Drug combination

药物组合
  • 文章类型: Journal Article
    肝纤维化是危害人类健康的病理过程,迄今为止,有效的治疗方法仍然难以捉摸。芍药苷(PAE),中药芍药中的松果烷型单萜化合物,和二甲双胍(MET),口服双胍降血糖药,两者都具有抗炎和保护肝脏的作用。在目前的工作中,我们首次发现,PAE和MET的联合治疗在两种不同的动物模型中协同抑制肝纤维化的进展:治疗性和预防性。肝纤维化标志物表达水平的降低和组织病理学特征的改善证明了这种治疗效果。机制探索进一步表明,这种联合疗法下调TGF-β1和p-Smad2的表达,同时上调两种模型中Smad7的表达。重要的是,我们还发现,这种组合方法在两种模型中均显著降低了肝毒性和肾毒性.我们的研究结果表明肝纤维化的有效联合治疗,并为肝纤维化患者提供治疗改善的可能性。
    Liver fibrosis is a pathological process that endangers human health, for which effective treatments remain elusive to date. Paeoniflorin (PAE), a pineane-type monoter penoid compound from the traditional Chinese medicine PaeoniaeRubra Radix, and metformin (MET), an oral biguanide hypoglycemic agent, both demonstrate anti-inflammatory and hepatoprotective effects. In current work, we first discovered that the combined treatment of PAE and MET synergistically inhibited the progression of liver fibrosis in two different animal models: therapeutic and preventive. This therapeutic effect is evidenced by a reduction in the expression levels of liver fibrosis markers and an improvement in histopathological characteristics. Mechanistic exploration further revealed that this combination therapy downregulated the expression of TGF-β1 and p-Smad2, while upregulating Smad7 expression in both models. Importantly, we also found that this combinatorial approach significantly reduced hepatotoxicity and nephrotoxicity in both models. Our findings suggest an effective combination therapy for liver fibrosis and provide the possibility of therapeutic improvement for patients with liver fibrosis.
    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
    抗生素耐药性是一个日益严重的全球性问题,需要创新的治疗方法和施用抗生素的策略。一种有希望的方法是联合治疗,其中两种或两种以上的药物联合来对抗感染。沿着这条线,抗菌肽(AMPs)与常规抗生素的组合主要由于AMPs和常规抗生素的互补作用机制而受到关注。在这篇文章中,我们回顾了探索AMPs与抗生素之间协同作用的体外和体内研究。我们强调了在体外实验中观察到协同作用的几种机制,包括增加膜的渗透性,破坏生物膜,直接增强抗生素功效,抑制抗性发展。此外,体内研究揭示了其他机制,如增强/调节的免疫反应,减少炎症,和改善组织再生。一起,目前的文献表明,AMP-抗生素组合可以大大提高抗生素治疗的疗效,包括抵抗细菌的疗法,这代表了对当前抗菌策略的有价值的增强。
    Antibiotic resistance is a growing global problem that requires innovative therapeutic approaches and strategies for administering antibiotics. One promising approach is combination therapy, in which two or more drugs are combined to combat an infection. Along this line, the combination of antimicrobial peptides (AMPs) with conventional antibiotics has gained attention mainly due to the complementary mechanisms of action of AMPs and conventional antibiotics. In this article, we review both in vitro and in vivo studies that explore the synergy between AMPs and antibiotics. We highlight several mechanisms through which synergy is observed in in vitro experiments, including increasing membrane permeability, disrupting biofilms, directly potentiating antibiotic efficacy, and inhibiting resistance development. Moreover, in vivo studies reveal additional mechanisms such as enhanced/modulated immune responses, reduced inflammation, and improved tissue regeneration. Together, the current literature demonstrates that AMP-antibiotic combinations can substantially enhance efficacy of antibiotic therapies, including therapies against resistant bacteria, which represents a valuable enhancement to current antimicrobial strategies.
    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:Olaparib是一种PARP抑制剂,在由BRCA1/2突变引起的同源重组缺陷(HRD)的肿瘤中诱导合成致死性。FDA已批准一线铂敏感药物的单药治疗,复发性高级别上皮性卵巢癌。与DNA损伤疗法一起的联合疗法是克服HRD患者的有限功效的有希望的解决方案。本研究旨在开发负载托泊替康和奥拉帕尼的脂质体(TLL和OLL),并评估其组合在患者来源的卵巢癌样品中的有效性。
    方法:我们使用HEOC,四种透明细胞肿瘤(EOC1-4),恶性腹水,和OCI-E1p子宫内膜样原发性卵巢癌细胞系,并对BRCA1/2突变状态进行NGS分析。用MTT测定法测定抗增殖活性。使用Chou-Talalay算法来研究TLL和OLL的体外药效学相互作用。
    结果:OLL在所有具有野生型BRCA1/2的卵巢癌类型中显示出比常规制剂明显更高的疗效,提示体内功效增加的潜力。与标准配方相比,TLL对EOC1,EOC3,腹水的毒性明显更高,对HEOC的毒性更低。提示更好的治疗效果和安全性。与单独使用的药物相比,所研究化合物的组合显示出更高的细胞活力降低,在大多数选定的浓度下表现出协同抗肿瘤作用。
    结论:不同卵巢癌细胞类型对联合治疗的浓度依赖性反应证实了需要体外优化以最大化药物的细胞毒性。OLL和TLL组合是进一步动物研究的有前途的配方,特别是用于消除野生型BRCA1/2的上皮性卵巢癌。
    BACKGROUND: Olaparib is a PARP inhibitor inducing synthetic lethality in tumors with deficient homologous recombination (HRD) caused by BRCA1/2 mutations. The FDA has approved monotherapy for first-line platinum-sensitive, recurrent high-grade epithelial ovarian cancer. Combination therapy alongside DNA-damaging therapeutics is a promising solution to overcome the limited efficacy in patients with HRD. The present study was designed to develop topotecan- and olaparib-loaded liposomes (TLL and OLL) and assess the effectiveness of their combination in patient-derived ovarian cancer samples.
    METHODS: We used HEOC, four clear-cell tumors (EOC 1-4), malignant ascites, and an OCI-E1p endometrioid primary ovarian cancer cell line and performed NGS analysis of BRCA1/2 mutation status. Antiproliferative activity was determined with the MTT assay. The Chou-Talalay algorithm was used to investigate the in vitro pharmacodynamic interactions of TLLs and OLLs.
    RESULTS: The OLL showed significantly higher efficacy in all ovarian cancer types with wild-type BRCA1/2 than a conventional formulation, suggesting potential for increased in vivo efficacy. The TLL revealed substantially higher toxicity to EOC 1, EOC 3, ascites and lower toxicity to HEOC than the standard formulation, suggesting better therapeutic efficacy and safety profile. The combination of studied compounds showed a higher reduction in cell viability than drugs used individually, demonstrating a synergistic antitumor effect at most of the selected concentrations.
    CONCLUSIONS: The concentration-dependent response of different ovarian cancer cell types to combination therapy confirms the need for in vitro optimization to maximize drug cytotoxicity. The OLL and TLL combination is a promising formulation for further animal studies, especially for eliminating epithelial ovarian cancer with wild-type BRCA1/2.
    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
    通过对食品和药物管理局不良事件报告系统(FAERS)的分析,我们探讨了与秋水仙碱和他汀类药物联合使用引起的肌病相关的不良反应(ADR)的信号强度,并深入了解了这些肌病相关ADR的特点.
    我们从FAERS数据库中提取了秋水仙碱和他汀类药物联合使用导致肌病患者不良反应的相关数据。该分析是在2004年1月至2023年12月期间进行的,使用报告的优势比(ROR)和信息成分(IC)方法评估肌肉相关的ADR信号。
    共有18,386例他汀类药物肌病相关不良反应报告,348秋水仙碱肌病相关不良反应,收集到461例因两者联合所致的肌肉相关不良反应;他汀类药物肌毒性事件的最强信号是坏死性肌炎(ROR50.47,95%CL41.74-61.01;IC3.7095%CL3.25-4.08);秋水仙碱肌毒性事件的最强信号是中毒性肌病(ROR32.50,95%CL5.89%CL1.51);他汀类药物联合秋水仙碱的最强信号是中毒性肌病(ROR159.85,95%CL111.60-228.98;IC7.2295%CL3.59-5.9);当两种药物按照秋水仙碱联合氟伐他汀的顺序联合使用时,肌肉相关的不良反应信号是有意义的(ROR187.38,95%CL96.68-363.17;IC6.9995%与colsim95%CL25.25-35.85;IC4.8095%CL3.96-5.12);秋水仙碱联合瑞舒伐他汀(ROR25.73,95%CL20.16-32.83;IC4.5995%CL3.38-4.98)与秋水仙碱联合阿托伐他汀(ROR25.73,95%CL22.33-29.66;IC4.5995%CL3.97-4.91其次是秋水仙碱联合普伐他汀(ROR13.67,95%CL9.17-20.37;IC3.7395%CL1.87-4.47),而洛伐他汀或匹伐他汀没有产生信号。
    当秋水仙碱和他汀类药物单独或联合使用时,会发生类似的不良反应;然而,这些反应的强度可能不同。为了最大限度地降低药物相互作用的风险,具有较少潜在相互作用的他汀类药物,比如洛伐他汀,匹伐他汀,还有普伐他汀,应该选择,在使用过程中应密切监测与肌病相关的指标和症状。
    UNASSIGNED: Through an analysis of the Food and Drug Administration Adverse Event Reporting System (FAERS), we explored the signal strength of adverse reactions (ADRs) related to myopathy caused by the combination of colchicine and statins and gained insight into the characteristics of these myopathy related ADRs.
    UNASSIGNED: We extracted data from the FAERS database about ADRs in individuals with myopathy resulting from the combination of colchicine and statins. The analysis was conducted for the period spanning from January 2004 to December 2023 using the reported odds ratio (ROR) and information component (IC) methods to assess muscle-related ADR signals.
    UNASSIGNED: A total of 18,386 reports of statin myopathy-associated adverse reactions, 348 colchicine myopathy-associated adverse reactions, and 461 muscle-associated adverse reactions due to the combination of the two were collected; the strongest signals of statin myotoxicity events were for necrotizing myositis (ROR 50.47, 95% CL 41.74-61.01; IC 3.70 95% CL 3.25-4.08); the strongest signal for colchicine myotoxicity events was toxic myopathy (ROR 32.50, 95% CL 19.74-53.51; IC 4.97 95% CL 1.89-5.10), and the strongest signal for statins combined with colchicine was toxic myopathy (ROR 159.85, 95% CL 111.60-228.98; IC 7.22 95% CL 3.59-5.9); muscle-related adverse reactions signals were meaningful when the two drugs were combined in the order of colchicine combined with fluvastatin (ROR 187.38, 95% CL 96.68-363.17; IC 6.99 95% CL 1.65-5.68); colchicine combined with simvastatin in 135 cases (ROR 30.08. 95% CL 25.25-35.85; IC 4.80 95% CL 3.96-5.12); and colchicine combined with rosuvastatin (ROR 25.73, 95% CL 20.16-32.83; IC 4.59 95% CL 3.38-4.98) versus colchicine combined with atorvastatin (ROR 25.73, 95% CL 22.33-29.66; IC 4.59 95% CL 3.97-4.91) with almost identical signal intensity, followed by colchicine combined with pravastatin (ROR 13.67, 95% CL 9.17-20.37; IC 3.73 95% CL 1.87-4.47), whereas no signals were generated for lovastatin or pitavastatin.
    UNASSIGNED: Similar ADRs can occur when colchicine and statins are used individually or in combination; however, the strength of these reactions may differ. To minimize the risk of drug interactions, statins with less potential interactions, such as lovastatin, pitavastatin, and pravastatin, should be chosen, and myopathy-related indices and symptoms should be closely monitored during use.
    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
    这项研究调查了瓦伦西亚KK4型花生皮乙醇提取物(KK4-PSE)联合顺铂或5-氟尿嘧啶(5-FU)在体外和异种移植模型中对HeLa细胞的影响。在24、48和72小时的暴露时间,KK4-PSE抑制HeLa细胞的生长,半数最大抑制浓度(IC50)为79.43±0.54、55.55±1.57和41.32±0.74µg/mL,分别。通过Chou-Talalay方法评估的药物相互作用表明,KK4-PSE增强了5-FU对HeLa细胞的抗增殖活性,其组合指数(CI)值为0.49(48h)和0.60(72h),表明协同效应,而KK4-PSE联合顺铂在72小时表现出累加效应(CI=1.02),以及在24和48小时暴露时的拮抗作用(CI分别为1.12和1.18)。在裸鼠异种移植模型中,与对照组和单一药物治疗组相比,5-FU和KK4-PSE的组合显著降低了HeLa肿瘤重量.KK4-PSE和5-FU的组合比KK4-PSE-顺铂组合实现更大的肿瘤生长抑制。KK4-PSE减轻了顺铂和5-FU在裸鼠中诱导的肝毒性。在5-FU和KK4-PSE的组合治疗中,脾浆膜炎显著减少。这些结果表明,KK4-PSE具有限制宫颈癌细胞增殖的潜力,同时降低顺铂和5-FU的毒性。
    This study investigated the impact of Valencia KK4-type peanut skin ethanolic extract (KK4-PSE) combined with cisplatin or 5-fluorouracil (5-FU) on HeLa cells in vitro and in xenograft models. At exposure times of 24, 48 and 72 h, KK4-PSE inhibited the growth of HeLa cells with a half maximal inhibitory concentration (IC50) of 79.43 ± 0.54, 55.55 ± 1.57 and 41.32 ± 0.74 µg/mL, respectively. Drug interactions evaluated by the Chou-Talalay method demonstrated that KK4-PSE enhanced antiproliferative activity of 5-FU against HeLa cells with combination index (CI) values of 0.49 (48 h) and 0.60 (72 h), indicating a synergistic effect, while KK4-PSE combined with cisplatin exhibited an additive effect (CI = 1.02) at 72 h, and an antagonistic effect at 24 and 48 h exposures (CI = 1.12 and 1.18, respectively). In nude mouse xenograft models, the combination of 5-FU and KK4-PSE markedly reduced HeLa tumor weights compared with the control and single agent treatments groups. The combination of KK4-PSE and 5-FU achieved greater tumor growth inhibition than that of the KK4-PSE-cisplatin combination. KK4-PSE mitigated hepatotoxicity induced by both cisplatin and 5-FU in nude mice. The spleen hyaloserositis was significantly reduced in the combination treatment of 5-FU and KK4-PSE. These results suggest that KK4-PSE has the potential to limit cervical cancer cell proliferation while reducing the toxicity of cisplatin and 5-FU.
    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
    在联合治疗中,细菌同时受到两种或多种抗生素的攻击。理想情况下,需要单独的突变来适应它们中的每一个,这是先验预期会阻碍全面抵抗的演变。然而,这种策略的成功最终取决于组合对有和没有抗性突变的细菌生长的控制程度。为了设计组合治疗,我们需要选择药物及其剂量,并决定混合多少种药物。哪些组合是好的?要回答这个问题,我们建立了一个随机药效学模型,并确定了成功根除细菌种群的概率。我们考虑抑菌和两种类型的杀菌药物-那些独立于复制而杀死的药物和那些在复制期间杀死的药物。要建立空模型的结果,我们考虑非相互作用药物,并实施两个最常见的药物独立性模型-Loewe可加性和Bliss独立性.我们的结果表明,联合治疗在限制耐药性发展方面几乎总是比只服用一种药物更好。即使我们保持药物总剂量恒定,以进行“公平”的比较。然而,对于剂量-反应曲线陡峭的药物存在例外情况.组合抑菌药物和可杀死非复制细胞的杀菌药物是特别有益的。我们的结果表明,50:50的药物比例-即使不总是最佳的-通常是一个好的和安全的选择。应用三种或四种药物对于具有大突变率的菌株的治疗是有益的,但是否则添加更多的药物仅提供边际益处或甚至缺点。通过系统地解决治疗设计的关键要素,我们的研究为将来考虑更多因素的模型提供了基础.它还强调了将传统的药物独立性概念转化为单细胞水平的概念挑战。
    In combination therapy, bacteria are challenged with two or more antibiotics simultaneously. Ideally, separate mutations are required to adapt to each of them, which is a priori expected to hinder the evolution of full resistance. Yet, the success of this strategy ultimately depends on how well the combination controls the growth of bacteria with and without resistance mutations. To design a combination treatment, we need to choose drugs and their doses and decide how many drugs get mixed. Which combinations are good? To answer this question, we set up a stochastic pharmacodynamic model and determine the probability to successfully eradicate a bacterial population. We consider bacteriostatic and two types of bactericidal drugs-those that kill independent of replication and those that kill during replication. To establish results for a null model, we consider non-interacting drugs and implement the two most common models for drug independence-Loewe additivity and Bliss independence. Our results show that combination therapy is almost always better in limiting the evolution of resistance than administering just one drug, even though we keep the total drug dose constant for a \'fair\' comparison. Yet, exceptions exist for drugs with steep dose-response curves. Combining a bacteriostatic and a bactericidal drug which can kill non-replicating cells is particularly beneficial. Our results suggest that a 50:50 drug ratio-even if not always optimal-is usually a good and safe choice. Applying three or four drugs is beneficial for treatment of strains with large mutation rates but adding more drugs otherwise only provides a marginal benefit or even a disadvantage. By systematically addressing key elements of treatment design, our study provides a basis for future models which take further factors into account. It also highlights conceptual challenges with translating the traditional concepts of drug independence to the single-cell level.
    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
    尽管在癌症预防方面取得了重大进展,筛选,和治疗,仍然有限的治疗选择是提高癌症治愈率的障碍。近年来,为了开发选择性靶向致癌Wnt/β-catenin信号通路的不同组分的治疗方法,人们进行了许多努力。这些包括小分子抑制剂,抗体,最近,基于基因的方法。尽管其中一些在临床试验中显示出有希望的结果,Wnt/β-catenin通路在癌症治疗的常规临床实践中仍未被靶向。至于大多数抗癌治疗,使用Wnt/β-连环蛋白抑制剂的一个关键限制是它们的治疗指数,即,将有效的抗癌活性与可接受的毒性结合起来的困难。保护健康组织免受Wnt/β-连环蛋白抑制剂的影响是一个主要问题,因为Wnt/β-连环蛋白信号通路在成人组织稳态和再生中的重要作用。在这次审查中,我们提供了关于Wnt/β-catenin途径抑制剂的临床试验的最新摘要,检查它们的抗肿瘤活性和相关的不良事件,并探索正在开发的策略,以改善这种治疗方法的益处/风险状况。
    Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach.
    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
    恶性肿瘤的临床治疗具有挑战性,往往导致严重的不良反应和死亡。耐药性(DR)拮抗治疗的有效性,增加药物剂量会使治疗指数(TI)恶化。目前克服DR的努力主要涉及药物组合的使用,包括应用多种抗癌药物,雇用药物致敏者,它们是增强药代动力学(PK)的化学试剂,包括靶向细胞通路和调节相关的膜转运蛋白。虽然组合多种化合物可能导致药物-药物相互作用(DDI)或多药效,使用药物致敏剂可以在疾病部位快速获得有效的治疗剂量,以预防早期DR并最大程度地减少副作用,并且由于需要较低的药物剂量,将减少DDI的机会。这篇综述强调了TI在评估癌症治疗药物剂量中的重要用途,并讨论了该领域缺乏统一的TI标准。总结了常用的收益-风险评估标准,以及对TI在制药工业部门当前使用的关键探索。具体来说,这篇综述引发了对药物致敏剂的讨论,以促进直接改善人体有效剂量与毒性剂量的比率。药物和敏化剂分子的组合可能会通过使用较低的药物剂量来消除有害的脱靶活性,从而重新点燃那些在后期临床试验中失败的药物。药物组合和使用药物致敏剂是对抗DR的潜在手段。综述了TI上药物组合和多重用药的演变。值得注意的是,引入了新颖的二元武器方法,作为改进TI的新机会。这篇综述强调迫切需要一个标准来系统地评估药物的安全性和效率,以便在该领域的实际实施。
    The clinical management of malignant tumours is challenging, often leading to severe adverse effects and death. Drug resistance (DR) antagonises the effectiveness of treatments, and increasing drug dosage can worsen the therapeutic index (TI). Current efforts to overcome DR predominantly involve the use of drug combinations, including applying multiple anti-cancerous drugs, employing drug sensitisers, which are chemical agents that enhance pharmacokinetics (PK), including the targeting of cellular pathways and regulating pertinent membrane transporters. While combining multiple compounds may lead to drug-drug interactions (DDI) or polypharmacy effect, the use of drug sensitisers permits rapid attainment of effective treatment dosages at the disease site to prevent early DR and minimise side effects and will reduce the chance of DDI as lower drug doses are required. This review highlights the essential use of TI in evaluating drug dosage for cancer treatment and discusses the lack of a unified standard for TI within the field. Commonly used benefit-risk assessment criteria are summarised, and the critical exploration of the current use of TI in the pharmaceutical industrial sector is included. Specifically, this review leads to the discussion of drug sensitisers to facilitate improved ratios of effective dose to toxic dose directly in humans. The combination of drug and sensitiser molecules might see additional benefits to rekindle those drugs that failed late-stage clinical trials by the removal of detrimental off-target activities through the use of lower drug doses. Drug combinations and employing drug sensitisers are potential means to combat DR. The evolution of drug combinations and polypharmacy on TI are reviewed. Notably, the novel binary weapon approach is introduced as a new opportunity to improve TI. This review emphasises the urgent need for a criterion to systematically evaluate drug safety and efficiency for practical implementation in the field.
    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
    粉防己碱(TET)是一种天然的双苄基异喹啉类生物碱,具有广泛的生物学和药理活性,在临床上主要用作抗炎药或抗肿瘤佐剂。然而,局限性,如突出的疏水性,严重的脱靶毒性,和低吸收导致次优的治疗结果阻止其广泛采用。纳米颗粒已被证明是靶向药物递送的有效装置,因为携带药物的纳米颗粒可以通过增强的渗透性和保留(EPR)效应被动地转运到肿瘤部位。从而在癌症治疗中占据一席之地。由于其突出的优点,如增加水溶性,在用于TET递送的纳米载体构建方面取得了巨大的进展,改善生物分布和血液循环,减少脱靶刺激,和组合疗法。在这里,我们系统回顾了载TET纳米粒子的最新进展及其各自的特性,以期为TET的未来研究和潜在应用提供视角和指南.
    Tetrandrine (TET) is a natural bis-benzylisoquinoline alkaloid isolated from Stephania species with a wide range of biological and pharmacologic activities; it mainly serves as an anti-inflammatory agent or antitumor adjuvant in clinical applications. However, limitations such as prominent hydrophobicity, severe off-target toxicity, and low absorption result in suboptimal therapeutic outcomes preventing its widespread adoption. Nanoparticles have proven to be efficient devices for targeted drug delivery since drug-carrying nanoparticles can be passively transported to the tumor site by the enhanced permeability and retention (EPR) effects, thus securing a niche in cancer therapies. Great progress has been made in nanocarrier construction for TET delivery due to their outstanding advantages such as increased water-solubility, improved biodistribution and blood circulation, reduced off-target irritation, and combinational therapy. Herein, we systematically reviewed the latest advancements in TET-loaded nanoparticles and their respective features with the expectation of providing perspective and guidelines for future research and potential applications of TET.
    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
    虽然药物联合疗法非常重要,特别是在癌症治疗中,确定新的协同药物组合一直是一个具有挑战性的冒险。在这种情况下,计算方法已成为一种有前途的工具,可以优先考虑药物组合以进行进一步评估。尽管他们表现有限,实用程序,和可解释性。这里,我们提出了一种新的预测工具,piscesCSM,利用基于图形的表示来模拟小分子化学结构,以准确预测对一种或多种癌细胞系具有有利抗癌协同作用的药物组合。利用这些见解,我们开发了一个通用的监督机器学习模型来指导30多个细胞系中抗癌协同药物组合的预测。在独立的非冗余盲测试中,接收器工作特性曲线(AUROC)下的面积高达0.89,在大规模肿瘤学筛查数据和阿斯利康产生的独立测试集上,均优于最先进的方法(预测准确性提高16%以上).此外,通过探索我们方法的可解释性,我们发现,简单的理化性质和基于图形的特征可以预测化疗的协同作用.为了提供一个简单和集成的平台,以快速筛选具有有利的协同抗癌作用的潜在候选对,我们在https://biosig在线免费提供piscesCSM。实验室。uq.edu.au/piscescsm/作为Web服务器和API。我们相信,我们的预测工具将为优化和扩大组合筛选库提供宝贵的资源,以确定有效和安全的协同抗癌药物组合。科学贡献:这项工作提出了piscesCSM,一个基于机器学习的框架,它依赖于建立良好的基于图的小分子表示来识别和提供更好的预测准确性的合成药物组合。我们的模型,piscesCSM,表明,将物理化学特性与基于图的签名相结合可以在分类预测任务上胜过当前的体系结构。此外,将我们的工具作为网络服务器实施,为研究人员提供了一个用户友好的平台,以筛选对一种或多种癌细胞系具有良好抗癌作用的潜在协同药物组合.
    While drug combination therapies are of great importance, particularly in cancer treatment, identifying novel synergistic drug combinations has been a challenging venture. Computational methods have emerged in this context as a promising tool for prioritizing drug combinations for further evaluation, though they have presented limited performance, utility, and interpretability. Here, we propose a novel predictive tool, piscesCSM, that leverages graph-based representations to model small molecule chemical structures to accurately predict drug combinations with favourable anticancer synergistic effects against one or multiple cancer cell lines. Leveraging these insights, we developed a general supervised machine learning model to guide the prediction of anticancer synergistic drug combinations in over 30 cell lines. It achieved an area under the receiver operating characteristic curve (AUROC) of up to 0.89 on independent non-redundant blind tests, outperforming state-of-the-art approaches on both large-scale oncology screening data and an independent test set generated by AstraZeneca (with more than a 16% improvement in predictive accuracy). Moreover, by exploring the interpretability of our approach, we found that simple physicochemical properties and graph-based signatures are predictive of chemotherapy synergism. To provide a simple and integrated platform to rapidly screen potential candidate pairs with favourable synergistic anticancer effects, we made piscesCSM freely available online at https://biosig.lab.uq.edu.au/piscescsm/ as a web server and API. We believe that our predictive tool will provide a valuable resource for optimizing and augmenting combinatorial screening libraries to identify effective and safe synergistic anticancer drug combinations. SCIENTIFIC CONTRIBUTION: This work proposes piscesCSM, a machine-learning-based framework that relies on well-established graph-based representations of small molecules to identify and provide better predictive accuracy of syngenetic drug combinations. Our model, piscesCSM, shows that combining physiochemical properties with graph-based signatures can outperform current architectures on classification prediction tasks. Furthermore, implementing our tool as a web server offers a user-friendly platform for researchers to screen for potential synergistic drug combinations with favorable anticancer effects against one or multiple cancer cell lines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号