targeted radionuclide therapy

靶向放射性核素治疗
  • 文章类型: Journal Article
    头颈部鳞状细胞癌(HNSCC)是一种侵袭性头颈部癌,难以治疗,常伴有不良预后。HNSCC是全球第六大最常见的癌症,强调需要开发新的治疗这种疾病。HNSCC的当前护理标准通常涉及手术切除的组合,放射治疗,和化疗。化疗因其有害的副作用而臭名昭著,包括恶心,疲劳,脱发,还有更多.由于头部和颈部区域的解剖结构以及正常组织的存在,放射治疗可能是一个挑战。除了化疗和放疗的缺点,HNSCC的高发病率和高死亡率凸显了对替代治疗方案的迫切需求.免疫疗法最近已成为包括HNSCC在内的癌症的可能治疗选择。其中单克隆抗体用于帮助免疫系统对抗疾病。结合美国食品和药物管理局批准的单克隆抗体,如西妥昔单抗和派姆单抗,对局部晚期患者进行放疗或铂类化疗,经常性,或转移性HNSCC是公认的一线治疗。靶向放射性核素治疗可能与一线治疗结合使用。或者作为额外的治疗选择,改善患者预后和生活质量。表皮生长因子受体是HNSCC的已知分子靶标;然而,其他靶标,例如人表皮生长因子受体2,人表皮生长因子受体3,程序性细胞死亡蛋白1和程序性死亡配体1,是诊断和治疗HNSCC的新兴分子靶标。为了开发成功的放射性药物,首先必须了解感兴趣的疾病的分子生物学。对于癌症,这种理解通常意味着分子靶标的检测和表征,如细胞表面受体,可以用作敏感的靶向剂。本文的目的是探索HNSCC的分子靶标,并剖析先前在核医学中进行的研究,并为开发用于HNSCC靶向放射性核素治疗的新型放射性药物提供可能的途径。到目前为止还没有得到充分的探索。
    Head and neck squamous cell carcinoma (HNSCC) is a type of head and neck cancer that is aggressive, difficult to treat, and often associated with poor prognosis. HNSCC is the sixth most common cancer worldwide, highlighting the need to develop novel treatments for this disease. The current standard of care for HNSCC usually involves a combination of surgical resection, radiation therapy, and chemotherapy. Chemotherapy is notorious for its detrimental side effects including nausea, fatigue, hair loss, and more. Radiation therapy can be a challenge due to the anatomy of the head and neck area and presence of normal tissues. In addition to the drawbacks of chemotherapy and radiation therapy, high morbidity and mortality rates for HNSCC highlight the urgent need for alternative treatment options. Immunotherapy has recently emerged as a possible treatment option for cancers including HNSCC, in which monoclonal antibodies are used to help the immune system fight disease. Combining monoclonal antibodies approved by the US Food and Drug Administration, such as cetuximab and pembrolizumab, with radiotherapy or platinum-based chemotherapy for patients with locally advanced, recurrent, or metastatic HNSCC is an accepted first-line therapy. Targeted radionuclide therapy can potentially be used in conjunction with the first-line therapy, or as an additional treatment option, to improve patient outcomes and quality of life. Epidermal growth factor receptor is a known molecular target for HNSCC; however, other targets such as human epidermal growth factor receptor 2, human epidermal growth factor receptor 3, programmed cell death protein 1, and programmed death-ligand 1 are emerging molecular targets for the diagnosis and treatment of HNSCC. To develop successful radiopharmaceuticals, it is imperative to first understand the molecular biology of the disease of interest. For cancer, this understanding often means detection and characterization of molecular targets, such as cell surface receptors, that can be used as sensitive targeting agents. The goal of this review article is to explore molecular targets for HNSCC and dissect previously conducted research in nuclear medicine and provide a possible path forward for the development of novel radiopharmaceuticals used in targeted radionuclide therapy for HNSCC, which has been underexplored to date.
    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
    目的:靶向放射性核素治疗(TRT)是一种在各种癌症类型中具有相对疗效的癌症治疗方法。我们在表达人FAP(hFAP)的肺癌小鼠模型中的免疫活性小鼠中,使用靶向用225Ac或131I标记的sdAb(4AH29)的成纤维细胞激活蛋白-α(FAP)研究了TRT的治疗潜力。我们进一步探索了TRT与程序性细胞死亡配体1(PD-L1)免疫检查点阻断(ICB)的组合。
    方法:我们通过离体γ计数研究了[131I]I-GMIB-4AH29和[225Ac]Ac-DOTA-4AH29的生物分布和肿瘤摄取。在免疫活性小鼠模型中评价[131I]I-GMIB-4AH29和[225Ac]Ac-DOTA-4AH29的治疗功效。进行来自[225Ac]Ac-DOTA-4AH29处理的小鼠的肿瘤的流式细胞术分析。与PD-L1ICB组合重复用[225Ac]Ac-DOTA-4AH29治疗。
    结果:生物分布显示[131I]I-GMIB-4AH29的高肿瘤摄取,注射后1h(p.i.)降低至0.9±0.1%IA/g24h后。[225Ac]Ac-DOTA-4AH29的肿瘤摄取与1hA25%Ac相比,高剂量A29和4GIA/g/g治疗后4此外,与溶媒溶液相比,我们观察到用[225Ac]Ac-DOTA-4AH29治疗的小鼠肿瘤中PD-L1表达显著更高.因此,我们将高剂量[225Ac]Ac-DOTA-4AH29与PD-L1ICB联合使用,显示出治疗协同作用。
    结论:[225Ac]Ac-DOTA-4AH29和[131I]I-GMIB-4AH29表现出高和持续的肿瘤靶向性,转化为在携带侵袭性肿瘤的小鼠中延长存活。此外,我们证明PD-L1ICB与[225Ac]Ac-DOTA-4AH29TRT的组合可增强其治疗功效.
    OBJECTIVE: Targeted radionuclide therapy (TRT) is a cancer treatment with relative therapeutic efficacy across various cancer types. We studied the therapeutic potential of TRT using fibroblast activation protein-α (FAP) targeting sdAbs (4AH29) labelled with 225Ac or 131I in immunocompetent mice in a human FAP (hFAP) expressing lung cancer mouse model. We further explored the combination of TRT with programmed cell death ligand 1 (PD-L1) immune checkpoint blockade (ICB).
    METHODS: We studied the biodistribution and tumour uptake of [131I]I-GMIB-4AH29 and [225Ac]Ac-DOTA-4AH29 by ex vivo γ-counting. Therapeutic efficacy of [131I]I-GMIB-4AH29 and [225Ac]Ac-DOTA-4AH29 was evaluated in an immunocompetent mouse model. Flow cytometry analysis of tumours from [225Ac]Ac-DOTA-4AH29 treated mice was performed. Treatment with [225Ac]Ac-DOTA-4AH29 was repeated in combination with PD-L1 ICB.
    RESULTS: The biodistribution showed high tumour uptake of [131I]I-GMIB-4AH29 with 3.5 ± 0.5% IA/g 1 h post-injection (p.i.) decreasing to 0.9 ± 0.1% IA/g after 24 h. Tumour uptake of [225Ac]Ac-DOTA-4AH29 was also relevant with 2.1 ± 0.5% IA/g 1 h p.i. with a less steep decrease to 1.7 ± 0.2% IA/g after 24 h. Survival was significantly improved after treatment with low and high doses [131I]I-GMIB-4AH29 or [225Ac]Ac-DOTA-4AH29 compared to vehicle solution. Moreover, we observed significantly higher PD-L1 expression in tumours of mice treated with [225Ac]Ac-DOTA-4AH29 compared to vehicle solution. Therefore, we combined high dose [225Ac]Ac-DOTA-4AH29 with PD-L1 ICB showing therapeutic synergy.
    CONCLUSIONS: [225Ac]Ac-DOTA-4AH29 and [131I]I-GMIB-4AH29 exhibit high and persistent tumour targeting, translating into prolonged survival in mice bearing aggressive tumours. Moreover, we demonstrate that the combination of PD-L1 ICB with [225Ac]Ac-DOTA-4AH29 TRT enhances its therapeutic efficacy.
    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
    癌症是人类最复杂、最具挑战性的疾病之一,尽管诊断和个性化治疗方案得到改善,但发病率和癌症相关死亡人数仍在上升。靶向α治疗(TαT)为癌症治疗提供了一种令人兴奋的策略,即使在对其他治疗产生抗药性的晚期转移性疾病患者中,这种策略也被证明是有效的。然而,在许多情况下,需要更复杂的策略来阻止疾病进展并克服对TαT的抵抗。历史上用作独立治疗的两种或更多种疗法的组合是近年来一直在追求的方法。这篇综述旨在概述TαT和癌症管理中治疗策略的四个主要支柱。即外部束放射治疗(EBRT),免疫检查点抑制剂(ICI),细胞抑制化疗(CCT),和近距离放射治疗(BT),并讨论它们与TαT结合的潜在用途。每种疗法的简要描述之后是对已知的生物学方面和最先进的治疗实践的回顾。重点,然而,与TαT结合的动机以及迄今为止进行的临床前和临床研究。
    Cancer is one of the most complex and challenging human diseases, with rising incidences and cancer-related deaths despite improved diagnosis and personalized treatment options. Targeted alpha therapy (TαT) offers an exciting strategy emerging for cancer treatment which has proven effective even in patients with advanced metastatic disease that has become resistant to other treatments. Yet, in many cases, more sophisticated strategies are needed to stall disease progression and overcome resistance to TαT. The combination of two or more therapies which have historically been used as stand-alone treatments is an approach that has been pursued in recent years. This review aims to provide an overview on TαT and the four main pillars of therapeutic strategies in cancer management, namely external beam radiation therapy (EBRT), immunotherapy with checkpoint inhibitors (ICI), cytostatic chemotherapy (CCT), and brachytherapy (BT), and to discuss their potential use in combination with TαT. A brief description of each therapy is followed by a review of known biological aspects and state-of-the-art treatment practices. The emphasis, however, is given to the motivation for combination with TαT as well as the pre-clinical and clinical studies conducted to date.
    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:胶质母细胞瘤(GBM)是一种高度侵袭性,侵入性,和不依赖生长因子的IV级胶质瘤。诊断后的生存率通常很差,中位生存期约为15个月,它被认为是最具侵袭性和致命性的中枢神经系统肿瘤。基于手术的常规治疗方法,化疗,放射治疗只能延缓进展,死亡是不可避免的。恶性胶质瘤细胞对传统疗法有抗药性,可能是由于具有侵袭性并能够快速再生的神经胶质瘤干细胞亚群。
    方法:这是一个文献综述。在PubMed上进行了系统的信息检索,Embase,谷歌学者。PubMed中使用了指定的关键词,检索到的文章发表在同行评审的科学期刊上,并与脑GBM癌症和碘化钠同向转运蛋白(NIS)相关。此外,“放射性核素治疗或间充质,或放射性碘或碘-131或分子成像或基因治疗或翻译成像或靶向或治疗或转运体或病毒或实体瘤或联合治疗或垂体或质粒和胶质母细胞瘤或GBM或GB或神经胶质瘤也用于PubMed和GoogleScholar的适当文献数据库。在这次关于间充质干细胞碘化钠转运体和GBM的搜索中,共发现了68,244篇文章。这些文章直到2024年才被发现。为了研究最新进展,添加了一个过滤器,仅包括2014年至2024年的文章,删除了重复项,与标题无关的文章被排除在外。共78篇。从这些,删除关键词不匹配的文章后,没有检索到9个,只选择了7个。适当的研究被隔离,和重要的信息从他们中的每一个被理解,并输入到数据库中的信息,从这篇文章中使用。
    结果:由于它们天然的识别恶性肿瘤的能力,MSC被用作肿瘤治疗载体。因为可以使用几种方法移植MSCs,它们已被提议作为NIS基因转移的理想载体。在许多肿瘤模型中,MSC已经被用作抗癌药物的递送载体,因为它们能够精确地移动到恶性肿瘤。此外,通过将放射性标记的MSCs直接注射到恶性肿瘤中,可以沉积β辐射的治疗剂量,额外的好处是肿瘤只会定位而不会扩散到周围的健康组织。
    结论:基于非侵入性成像的胶质瘤干细胞检测为监测肿瘤、诊断和评估复发提供了一种替代手段。碘化钠转运体基因是多种人类甲状腺疾病中的特异性基因,其功能是将碘移动到细胞中。近年来,越来越多的研究已经报道了与钠碘转运体基因相关的各种肿瘤以及作为成像和治疗的治疗载体。基因治疗和核医学治疗为GBM提供了新的方向。在所有的临床前研究中,图像引导的细胞治疗带来了更大的生存益处,因此,有可能转化为胶质母细胞瘤治疗试验中的技术。
    BACKGROUND: Glioblastoma (GBM) is a highly aggressive, invasive, and growth factor-independent grade IV glioma. Survival following the diagnosis is generally poor, with a median survival of approximately 15 months, and it is considered the most aggressive and lethal central nervous system tumor. Conventional treatments based on surgery, chemotherapy, and radiation therapy only delay progression, and death is inevitable. Malignant glioma cells are resistant to traditional therapies, potentially due to a subpopulation of glioma stem cells that are invasive and capable of rapid regrowth.
    METHODS: This is a literature review. The systematic retrieval of information was performed on PubMed, Embase, and Google Scholar. Specified keywords were used in PubMed and the articles retrieved were published in peer-reviewed scientific journals and were associated with brain GBM cancer and the sodium iodide symporter (NIS). Additionally, the words \'radionuclide therapy OR mesenchyma, OR radioiodine OR iodine-131 OR molecular imaging OR gene therapy OR translational imaging OR targeted OR theranostic OR symporter OR virus OR solid tumor OR combined therapy OR pituitary OR plasmid AND glioblastoma OR GBM OR GB OR glioma\' were also used in the appropriate literature databases of PubMed and Google Scholar. A total of 68,244 articles were found in this search on Mesenchymal Stem Cell Sodium Iodide Symporter and GBM. These articles were found till 2024. To study recent advances, a filter was added to include articles only from 2014 to 2024, duplicates were removed, and articles not related to the title were excluded. These came out to be 78 articles. From these, nine were not retrieved and only seven were selected after the removal of keyword mismatched articles. Appropriate studies were isolated, and important information from each of them was understood and entered into a database from which the information was used in this article.
    RESULTS: As a result of their natural capacity to identify malignancies, MSCs are employed as tumor therapy vehicles. Because MSCs may be transplanted using several methods, they have been proposed as the ideal vehicles for NIS gene transfer. MSCs have been used as a delivery vector for anticancer drugs in many tumor models due to their capacity to move precisely to malignancies. Also, by directly injecting radiolabeled MSCs into malignant tumors, a therapeutic dosage of beta radiation may be deposited, with the added benefit that the tumor would only localize and not spread to the surrounding healthy tissues.
    CONCLUSIONS: The non-invasive imaging-based detection of glioma stem cells presents an alternate means to monitor the tumor and diagnose and evaluate recurrence. The sodium iodide symporter gene is a specific gene in a variety of human thyroid diseases that functions to move iodine into the cell. In recent years, an increasing number of studies related to the sodium iodide symporter gene have been reported in a variety of tumors and as therapeutic vectors for imaging and therapy. Gene therapy and nuclear medicine therapy for GBM provide a new direction. In all the preclinical studies reviewed, image-guided cell therapy led to greater survival benefits and, therefore, has the potential to be translated into techniques in glioblastoma treatment trials.
    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
    转移性前列腺癌是一种常见且致命的疾病。自[177Lu]Lu-PSMA-617批准以来,靶向放射性核素治疗(TRT)已成为一种现成的治疗选择。目前正在研究各种分子用于前列腺癌中的TRT。我们回顾了用于靶向前列腺癌细胞和优化药代动力学的同位素和载体的各种组合。有希望的创新包括化学修饰以改善生物分布,识别新目标,以及新型放射性同位素如α发射体的使用。患者总结:我们的小型综述总结了靶向放射性药物治疗转移性前列腺癌的研究。几种有前途的放射性药物正在临床试验中进行评估,但更多的研究是必要的之前,这些可以用于常规临床实践。
    Metastatic prostate cancer is a frequent and fatal disease. Targeted radionuclide therapy (TRT) has become a readily available therapeutic option since the approval of [177Lu]Lu-PSMA-617. Various molecules are currently being studied for TRT in prostate cancer. We review various combinations of isotopes and vectors being used to target prostate cancer cells and optimize pharmacokinetics. Promising innovations include chemical modifications to improve biodistribution, identification of new targets, and the use of novel radioisotopes such as α emitters. PATIENT SUMMARY: Our mini review summarizes research on targeted radioactive drugs for treatment of metastatic prostate cancer. Several promising radioactive pharmaceuticals are being evaluated in clinical trials, but more studies are necessary before these can be used in routine clinical practice.
    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:肽受体放射性核素治疗(PRRT)使用[177Lu]Lu-[DOTA0-Tyr3]奥曲酯([177Lu]Lu-DOTA-TATE)治疗过度表达生长抑素受体2A(SSTR2A)的神经内分泌肿瘤(NETs)患者。它在生存和症状缓解方面显示出显着的短期改善,但仍有改进的余地。这里,我们研究了[177Lu]Lu-DOTA-TATE与化疗药联合使用是否能增强[177Lu]Lu-DOTA-TATE的体外疗效。
    结果:转染的人骨肉瘤(U2OS+SSTR2A,高SSTR2A表达)和胰腺网络(BON1+STTR2A,培养基SSTR2A表达)细胞接受羟基脲,吉西他滨或triapine在37℃和5%CO2下24小时。然后将细胞回收4小时,然后与0.7-1.03MBq[177Lu]Lu-DOTA-TATE(25nM)孵育24小时,用于摄取和代谢活力研究。U2OS+SSTR2A细胞与羟基脲的孵育,吉西他滨,和triapine增强[177Lu]Lu-DOTA-TATE的摄取从未处理细胞中的0.2±0.1增加到U2OSSSTR2A细胞中的0.4±0.1、1.1±0.2和0.9±0.2Bq/细胞,分别。与用[177Lu]Lu-DOTA-TATE单一疗法处理的细胞相比,用[177Lu]Lu-DOTA-TATE处理后的细胞活力降低。例如,用羟基脲预处理后,用[177Lu]Lu-DOTA-TATE孵育的U2OSSSTR2A细胞的活力从59.5±22.3%降低到18.8±5.2%。对照条件显示没有降低的代谢活力。还收获细胞以评估细胞周期进展。SSTR2A表达式,和细胞大小通过流式细胞术。化疗增加了U2OS+SSTR2A和BON1+STTR2A细胞中的SSTR2A表达和细胞大小。异步U2OS+SSTR2A细胞培养物的S期亚群从45.5±3.3%增加到84.8±2.5%,85.9±1.9%,用羟基脲处理时,86.6±2.2%,吉西他滨,和triapine,分别。
    结论:羟基脲,吉西他滨和曲平都增加了细胞大小,SSTR2A表达式,和[177Lu]Lu-DOTA-TATE摄取,与[177Lu]Lu-DOTA-TATE单一疗法相比,同时降低了U2OS+SSTR2A细胞中的细胞代谢活力。进一步的调查可以改变患者的护理,并积极增加接受[177Lu]Lu-DOTA-TATE治疗的患者的预后。
    BACKGROUND: Peptide receptor radionuclide therapy (PRRT) uses [177Lu]Lu-[DOTA0-Tyr3]octreotate ([177Lu]Lu-DOTA-TATE) to treat patients with neuroendocrine tumours (NETs) overexpressing the somatostatin receptor 2A (SSTR2A). It has shown significant short-term improvements in survival and symptom alleviation, but there remains room for improvement. Here, we investigated whether combining [177Lu]Lu-DOTA-TATE with chemotherapeutics enhanced the in vitro therapeutic efficacy of [177Lu]Lu-DOTA-TATE.
    RESULTS: Transfected human osteosarcoma (U2OS + SSTR2A, high SSTR2A expression) and pancreatic NET (BON1 + STTR2A, medium SSTR2A expression) cells were subjected to hydroxyurea, gemcitabine or triapine for 24 h at 37oC and 5% CO2. Cells were then recovered for 4 h prior to a 24-hour incubation with 0.7-1.03 MBq [177Lu]Lu-DOTA-TATE (25 nM) for uptake and metabolic viability studies. Incubation of U2OS + SSTR2A cells with hydroxyurea, gemcitabine, and triapine enhanced uptake of [177Lu]Lu-DOTA-TATE from 0.2 ± 0.1 in untreated cells to 0.4 ± 0.1, 1.1 ± 0.2, and 0.9 ± 0.2 Bq/cell in U2OS + SSTR2A cells, respectively. Cell viability post treatment with [177Lu]Lu-DOTA-TATE in cells pre-treated with chemotherapeutics was decreased compared to cells treated with [177Lu]Lu-DOTA-TATE monotherapy. For example, the viability of U2OS + SSTR2A cells incubated with [177Lu]Lu-DOTA-TATE decreased from 59.5 ± 22.3% to 18.8 ± 5.2% when pre-treated with hydroxyurea. Control conditions showed no reduced metabolic viability. Cells were also harvested to assess cell cycle progression, SSTR2A expression, and cell size by flow cytometry. Chemotherapeutics increased SSTR2A expression and cell size in U2OS + SSTR2A and BON1 + STTR2A cells. The S-phase sub-population of asynchronous U2OS + SSTR2A cell cultures was increased from 45.5 ± 3.3% to 84.8 ± 2.5%, 85.9 ± 1.9%, and 86.6 ± 2.2% when treated with hydroxyurea, gemcitabine, and triapine, respectively.
    CONCLUSIONS: Hydroxyurea, gemcitabine and triapine all increased cell size, SSTR2A expression, and [177Lu]Lu-DOTA-TATE uptake, whilst reducing cell metabolic viability in U2OS + SSTR2A cells when compared to [177Lu]Lu-DOTA-TATE monotherapy. Further investigations could transform patient care and positively increase outcomes for patients treated with [177Lu]Lu-DOTA-TATE.
    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
    尽管开发了各种新疗法,胶质母细胞瘤(GBM)仍然是一种毁灭性的疾病,中位生存期少于15个月。最近,靶向放射性核素治疗在治疗实体肿瘤方面取得了重大进展,经美国食品和药物管理局(FDA)和欧洲药品管理局(EMA)批准,Lutathera用于神经内分泌肿瘤,Pluvicto用于前列腺癌。这一成就揭示了靶向放射性核素治疗其他实体瘤的潜力,包括GBM。本文综述了GBM中放射性核素靶向治疗的现状。突出常用的治疗放射性核素发射α,β粒子,和俄歇电子可以诱导有效的分子和细胞损伤来治疗GBM。然后我们探索一系列靶向载体,包括小分子,肽,和抗体,选择性靶向表达抗原的肿瘤细胞,与健康组织的结合最小或不结合。考虑到GBM的放射性药物通常是局部给药以绕过血脑屏障(BBB),我们回顾了突出的交付方法,如对流增强交付,局部植入,和立体定向注射。最后,我们解决了GBM这种治疗方法的挑战,并提出了潜在的解决方案.
    Despite the development of various novel therapies, glioblastoma (GBM) remains a devastating disease, with a median survival of less than 15 months. Recently, targeted radionuclide therapy has shown significant progress in treating solid tumors, with the approval of Lutathera for neuroendocrine tumors and Pluvicto for prostate cancer by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This achievement has shed light on the potential of targeted radionuclide therapy for other solid tumors, including GBM. This review presents the current status of targeted radionuclide therapy in GBM, highlighting the commonly used therapeutic radionuclides emitting alpha, beta particles, and Auger electrons that could induce potent molecular and cellular damage to treat GBM. We then explore a range of targeting vectors, including small molecules, peptides, and antibodies, which selectively target antigen-expressing tumor cells with minimal or no binding to healthy tissues. Considering that radiopharmaceuticals for GBM are often administered locoregionally to bypass the blood-brain barrier (BBB), we review prominent delivery methods such as convection-enhanced delivery, local implantation, and stereotactic injections. Finally, we address the challenges of this therapeutic approach for GBM and propose potential solutions.
    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
    目的:本研究旨在比较使用不同成像方案和HERMESHybridViewer剂量测定软件在177Lu-PSMAI&T和177Lu-DOTATATE治疗中获得的靶器官和肿瘤的计算吸收剂量。
    方法:从18例患者中获取了多个时间点全身平面图像和一个SPECT/CT图像,包括177Lu-PSMAI&T(13例)和177Lu-DOTATATE治疗(5例)。注射3.80-8.58GBq的活动。感兴趣的区域被绘制在整个身体中,肾脏,肝脏,膀胱,唾液腺,和肿瘤来确定源器官的时间整合活性(TIA)。根据医学内部辐射剂量(MIRD)方案,使用与OLINDA/EXMV.2.1集成的HERMESHybridViewer剂量测定法计算靶器官的吸收剂量,该剂量利用非均匀有理B样条(NURBS)进行计算数字体模。
    结果:与基于混合的剂量测定法相比,基于平面的剂量测定法显示每次注射活动的剂量更高,主要是由于器官重叠。在脾脏中观察到成像方案之间吸收剂量的最高差异,变化高达51.6%,而其他靶器官和肿瘤的差异小于40%。
    结论:基于2D平面的方法得出的剂量测定计算始终表明,与基于混合的方法相比,器官和肿瘤的吸收剂量明显更高。然而,在肿瘤可视化和无重叠器官勾画方面,混合方法优于平面方法。
    OBJECTIVE: This study aims to compare the calculated absorbed dose in target organs and tumors obtained using the different imaging protocols and the calculation methodologies implemented by HERMES HybridViewer dosimetry software for 177Lu-PSMA I&T and 177Lu-DOTATATE therapy.
    METHODS: Multiple time-point whole-body planar images and one SPECT/CT image were acquired from 18 patients including 177Lu-PSMA I&T (13 patients) and 177Lu-DOTATATE treatment (5 patients) after administration of 3.80-8.58 GBq injected activity. The regions of interest were drawn in the whole body, kidneys, liver, urinary bladder, salivary glands, and tumors to determine the time-integrated activity (TIA) in source organs. Absorbed doses in target organs were calculated according to the Medical Internal Radiation Dose (MIRD) scheme using the HERMES HybridViewer dosimetry integrated with OLINDA/EXM V.2.1 that utilizes the non-uniform rational B-splines (NURBS) for computational digital phantom.
    RESULTS: The planar-based dosimetry showed a higher dose per injected activity compared to the hybrid-based dosimetry, primarily due to organ overlap. The highest difference in absorbed dose between the imaging scenarios was observed in the spleen with a variation of up to 51.6%, while the difference for other target organs and tumors was less than 40%.
    CONCLUSIONS: The dosimetry calculation derived from the 2D planar-based method consistently demonstrates a significantly higher absorbed dose in organs and tumors compared with the hybrid-based method. However, the hybrid method outperforms the planar method in terms of tumor visualization and overlap-free organ delineation.
    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
    靶向放射性核素治疗(TRT)是一种有效的肿瘤治疗方法。自缩合策略可以增强放射性核素在肿瘤中的保留并增强抗肿瘤效果。考虑到legumain在几种类型的人类癌症中过度表达,我们报道了一种131I标记的放射性药物([131I]MAAN),其基于2-氰基苯并噻唑(CBT)和半胱氨酸(Cys)的自缩合反应,用于体内治疗豆科蛋白酶过表达的肿瘤.然而,肝脏富集限制了它的应用。在这项研究中,通过将亲水肽序列His-Glu-His-Glu-His-Glu-Glu-Glu((HE)3)引入[131I]MAAN以优化药代动力学,合成了一种新的放射性药物[131I]IM(HE)3AAN.在还原环境下被生肉激活后,亲水性[131I]IM(HE)3AAN可以与其前体反应形成高度疏水的异源二聚体([131I]H-二聚体)。切伦科夫成像显示,与[131I]MAAN相比,[131I]IM(HE)3AAN显示出更高的肿瘤选择性和更长的肿瘤保留时间,肝脏摄取显着减少。在用[131I]IM(HE)3AAN治疗18天后,肿瘤增殖受到明显抑制,治疗过程中正常器官未见明显损伤。这些发现表明[131I]IM(HE)3AAN成为治疗豆科蛋白过表达肿瘤的有希望的候选者。
    Targeted radionuclide therapy (TRT) is an effective treatment for tumors. Self-condensation strategies can enhance the retention of radionuclides in tumors and enhance the anti-tumor effect. Considering legumain is overexpressed in multiple types of human cancers, a 131I-labeled radiopharmaceutical ([131I]MAAN) based on the self-condensation reaction between 2-cyanobenzothiazole (CBT) and cysteine (Cys) was developed by us recently for treating legumain-overexpressed tumors. However, liver enrichment limits its application. In this study, a new radiopharmaceutical [131I]IM(HE)3AAN was designed and synthesized by introducing a hydrophilic peptide sequence His-Glu-His-Glu-His-Glu ((HE)3) into [131I]MAAN to optimize the pharmacokinetics. Upon activation by legumain under a reducing environment, hydrophilic [131I]IM(HE)3AAN could react with its precursor to form heterologous dimer [131I]H-Dimer that is highly hydrophobic. Cerenkov imaging revealed that [131I]IM(HE)3AAN displayed superior tumor selectivity and longer tumor retention time as compared with [131I]MAAN, with a significant reduction in the liver uptake. After an 18-day treatment with [131I]IM(HE)3AAN, the tumor proliferation was obviously inhibited, while no obvious injury was observed in the normal organs. These findings suggest that [131I]IM(HE)3AAN could serve as a promising drug candidate for treating legumain-overexpressed 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:免疫检查点抑制剂(ICI)通常用于晚期透明细胞肾细胞癌(ccRCC)。然而,大量患者对ICI治疗无反应.辐射是增加ICI应答率的有希望的方法,因为它可以产生抗肿瘤免疫。靶向放射性核素治疗(TRT)是一种全身放射治疗,非常适合转移癌症的精确照射。因此,本研究的目的是探索联合TRT的潜力,靶向在ccRCC中过表达的碳酸酐酶IX(CAIX),使用[177Lu]Lu-DOTA-hG250和ICI治疗ccRCC。方法:在本研究中,我们评估了[177Lu]Lu-DOTA-hG250联合aPD-1/a-CTLA-4ICI的治疗和免疫学作用。首先,在携带Renca-CAIX或CT26-CAIX肿瘤的BALB/cAnNRj小鼠中研究了[177Lu]Lu-DOTA-hG250的生物分布。Renca-CAIX和CT26-CAIX肿瘤的特征是T细胞浸润差和广泛的T细胞浸润,PD-L1表达同质和异质。分别。通过剂量学估计肿瘤吸收的辐射剂量。随后,[177Lu]通过监测肿瘤生长和存活来评估有和没有ICI的Lu-DOTA-hG250TRT功效。通过收集治疗前和治疗后5或8天的肿瘤组织来研究治疗诱导的肿瘤微环境变化,并通过免疫组织化学进行分析。流式细胞术,和RNA分析。结果:生物分布研究显示[177Lu]Lu-DOTA-hG250在两种肿瘤模型中的高肿瘤摄取。Renca-CAIX荷瘤小鼠的剂量递增治疗研究表明[177Lu]Lu-DOTA-hG250的剂量依赖性抗肿瘤功效和显着的治疗协同作用,包括假定的亚治疗TRT剂量时的完全缓解(4MBq,作为单一疗法没有明显疗效)与aPD-1+aCTLA-4联合使用。对于4MBq[177Lu]Lu-DOTA-hG250+a-PD1,在CT26-CAIX模型中获得了类似的结果。治疗肿瘤的离体分析显示DNA损伤,T细胞浸润,并调节联合治疗后TME中的免疫信号通路。结论:亚治疗[177Lu]Lu-DOTA-hG250联合ICI显示出更好的治疗效果,并显着改变了TME。我们的结果强调了在临床环境中对晚期ccRCC患者进行这种联合治疗的重要性。进一步的研究应集中在未来如何最佳地应用联合疗法。
    Background: Immune checkpoint inhibitors (ICI) are routinely used in advanced clear cell renal cell carcinoma (ccRCC). However, a substantial group of patients does not respond to ICI therapy. Radiation is a promising approach to increase ICI response rates since it can generate anti-tumor immunity. Targeted radionuclide therapy (TRT) is a systemic radiation treatment, ideally suited for precision irradiation of metastasized cancer. Therefore, the aim of this study is to explore the potential of combined TRT, targeting carbonic anhydrase IX (CAIX) which is overexpressed in ccRCC, using [177Lu]Lu-DOTA-hG250, and ICI for the treatment of ccRCC. Methods: In this study, we evaluated the therapeutic and immunological action of [177Lu]Lu-DOTA-hG250 combined with aPD-1/a-CTLA-4 ICI. First, the biodistribution of [177Lu]Lu-DOTA-hG250 was investigated in BALB/cAnNRj mice bearing Renca-CAIX or CT26-CAIX tumors. Renca-CAIX and CT26-CAIX tumors are characterized by poor versus extensive T-cell infiltration and homogeneous versus heterogeneous PD-L1 expression, respectively. Tumor-absorbed radiation doses were estimated through dosimetry. Subsequently, [177Lu]Lu-DOTA-hG250 TRT efficacy with and without ICI was evaluated by monitoring tumor growth and survival. Therapy-induced changes in the tumor microenvironment were studied by collection of tumor tissue before and 5 or 8 days after treatment and analyzed by immunohistochemistry, flow cytometry, and RNA profiling. Results: Biodistribution studies showed high tumor uptake of [177Lu]Lu-DOTA-hG250 in both tumor models. Dose escalation therapy studies in Renca-CAIX tumor-bearing mice demonstrated dose-dependent anti-tumor efficacy of [177Lu]Lu-DOTA-hG250 and remarkable therapeutic synergy including complete remissions when a presumed subtherapeutic TRT dose (4 MBq, which had no significant efficacy as monotherapy) was combined with aPD-1+aCTLA-4. Similar results were obtained in the CT26-CAIX model for 4 MBq [177Lu]Lu-DOTA-hG250 + a-PD1. Ex vivo analyses of treated tumors revealed DNA damage, T-cell infiltration, and modulated immune signaling pathways in the TME after combination treatment. Conclusions: Subtherapeutic [177Lu]Lu-DOTA-hG250 combined with ICI showed superior therapeutic outcome and significantly altered the TME. Our results underline the importance of investigating this combination treatment for patients with advanced ccRCC in a clinical setting. Further investigations should focus on how the combination therapy should be optimally applied in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号