Hypoxia-activated prodrug

  • 文章类型: Journal Article
    三阴性乳腺癌(TNBC)是最致命的乳腺癌亚型。缺氧激活的前药(HAP)已显示出有望作为TNBC的潜在治疗剂。虽然增加缺氧水平可能会促进HAP的激活,它引起了人们对HIF1α依赖性耐药性的担忧。期望开发一种靶向方法,其增强肿瘤缺氧以激活HAP而不促进TNBC治疗中的HIF1α依赖性药物抗性。在这里,提出了一种多响应无载体自组装纳米药物AQ4N@CA4T1ASO。这种纳米药物首先通过TNBC靶向适体(T1)靶向肿瘤,然后在肿瘤内的还原性和酸性条件下分解。释放的Combrestatin4(CA4)可以加剧缺氧,从而促进无活性的Banoxantrone(AQ4N)向其活性形式的转化,AQ4.同时,所释放的反义寡核苷酸(ASO)可以减弱缺氧诱导的HIF1αmRNA表达,从而使肿瘤对化疗敏感。总的来说,这种智能纳米医学代表了一种深刻的靶向治疗策略,结合“增强缺氧,缺氧激活,TNBC治疗的化学致敏方法。体内研究证明了肿瘤生长的显著抑制,强调了这种纳米医学在未来临床翻译中的有希望的潜力。
    Triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer. Hypoxia-activated prodrugs (HAPs) have shown promise as potential therapeutic agents for TNBC. While increasing hypoxia levels may promote the HAP activation, it raises concerns regarding HIF1α-dependent drug resistance. It is desirable to develop a targeted approach that enhances tumor hypoxia for HAP activation without promoting HIF1α-dependent drug resistance in TNBC treatment. Herein, we proposed a multi-responsive carrier-free self-assembled nanomedicine named AQ4N@CA4T1ASO. This nanomedicine first targeted tumors by the TNBC-targeting aptamers (T1), and then disassembled in the reductive and acidic conditions within tumors. The released Combretastatin 4 (CA4) could exacerbate hypoxia, thereby promoting the conversion of inactive Banoxantrone (AQ4N) to its active form, AQ4. Simultaneously, the released antisense oligonucleotide (ASO) could attenuate hypoxia-induced HIF1α mRNA expression, thereby sensitizing the tumor to chemotherapy. Overall, this smart nanomedicine represents a profound targeted therapy strategy, combining \"hypoxia-potentiating, hypoxia-activated, chemo-sensitization\" approaches for TNBC treatment. In vivo study demonstrated significant suppression of tumor growth, highlighting the promising potential of this nanomedicine for future clinical translation.
    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
    优化化疗以提高抗肿瘤治疗效果势在必行。不受约束的肿瘤细胞增殖和持续的血管生成是癌症进展的关键。普那布林,血管破坏剂,选择性地破坏肿瘤血管。Tirapazamine(TPZ),缺氧激活的前药,在减少肿瘤细胞内的氧水平时增强细胞毒性。尽管完成了III期临床试验,由于剂量限制性毒性,两种药物均表现出适度的治疗效率.在这项研究中,我们使用甲氧基聚(乙二醇)-b-聚(D,L-丙交酯)(mPEG-b-PDLLA)将普那布林和TPZ共同递送至肿瘤部位,同时破坏血管和消除肿瘤细胞,解决症状和肿瘤进展的根本原因。普那布林转化为具有酯酶反应(PSM)的前药,和TPZ被合成为含己基链的衍生物(TPZHex)用于有效的共递送。PSM和TPZHex与mPEG-b-PDLLA共封装,形成纳米药物(PT-NP)。在肿瘤部位,PT-NP对酯酶过表达有反应,释放普那布林,破坏血管,导致营养和缺氧。TPZHex响应于缺氧增加而被激活,杀死肿瘤细胞。在治疗4T1肿瘤时,PT-NP表现出增强的治疗效果,达到92.9%的抑瘤率和20%的治愈率。这项研究提出了一种创新的策略,以增强联合化疗的协同功效并减少毒性。
    It is imperative to optimize chemotherapy for heightened anti-tumor therapeutic efficacy. Unrestrained tumor cell proliferation and sustained angiogenesis are pivotal for cancer progression. Plinabulin, a vascular disrupting agent, selectively destroys tumor blood vessels. Tirapazamine (TPZ), a hypoxia-activated prodrug, intensifies cytotoxicity in diminishing oxygen levels within tumor cells. Despite completing Phase III clinical trials, both agents exhibited modest treatment efficiency due to dose-limiting toxicity. In this study, we employed methoxy poly(ethylene glycol)-b-poly(D,L-lactide) (mPEG-b-PDLLA) to co-deliver Plinabulin and TPZ to the tumor site, concurrently disrupting blood vessels and eliminating tumor cells, addressing both symptoms and the root cause of tumor progression. Plinabulin was converted into a prodrug with esterase response (PSM), and TPZ was synthesized into a hexyl chain-containing derivative (TPZHex) for effective co-delivery. PSM and TPZHex were co-encapsulated with mPEG-b-PDLLA, forming nanodrugs (PT-NPs). At the tumor site, PT-NPs responded to esterase overexpression, releasing Plinabulin, disrupting blood vessels, and causing nutritional and oxygen deficiency. TPZHex was activated in response to increased hypoxia, killing tumor cells. In treating 4T1 tumors, PT-NPs demonstrated enhanced therapeutic efficacy, achieving a 92.9 % tumor suppression rate and a 20 % cure rate. This research presented an innovative strategy to enhance synergistic efficacy and reduce toxicity in combination chemotherapy.
    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
    Tirapazamine(TPZ)凭借其出色的抗癌潜力已被批准用于多项临床试验。然而,作为典型的缺氧激活前药(HAP),由于肿瘤患者缺氧水平不足,TPZ在III期临床试验中用于联合治疗时没有表现出生存优势。在这项研究中,为了提高TPZ的治疗效果,我们首先引入尿素合成了一系列含尿素的TPZ衍生物。与TPZ相比,所有含尿素的TPZ衍生物均显示出增加的缺氧细胞毒性(9.51-30.85倍),同时保持低氧选择性。TPZP,这些衍生物之一,显示比TPZ高20倍的细胞毒性,同时保持相似的缺氧细胞毒性比率。为了高效地将TPZP递送至肿瘤并减少其对健康组织的副作用,我们进一步将TPZP制成具有纤维蛋白靶向能力的纳米药物:FT11-TPZP-NP。CA4-NP,血管破坏剂,用于增加肿瘤内的纤维蛋白水平并加剧肿瘤缺氧。通过与CA4-NP结合,FT11-TPZP-NP可以在缺氧加重的肿瘤中积累并充分激活以杀死肿瘤细胞。单剂量治疗后,FT11-TPZP-NPs+CA4-NPs对初始体积为480mm3的CT26肿瘤模型显示出98.1%的高抑制率,六个肿瘤中有四个被完全消除;从而发挥了显著的抗肿瘤作用。本研究为提高TPZ和其他HAPs在抗癌治疗中的疗效提供了新的策略。
    Tirapazamine (TPZ) has been approved for multiple clinical trials relying on its excellent anticancer potential. However, as a typical hypoxia-activated prodrug (HAP), TPZ did not exhibit survival advantages in Phase III clinical trials when used in combination therapy due to the insufficient hypoxia levels in patients\' tumors. In this study, to improve the therapeutic effects of TPZ, we first introduced urea to synthesize a series of urea-containing derivatives of TPZ. All urea-containing TPZ derivatives showed increased hypoxic cytotoxicity (9.51-30.85-fold) compared with TPZ, while maintaining hypoxic selectivity. TPZP, one of these derivatives, showed 20-fold higher cytotoxicity than TPZ while maintaining a similar hypoxic cytotoxicity ratio. To highly efficiently deliver TPZP to the tumors and reduce its side effects on healthy tissues, we further prepared TPZP into a nanodrug with fibrin-targeting ability: FT11-TPZP-NPs. CA4-NPs, a vascular disrupting agent, was used to increase the fibrin level within tumors and exacerbate tumor hypoxia. By being combined with CA4-NPs, FT11-TPZP-NPs can accumulate in the hypoxia-aggravated tumors and activate sufficiently to kill tumor cells. After a single-dose treatment, FT11-TPZP-NPs + CA4-NPs showed a high inhibition rate of 98.1% against CT26 tumor models with an initial volume of ∼480 mm3 and four out of six tumors were completely eliminated; it thereby exerted a significant antitumor effect. This study provides a new strategy for improving the therapeutic effect of TPZ and other HAPs in anticancer 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
    6-重氮-5-氧代-L-正亮氨酸(DON)是一种有效的谷氨酰胺拮抗剂,具有毒副作用;为了减少这些影响,已经设计了多种前药。然而,目前还没有报道DON前药具有明确的实现高肿瘤选择性的机制.提高DON对肿瘤细胞的选择性毒性,同时降低全身毒性,缺氧激活的前药,称为HDON,是设计的。HDON在高缺氧的H22鼠肝癌模型中实现了76.4±5.2%的显着肿瘤抑制,而不会导致体重减轻。此外,为了增强HDON的治疗效果,康布他汀A4纳米粒用于加重MC38小鼠结肠癌和4个T1小鼠乳腺癌的肿瘤缺氧,将HDON激活为DON,并刺激强大的抗肿瘤免疫反应,同时在体内选择性杀死肿瘤细胞,达到98.3±3.4%和98.1±3.1%的显着升高的肿瘤抑制率,治愈率分别为80.0%和20.0%,分别。
    6-Diazo-5-oxo-L-norleucine (DON) is a potent glutamine antagonist with toxic side effects; in order to reduce these effects, multiple prodrugs have been designed. However, there are currently no reports of a DON prodrug with a defined mechanism to achieve high tumor selectivity. To improve the selective toxicity of DON to tumor cells while reducing systemic toxicity, a hypoxia-activated prodrug, termed HDON, was designed. HDON achieved remarkable tumor suppression of 76.4 ± 5.2% without leading to weight loss in an H22 murine liver cancer model with high hypoxia. Moreover, to augment the therapeutic efficacy of HDON, combretastatin A4 nanoparticles were used to aggravate tumor hypoxia of MC38 murine colon cancer and 4T1 murine breast cancer, activate HDON to DON, and stimulate a robust anti-tumor immune response while selectively killing in tumor cells in vivo, achieving significantly elevated tumor suppression rates of 98.3 ± 3.4% and 98.1 ± 3.1%, with cure rates of 80.0% and 20.0%, respectively.
    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
    简介:癌症选择性,包括肿瘤细胞的靶向内化和加速药物释放,设计新型刺激响应性纳米载体以提高治疗效果仍然是一个主要挑战。光动力疗法(PDT)产生的低氧微环境被认为在化学抗性中起关键作用。方法:我们构建了双响应载体(DANPCT),该载体封装了光敏剂二氢卟啉e6(Ce6)和缺氧激活的前药替拉嗪(TPZ),以实现有效的PDT和PDT增强的缺氧激活化疗。结果和讨论:由于TAT掩蔽,DANPCT延长了血液中的有效载荷循环,通过酸度触发的TAT呈递发生选择性肿瘤细胞摄取。使用空间控制的660nm激光进行PDT,以实现精确的细胞杀伤并加剧缺氧。疏水性NI部分的缺氧响应转化导致DANPCT的分解,促进TPZ释放。TPZ在缺氧条件下被还原成细胞毒性自由基,有助于化疗级联。这项工作为编程化学PDT提供了一种复杂的策略。
    Introduction: Cancer selectivity, including targeted internalization and accelerated drug release in tumor cells, remains a major challenge for designing novel stimuli-responsive nanocarriers to promote therapeutic efficacy. The hypoxic microenvironment created by photodynamic therapy (PDT) is believed to play a critical role in chemoresistance. Methods: We construct dual-responsive carriers (DANPCT) that encapsulate the photosensitizer chlorin e6 (Ce6) and hypoxia-activated prodrug tirapazamine (TPZ) to enable efficient PDT and PDT-boosted hypoxia-activated chemotherapy. Results and discussion: Due to TAT masking, DANPCT prolonged payload circulation in the bloodstream, and selective tumor cell uptake occurred via acidity-triggered TAT presentation. PDT was performed with a spatially controlled 660-nm laser to enable precise cell killing and exacerbate hypoxia. Hypoxia-responsive conversion of the hydrophobic NI moiety led to the disassembly of DANPCT, facilitating TPZ release. TPZ was reduced to cytotoxic radicals under hypoxic conditions, contributing to the chemotherapeutic cascade. This work offers a sophisticated strategy for programmed chemo-PDT.
    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
    缺氧是实体瘤的突出特征之一。缺氧激活前药(HAP),选择性地杀死缺氧细胞,在精确治疗中具有将缺氧从令人讨厌转变为优势的潜力。表现出更显著的缺氧微环境,胶质瘤,作为最常见和无法治愈的神经肿瘤,为HAP提供了更有吸引力的治疗前景。然而,缺氧不足和血脑屏障(BBB)的阻塞严重限制了HAP的激活和生物利用度。在这里,设计并合成了一种新型的纳米颗粒iRGD@ZnPcTPZ,通过封装作为HAP的替拉帕明(TPZ)和作为光敏剂的酞菁锌(ZnPc)来增强缺氧,从而实现胶质瘤的抑制。iRGD@ZnPc+TPZ可以实现突破性BBB,深层渗透,并在神经胶质瘤中显著保留,这归因于iRGD介导的受体靶向和主动转运。被肿瘤细胞内化并辐射后,ZnPc有效消耗肿瘤内O2产生活性氧,不仅实现了肿瘤抑制,但也加强缺氧激活TPZ放大化疗。光敏剂增强的HAP激活抑制神经胶质瘤的生长。本研究为临床胶质瘤的治疗提供了一种新的致敏和激活HAP的策略。
    Hypoxia is one of the prominent features of solid tumors. Hypoxia activated prodrugs (HAPs), selectively killing hypoxic cells, possess the potential to transform hypoxia from a nuisance to an advantage in precision therapy. Exhibiting a more significant hypoxic microenvironment, gliomas, as the most frequent and incurable neurological tumors, provide HAPs a more attractive therapeutic prospect. However, the insufficient hypoxia and the obstruction of the blood-brain barrier (BBB) severely limit the activation and bio-availability of HAPs. Herein, a novel nanoparticle iRGD@ZnPc + TPZ was designed and synthesized to achieve gliomas inhibition by encapsulating tirapazamine (TPZ) as a HAP and zinc phthalocyanine (ZnPc) as a photosensitizer to enhance hypoxia. iRGD@ZnPc + TPZ can realize breakthrough BBB, deep penetration, and significant retention in gliomas, which is attributed to the iRGD-mediated receptor targeting and active transport. After being internalized by tumor cells and radiated, ZnPc efficiently consumes intratumoral O2 to produce reactive oxygen species, which not only implements tumor suppression, but also intensify hypoxia to activate TPZ for amplifying chemotherapy. The photosensitizer-enhanced activation of HAPs inhibits gliomas growth. This study provides a new strategy with sensitizing and activating HAPs for gliomas treatment in clinical.
    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
    缺氧激活的前药(HAP)在提高抗肿瘤效果的同时将副作用降至最低方面已引起越来越多的关注。然而,肿瘤中缺氧区域的分布不均严重阻碍了HAP的疗效。此外,大多数HAP不适合光学成像,很难在组织中精确追踪它们。在这里,我们精心设计并合成了一种多功能治疗性BAC前药,方法是将化疗药物喜树碱(CPT)和荧光光热剂硼二吡咯亚甲基(BODIPY)通过缺氧响应性偶氮苯接头连接。为了提高溶解度和肿瘤的积累,将制备的BAC进一步包封到基于人血清白蛋白(HSA)的药物递送系统中以形成HSA@BAC纳米颗粒。由于CPT在活性位点被基于BODIPY的分子笼住,BAC表现出优异的生物安全性。重要的是,激活的CPT可以从BAC中快速释放,并可以在缺氧的癌细胞中进行化疗,归因于过表达的偶氮还原酶对偶氮苯接头的裂解。用730nm激光照射后,HSA@BAC可以有效地产生热疗,通过不依赖氧的光热疗法实现不可逆的癌细胞死亡。在荧光成像引导的局部照射下,体外和体内研究均表明,HSA@BAC具有优异的抗肿瘤作用,副作用最小。
    Hypoxia-activated prodrugs (HAPs) have drawn increasing attention for improving the antitumor effects while minimizing side effects. However, the heterogeneous distribution of the hypoxic region in tumors severely impedes the curative effect of HAPs. Additionally, most HAPs are not amenable to optical imaging, and it is difficult to precisely trace them in tissues. Herein, we carefully designed and synthesized a multifunctional therapeutic BAC prodrug by connecting the chemotherapeutic drug camptothecin (CPT) and the fluorescent photothermal agent boron dipyrromethene (BODIPY) via hypoxia-responsive azobenzene linkers. To enhance the solubility and tumor accumulation, the prepared BAC was further encapsulated into a human serum albumin (HSA)-based drug delivery system to form HSA@BAC nanoparticles. Since the CPT was caged by a BODIPY-based molecule at the active site, the BAC exhibited excellent biosafety. Importantly, the activated CPT could be quickly released from BAC and could perform chemotherapy in hypoxic cancer cells, which was ascribed to the cleavage of the azobenzene linker by overexpressed azoreductase. After irradiation with a 730 nm laser, HSA@BAC can efficiently generate hyperthermia to achieve irreversible cancer cell death by oxygen-independent photothermal therapy. Under fluorescence imaging-guided local irradiation, both in vitro and in vivo studies demonstrated that HSA@BAC exhibited superior antitumor effects with minimal side effects.
    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
    胰腺导管腺癌(PDAC)由于其独特的缺氧生物学特性而成为药物递送的未满足的临床挑战。长春碱-N-氧化物(CPD100)是一种缺氧激活的前药(HAP),可选择性地转化为其母体化合物,长春碱,一种有效的细胞毒性剂,在氧气梯度下。该研究评估了微流体配制的脂质体CPD100(CPD100Li)在PDAC中的功效。CPD100Li被配制为具有95nm的尺寸和0.2的多分散指数。当浓度为3.55mg/mL时,CPD100Li在18个月内是稳定的。CPD100和CPD100Li证实了在胰腺癌细胞系中低氧水平下的选择性激活。此外,在3D球体中,与CPD100相比,CPD100Li显示出更高的穿透性和破坏。在患者衍生的3D类器官中,与CPD100相比,CPD100Li在表现出更高的缺氧诱导因子1α(HIF1A)表达的类器官中表现出更高的细胞抑制。在原位模型中,CPD100Li联合吉西他滨(GEM)(PDAC的标准治疗)在90天内显示出比单独使用CPD100Li更高的疗效.总之,CPD100Li在多种细胞模型中的评估为其在PDAC中的临床应用提供了坚实的基础。
    Pancreatic ductal adenocarcinoma (PDAC) presents as an unmet clinical challenge for drug delivery due to its unique hypoxic biology. Vinblastine-N-Oxide (CPD100) is a hypoxia-activated prodrug (HAP) that selectively converts to its parent compound, vinblastine, a potent cytotoxic agent, under oxygen gradient. The study evaluates the efficacy of microfluidics formulated liposomal CPD100 (CPD100Li) in PDAC. CPD100Li were formulated with a size of 95 nm and a polydispersity index of 0.2. CPD100Li was stable for a period of 18 months when freeze-dried at a concentration of 3.55 mg/mL. CPD100 and CPD100Li confirmed selective activation at low oxygen levels in pancreatic cancer cell lines. Moreover, in 3D spheroids, CPD100Li displayed higher penetration and disruption compared to CPD100. In patient-derived 3D organoids, CPD100Li exhibited higher cell inhibition in the organoids that displayed higher expression of hypoxia-inducible factor 1 alpha (HIF1A) compared to CPD100. In the orthotopic model, the combination of CPD100Li with gemcitabine (GEM) (standard of care for PDAC) showed higher efficacy than CPD100Li alone for a period of 90 days. In summary, the evaluation of CPD100Li in multiple cellular models provides a strong foundation for its clinical application in PDAC.
    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
    光动力疗法(PDT)可加重缺氧加重,可进一步用于缺氧激活前药(HAP)的激活。理想情况下,光敏剂(PSs)主要作用于邻近高氧区的肿瘤血管,有效产生活性氧(ROS),进一步加重肿瘤缺氧,而HAP尽可能输送到内部肿瘤以有效激活。然而,迫切需要能够将PS和HAP分别运输到期望区域以获得最佳效果的递送系统。这里,我们开发了一种生物正交点击化学和光照控制的可编程尺寸可变纳米药物,用于协同光动力和缺氧激活治疗。它利用肿瘤酸度响应性生物正交点击反应来交联纳米颗粒,以构建具有肿瘤血管邻近区域的药物储库,以在常氧下保留PDT。在激光照明下,裂解ROS响应性硫酮(TK)交联剂以释放与HAP缀合的小尺寸聚(酰胺胺)(PAMAM)树枝状聚合物,以增强肿瘤向低氧区域的渗透。因此,这种策略可以在所需的空间分布中不同地交付PS和HAP,最终在联合PDT和缺氧激活疗法中实现增强的协同增强。
    Photodynamic therapy (PDT) can aggravate the hypoxia aggravation and be further utilized for the activation of hypoxia-activated prodrug (HAP). Ideally, photosensitizers (PSs) are mainly administrated to tumor vasculatures adjacent to regions with high oxygen to effectively generate reactive oxygen species (ROS) effectively and further aggravate tumor hypoxia, while the HAP is delivered to the inner tumor as far as possible for efficient activation. However, a delivery system capable of transporting PSs and HAP to the desired region respectively for the optimum effect is urgently needed. Here, we developed a bioorthogonal click chemistry and illumination controlled programmed size-changeable nanomedicine for synergistic photodynamic and hypoxia-activated therapy. It utilized tumor acidity responsive bioorthogonal click reaction for crosslinking nanoparticles to construct a drug depot with tumor vasculatures adjacent region retention for PDT in normoxia. Under laser illumination, cleavage of the ROS-responsive thioketal (TK) crosslinker to release small sized poly(amidoamine) (PAMAM) dendrimer conjugated with HAP for enhanced tumor penetration into the hypoxic region. Therefore, this strategy could differentially deliver PSs and HAP in desired spatial distribution, eventually achieving the enhanced synergistic enhancement in the combined PDT and hypoxia-activated therapy.
    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
    缺氧激活的前药在缺氧的肿瘤区域被生物激活,代表了一种利用这种药理保护区获得治疗收益的新策略。该方法依赖于前药在病理性缺氧下的选择性代谢,以产生具有扩散到整个肿瘤微环境并通过“旁观者效应”增强细胞杀伤能力的活性代谢物。在本研究中,我们使用计算机空间分辨的药代动力学/药效学(SR-PK/PD)模型研究了氮芥前药CP-506在肿瘤组织中的药理特性。该方法采用了许多实验模型系统来定义细胞摄取的参数,前药及其代谢物的代谢和扩散。该模型预测CP-506的快速摄取到高细胞内浓度,其长血浆半衰期驱动组织扩散到190µm的穿透深度,在低氧激活区域的深处。虽然生物还原性代谢仅限于严重病理性缺氧(<1µMO2)的区域,它的活性代谢物显示出相当大的旁观者潜力,从起源细胞释放到细胞外空间。使用球体共培养验证了旁观者效率的模型预测,其中代谢缺陷的“目标”细胞的克隆杀伤作用随着代谢能力的“激活剂”细胞的比例而增加。我们的模拟预测,在组织样密度下,双氯芥子气胺代谢物(CP-506M-Cl2)被确定为主要的扩散代谢物,具有惊人的旁观者效率。总的来说,这项研究表明,CP-506在肿瘤组织中具有良好的药理学特性,并支持其正在开发用于治疗晚期实体恶性肿瘤患者的方法.
    Hypoxia-activated prodrugs are bioactivated in oxygen-deficient tumour regions and represent a novel strategy to exploit this pharmacological sanctuary for therapeutic gain. The approach relies on the selective metabolism of the prodrug under pathological hypoxia to generate active metabolites with the potential to diffuse throughout the tumour microenvironment and potentiate cell killing by means of a \"bystander effect\". In the present study, we investigate the pharmacological properties of the nitrogen mustard prodrug CP-506 in tumour tissues using in silico spatially-resolved pharmacokinetic/pharmacodynamic (SR-PK/PD) modelling. The approach employs a number of experimental model systems to define parameters for the cellular uptake, metabolism and diffusion of both the prodrug and its metabolites. The model predicts rapid uptake of CP-506 to high intracellular concentrations with its long plasma half-life driving tissue diffusion to a penetration depth of 190 µm, deep within hypoxic activating regions. While bioreductive metabolism is restricted to regions of severe pathological hypoxia (<1 µM O2), its active metabolites show substantial bystander potential with release from the cell of origin into the extracellular space. Model predictions of bystander efficiency were validated using spheroid co-cultures, where the clonogenic killing of metabolically defective \"target\" cells increased with the proportion of metabolically competent \"activator\" cells. Our simulations predict a striking bystander efficiency at tissue-like densities with the bis-chloro-mustard amine metabolite (CP-506M-Cl2) identified as a major diffusible metabolite. Overall, this study shows that CP-506 has favourable pharmacological properties in tumour tissue and supports its ongoing development for use in the treatment of patients with advanced solid malignancies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号