synthetic lethality

合成杀伤力
  • 文章类型: Journal Article
    在这里,我们报告了同时抑制三种主要的DNA损伤识别PI3激酶样激酶(PIKKs)-ATM,ATR,和DNA-PK-在哺乳动物细胞中诱导严重的组合合成致死性。利用中国仓鼠细胞系CHO和V79及其各自的PIKK突变体,我们评估了抑制这三种激酶对细胞活力的影响,DNA损伤反应,和染色体完整性。我们的结果表明,虽然单或双激酶抑制增加细胞毒性,抑制所有三种PIKK导致显著更高的协同致死率,染色体畸变,和DNA双链断裂(DSB)诱导,如通过它们的协同作用评分计算的。这些发现表明,ATM的重叠冗余,ATR,DNA-PK功能对细胞存活至关重要,它们的联合抑制作用极大地破坏了DNA损伤信号和修复过程,导致细胞死亡。这项研究为多靶向DDR激酶抑制作为一种有效的抗癌策略的潜力提供了见解。需要进一步研究以阐明潜在的机制和治疗应用。
    Here we report that simultaneous inhibition of the three primary DNA damage recognition PI3 kinase-like kinases (PIKKs) -ATM, ATR, and DNA-PK- induces severe combinatorial synthetic lethality in mammalian cells. Utilizing Chinese hamster cell lines CHO and V79 and their respective PIKK mutants, we evaluated effects of inhibiting these three kinases on cell viability, DNA damage response, and chromosomal integrity. Our results demonstrate that while single or dual kinase inhibition increased cytotoxicity, inhibition of all three PIKKs results in significantly higher synergistic lethality, chromosomal aberrations, and DNA double-strand break (DSB) induction as calculated by their synergy scores. These findings suggest that the overlapping redundancy of ATM, ATR, and DNA-PK functions is critical for cell survival, and their combined inhibition greatly disrupts DNA damage signaling and repair processes, leading to cell death. This study provides insights into the potential of multi-targeted DDR kinase inhibition as an effective anticancer strategy, necessitating further research to elucidate underlying mechanisms and therapeutic applications.
    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
    晚期上皮性卵巢癌是妇科癌症死亡的最常见原因。晚期疾病的一线治疗包括铂-紫杉烷化疗(术后或围手术期)和最大减积手术的组合。对化疗的初始反应率很高(高达80%),但大多数患者会复发(约70-90%)并死于该疾病。最近,聚ADP-核糖聚合酶(PARP)抑制(通过Olaparib等药物,Niraparib或Rucaparib)在BRCA种系突变或铂敏感疾病中的定向合成致死方法为患者带来了真正的希望。PARP抑制剂(PARPi)维持治疗可以延长生存期,但由于对PARPi治疗的内在或获得性继发性耐药性,治疗反应无法持续。BRCA1/2突变的逆转可导致BRCA种系突变卵巢癌的临床PARPi耐药。然而,在更常见的铂敏感的零星HGSOC中,PARPi耐药的临床机制尚待确定.在这里,我们对PARPi的现状和耐药机制进行了全面的综述。
    Advanced epithelial ovarian cancer is the commonest cause of gynaecological cancer deaths. First-line treatment for advanced disease includes a combination of platinum-taxane chemotherapy (post-operatively or peri-operatively) and maximal debulking surgery whenever feasible. Initial response rate to chemotherapy is high (up to 80%) but most patients will develop recurrence (approximately 70-90%) and succumb to the disease. Recently, poly-ADP-ribose polymerase (PARP) inhibition (by drugs such as Olaparib, Niraparib or Rucaparib) directed synthetic lethality approach in BRCA germline mutant or platinum sensitive disease has generated real hope for patients. PARP inhibitor (PARPi) maintenance therapy can prolong survival but therapeutic response is not sustained due to intrinsic or acquired secondary resistance to PARPi therapy. Reversion of BRCA1/2 mutation can lead to clinical PARPi resistance in BRCA-germline mutated ovarian cancer. However, in the more common platinum sensitive sporadic HGSOC, the clinical mechanisms of development of PARPi resistance remains to be defined. Here we provide a comprehensive review of the current status of PARPi and the mechanisms of resistance to therapy.
    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
    1型神经纤维瘤病(NF1)是由NF1基因突变引起的遗传性疾病,与各种症状有关。包括良性肿瘤的形成,叫做神经纤维瘤,在神经内。药物治疗目前是有限的。丝裂原激活的蛋白激酶激酶(MEK)抑制剂司美替尼用于丛状神经纤维瘤(PNs)的一部分,但并不总是有效的,并且可能引起副作用。因此,显然需要发现靶向NF1缺陷肿瘤细胞的新药.使用NF1的果蝇细胞模型,我们进行了合成致死筛选以鉴定新的药物靶标。我们鉴定了54个候选基因,通过可变剂量分析作为二次筛选进行验证。可以使用现有药物靶向与五个候选物相关的途径。其中,氯喹(CQ)和巴弗洛霉素A1,已知靶向自噬途径,显示了选择性杀死NF1缺陷果蝇细胞的最大潜力。当进一步研究自噬相关基因时,我们发现,在30个测试基因中,有14个基因与NF1有合成致死相互作用.这14个基因涉及自噬途径的多个方面,并且可以用介导自噬途径的其他药物靶向,虽然CQ是最有效的。自噬抑制剂的致死作用在一组缺乏NF1的人Schwann细胞系中是保守的,强调他们的翻译潜力。在果蝇NF1体内模型和在小鼠中生长的异种移植NF1缺陷肿瘤细胞系中,CQ的作用也得到了保留。CQ治疗比司美替尼治疗更显著地降低了肿瘤生长。此外,CQ和司美替尼联合治疗导致NF1缺陷型细胞活力进一步降低.总之,NF1缺陷型细胞易受自噬途径破坏的影响。该途径代表了治疗NF1相关肿瘤的有希望的靶标,我们确定CQ是治疗NF1肿瘤的候选药物。
    Neurofibromatosis type 1 (NF1) is a genetic disorder caused by mutation of the NF1 gene that is associated with various symptoms, including the formation of benign tumors, called neurofibromas, within nerves. Drug treatments are currently limited. The mitogen-activated protein kinase kinase (MEK) inhibitor selumetinib is used for a subset of plexiform neurofibromas (PNs) but is not always effective and can cause side effects. Therefore, there is a clear need to discover new drugs to target NF1-deficient tumor cells. Using a Drosophila cell model of NF1, we performed synthetic lethal screens to identify novel drug targets. We identified 54 gene candidates, which were validated with variable dose analysis as a secondary screen. Pathways associated with five candidates could be targeted using existing drugs. Among these, chloroquine (CQ) and bafilomycin A1, known to target the autophagy pathway, showed the greatest potential for selectively killing NF1-deficient Drosophila cells. When further investigating autophagy-related genes, we found that 14 out of 30 genes tested had a synthetic lethal interaction with NF1. These 14 genes are involved in multiple aspects of the autophagy pathway and can be targeted with additional drugs that mediate the autophagy pathway, although CQ was the most effective. The lethal effect of autophagy inhibitors was conserved in a panel of human NF1-deficient Schwann cell lines, highlighting their translational potential. The effect of CQ was also conserved in a Drosophila NF1 in vivo model and in a xenografted NF1-deficient tumor cell line grown in mice, with CQ treatment resulting in a more significant reduction in tumor growth than selumetinib treatment. Furthermore, combined treatment with CQ and selumetinib resulted in a further reduction in NF1-deficient cell viability. In conclusion, NF1-deficient cells are vulnerable to disruption of the autophagy pathway. This pathway represents a promising target for the treatment of NF1-associated tumors, and we identified CQ as a candidate drug for the treatment of NF1 tumors.
    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:癌症是全球主要的公共卫生挑战。然而,对癌症研究社区的演变模式及其研究能力和影响的影响因素知之甚少,这不仅受到通过研究合作建立的社交网络的影响,也受到研究项目嵌入的知识网络的影响。
    方法:本研究的重点是癌症研究中的一个特定主题——“合成致死性”。在过去的十年中,这一领域取得了蓬勃发展和多学科合作。根据与“合成致死性”相关的癌症研究论文的文献计量数据,建立并分析了多层次的合作和知识网络。负二项回归分析被进一步应用于探索这些网络中的节点属性,以及其他潜在因素,受影响的论文引用,被广泛接受为评估研究能力和影响的代理。
    结果:我们的研究表明,基于合成致死性的癌症研究领域的特征是具有高度集成的知识网络,除了展示一些集群的协作网络。我们发现某些因素与引文计数之间存在显着相关性。具体来说,在国家级国际合作网络中的领先地位和行业参与都被发现与更高的引用显着相关。在个人层面的协作网络中,主要作者的学位中心性与引文呈倒U型关系,而它们的结构孔表现出积极而显著的影响。在知识网络中,然而,只有结构孔的度量对引用次数有积极和显著的影响。
    结论:为了提高癌症研究能力和影响力,非领导国应采取措施提高其国际合作地位。对于早期职业研究人员来说,增加合作者的数量似乎更有效。还应鼓励产学合作,加强人力资源的整合,技术,资金,研究平台和医疗资源。通过这项研究获得的见解也为研究人员或管理人员从知识网络的角度设计未来的研究方向提供了建议。专注于独特的问题,特别是跨学科领域将提高产出并影响他们的研究工作。
    BACKGROUND: Cancer is a major public health challenge globally. However, little is known about the evolution patterns of cancer research communities and the influencing factors of their research capacity and impact, which is affected not only by the social networks established through research collaboration but also by the knowledge networks in which the research projects are embedded.
    METHODS: The focus of this study was narrowed to a specific topic - \'synthetic lethality\' - in cancer research. This field has seen vibrant growth and multidisciplinary collaboration in the past decade. Multi-level collaboration and knowledge networks were established and analysed on the basis of bibliometric data from \'synthetic lethality\'-related cancer research papers. Negative binomial regression analysis was further applied to explore how node attributes within these networks, along with other potential factors, affected paper citations, which are widely accepted as proxies for assessing research capacity and impact.
    RESULTS: Our study revealed that the synthetic lethality-based cancer research field is characterized by a knowledge network with high integration, alongside a collaboration network exhibiting some clustering. We found significant correlations between certain factors and citation counts. Specifically, a leading status within the nation-level international collaboration network and industry involvement were both found to be significantly related to higher citations. In the individual-level collaboration networks, lead authors\' degree centrality has an inverted U-shaped relationship with citations, while their structural holes exhibit a positive and significant effect. Within the knowledge network, however, only measures of structural holes have a positive and significant effect on the number of citations.
    CONCLUSIONS: To enhance cancer research capacity and impact, non-leading countries should take measures to enhance their international collaboration status. For early career researchers, increasing the number of collaborators seems to be more effective. University-industry cooperation should also be encouraged, enhancing the integration of human resources, technology, funding, research platforms and medical resources. Insights gained through this study also provide recommendations to researchers or administrators in designing future research directions from a knowledge network perspective. Focusing on unique issues especially interdisciplinary fields will improve output and influence their research work.
    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
    胰腺导管腺癌(PDAC)是最致命的恶性肿瘤之一。基于表观遗传的合成致死策略为PDAC治疗提供了新的机会。寻找更多的具有异常表观遗传变化的DNA损伤修复(DDR)相关或细胞命运相关分子变得非常重要。具有序列相似性的家族110C(FAM110C)是细胞命运相关基因,其在癌症中的功能尚不清楚。
    七个细胞系,导管内乳头状黏液性肿瘤(IPMN)34例,采用15例黏液性囊性肿瘤(MCN)和284例PDAC样本。甲基化特异性PCR,westernblot,CRISPR基因敲除,本研究使用免疫沉淀和异种移植小鼠模型。
    FAM110C在41.18%(14/34)的IPMN中甲基化,MCN的46.67%(7/15)和PDAC的72.89%(207/284),IPMN/MCN有进展趋势(P=0.0001,P=0.0389)。FAM110C甲基化与不良总生存期(OS)显着相关(P=0.0065),并且是不良OS的独立预后指标(P=0.0159)。FAM110C在体外和体内抑制PDAC细胞生长,作为一种新型的肿瘤抑制剂。FAM110C通过与HMGB1相互作用激活ATM和NHEJ信号通路。FAM110C表达的缺失使PDAC细胞对VE-822(ATR抑制剂)和MK-8776(CHK1抑制剂)敏感。
    FAM110C甲基化是PDAC中潜在的诊断和预后标志物,其表观遗传沉默使PDAC细胞对ATR/CHK1抑制剂敏感。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. An epigenetic-based synthetic lethal strategy provides a novel opportunity for PDAC treatment. Finding more DNA damage repair (DDR)-related or cell fate-related molecules with aberrant epigenetic changes is becoming very important. Family with sequence similarity 110C (FAM110C) is a cell fate-related gene and its function in cancer remains unclear.
    UNASSIGNED: Seven cell lines, 34 cases of intraductal papillary mucinous neoplasm (IPMN), 15 cases of mucinous cystic neoplasm (MCN) and 284 cases of PDAC samples were employed. Methylation-specific PCR, western blot, CRISPR knockout, immunoprecipitation and a xenograft mouse model were used in this study.
    UNASSIGNED: FAM110C is methylated in 41.18% (14/34) of IPMN, 46.67% (7/15) of MCN and 72.89% (207/284) of PDAC, with a progression trend from IPMN/MCN to pancreatic cancer (P = 0.0001, P = 0.0389). FAM110C methylation is significantly associated with poor overall survival (OS) (P = 0.0065) and is an independent prognostic marker for poor OS (P = 0.0159). FAM110C inhibits PDAC cells growth both in vitro and in vivo, serving as a novel tumor suppressor. FAM110C activates ATM and NHEJ signaling pathways by interacting with HMGB1. Loss of FAM110C expression sensitizes PDAC cells to VE-822 (an ATR inhibitor) and MK-8776 (a CHK1 inhibitor).
    UNASSIGNED: FAM110C methylation is a potential diagnostic and prognostic marker in PDAC, and its epigenetic silencing sensitizes PDAC cells to ATR/CHK1 inhibitors.
    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
    载脂蛋白BmRNA编辑催化多肽样(APOBEC)家族的酶是在DNA和RNA中将胞嘧啶转化为尿嘧啶的胞嘧啶脱氨酶。在这些蛋白质中,APOBEC3亚家族成员,APOBEC3A(A3A)和APOBEC3B(A3B),是癌细胞突变的主要来源。癌细胞中A3A和A3B的异常表达导致具有特定单碱基取代(SBS)特征的突变积累,以C→T和C→G变化为特征,在许多肿瘤类型中。除了助长诱变,A3A和A3B,特别是A3A,诱导DNA复制应激,DNA损伤,通过它们的催化活性和染色体的不稳定性,触发一系列细胞反应。因此,A3A/B已成为癌症发展过程中基因组进化的关键驱动因素,有助于肿瘤发生,肿瘤异质性,和治疗抗性。然而,A3A/B在癌细胞中的表达呈现了可用于治疗的癌症脆弱性。在这次审查中,我们讨论了最近的研究,阐明了调节A3A表达的机制和A3A在癌症中的影响。我们还回顾了A3A抑制剂的最新进展,并对A3A的未来研究方向进行了展望。
    Enzymes of the apolipoprotein B mRNA editing catalytic polypeptide like (APOBEC) family are cytosine deaminases that convert cytosine to uracil in DNA and RNA. Among these proteins, APOBEC3 sub-family members, APOBEC3A (A3A) and APOBEC3B (A3B), are prominent sources of mutagenesis in cancer cells. The aberrant expression of A3A and A3B in cancer cells leads to accumulation of mutations with specific single-base substitution (SBS) signatures, characterized by C→T and C→G changes, in a number of tumor types. In addition to fueling mutagenesis, A3A and A3B, particularly A3A, induce DNA replication stress, DNA damage, and chromosomal instability through their catalytic activities, triggering a range of cellular responses. Thus, A3A/B have emerged as key drivers of genome evolution during cancer development, contributing to tumorigenesis, tumor heterogeneity, and therapeutic resistance. Yet, the expression of A3A/B in cancer cells presents a cancer vulnerability that can be exploited therapeutically. In this review, we discuss the recent studies that shed light on the mechanisms regulating A3A expression and the impact of A3A in cancer. We also review recent advances in the development of A3A inhibitors and provide perspectives on the future directions of A3A research.
    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
    与广谱抗生素相比,具有相对窄谱的选择性药物可以通过特异性靶向引起感染的病原体来减少治疗的副作用。此外,对抗传染性病原体,尤其是耐药微生物,通过攻击多个目标更有效。这里,我们通过系统分析6种不同微生物的基因组尺度代谢模型,将合成致死性与选择性药物靶向相结合,以鉴定多靶点和生物体特异性的潜在候选药物.通过将微生物视为靶向的或保守的,在一到六个成员的群体中,我们设计了665个案例研究。对于每种情况,我们确定了单一的基本反应以及双重反应,三重,和四重合成致死反应集,对目标微生物是致死的,对保守微生物是中性的。不出所料,每种情况下获得的解决方案的数量取决于所研究微生物之间的基因组相似性。将确定的潜在药物靶标映射到其相应的途径,突出了关键子系统的重要性,如细胞包膜生物合成。甘油磷脂代谢,膜脂代谢,和核苷酸补救途径。为了协助验证和进一步调查我们提出的潜在药物靶点,我们引入了两组目标,理论上可以解决665例病例中的很大一部分。我们期望所获得的解决方案为设计窄谱药物提供了有价值的见解,这些药物选择性地仅对目标微生物造成全系统损害。
    Selective drugs with a relatively narrow spectrum can reduce the side effects of treatments compared to broad-spectrum antibiotics by specifically targeting the pathogens responsible for infection. Furthermore, combating an infectious pathogen, especially a drug-resistant microorganism, is more efficient by attacking multiple targets. Here, we combined synthetic lethality with selective drug targeting to identify multi-target and organism-specific potential drug candidates by systematically analyzing the genome-scale metabolic models of six different microorganisms. By considering microorganisms as targeted or conserved in groups ranging from one to six members, we designed 665 individual case studies. For each case, we identified single essential reactions as well as double, triple, and quadruple synthetic lethal reaction sets that are lethal for targeted microorganisms and neutral for conserved ones. As expected, the number of obtained solutions for each case depends on the genomic similarity between the studied microorganisms. Mapping the identified potential drug targets to their corresponding pathways highlighted the importance of key subsystems such as cell envelope biosynthesis, glycerophospholipid metabolism, membrane lipid metabolism, and the nucleotide salvage pathway. To assist in the validation and further investigation of our proposed potential drug targets, we introduced two sets of targets that can theoretically address a substantial portion of the 665 cases. We expect that the obtained solutions provide valuable insights into designing narrow-spectrum drugs that selectively cause system-wide damage only to the target microorganisms.
    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
    卵巢癌是妇科恶性肿瘤中死亡的主要原因。卡铂和聚(ADP-核糖)聚合酶抑制剂(PARPi)通常用于治疗卵巢癌。同源重组缺陷(HRD)肿瘤对这些治疗的敏感性增加;然而,许多卵巢癌患者是同源重组(HRP)。TTField是在各种癌症类型中诱导HRD样表型的非侵入性电场。本研究旨在研究TTField与卡铂或PARPi(奥拉帕尼或尼拉帕尼)一起应用于临床前卵巢癌模型的影响。
    A2780(HRP),OVCAR3(HRD),和A2780cis(铂抗性)人卵巢癌细胞在体外用TTField(1V/cmRMS,200kHz,72小时),单独或与各种药物浓度。测量处理过的细胞的细胞计数,菌落形成,凋亡,DNA损伤,DNA修复蛋白的表达,和细胞周期。在体内,将ID8-fLuc(HRP)卵巢癌细胞腹膜内接种于C57BL/6小鼠,然后用任何一个假的治疗,TTFelds(200kHz),奥拉帕尼(50mg/kg),或TTFields加olaparib;为期四周。在治疗停止时使用生物发光成像分析肿瘤生长;并进行存活分析。
    TTFields-药物相互作用的性质取决于药物的潜在作用机制和细胞的遗传背景,在HRP和耐药细胞中观察到TTField与卡铂或PARPi之间的协同相互作用。处理过的细胞表现出高水平的DNA损伤,伴随着G2/M逮捕,和HRD样表型的诱导。在荷瘤小鼠中,相对于奥拉帕尼单一疗法和对照,TTField和奥拉帕尼共同治疗导致肿瘤体积减小和存活益处。
    通过诱导HRD样表型,TTFelds使HRP和耐药卵巢癌细胞对卡铂或PARPi治疗敏感,可能减轻先验和从头耐药性,卵巢癌治疗的主要限制。
    UNASSIGNED: Ovarian cancer is the leading cause of mortality among gynecological malignancies. Carboplatin and poly (ADP-ribose) polymerase inhibitors (PARPi) are often implemented in the treatment of ovarian cancer. Homologous recombination deficient (HRD) tumors demonstrate increased sensitivity to these treatments; however, many ovarian cancer patients are homologous recombination proficient (HRP). TTFields are non-invasive electric fields that induce an HRD-like phenotype in various cancer types. The current study aimed to investigate the impact of TTFields applied together with carboplatin or PARPi (olaparib or niraparib) in preclinical ovarian cancer models.
    UNASSIGNED: A2780 (HRP), OVCAR3 (HRD), and A2780cis (platinum-resistant) human ovarian cancer cells were treated in vitro with TTFields (1 V/cm RMS, 200 kHz, 72 h), alone or with various drug concentrations. Treated cells were measured for cell count, colony formation, apoptosis, DNA damage, expression of DNA repair proteins, and cell cycle. In vivo, ID8-fLuc (HRP) ovarian cancer cells were inoculated intraperitoneally to C57BL/6 mice, which were then treated with either sham, TTFields (200 kHz), olaparib (50 mg/kg), or TTFields plus olaparib; over a period of four weeks. Tumor growth was analyzed using bioluminescent imaging at treatment cessation; and survival analysis was performed.
    UNASSIGNED: The nature of TTFields-drug interaction was dependent on the drug\'s underlying mechanism of action and on the genetic background of the cells, with synergistic interactions between TTFields and carboplatin or PARPi seen in HRP and resistant cells. Treated cells demonstrated elevated levels of DNA damage, accompanied by G2/M arrest, and induction of an HRD-like phenotype. In the tumor-bearing mice, TTFields and olaparib co-treatment resulted in reduced tumor volume and a survival benefit relative to olaparib monotherapy and to control.
    UNASSIGNED: By inducing an HRD-like phenotype, TTFields sensitize HRP and resistant ovarian cancer cells to treatment with carboplatin or PARPi, potentially mitigating a-priori and de novo drug resistance, a major limitation in ovarian cancer 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
    基因对中的互斥功能丧失改变是那些一起发生的频率低于预期的频率,并且可能表示基因之间的合成致死关系(SSL)。可以在治疗上利用SSL选择性地杀死癌细胞。这里,我们分析了突变,拷贝数变化,和来自癌症基因组图谱的样本中的甲基化水平,使用超几何和泊松二项测试来鉴定互斥的灭活基因。我们专注于基因对,其中一个是灭活的肿瘤抑制因子,另一个是其蛋白质产物可以被已知药物抑制的基因。这为几种癌症提供了丰富的潜在靶向治疗和重新定位机会。这些数据可在MexDrugs网站上获得,https://生物信息板.Sussex.AC.英国/麦克斯药。
    Mutually exclusive loss-of-function alterations in gene pairs are those that occur together less frequently than may be expected and may denote a synthetically lethal relationship (SSL) between the genes. SSLs can be exploited therapeutically to selectively kill cancer cells. Here, we analysed mutation, copy number variation, and methylation levels in samples from The Cancer Genome Atlas, using the hypergeometric and the Poisson binomial tests to identify mutually exclusive inactivated genes. We focused on gene pairs where one is an inactivated tumour suppressor and the other a gene whose protein product can be inhibited by known drugs. This provided an abundance of potential targeted therapeutics and repositioning opportunities for several cancers. These data are available on the MexDrugs website, https://bioinformaticslab.sussex.ac.uk/mexdrugs.
    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
    包括聚(ADP-核糖)聚合酶(PARP)抑制剂在治疗无法手术的肿瘤患者中具有显着改善的结果。PARP抑制剂通过在DNA损伤位点捕获聚(ADP-核糖)聚合酶(PARP)来阻碍单链脱氧核糖核酸(DNA)修复。形成无功能的“PARP酶抑制剂复合物”,导致细胞毒性。在PARP上调和同源重组(HR)缺陷如乳腺癌相关基因(BRCA1/2)的存在下,该效应更显著。因此,通过基因组分析识别HR缺陷-例如,用于三阴性乳腺癌的BRCA1/2应该是PARP抑制剂治疗选择过程的一部分。发表的数据表明,BRCA1/2种系突变并不能一致地预测对PARP抑制剂的有利反应。表明肿瘤突变状态以外的其他因素可能在起作用。各种因素,包括PARP表达的肿瘤异质性以及对PARP抑制剂的内在和/或获得性抗性,可能是促成因素。这证明了使用额外的工具进行适当的患者选择,是非侵入性的,并且能够评估全身体内PARP表达并评估PARP抑制剂的药代动力学,作为目前可用的BRCA1/2分析的补充。在这次审查中,我们讨论了[18F]氟PARP抑制剂放射性示踪剂及其在PARP表达和PARP抑制剂药代动力学成像中的潜力。为了提供背景,我们还简要讨论了PARP抑制剂耐药或无效的可能原因。讨论的重点是TNBC,这是一种肿瘤类型,其中PARP抑制剂被用作标准治疗策略的一部分。
    Including poly(ADP-ribose) polymerase (PARP) inhibitors in managing patients with inoperable tumors has significantly improved outcomes. The PARP inhibitors hamper single-strand deoxyribonucleic acid (DNA) repair by trapping poly(ADP-ribose)polymerase (PARP) at sites of DNA damage, forming a non-functional \"PARP enzyme-inhibitor complex\" leading to cell cytotoxicity. The effect is more pronounced in the presence of PARP upregulation and homologous recombination (HR) deficiencies such as breast cancer-associated gene (BRCA1/2). Hence, identifying HR-deficiencies by genomic analysis-for instance, BRCA1/2 used in triple-negative breast cancer-should be a part of the selection process for PARP inhibitor therapy. Published data suggest BRCA1/2 germline mutations do not consistently predict favorable responses to PARP inhibitors, suggesting that other factors beyond tumor mutation status may be at play. A variety of factors, including tumor heterogeneity in PARP expression and intrinsic and/or acquired resistance to PARP inhibitors, may be contributing factors. This justifies the use of an additional tool for appropriate patient selection, which is noninvasive, and capable of assessing whole-body in vivo PARP expression and evaluating PARP inhibitor pharmacokinetics as complementary to the currently available BRCA1/2 analysis. In this review, we discuss [18F]Fluorine PARP inhibitor radiotracers and their potential in the imaging of PARP expression and PARP inhibitor pharmacokinetics. To provide context we also briefly discuss possible causes of PARP inhibitor resistance or ineffectiveness. The discussion focuses on TNBC, which is a tumor type where PARP inhibitors are used as part of the standard-of-care treatment strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号