Panobinostat

Panobinostat
  • 文章类型: Journal Article
    目前胰腺癌的5年生存率约为12%,使其成为最致命的恶性肿瘤之一.快速转移,获得性耐药性,和不良的患者预后需要更好的胰腺导管腺癌(PDAC)的治疗策略。多项研究表明,联合化疗治疗实体瘤是成功的。针对两个不同的新兴标志,如panobinostat(Pan)的非突变表观遗传变化和abemaciclib(Abe)的细胞周期进展延迟,抑制胰腺癌的生长.HDAC和CDK4/6抑制剂是有效的,但作为单一药剂容易产生耐药性和失效。因此,我们假设,Abe和Pan的联合用药可以协同和致命地影响PDAC的存活和增殖.多种基于细胞的检测,酶活性实验,并进行流式细胞术实验以确定Abe的影响,Pan,以及它们在PDAC细胞和人真皮成纤维细胞上的组合。Western印迹用于确定细胞周期的表达,表观遗传,和凋亡标志物。Abe-Pan组合表现出优异的疗效并产生协同作用,改变细胞周期蛋白和表观遗传标记的表达。Pan,单独和安倍一起,引起胰腺癌细胞凋亡。Abe-Pan共治疗在正常人真皮成纤维细胞中显示出相对安全性。我们的Abe和Pan的新型组合治疗对PDAC细胞显示出协同作用。该组合诱导细胞凋亡,显示相对安全,由于其在PDAC治疗中的治疗潜力,值得进一步研究。
    The current 5-year survival rate of pancreatic cancer is about 12%, making it one of the deadliest malignancies. The rapid metastasis, acquired drug resistance, and poor patient prognosis necessitate better therapeutic strategies for pancreatic ductal adenocarcinoma (PDAC). Multiple studies show that combining chemotherapeutics for solid tumors has been successful. Targeting two distinct emerging hallmarks, such as non-mutational epigenetic changes by panobinostat (Pan) and delayed cell cycle progression by abemaciclib (Abe), inhibits pancreatic cancer growth. HDAC and CDK4/6 inhibitors are effective but are prone to drug resistance and failure as single agents. Therefore, we hypothesized that combining Abe and Pan could synergistically and lethally affect PDAC survival and proliferation. Multiple cell-based assays, enzymatic activity experiments, and flow cytometry experiments were performed to determine the effects of Abe, Pan, and their combination on PDAC cells and human dermal fibroblasts. Western blotting was used to determine the expression of cell cycle, epigenetic, and apoptosis markers. The Abe-Pan combination exhibited excellent efficacy and produced synergistic effects, altering the expression of cell cycle proteins and epigenetic markers. Pan, alone and in combination with Abe, caused apoptosis in pancreatic cancer cells. Abe-Pan co-treatment showed relative safety in normal human dermal fibroblasts. Our novel combination treatment of Abe and Pan shows synergistic effects on PDAC cells. The combination induces apoptosis, shows relative safety, and merits further investigation due to its therapeutic potential in the treatment of PDAC.
    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:食管癌仍然是一种具有挑战性的疾病,死亡率高,治疗选择少。鉴于这些困难,表观遗传药物已成为患者护理的潜在替代品。这项研究的目标是评估Panobinostat治疗的效果和生物学后果,HDAC(组蛋白去乙酰化酶)抑制剂已被批准用于多发性骨髓瘤患者的治疗,在正常和恶性来源的食管细胞系中,后者代表了两种主要的组织学亚型:腺癌和鳞状细胞癌。
    结果:Panobinostat治疗抑制生长并阻碍增殖,食管癌细胞的集落形成和侵袭。考虑HDAC组织表达,与肿瘤组织相比,HDAC1在正常食管上皮中显著上调,而HDAC3与非恶性粘膜相比在食管癌中过度表达。在正常组织和肿瘤组织之间没有观察到HDAC2和HDAC8表达的差异。
    结论:Panobinostat暴露可有效损害食管癌细胞的恶性特征。因为HDAC3在食道肿瘤样本中显示过表达,这种表观遗传药物可能是食管癌患者的替代治疗选择.
    BACKGROUND: Oesophageal cancer remains a challenging disease with high mortality rates and few therapeutic options. In view of these difficulties, epigenetic drugs have emerged as potential alternatives for patient care. The goal of this study was to evaluate the effect and biological consequences of Panobinostat treatment, an HDAC (histone deacetylase) inhibitor already approved for treatment of patients with multiple myeloma, in oesophageal cell lines of normal and malignant origin, with the latter being representative of the two main histological subtypes: adenocarcinoma and squamous cell carcinoma.
    RESULTS: Panobinostat treatment inhibited growth and hindered proliferation, colony formation and invasion of oesophageal cancer cells. Considering HDAC tissue expression, HDAC1 was significantly upregulated in normal oesophageal epithelium in comparison with tumour tissue, whereas HDAC3 was overexpressed in oesophageal cancer compared to non-malignant mucosa. No differences between normal and tumour tissue were observed for HDAC2 and HDAC8 expression.
    CONCLUSIONS: Panobinostat exposure effectively impaired malignant features of oesophageal cancer cells. Because HDAC3 was shown to be overexpressed in oesophageal tumour samples, this epigenetic drug may represent an alternative therapeutic option for oesophageal cancer patients.
    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
    由于多发性骨髓瘤的难治性,非常需要进行基础研究以扩大多发性骨髓瘤的治疗选择。组蛋白去乙酰化酶(HDAC)抑制剂,它们是表观遗传调节因子,有吸引力,但应用有限。microRNAs(miRNAs),它们也是表观遗传调节因子,是可能导致未来治疗突破的重要分子。在这项研究中,我们全面搜索了骨髓瘤细胞中HDAC抑制剂改变的miRNA。我们将miR-7-5p(miR-7)鉴定为由HDAC抑制剂下调的miRNA。用miR-7转染骨髓瘤细胞系抑制细胞增殖,诱导细胞凋亡,并增强了HDAC抑制剂帕比司他的作用。抑制c-Myc下调miR-7的表达,但被硼替佐米上调.miR-7靶标的综合检查揭示了四种候选物:SLC6A9、LRRC59、EXOSC2和PSME3。其中,我们关注的是PSME3,一种与骨髓瘤细胞蛋白酶体能力有关的癌基因.PSME3敲低增加骨髓瘤细胞死亡和panobinostat敏感性。总之,miR-7被HDAC抑制剂下调,是一种靶向PSME3的肿瘤抑制剂。这种miR-7下调可能与HDAC抑制剂抗性有关。此外,应考虑补体miRNA表达变化的抗骨髓瘤药物组合.
    Basic research to expand treatment options for multiple myeloma is greatly needed due to the refractory nature of the disease. Histone deacetylase (HDAC) inhibitors, which are epigenetic regulators, are attractive but have limited applications. MicroRNAs (miRNAs), which are also epigenetic regulators, are important molecules that may lead to future therapeutic breakthroughs. In this study, we comprehensively searched for miRNAs that are altered by HDAC inhibitors in myeloma cells. We identified miR-7-5p (miR-7) as a miRNA downregulated by HDAC inhibitors. Transfection of myeloma cell lines with miR-7 suppressed cell proliferation, induced apoptosis, and enhanced the effects of the HDAC inhibitor panobinostat. Expression of miR-7 was downregulated by c-Myc inhibition, but upregulated by bortezomib. Comprehensive examination of miR-7 targets revealed four candidates: SLC6A9, LRRC59, EXOSC2, and PSME3. Among these, we focused on PSME3, an oncogene involved in proteasome capacity in myeloma cells. PSME3 knockdown increases myeloma cell death and panobinostat sensitivity. In conclusion, miR-7, which is downregulated by HDAC inhibitors, is a tumor suppressor that targets PSME3. This miR-7 downregulation may be involved in HDAC inhibitor resistance. In addition, combinations of anti-myeloma drugs that complement changes in miRNA expression should be considered.
    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
    小胶质细胞,中枢神经系统(CNS)中的固有免疫细胞,在维持中枢神经系统稳态中起关键作用。然而,响应脑损伤激活的小胶质细胞产生各种炎症介质,包括一氧化氮(NO)和促炎细胞因子,导致相当大的神经元损伤。诱导型NO合酶(iNOS)产生的NO与超氧化物迅速反应形成剧毒产物,过氧亚硝酸盐.因此,iNOS被认为是脑缺血的推定治疗靶点。这里,我们检查了帕比司他(Pano)的作用,组蛋白脱乙酰酶抑制剂,使用大鼠永生化小胶质细胞HAPI细胞对脂多糖(LPS)诱导的iNOS表达的影响。Pano以剂量依赖的方式抑制LPS诱导的iNOSmRNA表达和NO产生;然而,它对LPS诱导的c-Jun氨基末端激酶(JNK)和p38的激活或核因子-κB(NF-κB)的核易位影响很小。干扰素-β(IFN-β)/信号转导和转录激活因子(STAT)途径对于LPS诱导的巨噬细胞/小胶质细胞中iNOS的表达至关重要。我们还检查了Pano对LPS诱导的IFN-β信号传导的影响。Pano显著抑制LPS诱导的IFN-β表达和随后的STAT1酪氨酸磷酸化。然而,IFN-β的添加恢复了降低的STAT1磷酸化,但没有降低的iNOS表达。此外,Pano抑制了LPS增加的八聚体结合蛋白2和干扰素调节因子9的表达,但是IFN-β的添加也未能恢复这些因子的表达降低。因此,我们得出的结论是,Pano的抑制作用不仅归因于IFN-β/STAT轴的抑制,而且还归因于该轴未涉及的其他因子的下调。
    Microglia, resident immune cells in the central nervous system (CNS), play a critical role in maintaining CNS homeostasis. However, microglia activated in response to brain injury produce various inflammatory mediators, including nitric oxide (NO) and proinflammatory cytokines, leading to considerable neuronal damage. NO generated by inducible NO synthase (iNOS) rapidly reacts with superoxide to form a highly toxic product, peroxynitrite. Therefore, iNOS is considered to be a putative therapeutic target for cerebral ischemia. Here, we examined the effects of panobinostat (Pano), a histone deacetylase inhibitor, on lipopolysaccharide (LPS)-induced iNOS expression using rat immortalized microglia HAPI cells. Pano inhibited LPS-induced expression of iNOS mRNA and NO production in a dose-dependent manner; however, it had little effect on the LPS-induced activation of c-Jun N-terminal kinase (JNK) and p38 or nuclear translocation of nuclear factor-κB (NF-κB). The interferon-β (IFN-β)/signal transducer and activator of transcription (STAT) pathway is essential for LPS-induced iNOS expression in macrophages/microglia. We also examined the effects of Pano on LPS-induced IFN-β signaling. Pano markedly inhibited LPS-induced IFN-β expression and subsequent tyrosine phosphorylation of STAT1. However, the addition of IFN-β restored the decreased STAT1 phosphorylation but not the decreased iNOS expression. In addition, Pano inhibited the LPS-increased expression of octamer binding protein-2 and interferon regulatory factor 9 responsible for iNOS expression, but IFN-β addition also failed to restore the decreased expression of these factors. Thus, we conclude that the inhibitory effects of Pano are due not only to the inhibition of the IFN-β/STAT axis but also to the downregulation of other factors not involved in this axis.
    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
    背景:软组织肉瘤(STS)是罕见的疾病,通常由儿童和成人的结缔组织引起。然而,参与STS治疗的化疗可能会引起毒副作用和多药耐药,使治疗更具挑战性。组蛋白脱乙酰酶抑制剂(HDACi)是表观遗传剂,已显示出作为单一药物以及与其他药物联合使用的抗肿瘤作用。我们的项目旨在证明STS中的相同效果。
    方法:根据我们以前的研究,选择帕比司他(LBH589)加阿霉素进行研究。在上皮样肉瘤模型中尝试肿瘤异种移植物以验证体内良好的协同作用,并且使用平滑肌肉瘤模型作为阴性比较组。随后研究了基因谱变化。对HDACi可能引起的通路变化进行了探索和验证。
    结果:在STS细胞系和上皮样肉瘤小鼠模型中成功验证了LBH589加多柔比星的协同作用。我们试图将阿霉素的剂量降低到较低水平,发现该药物组合仍然可以抑制小鼠的肿瘤大小。此外,通过RNA测序分析研究了由LBH589引起的基因谱变化。结果表明,LBH589可以对一组靶基因发挥作用,这些靶基因可以调节潜在的生物学功能,尤其是在细胞周期通路中。
    BACKGROUND: Soft tissue sarcomas (STS) are rare diseases typically arising from connective tissues in children and adults. However, chemotherapies involved in the treatment of STS may cause toxic side effects and multi-drug chemoresistance, making the treatment even more challenging. Histone deacetylase inhibitors (HDACi) are epigenetic agents which have shown anti-tumor effects as single agent as well as combination use with other drugs. Our project intends to prove the same effects in STS.
    METHODS: Panobinostat (LBH589) plus doxorubicin was selected for investigations based on our previous research. Tumor xenografts were tried in an epithelioid sarcoma model to validate good synergy effects in vivo and a leiomyosarcoma model was used as a negative comparison group. Gene profile changes were studied afterwards. The possible pathway changes caused by HDACi were explored and validated by several assays.
    RESULTS: Synergy effect of LBH589 plus doxorubicin was successfully validated in STS cell lines and an epithelioid sarcoma mice model. We tried to reduce the dose of doxorubicin to a lower level and found the drug combination can still inhibit tumor size in mice. Furthermore, gene profile changes caused by LBH589 was studied by RNA-Sequencing analysis. Results showed LBH589 can exert effects on a group of target genes which can regulate potential biological functions especially in the cell cycle pathway.
    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
    过度表达人表皮生长因子受体2(HER2)的乳腺癌预后不良。此外,可用的化学疗法由于选择性差而引起许多副作用。为了推进更有效和更安全的HER2阳性乳腺癌治疗,我们探索了药物递送技术和免疫治疗的融合。我们的研究导致了加载帕比司他和曲妥珠单抗抗体功能化的免疫囊体的设计,能够精确靶向过表达HER2的乳腺癌细胞。我们使用小角度X射线散射(SAXS)表征了立方体的纳米结构,低温透射电子显微镜(cryo-TEM),和动态光散射(DLS)。此外,我们通过傅里叶变换红外光谱(FTIR)和十二烷基硫酸钠聚丙烯酰胺凝胶电泳(SDS-PAGE)证实了免疫囊体上曲妥珠单抗抗体的完整性.此外,我们发现panobinostat负载的免疫囊体细胞毒性更大,以吸收依赖的方式,与代表健康细胞的细胞系(L929)相比,HER2阳性乳腺癌细胞系(SKBR3)。这些结果支持具有抗体的立方体的功能化增强了负载药物的有效性及其靶向HER2阳性乳腺癌细胞的选择性。
    Breast cancers that overexpress human epidermal growth factor receptor 2 (HER2) have poor prognosis. Moreover, available chemotherapies cause numerous side effects due to poor selectivity. To advance more effective and safer therapies for HER2-positive breast cancer, we explored the fusion of drug delivery technology and immunotherapy. Our research led to the design of immunocubosomes loaded with panobinostat and functionalized with trastuzumab antibodies, enabling precise targeting of breast cancer cells that overexpress HER2. We characterised the nanostructure of cubosomes using small-angle X-ray scattering (SAXS), cryo-transmission electron microscopy (cryo-TEM), and dynamic light scattering (DLS). Moreover, we confirmed the integrity of the trastuzumab antibodies on the immunocubosomes by Fourier-transform infrared spectroscopy (FTIR) and sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Additionally, we found that panobinostat-loaded immunocubosomes were more cytotoxic, and in an uptake-dependant manner, towards a HER2-positive breast cancer cell line (SKBR3) compared to a cell line representing healthy cells (L929). These results support that the functionalization of cubosomes with antibodies enhances both the effectiveness of the loaded drug and its selectivity for targeting HER2-positive breast cancer cells.
    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
    背景:哺乳动物或雷帕霉素复合物1(mTORC1)的机制靶标是癌症等疾病的有效治疗靶标,糖尿病,老化,和神经变性。然而,缺乏监测活细胞或组织中mTORC1抑制的有效工具。
    结果:我们开发了一种称为TORSEL的基因编码mTORC1传感器。当4EBP1去磷酸化发生并与eIF4E相互作用时,该传感器将其荧光模式从扩散变为点状。TORSEL可以特别感知生理,药理学,以及对活细胞和组织中mTORC1信号传导的遗传抑制。重要的是,TORSEL是用于mTORC1抑制剂的基于成像的视觉筛选的有价值的工具。使用TORSEL,我们鉴定了组蛋白去乙酰化酶抑制剂,其选择性阻断营养敏感信号以抑制mTORC1.
    结论:TORSEL是一种独特的活细胞传感器,可有效检测mTORC1活性的抑制,组蛋白脱乙酰酶抑制剂如帕比司他通过氨基酸传感靶向mTORC1信号。
    BACKGROUND: Mammalian or mechanistic target of rapamycin complex 1 (mTORC1) is an effective therapeutic target for diseases such as cancer, diabetes, aging, and neurodegeneration. However, an efficient tool for monitoring mTORC1 inhibition in living cells or tissues is lacking.
    RESULTS: We developed a genetically encoded mTORC1 sensor called TORSEL. This sensor changes its fluorescence pattern from diffuse to punctate when 4EBP1 dephosphorylation occurs and interacts with eIF4E. TORSEL can specifically sense the physiological, pharmacological, and genetic inhibition of mTORC1 signaling in living cells and tissues. Importantly, TORSEL is a valuable tool for imaging-based visual screening of mTORC1 inhibitors. Using TORSEL, we identified histone deacetylase inhibitors that selectively block nutrient-sensing signaling to inhibit mTORC1.
    CONCLUSIONS: TORSEL is a unique living cell sensor that efficiently detects the inhibition of mTORC1 activity, and histone deacetylase inhibitors such as panobinostat target mTORC1 signaling through amino acid sensing.
    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
    我们旨在使用高通量药物筛选(HTS)和基因组测序方法确定PitNETs的可药用细胞固有漏洞和基于靶标的药物治疗。我们检查了HTS中9个患者来源的PitNET原代细胞。根据筛选结果,通过基因组测序分析了180个PitNETs的潜在靶基因.我们确定并验证了最有效的药物之一,在体外和体内PitNET模型中均靶向组蛋白脱乙酰酶(HDACs)。进一步的RNA测序揭示了用代表性的HDACs抑制剂治疗后的潜在分子机制,Panobinostat.HTS产生了总共20,736种单药剂量反应,这些反应在多种致癌靶标的多种抑制剂中富集。包括HDAC,PI3K,mTOR,和蛋白酶体。在这些药物中,HDAC抑制剂(HDACIs),平均而言,最有效的药物类别。使用体外的进一步研究,在体内,和隔离的PitNET主细胞模型验证HDACIs,尤其是Panobinostat,作为一种有前途的治疗剂。转录调查显示,Panobinostat治疗后Nrf2信号发生了实质性变化。此外,Nrf2在PitNETs中高度表达。Panobinostat和Nrf2抑制剂ML385的组合对PitNET抑制具有协同作用。目前的研究揭示了一类有效的抗PitNET药物,HDACIs,基于HTS和基因组测序。代表性化合物之一,Panobinostat,可能是通过Nrf2介导的氧化还原调节PitNET治疗的潜在药物。Panobinostat和ML385的组合进一步增强了PitNET治疗的有效性。
    We aimed to identify the druggable cell-intrinsic vulnerabilities and target-based drug therapies for PitNETs using the high-throughput drug screening (HTS) and genomic sequencing methods. We examined 9 patient-derived PitNET primary cells in HTS. Based on the screening results, the potential target genes were analyzed with genomic sequencing from a total of 180 PitNETs. We identified and verified one of the most potentially effective drugs, which targeted the Histone deacetylases (HDACs) both in in vitro and in vivo PitNET models. Further RNA sequencing revealed underlying molecular mechanisms following treatment with the representative HDACs inhibitor, Panobinostat. The HTS generated a total of 20,736 single-agent dose responses which were enriched among multiple inhibitors for various oncogenic targets, including HDACs, PI3K, mTOR, and proteasome. Among these drugs, HDAC inhibitors (HDACIs) were, on average, the most potent drug class. Further studies using in vitro, in vivo, and isolated PitNET primary cell models validated HDACIs, especially Panobinostat, as a promising therapeutic agent. Transcriptional surveys revealed substantial alterations to the Nrf2 signaling following Panobinostat treatment. Moreover, Nrf2 is highly expressed in PitNETs. The combination of Panobinostat and Nrf2 inhibitor ML385 had a synergistic effect on PitNET suppression. The current study revealed a class of effective anti-PitNET drugs, HDACIs, based on the HTS and genomic sequencing. One of the representative compounds, Panobinostat, may be a potential drug for PitNET treatment via Nrf2-mediated redox modulation. Combination of Panobinostat and ML385 further enhance the effectiveness for PitNET treatment.
    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
    蛋白质合成和加工的高速率使多发性骨髓瘤(MM)细胞易受蛋白质稳态的干扰。通过用蛋白酶体抑制剂(PI)靶向蛋白质降解来诱导蛋白毒性应激已经彻底改变了MM的治疗。然而,对PI的耐药性是不可避免的,并且是一个持续的临床挑战。我们首次研究核糖体RNA基因的RNA聚合酶I转录的选择性抑制剂,CX-5461已在六名严重预处理的MM患者中的三名中证明了抗肿瘤活性的潜在信号。这里,我们表明,CX-5461在PI耐药的MM临床前模型中在体外和体内具有有效的抗骨髓瘤活性。除了抑制核糖体生物发生,CX-5461导致拓扑异构酶II捕获和复制依赖性DNA损伤,导致G2/M细胞周期停滞和凋亡细胞死亡。将CX-5461与PI组合不会进一步增强CX-5461的体内抗骨髓瘤活性。相比之下,CX-5461通过不同的机制靶向核糖体生物发生和蛋白质合成,在MM的Vk*MYC和5T33-KaLwRij小鼠模型中都显示出与组蛋白脱乙酰酶抑制剂panobinostat的协同相互作用。因此,我们的发现提供了强有力的证据来促进靶向核糖体治疗复发和难治性MM的临床发展。
    The high rates of protein synthesis and processing render multiple myeloma (MM) cells vulnerable to perturbations in protein homeostasis. The induction of proteotoxic stress by targeting protein degradation with proteasome inhibitors (PIs) has revolutionized the treatment of MM. However, resistance to PIs is inevitable and represents an ongoing clinical challenge. Our first-in-human study of the selective inhibitor of RNA polymerase I transcription of ribosomal RNA genes, CX-5461, has demonstrated a potential signal for anti-tumor activity in three of six heavily pre-treated MM patients. Here, we show that CX-5461 has potent anti-myeloma activity in PI-resistant MM preclinical models in vitro and in vivo. In addition to inhibiting ribosome biogenesis, CX-5461 causes topoisomerase II trapping and replication-dependent DNA damage, leading to G2/M cell-cycle arrest and apoptotic cell death. Combining CX-5461 with PI does not further enhance the anti-myeloma activity of CX-5461 in vivo. In contrast, CX-5461 shows synergistic interaction with the histone deacetylase inhibitor panobinostat in both the Vk∗MYC and the 5T33-KaLwRij mouse models of MM by targeting ribosome biogenesis and protein synthesis through distinct mechanisms. Our findings thus provide strong evidence to facilitate the clinical development of targeting the ribosome to treat relapsed and refractory 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
    组蛋白去乙酰化酶(HDAC)抑制剂正在成为血液恶性肿瘤的有希望的治疗方法。具有潜在的应用扩展到实体瘤的未来。鉴于其广泛的生物学效应,迫切需要彻底了解与HDAC抑制相关的毒性。在这项研究中,使用FDA不良事件报告系统数据库进行药物警戒分析.通过各种统计方法检测到与HDAC抑制剂相关的可疑不良事件,包括报告赔率比,比例报告比率,信息组件,和经验贝叶斯几何均值。我们的研究结果表明,在报告的所有案件中,胃肠道疾病占队列患者的13%,而淋巴系统疾病占队列的8%,所有这些都表现为HDAC抑制剂诱导的不良事件。重要的是,发现HDAC抑制剂的使用与房颤事件有关,心力衰竭,呼吸衰竭,肝功能障碍,和急性肾损伤。Romidepsin和belinostat与panobinostat和vorinostat相比,表现出更明显的不良事件信号,强调需要对这一特定人群的不良事件进行警惕监测。此外,心房颤动(临床优先评分为7分)是最重要的医学事件,值得临床关注.最终,观察到在治疗开始后的第1个月和第2个月内出现多个不良事件.警惕监测和支持护理策略对于解决与HDAC抑制剂相关的毒性至关重要。特别是那些关于心脏毒性的,呼吸毒性,肾毒性,和肝毒性。
    Histone deacetylase (HDAC) inhibitors are emerging as promising treatments for hematological malignancies, with potential applications extending to solid tumors in the future. Given their wide-ranging biological effects, there is a pressing need for a thorough understanding of the toxicities linked to HDAC inhibition. In this study, a pharmacovigilance analysis was conducted using the FDA Adverse Event Reporting System database. Suspected adverse events linked to HDAC inhibitors were detected through various statistical methodologies, including reporting odds ratio, proportional reporting ratio, information component, and Empirical Bayes Geometric Mean. Our study findings have illuminated that, among the total reported cases examined, gastrointestinal disorders accounted for 13% patients of the cohort, while lymphatic system disorders comprised 8% cases of the cohort, all of which manifested as adverse events induced by HDAC inhibitors. Importantly, the usage of HDAC inhibitors was found to be associated with incidents of atrial fibrillation, heart failure, respiratory failure, hepatic dysfunction, and acute kidney injury. Romidepsin and belinostat demonstrated more pronounced signals of adverse events compared to panobinostat and vorinostat, emphasizing the need for vigilant monitoring of adverse events in this particular population. Furthermore, atrial fibrillation (clinical priority score of 7 points) emerged as the paramount medical event warranting utmost clinical attention. Eventually, multiple adverse events were observe to emerge within the initial and second months following the initiation of treatment. Vigilant monitoring and supportive care strategies are critical in addressing the toxicities associated with HDAC inhibitors, particularly those concerning cardiotoxicity, respiratory toxicity, renal toxicity, and hepatotoxicity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号