DNA-PK inhibitor

DNA - PK 抑制剂
  • 文章类型: Journal Article
    (1)头颈部鳞状细胞癌(HNSCC)是常见的,虽然治疗很困难,死亡率很高。激酶抑制剂有望增强放射治疗的效果。我们比较了PARP抑制剂talazoparib和niraparib以及DNA-PKcs抑制剂AZD7648联合电离辐射的效果。(2)七个HNSCC细胞系,包括Cal33,CLS-354,底特律562,HSC4,RPMI2650(HPV阴性),UD-SCC-2和UM-SCC-47(HPV阳性),和两个健康的成纤维细胞细胞系,研究了SBLF8和SBLF9。流式细胞术用于分析凋亡和坏死诱导(AnnexinV/7AAD)和细胞周期分布(Hoechst)。通过集落形成测定法研究细胞失活。(3)AZD7648效应最强,放射增敏所有HNSCC细胞系,几乎总是以超相加的方式。Talazoparib和niraparib在两种HPV阳性细胞系中均有效,但仅在一种和两种HPV阴性细胞系中均有效。分别。健康的成纤维细胞不受凋亡和坏死诱导或G2/M期停滞的任何联合治疗的影响。AZD7648单独对健康成纤维细胞没有毒性,而与电离辐射的结合降低了克隆性。(4)总之,talazoparib,尼拉帕利和,最有力的,AZD7648可以改善HNSCC的放射治疗。健康的成纤维细胞单独耐受AZD7648非常好,但是辐射诱导的效应可能会发生。我们的结果证明了体内研究的正确性。
    (1) Head and neck squamous cell carcinoma (HNSCC) is common, while treatment is difficult, and mortality is high. Kinase inhibitors are promising to enhance the effects of radiotherapy. We compared the effects of the PARP inhibitors talazoparib and niraparib and that of the DNA-PKcs inhibitor AZD7648, combined with ionizing radiation. (2) Seven HNSCC cell lines, including Cal33, CLS-354, Detroit 562, HSC4, RPMI2650 (HPV-negative), UD-SCC-2 and UM-SCC-47 (HPV-positive), and two healthy fibroblast cell lines, SBLF8 and SBLF9, were studied. Flow cytometry was used to analyze apoptosis and necrosis induction (AnnexinV/7AAD) and cell cycle distribution (Hoechst). Cell inactivation was studied by the colony-forming assay. (3) AZD7648 had the strongest effects, radiosensitizing all HNSCC cell lines, almost always in a supra-additive manner. Talazoparib and niraparib were effective in both HPV-positive cell lines but only consistently in one and two HPV-negative cell lines, respectively. Healthy fibroblasts were not affected by any combined treatment in apoptosis and necrosis induction or G2/M-phase arrest. AZD7648 alone was not toxic to healthy fibroblasts, while the combination with ionizing radiation reduced clonogenicity. (4) In conclusion, talazoparib, niraparib and, most potently, AZD7648 could improve radiation therapy in HNSCC. Healthy fibroblasts tolerated AZD7648 alone extremely well, but irradiation-induced effects might occur. Our results justify in vivo 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
    放疗的有效性取决于“正常”细胞和癌细胞对所施用的辐射剂量的敏感性。通过抑制DNA损伤修复来提高癌症的放射敏感性是当前许多研究的目标。然而,成功取决于避免在治疗期间不可避免地照射正常组织的伴随致敏。在这项研究中,我们通过检查对增殖和静止细胞群体的影响,研究了DNA-PK和PARP抑制剂的放射增敏机制。在体外和体内在BRCA1/2null和野生型亲本癌症模型中进行实验。总体上,AZD7648相对于Olaparib具有更大的放射增敏活性,BRCA2缺陷模型显示出最大的敏感性。然而,DNA-PK抑制剂AZD7648在所有照射的小鼠中也产生更大的毒性。虽然DNA-PK和PARP抑制使野生型肿瘤细胞对辐射敏感,在BRCA1/2缺陷型细胞中,奥拉帕尼对PARP的抑制具有有限的放射增敏能力.静止细胞比增殖细胞更耐辐射,这些也被AZD7648有效致敏,而Olaparib无法增加辐射诱导的细胞杀伤,即使在BRCA1/2null单元格中。这些发现强调了DNA-PK和PARP抑制剂的放射增敏的不同机制。虽然DNA-PK抑制剂能够靶向增殖和非增殖肿瘤细胞,以获得更大的整体抗癌益处,它们的应用受到正常组织毒性恶化的限制。相反,PARP抑制剂对增殖细胞具有选择性活性,提供了一种靶向癌症的机制,但是由于非增殖细胞的活性差,它们对肿瘤生长控制的影响总体上降低了。这项研究强调了用DNA损伤修复抑制辐射致敏策略创建治疗比例的重要性。
    The effectiveness of radiotherapy depends on the sensitivities of \'normal\' and cancer cells to the administered radiation dose. Increasing the radiosensitivity of cancers by inhibiting DNA damage repair is a goal of much current research, however success depends on avoiding concomitant sensitization of normal tissues inevitably irradiated during therapy. In this study we investigated the mechanisms of radiosensitization for DNA-PK and PARP inhibitors by examining the impacts on proliferating vs quiescent cell populations. Experiments were performed in BRCA1/2null and wild-type parental cancer models in vitro and in vivo. Overall AZD7648 has greater radiosensitizing activity relative to Olaparib, with BRCA2-deficient models showing the greatest sensitivity. However, DNA-PK inhibitor AZD7648 also produced greater toxicity in all irradiated mice. While both DNA-PK and PARP inhibition sensitizes wild type tumor cells to radiation, in BRCA1/2 deficient cells PARP inhibition by Olaparib had limited radiosensitization capacity. Quiescent cells are more radioresistant than proliferating cells, and these were also effectively sensitized by AZD7648 while Olaparib was unable to increase radiation-induced cell kill, even in BRCA1/2null cells. These findings underscore the distinct mechanisms of radiosensitization for DNA-PK and PARP inhibitors. While DNA-PK inhibitors are able to target both proliferating and non-proliferating tumor cells for greater overall anti-cancer benefit, their application is limited by exacerbation of normal tissue toxicities. Conversely, PARP inhibitors exhibit selective activity for proliferating cells, providing a mechanism for targeting activity to cancers, but due to poor activity in non-proliferating cells they have an overall reduced impact on tumor growth control. This study highlights the importance of creating a therapeutic ratio with DNA damage repair inhibition radiation sensitizing strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    据报道,在非同源末端连接修复途径中抑制DNA依赖性蛋白激酶(DNA-PK)会增加癌细胞的辐射敏感性。我们最近报道了BR101801,PI3K-γ(γ)的新型三联抑制剂,delta(δ),和DNA-PK,在各种人类实体癌细胞和异种移植小鼠模型中,充当辐射诱导的DNA损伤的有效敏化剂。鉴于p53抑癌基因在放疗疗效中起着重要作用,在目前的研究中,我们使用等基因HCT116p53+/+和HCT116p53-/-人结直肠癌细胞系,重点研究了p53状态对BR101801诱导的放射增敏作用的影响.体外,HCT116p53+/+和HCT116p53-/-人结直肠癌细胞用1μMBR101801预处理24小时,然后暴露于电离辐射(IR)。然后是分析集落形成的方法,DNA损伤,细胞周期的变化,衰老,自噬,凋亡,和DNA损伤反应相关蛋白。构建异种移植小鼠模型以检查BR101801(50mg/kg,每天口服一次)和对体内肿瘤生长抑制作用的IR(2Gy×3天)。BR101801抑制HCT116p53+/+和HCT116p53-/-人结直肠癌细胞的细胞增殖并延长DNA损伤。BR101801和IR联合治疗强烈诱导G2/M期细胞周期停滞,凋亡,与单独IR治疗相比,HCT116p53-/-细胞的细胞衰老。此外,BR101801协同抑制HCT116p53-/-异种移植小鼠模型中的肿瘤生长。BR101801增强了HCT116人结直肠癌细胞的放射敏感性,无论其p53状态如何。此外,BR101801对IR诱导的细胞周期阻滞发挥了强大的协同作用,凋亡,和肿瘤生长抑制,甚至在耐放射性HCT116p53-/-细胞中。总的来说,这些发现为将BR101801与IR结合作为克服p53缺陷引起的放射抗性的新治疗策略提供了科学依据.
    Inhibition of DNA-dependent protein kinase (DNA-PK) in the non-homologous end-joining repair pathway reportedly increases the radiation sensitivity of cancer cells. We have recently reported that BR101801, a novel triple inhibitor of PI3K-gamma (γ), delta (δ), and DNA-PK, functions as an efficient sensitizer of radiation-induced DNA damage in various human solid cancer cells and a xenograft mouse model. Given that the p53 tumor suppressor gene plays an important role in radiotherapeutic efficacy, in the current study, we focused on the impact of the p53 status on BR101801-induced radiosensitization using isogenic HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cell lines. In vitro, HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cells were pretreated with 1 μM BR101801 for 24 h before exposure to ionizing radiation (IR), followed by assays to analyze colony formation, DNA damage, cell cycle changes, senescence, autophagy, apoptosis, and DNA damage response-related proteins. Xenograft mouse models were constructed to examine the potential synergistic effects of BR101801 (50 mg/kg, orally administered once daily) and fractionated IR (2 Gy × 3 days) on tumor growth inhibition in vivo. BR101801 inhibited cell proliferation and prolonged DNA damage in both HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cells. Combined treatment with BR101801 and IR robustly induced G2/M phase cell cycle arrest, apoptosis, and cellular senescence in HCT116 p53-/- cells when compared with treatment with IR alone. Furthermore, BR101801 synergistically inhibited tumor growth in the HCT116 p53-/- xenograft mouse model. BR101801 enhanced the radiosensitivity of HCT116 human colorectal cancer cells regardless of their p53 status. Moreover, BR101801 exerted robust synergistic effects on IR-induced cell cycle arrest, apoptosis, and tumor growth inhibition, even in radioresistant HCT116 p53-/- cells. Overall, these findings provide a scientific rationale for combining BR101801 with IR as a new therapeutic strategy to overcome radioresistance induced by p53 deficiency.
    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
    非同源末端连接途径对于修复DNA双链断裂(DSB)至关重要,DNA依赖性蛋白激酶(DNA-PK)起着关键作用。慢性(CML)和急性髓细胞性白血病(AML)中改变的DNA损伤反应(DDR)提供了潜在的治疗机会。我们研究了AZD-7648(DNA-PK抑制剂)在CML和AML细胞系中的治疗潜力。本研究使用了两种CML(K-562和LAMA-84)和五种AML(HEL,HL-60,KG-1,NB-4和THP-1)细胞系。DDR基因突变来自COSMIC数据库。使用MS-MLPA和DDR基因评估拷贝数和甲基化谱,和端粒长度使用qPCR。使用WesternBlot评估p53蛋白表达,通过胞质分裂-阻断微核试验对染色体的损伤,和γH2AX水平和DSB修复动力学使用流式细胞术。在用浓度范围为10至200μM的AZD-7648处理后,使用锥虫蓝测定法分析细胞密度和活力。细胞死亡,细胞周期分布,使用流式细胞术评估细胞增殖率。细胞表现出不同的DNA基线损伤,DDR基因表达,突变,遗传/表观遗传变化,和p53表达。只有HEL单元显示低效的DSB修复。LAMA-84HEL,和KG-1细胞对AZD-7648最敏感,而HL-60和K-562对密度和活力的影响较低。除了细胞增殖的减少,AZD-7648诱导细胞凋亡,细胞周期停滞,和DNA损伤。总之,这些结果表明,AZD-7648有望成为髓系白血病的潜在治疗方法,然而,随着测试细胞系中药物敏感性的变化,因此支持进一步研究以确定影响该DNA-PK抑制剂敏感性的具体因素.
    The non-homologous end joining pathway is vital for repairing DNA double-strand breaks (DSB), with DNA-dependent protein kinase (DNA-PK) playing a critical role. Altered DNA damage response (DDR) in chronic (CML) and acute myeloid leukemia (AML) offers potential therapeutic opportunities. We studied the therapeutic potential of AZD-7648 (DNA-PK inhibitor) in CML and AML cell lines. This study used two CML (K-562 and LAMA-84) and five AML (HEL, HL-60, KG-1, NB-4, and THP-1) cell lines. DDR gene mutations were obtained from the COSMIC database. The copy number and methylation profile were evaluated using MS-MLPA and DDR genes, and telomere length using qPCR. p53 protein expression was assessed using Western Blot, chromosomal damage through cytokinesis-block micronucleus assay, and γH2AX levels and DSB repair kinetics using flow cytometry. Cell density and viability were analyzed using trypan blue assay after treatment with AZD-7648 in concentrations ranging from 10 to 200 µM. Cell death, cell cycle distribution, and cell proliferation rate were assessed using flow cytometry. The cells displayed different DNA baseline damage, DDR gene expressions, mutations, genetic/epigenetic changes, and p53 expression. Only HEL cells displayed inefficient DSB repair. The LAMA-84, HEL, and KG-1 cells were the most sensitive to AZD-7648, whereas HL-60 and K-562 showed a lower effect on density and viability. Besides the reduction in cell proliferation, AZD-7648 induced apoptosis, cell cycle arrest, and DNA damage. In conclusion, these results suggest that AZD-7648 holds promise as a potential therapy for myeloid leukemias, however, with variations in drug sensitivity among tested cell lines, thus supporting further investigation to identify the specific factors influencing sensitivity to this DNA-PK inhibitor.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase I
    背景:DNA依赖性蛋白激酶(DNA-PK)在通过非同源末端连接修复DNA双链断裂(DSB)中起关键作用。抑制DNA-PK可以增强DNADSB诱导抗癌疗法的效果。Peposertib(以前称为M3814)是一种口服给药,强力,和选择性小分子DNA-PK抑制剂,在异种移植模型中已证明具有放射增敏和抗肿瘤活性,并且在单一疗法中具有良好的耐受性。这项I期试验(NCT02516813)研究了最大耐受剂量,推荐的II期剂量(RP2D),安全,胸或头颈部肿瘤患者(A组)中peposertib联合姑息性放疗(RT)的耐受性和头颈部鳞状细胞癌患者(B组)中peposertib联合顺铂和根治性RT的耐受性。
    方法:患者每天一次(QD)以片剂或胶囊的递增剂量分组,联合姑息性RT(A组)或联合强度调节的治愈性RT和顺铂(B组)。
    结果:最常见的因治疗引起的不良事件(TEAE)是放射性皮肤损伤,疲劳,A组恶心(n=34)和口腔炎,恶心,放射性皮肤损伤,和B臂的味觉障碍(n=11)。根据对剂量限制性毒性的评估,耐受性,和药代动力学数据,A组的RP2D被宣布为200mgpeposertib片剂QD与RT组合。在B组(n=11)中,50mgpeposertib与治愈性RT和顺铂联合使用被宣布为可耐受。然而,由于该剂量的暴露量不足,因此终止了入组,并且未正式宣布RP2D.
    结论:Peposertib与姑息性RT联合使用,对于每个RT部分的200mgQD剂量的片剂,耐受性良好。当与RT和顺铂联合使用时,可耐受的peposertib剂量导致暴露不足。
    OBJECTIVE: DNA-dependent protein kinase (DNA-PK) plays a key role in the repair of DNA double strand breaks via nonhomologous end joining. Inhibition of DNA-PK can enhance the effect of DNA double strand break inducing anticancer therapies. Peposertib (formerly \"M3814\") is an orally administered, potent, and selective small molecule DNA-PK inhibitor that has demonstrated radiosensitizing and antitumor activity in xenograft models and was well-tolerated in monotherapy. This phase 1 trial (National Clinical Trial 02516813) investigated the maximum tolerated dose, recommended phase 2 dose (RP2D), safety, and tolerability of peposertib in combination with palliative radiation therapy (RT) in patients with thoracic or head and neck tumors (arm A) and of peposertib in combination with cisplatin and curative-intent RT in patients with squamous cell carcinoma of the head and neck (arm B).
    METHODS: Patients received peposertib once daily in ascending dose cohorts as a tablet or capsule in combination with palliative RT (arm A) or in combination with intensity modulated curative-intent RT and cisplatin (arm B).
    RESULTS: The most frequently observed treatment-emergent adverse events were radiation skin injury, fatigue, and nausea in arm A (n = 34) and stomatitis, nausea, radiation skin injury, and dysgeusia in arm B (n = 11). Based on evaluations of dose-limiting toxicities, tolerability, and pharmacokinetic data, RP2D for arm A was declared as 200 mg peposertib tablet once daily in combination with RT. In arm B (n = 11), 50 mg peposertib was declared tolerable in combination with curative-intent RT and cisplatin. However, enrollment was discontinued because of insufficient exposure at that dose, and the RP2D was not formally declared.
    CONCLUSIONS: Peposertib in combination with palliative RT was well-tolerated up to doses of 200 mg once daily as tablet with each RT fraction. When combined with RT and cisplatin, a tolerable peposertib dose yielded insufficient exposure.
    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
    局部晚期直肠癌的治疗包括放化疗和手术,但是患者对治疗的反应是可变的。有完全反应的患者有改善的结果;因此,迫切需要确定抵抗机制以规避它们。DNA-PK参与由辐射引起的DNA双链断裂的修复,我们发现直肠癌在治疗后会增加。我们假设用DNA-PK抑制剂抑制这种复合物,Peposertib(M3814),会改善治疗反应。我们使用蛋白质印迹法评估了直肠癌细胞系和小鼠模型中的pDNA-PK,活力测定,γH2AX染色,和治疗反应。三个治疗组是:标准护理(SOC)(5-氟尿嘧啶(5FU)与辐射),M3814带辐射,和带SOC的M3814。SOC治疗直肠癌细胞增加pDNA-PK蛋白和增加γH2AX灶,但这被M3814的加入废除了。用具有SOC的M3814处理的具有CT26肿瘤的小鼠在平均肿瘤大小上没有差异,但是个体肿瘤应答变化。添加M3814后,临床完全缓解率显着提高,但病理完全缓解却没有。我们研究了DNA修复的改变,发现添加M3814后Kap1和pATM增加,表明这可能介导抗性。当DNA-PK抑制剂,M3814,结合SOC治疗,在某些直肠癌模型中,反应有所改善,但其他修复机制的增加可能会降低效果。正在进行一项临床试验,以进一步探索DNA-PK抑制在直肠癌治疗中的作用。
    Treatment of locally advanced rectal cancer includes chemoradiation and surgery, but patient response to treatment is variable. Patients who have a complete response have improved outcomes; therefore, there is a critical need to identify mechanisms of resistance to circumvent them. DNA-PK is involved in the repair of DNA double-strand breaks caused by radiation, which we found to be increased in rectal cancer after treatment. We hypothesized that inhibiting this complex with a DNA-PK inhibitor, Peposertib (M3814), would improve treatment response. We assessed pDNA-PK in a rectal cancer cell line and mouse model utilizing western blotting, viability assays, γH2AX staining, and treatment response. The three treatment groups were: standard of care (SOC) (5-fluorouracil (5FU) with radiation), M3814 with radiation, and M3814 with SOC. SOC treatment of rectal cancer cells increased pDNA-PK protein and increased γH2AX foci, but this was abrogated by the addition of M3814. Mice with CT26 tumors treated with M3814 with SOC did not differ in average tumor size but individual tumor response varied. The clinical complete response rate improved significantly with the addition of M3814 but pathological complete response did not. We investigated alterations in DNA repair and found that Kap1 and pATM are increased after M3814 addition suggesting this may mediate resistance. When the DNA-PK inhibitor, M3814, is combined with SOC treatment, response improved in some rectal cancer models but an increase in other repair mechanisms likely diminishes the effect. A clinical trial is ongoing to further explore the role of DNA-PK inhibition in rectal cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    DNA-dependent protein kinase (DNA-PK), an essential component of the non-homologous end-joining (NHEJ) repair pathway, plays an important role in DNA damage repair (DDR). Therefore, DNA-PK inhibition is a promising approach for overcoming radiotherapy or chemotherapy resistance in cancers. In this study, we demonstrated that BR101801, a potent DNA-PK inhibitor, acted as an effective radiosensitizer in various human solid cancer cells and an in vivo xenograft model. Overall, BR101801 strongly elevated ionizing radiation (IR)-induced genomic instability via induction of cell cycle G2/M arrest, autophagic cell death, and impairment of DDR pathway in human solid cancer cells. Interestingly, BR101801 inhibited not only phosphorylation of DNA-PK catalytic subunit in NHEJ factors but also BRCA2 protein level in homologous recombination (HR) factors. In addition, combination BR101801 and IR suppressed tumor growth compared with IR alone by reducing phosphorylation of DNA-PK in human solid cancer xenografts. Our findings suggested that BR101801 is a selective DNA-PK inhibitor with a synergistic radiosensitizing effect in human solid cancers, providing evidence for clinical applications.
    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
    OBJECTIVE: Inhibitors of DNA-dependent protein kinase (DNA-PK) are effective radiation sensitisers in preclinical tumours, but little is known about risks of normal tissue radiosensitisation. Here, we evaluate radiosensitisation of head and neck squamous cell carcinoma (HNSCC) cells by DNA-PK inhibitor AZD7648 under oxia and anoxia in vitro, and tumour (SCCVII), oral mucosa and small intestine in mice.
    METHODS: Radiosensitisation of human (UT-SCC-54C) and murine (SCCVII) HNSCC cells by AZD7648 under oxia and anoxia was evaluated by clonogenic assay. Radiosensitisation of SCCVII tumours in C3H mice by oral AZD7648 (75 mg/kg) was determined by ex vivo clonogenic assay 3.5 days post-irradiation, with evaluation of normal tissue surrogate endpoints using 5-ethynyl-2\'-deoxyuridine to facilitate detection of regenerating crypts in the ileum and repopulating S-phase cells in the ileum and oral mucosa of the same animals.
    RESULTS: AZD7648 potently radiosensitised both cell lines, with similar sensitiser enhancement ratios for 10% survival (SER10) under oxia and anoxia. AZD7648 diffused rapidly through multicellular layers, suggesting rapid equilibration between plasma and hypoxic zones in tumours. SCCVII tumours were radiosensitised by AZD7648 (SER10 2.5). AZD7648 also enhanced radiation-induced body weight loss and suppressed regenerating intestinal crypts and repopulating S-phase cells in the ileum and tongue epithelium with SER values similar to SCCVII tumours.
    CONCLUSIONS: AZD7648 is a potent radiation sensitiser of both oxic and anoxic tumour cells, but also markedly radiosensitises stem cells in the small intestine and oral mucosa.
    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
    UNASSIGNED: This first-in-human Phase I study investigated the safety, pharmacokinetics (PK), pharmacodynamic profile, and preliminary efficacy of CC-115, a dual inhibitor of mammalian target of rapamycin (mTOR) kinase and DNA-dependent protein kinase.
    UNASSIGNED: Patients with advanced solid or hematologic malignancies were enrolled in dose-finding and cohort expansion phases. In dose-finding, once-daily or twice-daily (BID) ascending oral doses of CC-115 (range: 0.5-40 mg/day) in 28-day continuous cycles identified the maximum-tolerated dose for cohort expansion in 5 specified tumor types. Twelve additional patients with mixed solid tumors participated in a bioavailability substudy.
    UNASSIGNED: Forty-four patients were enrolled in the dose-finding cohort. Dose-limiting toxicity included thrombocytopenia, stomatitis, hyperglycemia, asthenia/fatigue, and increased transaminases. CC-115 10 mg BID was selected for cohort expansion (n=74) in which fatigue, nausea, and decreased appetite were the most frequent toxicities. Dose-proportional PK was found. CC-115 distributed to glioblastoma tissue (mean tumor/plasma concentration ratio: 0.713). Total exposure of CC-115 was similar under fasting and fed conditions. A patient with endometrial carcinoma remained in complete remission >4 years. Partial response (PR; n=2) and stable disease (SD; n=4) were reported in the bioavailability substudy; SD was reached in 53%, 22%, 21%, and 64% of patients with head and neck squamous cell carcinoma, Ewing sarcoma, glioblastoma multiforme, and castration-resistant prostate cancer, respectively. Chronic lymphocytic leukemia/small lymphocytic lymphoma showed 38% PR and 25% SD.
    UNASSIGNED: CC-115 was well-tolerated, with toxicities consistent with mTOR inhibitors. Together with biomarker inhibition and preliminary efficacy, oral CC-115 10 mg BID is a promising novel anticancer treatment.
    UNASSIGNED: NCT01353625.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: DNA-PK and PARP inhibitors sensitize cancer cells to chemo- and radiotherapy. ETS transcription factors (EWS-FLI1) have been described as biomarkers for PARP-inhibitor sensitivity. Sensitivity to single agent PARP inhibitors has so far been limited to homologous recombination repair (HRR) deficient tumors, exploiting synthetic lethality.
    RESULTS: In clonogenic assays, single agent rucaparib LD50 values for continuously exposed cells were similar to those observed in HRR-defective cells (CAPAN-1 cell line, BRCA2 defective); however, both ES cell lines (TC-71, CADO-ES1) had functional HRR. In vivo rucaparib administration (10 mg/kg daily) showed no responses. In clonogenic assays, rucaparib enhanced temozolomide, camptothecin and radiation cytotoxicity, which was most profound for temozolomide (15-29 fold enhancement). NU7441 increased the cytotoxicity of etoposide, doxorubicin and radiation.
    METHODS: We assessed PARP1/2 (rucaparib) and DNA-PK (NU7441) inhibitors in Ewing sarcoma (ES) cell lines by performing growth inhibition and clonogenic assays. HRR was measured by RAD51 focus formation. Single agent rucaparib was assessed in an in vivo orthotopic model.
    CONCLUSIONS: Single agent rucaparib ES sensitivity in vitro was not replicated in vivo. DNA-PK and PARP inhibitors are good chemo-/radiosensitizers in ES. The future of these inhibitors lies in their combination with chemo-/radiotherapy, which needs to be evaluated in clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号