Hypoxia-activated prodrug

  • 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
    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)

  • 文章类型: Journal Article
    尽管在缺氧相关的抗肿瘤治疗方面取得了相当大的进展,由于肿瘤区域内的缺氧不足,单独使用缺氧激活的前药的功效无法满足。在这项工作中,整合光动力疗法的线粒体靶向纳米平台,光热疗法和缺氧激活的化学疗法已被开发用于协同治疗癌症并最大化治疗窗口。以聚多巴胺包覆的中空硫化铜纳米颗粒作为光热纳米剂和热敏药物载体,负载缺氧激活的前药,TH302,在我们的研究中。将氯元素e6(Ce6)和三苯基鳞(TPP)缀合到纳米平台的表面上。在TPP的作用下,获得的纳米平台优先积累在线粒体中,以恢复药物活性并避免耐药性。使用660nm激光激发Ce6可产生ROS,同时加剧细胞缺氧。在808nm激光照射下,纳米平台产生局部热量,可以增加肿瘤细胞中TH302的释放,消融癌细胞以及增强肿瘤缺氧水平。然后,加重的肿瘤缺氧显着提高了TH302的抗肿瘤效率。体外和体内研究均表明,与常规缺氧相关化疗相比,抗癌活性得到了极大改善。这项工作强调了使用缺氧激活的前药和光疗组合用于协同癌症治疗的潜力。
    Despite considerable progress has been achieved in hypoxia-associated anti-tumor therapy, the efficacy of utilizing hypoxia-activated prodrugs alone is not satisfied owing to the inadequate hypoxia within the tumor regions. In this work, a mitochondrial targeted nanoplatform integrating photodynamic therapy, photothermal therapy and hypoxia-activated chemotherapy has been developed to synergistically treat cancer and maximize the therapeutic window. Polydopamine coated hollow copper sulfide nanoparticles were used as the photothermal nanoagents and thermosensitive drug carriers for loading the hypoxia-activated prodrug, TH302, in our study. Chlorin e6 (Ce6) and triphenyl phosphonium (TPP) were conjugated onto the surface of the nanoplatform. Under the action of TPP, the obtained nanoplatform preferentially accumulated in mitochondria to restore the drug activity and avoid drug resistance. Using 660 nm laser to excite Ce6 can generate ROS and simultaneously exacerbate the cellular hypoxia. While under the irradiation of 808 nm laser, the nanoplatform produced local heat which can increase the release of TH302 in tumor cells, ablate cancer cells as well as intensify the tumor hypoxia levels. The aggravated tumor hypoxia then significantly boosted the anti-tumor efficiency of TH302. Both in vitro and in vivo studies demonstrated the greatly improved anti-cancer activity compared to conventional hypoxia-associated chemotherapy. This work highlights the potential of using a combination of hypoxia-activated prodrugs plus phototherapy for synergistic 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
    缺氧可以说是第一个公认的癌症微环境标志,并影响肿瘤中存在的几乎所有细胞群体。在过去的几十年中,复杂的适应性细胞对氧剥夺的反应已被大量阐明,为新的抗癌药物带来了希望。尽管取得了不可否认的临床前进展,肿瘤缺氧的治疗靶向尚未从工作台过渡到床边。这篇综述集中于利用肿瘤缺氧或干扰缺氧信号的新药物,并讨论了最大化其治疗效果的策略。
    Hypoxia is arguably the first recognized cancer microenvironment hallmark and affects virtually all cellular populations present in tumors. During the past decades the complex adaptive cellular responses to oxygen deprivation have been largely elucidated, raising hope for new anti cancer agents. Despite undeniable preclinical progress, therapeutic targeting of tumor hypoxia is yet to transition from bench to bedside. This review focuses on new pharmacological agents that exploit tumor hypoxia or interfere with hypoxia signaling and discusses strategies to maximize their therapeutic impact.
    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
    Photodynamic therapy (PDT), where a photosensitizer (under light irradiation) converts molecular oxygen to singlet oxygen to elicit programmed cell death, is a promising cancer treatment modality with a high temporal and spatial resolution. However, only limited cancer treatment efficacy has been achieved in clinical PDT due to the hypoxic conditions of solid tumor microenvironment that limits the generation of singlet oxygen, and PDT process often leads to even more hypoxic microenvironment due to the consumption of oxygens during therapy. Herein, we designed novel supramolecular micelles to co-deliver photosensitizer and hypoxia-responsive prodrug to improve the overall therapeutic efficacy. The supramolecular micelles (CPC) were derived from a polyethylene glycol (PEG) system dually tagged with hydrophilic cucurbit[7]uril (CB[7]) and hydrophobic Chlorin e6 (Ce6), respectively on each end, for synergistic antitumor therapy via PDT of Ce6 and chemotherapy of a hypoxia-responsive prodrug, banoxantrone (AQ4N), loaded into the cavity of CB[7]. In addition, CPC was further modularly functionalized by folate (FA) via strong host-guest interaction between folate-amantadine (FA-ADA) and CB[7] to produce a novel nanoplatform, AQ4N@CPC-FA, for targeted delivery. AQ4N@CPC-FA exhibited enhanced cellular uptake, negligible cytotoxicity and good biocompatibility, and improved intracellular reactive oxygen species (ROS) generation efficiency. More importantly, in vivo evaluation of AQ4N@CPC-FA revealed a synergistic antitumor efficacy between PDT of Ce6 and hypoxia-activated chemotherapy of AQ4N (that can be converted to chemotherapeutic AQ4 for tumor chemotherapy in response to the strengthened hypoxic tumor microenvironment during PDT treatment). This study not only provides a new nanoplatform for synergistic photodynamic-chemotherapeutic treatment, but also offers important new insights to design and development of multifunctional supramolecular drug delivery system. STATEMENT OF SIGNIFICANCE: Photodynamic therapy (PDT) has exhibited a variety of advantages for cancer phototherapy as compared to traditional chemotherapy. However, the unsatisfactory therapeutic efficacy by PDT alone as a result of the enhanced tumor hypoxia during PDT has limited its clinical application. Herein, we designed multifunctional supramolecular micelles to co-deliver photosensitizer and hypoxia-responsive prodrug to improve the overall therapeutic efficacy. The supramolecular micelles are biocompatible and possess strong red absorption, controlled drug release profile, and ultimately enhanced therapeutic outcome via PDT-chemotherapy. This study not only provides a new nanoplatform for synergistic photodynamic-chemotherapeutic treatment of cancer, but also offers important new insights to design and development of multifunctional supramolecular drug delivery tool for multi-modality cancer therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    UNASSIGNED: The hypoxic tumor microenvironment represents a persistent obstacle in the treatment of most solid tumors. In the past years, significant efforts have been made to improve the efficacy of anti-cancer drugs. Therefore, hypoxia-activated prodrugs (HAPs) of chemotherapeutic compounds have attracted widespread interest as a therapeutic means to treat hypoxic tumors.
    UNASSIGNED: This updated review paper covers key patents published between 2006 and 2021 on the developments of HAP derivatives of anti-cancer compounds.
    UNASSIGNED: Despite significant achievements in the development of HAP derivatives of anti-cancer compounds and although many clinical trials have been performed or are ongoing both as monotherapies and as part of combination therapies, there has currently no HAP anti-cancer agent been commercialized into the market. Unsuccessful clinical translation is partly due to the lack of patient stratification based on reliable biomarkers that are predictive of a positive response to hypoxia-targeted therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    宿主免疫应答的幅度可以通过刺激性或抑制性免疫检查点分子来调节。抑制性分子之间的受体-配体结合经常被肿瘤利用以抑制抗肿瘤免疫应答。阻断这些抑制性相互作用的免疫检查点抑制剂可以减轻T细胞的负调节,并在临床上产生了显著的活动。尽管取得了这样的成功,临床数据显示,持久的反应仅限于少数患者和恶性肿瘤,表明存在潜在的抗性机制。越来越多的证据表明肿瘤缺氧,许多实体癌的普遍特征,是抑制检查点抑制剂产生的抗肿瘤免疫应答的关键现象。在这次审查中,我们讨论了与缺氧介导的免疫抑制相关的机制,并将重点放在调节肿瘤缺氧作为改善免疫治疗反应性的方法上。
    The magnitude of the host immune response can be regulated by either stimulatory or inhibitory immune checkpoint molecules. Receptor-ligand binding between inhibitory molecules is often exploited by tumours to suppress anti-tumour immune responses. Immune checkpoint inhibitors that block these inhibitory interactions can relieve T-cells from negative regulation, and have yielded remarkable activity in the clinic. Despite this success, clinical data reveal that durable responses are limited to a minority of patients and malignancies, indicating the presence of underlying resistance mechanisms. Accumulating evidence suggests that tumour hypoxia, a pervasive feature of many solid cancers, is a critical phenomenon involved in suppressing the anti-tumour immune response generated by checkpoint inhibitors. In this review, we discuss the mechanisms associated with hypoxia-mediate immunosuppression and focus on modulating tumour hypoxia as an approach to improve immunotherapy responsiveness.
    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
    Hypoxia is an adverse prognostic feature of solid cancers that may be overcome with hypoxia-activated prodrugs (HAPs). Tirapazamine (TPZ) is a HAP which has undergone extensive clinical evaluation in this context and stimulated development of optimized analogues. However the subcellular localization of the oxidoreductases responsible for mediating TPZ-dependent DNA damage remains unclear. Some studies conclude only nuclear-localized oxidoreductases can give rise to radical-mediated DNA damage and thus cytotoxicity, whereas others identify a broader role for endoplasmic reticulum and cytosolic oxidoreductases, indicating the subcellular location of TPZ radical formation is not a critical requirement for DNA damage. To explore this question in intact cells we engineered MDA-231 breast cancer cells to express the TPZ reductase human NADPH: cytochrome P450 oxidoreductase (POR) harboring various subcellular localization sequences to guide this flavoenzyme to the nucleus, endoplasmic reticulum, cytosol or inner surface of the plasma membrane. We show that all POR variants are functional, with differences in rates of metabolism reflecting enzyme expression levels rather than intracellular TPZ concentration gradients. Under anoxic conditions, POR expression in all subcellular compartments increased the sensitivity of the cells to TPZ, but with a fall in cytotoxicity per unit of metabolism (termed \'metabolic efficiency\') when POR is expressed further from the nucleus. However, under aerobic conditions a much larger increase in cytotoxicity was observed when POR was directed to the nucleus, indicating very high metabolic efficiency. Consequently, nuclear metabolism results in collapse of hypoxic selectivity of TPZ, which was further magnified to the point of reversing O2 dependence (oxic > hypoxic sensitivity) by employing a DNA-affinic TPZ analogue. This aerobic hypersensitivity phenotype was partially rescued by cellular copper depletion, suggesting the possible involvement of Fenton-like chemistry in generating short-range effects mediated by the hydroxyl radical. In addition, the data suggest that under aerobic conditions reoxidation strictly limits the TPZ radical diffusion range resulting in site-specific cytotoxicity. Collectively these novel findings challenge the purported role of intra-nuclear reductases in orchestrating the hypoxia selectivity of TPZ.
    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
    Immunogenic photodynamic therapy (PDT) has the potential to moderate the shortfalls of cancer immunotherapy. However, its efficacy is severely limited particularly because of the lack of optimal photosensitizers and smart delivery processes and the inherent shortcomings of PDT (e.g., hypoxia resistance). Here, we demonstrate a clinically promising approach that utilizes a water-soluble phthalocyanine derivative (PcN4) concomitantly delivered with a hypoxia-activated prodrug (AQ4N) to amplify the effect of PDT and enhance cancer immunotherapy. After intravenous injection, PcN4 selectively interacted with endogenous albumin dimers and formed supramolecular complexes, providing a facile and green approach for tumor-targeted PDT. The concomitant delivery of AQ4N overcame the limitations of hypoxia in PDT and improved the antitumor activity of PDT. Treatment with PcN4-mediated and AQ4N-amplified PDT almost completely eradicated sizable primary tumors in a triple-negative breast cancer model and significantly activated CD8+ T cells. As the majority of tumor infiltrating CD8+ T cells were both PD-1- and TIM3-positive, additional combination therapy using PD-L1/PD-1 pathway blockade was warranted. After combination with immune checkpoint blockade treatment, an enhanced abscopal effect was achieved in both distant and metastatic tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号