targeted alpha therapy

靶向 α 治疗
  • 文章类型: Journal Article
    Astatine-211(211At)由于其有利的核性质,已成为有前途的放射性核素用于癌症的靶向α治疗。然而,211At标记的放射性药物的有限的体内稳定性仍然是一个主要挑战。这篇综述全面概述了211At放射性标记的当前策略,包括亲核和亲电取代反应,以及新型双功能偶联剂和标记方法的最新进展,以增强211At标记化合物的稳定性。211At标记放射性药物的临床前和临床应用,包括小分子,肽,和抗体,也讨论了。展望未来,识别新的分子靶标,211At生产和质量控制方法的优化,在临床前和临床环境中对211At标记的放射性药物的持续评估将是实现基于211At的靶向α治疗的全部潜力的关键。随着人们对这一领域的兴趣和投资的增长,211At标记的放射性药物有望在未来的癌症治疗中发挥越来越重要的作用。
    Astatine-211 (211At) has emerged as a promising radionuclide for targeted alpha therapy of cancer by virtue of its favorable nuclear properties. However, the limited in vivo stability of 211At-labeled radiopharmaceuticals remains a major challenge. This review provides a comprehensive overview of the current strategies for 211At radiolabeling, including nucleophilic and electrophilic substitution reactions, as well as the recent advances in the development of novel bifunctional coupling agents and labeling approaches to enhance the stability of 211At-labeled compounds. The preclinical and clinical applications of 211At-labeled radiopharmaceuticals, including small molecules, peptides, and antibodies, are also discussed. Looking forward, the identification of new molecular targets, the optimization of 211At production and quality control methods, and the continued evaluation of 211At-labeled radiopharmaceuticals in preclinical and clinical settings will be the key to realizing the full potential of 211At-based targeted alpha therapy. With the growing interest and investment in this field, 211At-labeled radiopharmaceuticals are poised to play an increasingly important role in future 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
    癌症的持续威胁需要开发改进和更有效的治疗策略,以限制对健康组织的损害。靶向α治疗(TαT),一种新形式的放射免疫疗法(RIT),利用瞄准车辆,通常是抗体,为了提供高能量,但是短距离,专门针对癌细胞的α发射粒子,从而降低对周围正常组织的毒性。尽管全长抗体通常用作TαT的靶向载体,它们的高分子量和Fc区的存在导致长的血液半衰期,增加骨髓毒性,在肾脏等其他组织中积累,肝脏,还有脾脏.单结构域抗体(sdAb)的发现,或者纳米抗体,自然发生在骆驼和鲨鱼中,已经引入了一种用于分子成像和TαT的新型抗原特异性载体。鉴于纳米抗体是最小的天然抗原结合片段,它们的相对血液半衰期较短,增强肿瘤摄取,和等效或优越的结合亲和力和特异性。纳米抗体技术可以为基于全长抗体的TαT观察到的脱靶毒性提供可行的解决方案。值得注意的是,纳米抗体的药代动力学特性与许多短寿命α发射放射性核素的衰变特性更好。这篇综述旨在囊括纳米体作为TαT载体的最新进展。
    The persistent threat of cancer necessitates the development of improved and more efficient therapeutic strategies that limit damage to healthy tissues. Targeted alpha therapy (TαT), a novel form of radioimmuno-therapy (RIT), utilizes a targeting vehicle, commonly antibodies, to deliver high-energy, but short-range, alpha-emitting particles specifically to cancer cells, thereby reducing toxicity to surrounding normal tissues. Although full-length antibodies are often employed as targeting vehicles for TαT, their high molecular weight and the presence of an Fc-region lead to a long blood half-life, increased bone marrow toxicity, and accumulation in other tissues such as the kidney, liver, and spleen. The discovery of single-domain antibodies (sdAbs), or nanobodies, naturally occurring in camelids and sharks, has introduced a novel antigen-specific vehicle for molecular imaging and TαT. Given that nanobodies are the smallest naturally occurring antigen-binding fragments, they exhibit shorter relative blood half-lives, enhanced tumor uptake, and equivalent or superior binding affinity and specificity. Nanobody technology could provide a viable solution for the off-target toxicity observed with full-length antibody-based TαT. Notably, the pharmacokinetic properties of nanobodies align better with the decay characteristics of many short-lived α-emitting radionuclides. This review aims to encapsulate recent advancements in the use of nanobodies as a vehicle for TαT.
    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
    我们最近发现CD46是一种新的前列腺癌细胞表面抗原,在转移性去势抵抗性前列腺癌(mCRPC)的腺癌和小细胞神经内分泌亚型中均显示出谱系独立表达。发现了一种内在化的人单克隆抗体YS5,它与肿瘤选择性CD46表位结合,并开发了一种基于微管抑制剂的抗体药物缀合物,该药物正在mCRPC的多中心I期试验中(NCT03575819)。在这里,我们报道了基于YS5的新型CD46靶向α治疗的发展。我们共轭212Pb,体内产生α发射的212Bi和212Po,YS5通过螯合剂TCMC产生放射免疫缀合物,212Pb-TCMC-YS5。我们在体外对212Pb-TCMC-YS5进行了表征,并在体内建立了安全的剂量。接下来,我们使用三种前列腺癌小动物模型研究了单剂量212Pb-TCMC-YS5的治疗效果:皮下mCRPC细胞系衍生的异种移植(CDX)模型(subcu-CDX),原位移植mCRPCCDX模型(ortho-CDX),和前列腺癌患者来源的异种移植模型(PDX)。在所有三个模型中,单剂量0.74MBq(20µCi)212Pb-TCMC-YS5耐受性良好,对已建立的肿瘤产生有效和持续的抑制作用,在治疗的动物中存活率显著增加。在PDX模型上还研究了较低剂量(0.37MBq或10µCi212Pb-TCMC-YS5),对肿瘤生长抑制和延长动物生存期也有显著作用。这些结果表明,212Pb-TCMC-YS5在包括PDX在内的临床前模型中具有出色的治疗窗口,为这种新型CD46靶向alpha放射免疫疗法用于mCRPC治疗的临床转化开辟了直接途径。
    We recently identified CD46 as a novel prostate cancer cell surface antigen that shows lineage independent expression in both adenocarcinoma and small cell neuroendocrine subtypes of metastatic castration resistant prostate cancer (mCRPC), discovered an internalizing human monoclonal antibody YS5 that binds to a tumor selective CD46 epitope, and developed a microtubule inhibitor-based antibody drug conjugate that is in a multi-center phase I trial for mCRPC (NCT03575819). Here we report the development of a novel CD46-targeted alpha therapy based on YS5. We conjugated 212Pb, an in vivo generator of alpha-emitting 212Bi and 212Po, to YS5 through the chelator TCMC to create the radioimmunoconjugate, 212Pb-TCMC-YS5. We characterized 212Pb-TCMC-YS5 in vitro and established a safe dose in vivo. We next studied therapeutic efficacy of a single dose of 212Pb-TCMC-YS5 using three prostate cancer small animal models: a subcutaneous mCRPC cell line-derived xenograft (CDX) model (subcu-CDX), an orthotopically grafted mCRPC CDX model (ortho-CDX), and a prostate cancer patient-derived xenograft model (PDX). In all three models, a single dose of 0.74 MBq (20 µCi) 212Pb-TCMC-YS5 was well tolerated and caused potent and sustained inhibition of established tumors, with significant increases of survival in treated animals. A lower dose (0.37 MBq or 10 µCi 212Pb-TCMC-YS5) was also studied on the PDX model, which also showed a significant effect on tumor growth inhibition and prolongation of animal survival. These results demonstrate that 212Pb-TCMC-YS5 has an excellent therapeutic window in preclinical models including PDXs, opening a direct path for clinical translation of this novel CD46-targeted alpha radioimmunotherapy for mCRPC 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
    背景:Astatine-211是一种α发射器,具有高能α射线和对癌细胞的高细胞毒性。然而,靶向α治疗(TAT)也存在系统性免疫激活不足的问题,导致肿瘤转移和复发。联合免疫检查点阻断(ICB)与化学动力学疗法(CDT)可以增强抗肿瘤免疫力,这可能会放大TAT的免疫反应。本研究旨在通过三模型TAT-CDT-ICB策略阻止肿瘤转移和复发。
    方法:我们成功设计了基于Mn的放射免疫疗法启动子(211At-ATE-MnO2-BSA),由211At组成,MnO2和牛血清白蛋白(BSA)。211At-ATE-MnO2-BSA在转移和复发模型中作为单一疗法或与抗PD-L1组合进行了研究。通过流式细胞术分析放射免疫疗法启动子对细胞毒性T淋巴细胞和树突状细胞(DC)的免疫作用。采用酶联免疫吸附法和免疫荧光法探讨其作用机制。
    结果:这种放射免疫疗法启动子不仅可以增强TAT和CDT的治疗效果,而且还通过激活树突状细胞诱导强大的抗癌免疫活性。更有趣的是,当与免疫检查点抑制剂组合时,211At-ATE-MnO2-BSA可以有效地抑制原发性肿瘤和远端肿瘤的生长。
    结论:三模型TAT-CDT-ICB策略提供了长期免疫记忆,消除原始肿瘤后可以防止肿瘤再攻击。因此,这项工作为TAT-CDT-ICB三模态癌症治疗提供了一种新的方法,在临床上抑制了转移和复发。
    BACKGROUND: Astatine-211 is an α-emitter with high-energy α-ray and high cytotoxicity for cancer cells. However, the targeted alpha therapy (TAT) also suffers from insufficient systematic immune activation, resulting in tumor metastasis and relapse. Combined immune checkpoint blockade (ICB) with chemodynamic therapy (CDT) could boost antitumor immunity, which may magnify the immune responses of TAT. This study aims to discourage tumor metastasis and relapse by tri-model TAT-CDT-ICB strategy.
    METHODS: We successfully designed Mn-based radioimmunotherapy promoters (211At-ATE-MnO2-BSA), which are consisting of 211At, MnO2 and bovine serum albumin (BSA). The efficacy of 211At-ATE-MnO2-BSA was studied as monotherapy or in combination with anti-PD-L1 in both metastatic and relapse models. The immune effects of radioimmunotherapy promoters on cytotoxic T lymphocytes and dendritic cells (DCs) were analyzed by flow cytometry. Enzyme-linked immunosorbent assay and immunofluorescence were used to explore the underlying mechanism.
    RESULTS: Such radioimmunotherapy promoters could not only enhance the therapeutic outcomes of TAT and CDT, but also induce robust anti-cancer immune activity by activating dendritic cells. More intriguingly, 211At-ATE-MnO2-BSA could effectively suppress the growths of primary tumors and distant tumors when combined with immune checkpoint inhibitors.
    CONCLUSIONS: The tri-model TAT-CDT-ICB strategy provides a long-term immunological memory, which can protect against tumor rechallenge after eliminating original tumors. Therefore, this work presents a novel approach for TAT-CDT-ICB tri-modal cancer therapy with repressed metastasis and relapse in clinics.
    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
    Background: BAY 1862864 is an α-particle emitting 227Th-labeled CD22-targeting antibody. This first-in-human dose-escalation phase I study evaluated BAY 1862864 in patients with CD22-positive relapsed/refractory B cell non-Hodgkin lymphoma (R/R-NHL). Materials and Methods: BAY 1862864 intravenous injections were administered at the starting 227Th radioactivity dose of 1.5 MBq (2 or 10 mg antibody), and the radioactivity dose escalated in ∼1.5 MBq increments (10 mg antibody) until the maximum tolerated dose (MTD) was reported. The primary objective was to determine the safety, tolerability, and MTD. Results: Twenty-one patients received BAY 1862864. Two dose-limiting toxicities (grade 3 febrile neutropenia and grade 4 thrombocytopenia) were reported in one patient in the 4.6 MBq (10 mg antibody) cohort. The MTD was not reached. Ten (48%) patients reported grade ≥3 treatment-emergent adverse events, with the most common being neutropenia, thrombocytopenia, and leukopenia, each occurring in 3 (14%) patients. Pharmacokinetics demonstrated the dose proportionality and stability of BAY 1862864 in the blood. The objective response rate (ORR) was 25% (5/21 patients) according to the LUGANO 2014 criteria, including 1 complete and 4 partial responses. The ORR was 11% (1/9) and 30% (3/10) in patients with relapsed high- and low-grade lymphomas, respectively. Conclusions: BAY 1862864 was safe and tolerated in patients with R/R-NHL. Clinical Trial Registration numbers: NCT02581878 and EudraCT 2014-004140-36.
    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: Clinical Trial, Phase III
    OBJECTIVE: Radium-223, a targeted alpha therapy, is approved widely for the treatment of patients with metastatic castrate-resistant prostate cancer, based on a pivotal phase 3 study in predominantly white patients. We investigated the efficacy and safety of radium-223 in Asian patients with castrate-resistant prostate cancer and metastatic bone disease.
    METHODS: This multicenter, prospective, single-arm, open-label phase 3 trial evaluated the efficacy and safety of the standard radium-223 regimen (55 kBq/kg every 4 weeks for six cycles) in patients from Asian countries. The primary endpoints were the safety and overall survival.
    RESULTS: A total of 226 patients were enrolled and received at least one dose of radium-223. Median overall survival was 14.0 months (95% confidence interval [CI], 11.2-17.4). Median time to total alkaline phosphatase and prostate-specific antigen progression were 7.5 (95% CI, 6.8-7.7) and 3.6 (95% CI, 3.1-3.7) months, respectively. Median skeletal-related event-free survival was 26.0 months (95% CI, 12.6-not reached). Grade ≥3 treatment-emergent adverse events were reported in 103 (46%) of 226 patients, with anemia being the most common event (34 [15%] patients). Grade ≥3 drug-related treatment-emergent adverse events occurred in 39 (17%) of 226 patients. Serious treatment-emergent adverse events were reported in 65 (29%) of 226 patients. Seven (3%) patients had an adverse event leading to death; none were considered to be related to radium-223.
    CONCLUSIONS: The results of this study support the use of the standard radium-223 regimen for the treatment of Asian patients with castrate-resistant prostate cancer and symptomatic bone metastases.
    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
    Targeted alpha therapy (TAT) is a promising tumor therapy that can specifically transport α particle to the vicinity of tumor cells while the normal cells are only slightly irradiated. Mesothelin is a highly promising molecular signature for many types of solid tumors including malignant mesothelioma, pancreatic cancer, ovarian cancer and lung adenocarcinoma etc., while the expression in normal human tissues are limited, thus making mesothelin a promising antigen for TAT. Previously we developed a theoretical model that could predict and optimize in vitro screening of potential TAT drugs. The aim of the study is construction and preclinical evaluation of 211At labeled anti-mesothelin antibodies as potential TAT drugs. Mesothelin expression of two tumor cell lines were confirmed by flow cytometry, and their radiosensitivities were also evaluated. We used two kinds of anti-mesothelin antibodies, ET210-6 and ET210-28, to construct TAT drugs. Then, radiochemical purity, stability in vitro, affinity of the conjugates and mesothelin expression level were assessed. The specific killing of mesothelin-positive cancer cells treated by 211At-ET210-28 and 211At-ET210-6 were studied via Cell Counting Kit-8 assay and colony formation assay. 211At-ET210-28 and 211At-ET210-6 revealed excellent affinity and stability in both phosphate buffer saline and fetal bovine serum environment. Radiolabeled antibody conjugates bound specifically to mesothelin-positive cells in vitro. Both 211At-ET210-28 and 211At-ET210-6 could specifically kill mesothelin-positive cells with negligible damages to mesothelin-negative cells. Our findings provide initial proof-of-concept for the potential use of 211At labeled ET210-28/ET210-6 anti-mesothelin antibody in specific killings of mesothelin-positive tumor cells.
    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
    靶向α疗法(TAT)通过向肿瘤提供聚焦和致命的辐射剂量,具有治疗广泛的晚期癌症的巨大潜力。Actinium-225(225Ac)是适用于TAT的新兴α发射体;但是,用于Ac的螯合剂的可用性仍然限于少数实例(DOTA和macropa)。在这里,我们报道了一种名为“冠”的新型Ac大环螯合剂,在环境温度下与Ac定量和快速结合(<10分钟)。我们合成了225个Ac-冠-αMSH,靶向黑皮质素1受体(MC1R)的肽,在原发性和转移性黑色素瘤中特异性表达。在临床前模型中,注射后2小时,225Ac-crown-αMSH的生物分布显示出有利的肿瘤背景比。此外,当注射前经历不同的潜伏期时,我们证明了225Ac-crown-αMSH的生物结构模式截然不同。涉及HPLC的联合质量控制方法,建议使用γ光谱和放射性TLC。
    Targeted alpha-therapy (TAT) has great potential for treating a broad range of late-stage cancers by delivering a focused and lethal radiation dose to tumors. Actinium-225 (225 Ac) is an emerging alpha emitter suitable for TAT; however, the availability of chelators for Ac remains limited to a small number of examples (DOTA and macropa). Herein, we report a new Ac macrocyclic chelator named \'crown\', which binds quantitatively and rapidly (<10 min) to Ac at ambient temperature. We synthesized 225 Ac-crown-αMSH, a peptide targeting the melanocortin 1 receptor (MC1R), specifically expressed in primary and metastatic melanoma. Biodistribution of 225 Ac-crown-αMSH showed favorable tumor-to-background ratios at 2 h post injection in a preclinical model. In addition, we demonstrated dramatically different biodistrubution patterns of 225 Ac-crown-αMSH when subjected to different latency times before injection. A combined quality control methodology involving HPLC, gamma spectroscopy and radioTLC is recommended.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号