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
    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
    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
    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)

  • 文章类型: Journal Article
    [177Lu]Lu-PSMA-617最近成功获得FDA批准,MHRA,加拿大卫生部和EMA作为Plovicto®。然而,唾液腺(SG)和肾脏毒性是其主要的剂量限制性副作用,而其相应的吸收和保留机制仍然难以捉摸。最近,存在不同的ATP结合盒(ABC)转运蛋白,如人类乳腺癌耐药蛋白(BCRP),多药耐药蛋白(MDR1),多药耐药相关蛋白(MRP1,MRP4)和溶质盒(SLC)转运体,如多药和毒素挤出蛋白(MATE1,MATE2-K),有机阴离子转运蛋白(OAT1,OAT2v1,OAT3,OAT4)和肽转运蛋白(PEPT2),已在人类SGs和肾脏中的不同丰度得到验证。因此,我们的目的是评估[177Lu]Lu-PSMA-617和[225Ac]Ac-PSMA-617是否是这些ABC和SLC转运蛋白的底物。对于体外研究,新型同位素([α,β-3H]Nal)Lu-PSMA-617用于表达上述人ABC和SLC转运蛋白的细胞系或囊泡,用于抑制和摄取研究,分别。相应的探针底物和参考抑制剂用作对照。我们的结果表明[177Lu]Lu-PSMA-617和[225Ac]Ac-PSMA-617既不是所检查转运蛋白的抑制剂也不是底物。因此,我们的结果表明,人类ABC和SLC转运蛋白在SGs和肾脏中吸收和保留[177Lu]Lu-PSMA-617和[225Ac]Ac-PSMA-617以及观察到的毒性中没有核心作用。
    [177Lu]Lu-PSMA-617 has recently been successfully approved by the FDA, the MHRA, Health Canada and the EMA as Pluvicto®. However, salivary gland (SG) and kidney toxicities account for its main dose-limiting side-effects, while its corresponding uptake and retention mechanisms still remain elusive. Recently, the presence of different ATP-binding cassette (ABC) transporters, such as human breast cancer resistance proteins (BCRP), multidrug resistance proteins (MDR1), multidrug-resistance-related proteins (MRP1, MRP4) and solute cassette (SLC) transporters, such as multidrug and toxin extrusion proteins (MATE1, MATE2-K), organic anion transporters (OAT1, OAT2v1, OAT3, OAT4) and peptide transporters (PEPT2), has been verified at different abundances in human SGs and kidneys. Therefore, our aim was to assess whether [177Lu]Lu-PSMA-617 and [225Ac]Ac-PSMA-617 are substrates of these ABC and SLC transporters. For in vitro studies, the novel isotopologue ([α,β-3H]Nal)Lu-PSMA-617 was used in cell lines or vesicles expressing the aforementioned human ABC and SLC transporters for inhibition and uptake studies, respectively. The corresponding probe substrates and reference inhibitors were used as controls. Our results indicate that [177Lu]Lu-PSMA-617 and [225Ac]Ac-PSMA-617 are neither inhibitors nor substrates of the examined transporters. Therefore, our results show that human ABC and SLC transporters play no central role in the uptake and retention of [177Lu]Lu-PSMA-617 and [225Ac]Ac-PSMA-617 in the SGs and kidneys nor in the observed toxicities.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    嗜铬细胞瘤和副神经节瘤(PPGLs)是罕见的产生儿茶酚胺的肿瘤,表达生长抑素受体(SSTR),可以用of177DOTATATE(Lu-177-TRT)治疗;然而,治疗可能与危及生命的心血管事件有关.描述了接受Lu-177-TRT的高风险PPGL患者的管理策略。该78岁的转移性副神经节瘤患者被纳入并在NCT03206060下治疗。被认为是高风险的,患者提前入住重症监护病房(ICU),并设置了中央导管通道.由于合并症,该患者使用减少剂量的100mCi×4个周期.生命体征,血液检查,在不同时间点获得血清儿茶酚胺水平。尽管剂量减少,患者在输注Lu-177-TRT后数分钟内仍出现严重的高血压反应,收缩压为240mmHg,用静脉注射尼卡地平控制。患者在Lu-177-TRT后在ICU中停留24小时,然后再转移到住院病房24小时。所有随后的输注均使用减少的剂量和选择性ICU入院进行,并且耐受性良好。尽管风险增加,转移性PPGL患者可以通过适当的人员培训安全治疗,监测,并准备静脉注射药物,尤其是尼卡地平.
    Pheochromocytomas and paragangliomas (PPGLs) are rare catecholamine-producing tumors that express somatostatin receptors (SSTR) that can be treated with lutetium-177 DOTATATE (Lu-177-TRT); however, treatment can be associated with life-threatening cardiovascular events. A patient case with management strategies for high-risk PPGL patients receiving Lu-177-TRT is described. The 78-year-old patient with metastatic paraganglioma was enrolled and treated under NCT03206060. Deemed to be at high risk, the patient was preemptively admitted to the intensive care unit (ICU) with central line access placed. Due to comorbidities, a reduced dose of 100 mCi x 4 cycles was used for this patient. Vital signs, blood work, and serum catecholamine levels were obtained at various time points. Despite reduced dosing, the patient still developed a severe hypertensive reaction with systolic blood pressure of 240 mmHg within minutes of Lu-177-TRT infusion, which was controlled with an intravenous nicardipine drip. The patient remained in the ICU for 24 hours post Lu-177-TRT before moving to an inpatient ward for an additional 24 hours. All subsequent infusions were performed using reduced doses with elective ICU admissions and were well-tolerated. Despite the increased risk, metastatic PPGL patients can be safely treated with proper staff training, monitoring, and preparation for intravenous medications, especially nicardipine.
    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
    基于放射性核素的治疗代表了一种新的肿瘤治疗方案。在这些疗法中,Lutetium-177(177Lu)因其稳定性和安全性而受到广泛关注,以及它发射γ和β射线的能力,允许单光子发射计算机断层扫描和肿瘤治疗成像。因此,177Lu可用于前列腺癌和胃癌等疾病的诊断和治疗。因此,根据现有数据,本文简要概述了177Lu靶向放射性核素治疗转移性前列腺癌的临床应用。神经内分泌肿瘤和其他类型的实体瘤,并强调了目前的治疗效果,减少对正常组织的损伤和未来的研究方向,包括新核素的开发和更多核素在不同肿瘤中的应用。在未来,这种治疗方法可以用于更多的肿瘤。
    Radionuclide-based therapy represents a novel treatment regimen for tumors. Among these therapies, lutetium-177 (177Lu) has gained significant attention due to its stability and safety, as well as its ability to emit both γ and β rays, allowing for both imaging with single photon emission computed tomography and tumor treatment. As a result, 177Lu can be used for both diagnosis and treatment for diseases such as prostatic and gastric cancer. Therefore, based on the available data, the present review provides a brief overview of the clinical applications of 177Lu-targeted radionuclide therapy in metastatic prostate cancer, neuroendocrine tumors and other types of solid tumors, and highlights the current therapeutic effect, reduction in damage to normal tissues and future research directions, including the development of new nuclides and the application of more nuclides in different tumors. In the future, such treatments could be used in more tumors.
    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
    由于其潜在的免疫刺激作用,电离辐射作为免疫疗法的组合伙伴已经引起了相当大的关注。与更常用的外部光束辐射相比,我们探讨了将嵌合抗原受体(CAR)T细胞治疗与靶向放射性核素治疗(TRT)相结合的可行性,这是通过通过表达生长抑素受体2(SSTR2)的肿瘤浸润性CART细胞向肿瘤递送β-发射177Lu-DOTATATE来实现的。我们假设向肿瘤输送辐射可以与CAR-T治疗协同作用,导致增强的抗肿瘤免疫和肿瘤反应。为了确定177Lu-DOTATATE治疗的最佳剂量和时机,我们使用SSTR2特异性正电子发射放射性示踪剂通过正电子发射断层扫描(PET)纵向测量了肿瘤中的CART细胞浸润和扩增,18F-NOTA-奥曲肽。在给予177Lu-DOTATATE后接受CART细胞和低剂量(2.5Gy)TRT的动物中,我们观察到大皮下肿瘤的快速消退,这与血清促炎细胞因子的急剧增加相吻合。当将更高的辐射剂量(6Gy)递送至肿瘤时,肿瘤负荷也降低。然而,这种较高的剂量导致肿瘤和CART细胞的细胞死亡。我们的研究表明,可能存在TRT剂量的最佳范围,可以增强T细胞活性并使肿瘤细胞对T细胞杀伤敏感,与更高的辐射剂量相比,这可能会导致更持久的肿瘤控制。
    Ionizing radiation has garnered considerable attention as a combination partner for immunotherapy due to its potential immunostimulatory effects. In contrast to the more commonly used external beam radiation, we explored the feasibility of combining chimeric antigen receptor (CAR) T cell therapy with targeted radionuclide therapy (TRT), which is achieved by delivering β-emitting 177Lu-DOTATATE to tumor via tumor-infiltrating CAR T cells that express somatostatin receptor 2 (SSTR2). We hypothesized that the delivery of radiation to tumors could synergize with CAR T therapy, resulting in enhanced antitumor immunity and tumor response. To determine the optimal dosage and timing of 177Lu-DOTATATE treatment, we measured CAR T cell infiltration and expansion in tumors longitudinally through positron emission tomography (PET) using a SSTR2-specific positron-emitting radiotracer,18F-NOTA-Octreotide. In animals receiving CAR T cells and a low-dose (2.5 Gy) of TRT following the administration of 177Lu-DOTATATE, we observed a rapid regression of large subcutaneous tumors, which coincided with a dramatic increase in serum proinflammatory cytokines. Tumor burden was also reduced when a higher radiation dose (6 Gy) was delivered to the tumor. However, this higher dose led to cell death in both the tumor and CAR T cells. Our study suggests that there may exist an optimum range of TRT dosage that can enhance T cell activity and sensitize tumor cells to T cell killing, which may result in more durable tumor control compared to a higher radiation dose.
    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
    这篇综述文章探讨了分子放射疗法(MRT)的演变景观,强调肽受体放射性核素治疗(PRRT)的神经内分泌肿瘤(NET)。主要重点是PRRT中从β发射放射性药物到α发射剂的过渡,提供放射生物学基础的批判性分析,临床应用,以及靶向α治疗(TAT)的持续发展。通过广泛的文献综述,本文探讨了PRRT靶向生长抑素2亚型受体的机制和有效性,强调它的成功和局限性。讨论延伸到TAT的新兴范式,强调其具有α粒子发射的更高效力和特异性,这有望增强治疗效果和降低毒性。审查严格评估临床前和临床数据,强调需要标准化剂量测定和对TAT中剂量-反应关系的更深入理解。该综述最后强调了TAT在治疗SSTR2过度表达的癌症方面的巨大潜力,特别是在β-PRRT难治性患者中,同时也承认当前的挑战和进一步研究以优化治疗方案的必要性。
    This review article explores the evolving landscape of Molecular Radiotherapy (MRT), emphasizing Peptide Receptor Radionuclide Therapy (PRRT) for neuroendocrine tumours (NETs). The primary focus is on the transition from β-emitting radiopharmaceuticals to α-emitting agents in PRRT, offering a critical analysis of the radiobiological basis, clinical applications, and ongoing developments in Targeted Alpha Therapy (TAT). Through an extensive literature review, the article delves into the mechanisms and effectiveness of PRRT in targeting somatostatin subtype 2 receptors, highlighting both its successes and limitations. The discussion extends to the emerging paradigm of TAT, underlining its higher potency and specificity with α-particle emissions, which promise enhanced therapeutic efficacy and reduced toxicity. The review critically evaluates preclinical and clinical data, emphasizing the need for standardised dosimetry and a deeper understanding of the dose-response relationship in TAT. The review concludes by underscoring the significant potential of TAT in treating SSTR2-overexpressing cancers, especially in patients refractory to β-PRRT, while also acknowledging the current challenges and the necessity for further research to optimize treatment protocols.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号