrow. Due to the emergence of drug resistance, MM is still an incurable malignancy, which requires further exploration of pathogenesis and effective therapeutic targets.
    In this paper, the method of literature review is adopted to obtain the information about MM. Based on the literature, comprehensive and systematic review is made.
    MM is a complex pathophysiological process with great heterogeneity, mainly reflected in genomic instability and bone marrow microenvironment. At present, the treatment of MM has made great progress, proteasome inhibitors and immunomodulatory drugs are widely used in clinic. Allogeneic stem cell transplantation may be the only promising cure for MM, and its high transplant-related mortality limits its clinical application.
    The future of MM treatment lies in the development of more targeted therapies, novel immunotherapies, and a better understanding of the disease\'s molecular and genetic basis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号