targeted radionuclide therapy

靶向放射性核素治疗
  • 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)

  • 文章类型: Journal Article
    在过去的十年中,膀胱癌治疗的努力已经从广泛的手术转向器官保存。为此,本文中,我们通过整合粘膜渗透来开发用于膀胱癌降级和膀胱保留治疗的多功能纳米剂,减少脱靶效应,和内照射治疗纳米药物。具体来说,氧化铁纳米颗粒用作涂覆有透明质酸(HA)以促进粘膜渗透的载体。将二苯并环辛炔(DBCO)引入HA涂层中,通过生物正交反应与叠氮化物作为膀胱癌细胞的人工受体进行反应,以提高177Lu标记的纳米探针的细胞内化。通过磁共振成像,非肌肉浸润性膀胱癌(NMIBC)和肌肉浸润性膀胱癌(MIBC)的靶向成像是在膀胱内滴注多功能探针后实现的,NMIBC和MIBC都被发现降级,转移得到抑制,这证明了多功能纳米探针在膀胱癌治疗中保留膀胱的潜力。
    Efforts on bladder cancer treatment have been shifting from extensive surgery to organ preservation in the past decade. To this end, we herein develop a multifunctional nanoagent for bladder cancer downstaging and bladder-preserving therapy by integrating mucosa penetration, reduced off-target effects, and internal irradiation therapy into a nanodrug. Specifically, an iron oxide nanoparticle was used as a carrier that was coated with hyaluronic acid (HA) for facilitating mucosa penetration. Dibenzocyclooctyne (DBCO) was introduced into the HA coating layer to react through bioorthogonal reaction with azide as an artificial receptor of bladder cancer cells, to improve the cellular internalization of the nanoprobe labeled with 177Lu. Through magnetic resonance imaging, the targeted imaging of both nonmuscle-invasive bladder cancer (NMIBC) and muscle-invasive bladder cancer (MIBC) was realized after intravesical instillation of the multifunctional probe, both NMIBC and MIBC were found downstaged, and the metastasis was inhibited, which demonstrates the potential of the multifunctional nanoprobe for bladder preservation in bladder cancer treatment.
    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:为使用放射性标记的生长抑素受体(SSTR)配体进行脑膜瘤的诊断和治疗(治疗)提供实践指南/程序标准。
    方法:该联合实践指南/程序标准由欧洲核医学协会(EANM)共同开发,核医学与分子影像学会(SNMMI),欧洲神经肿瘤学协会(EANO),和神经肿瘤学工作组反应评估的PET工作组(PET/RANO)。
    结果:使用生长抑素受体(SSTR)配体的正电子发射断层扫描(PET)可以高灵敏度和特异性地检测脑膜瘤组织,并且可以提供超出仅从结构磁共振成像(MRI)或计算机断层扫描(CT)成像获得的临床相关信息。SSTR导向的PET成像对于鉴别诊断特别有用,脑膜瘤范围的描绘,骨受累的检测,以及治疗后瘢痕组织和肿瘤复发之间的区别。此外,SSTR肽受体放射性核素治疗(PRRT)是一种新兴的脑膜瘤研究性治疗方法。
    结论:这些实践指南将为脑膜瘤患者和相关的SSTR靶向PRRT在常规实践和临床试验中的PET成像应用定义程序标准,并将有助于协调数据采集和跨中心解释,促进研究的可比性,收集更大的数据库。当前文件为PET/RANO工作组关于在脑膜瘤Galldiks(NeuroOncol。2017;19(12):1576-87)。应在当地条件和法规的背景下考虑所提供的信息。
    OBJECTIVE: To provide practice guideline/procedure standards for diagnostics and therapy (theranostics) of meningiomas using radiolabeled somatostatin receptor (SSTR) ligands.
    METHODS: This joint practice guideline/procedure standard was collaboratively developed by the European Association of Nuclear Medicine (EANM), the Society of Nuclear Medicine and Molecular Imaging (SNMMI), the European Association of Neurooncology (EANO), and the PET task force of the Response Assessment in Neurooncology Working Group (PET/RANO).
    RESULTS: Positron emission tomography (PET) using somatostatin receptor (SSTR) ligands can detect meningioma tissue with high sensitivity and specificity and may provide clinically relevant information beyond that obtained from structural magnetic resonance imaging (MRI) or computed tomography (CT) imaging alone. SSTR-directed PET imaging can be particularly useful for differential diagnosis, delineation of meningioma extent, detection of osseous involvement, and the differentiation between posttherapeutic scar tissue and tumour recurrence. Moreover, SSTR-peptide receptor radionuclide therapy (PRRT) is an emerging investigational treatment approach for meningioma.
    CONCLUSIONS: These practice guidelines will define procedure standards for the application of PET imaging in patients with meningiomas and related SSTR-targeted PRRTs in routine practice and clinical trials and will help to harmonize data acquisition and interpretation across centers, facilitate comparability of studies, and to collect larger databases. The current document provides additional information to the evidence-based recommendations from the PET/RANO Working Group regarding the utilization of PET imaging in meningiomas Galldiks (Neuro Oncol. 2017;19(12):1576-87). The information provided should be considered in the context of local conditions and regulations.
    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:在这项工作中,我们提出并评估了一种用于执行溶液中β粒子发射放射性药物治疗剂的界面 测量的技术。 方法:使用NIST 匹配的X射线束校准未层压的EBT3膜对水的吸收剂量。定制丙烯酸源模被构建并放置在由骨骼组成的接口上方。肺,和水等效材料。将薄膜放置在与这些界面垂直的位置,并使用90Y和177Lu的溶液对水中的吸收剂量进行测量,并与用EGSnrc模拟的MonteCarlo对水的吸收剂量估计值进行比较。还进行了表面和深度剂量分布测量。 主要结果:177Lu的表面吸收剂量与预测结果 在3.6%和90Y的2.2%内一致。对于深度剂量和界面剖面,90Y的预测和测量的吸收 剂量对水的一致性优于177Lu。总的来说, 对于放射性核素和所有界面都观察到k=1不确定性范围内的一致性。由于测量对材料表面缺陷的敏感性增加,因此对于177Lu的骨-水界面发现了一个例外。
重要意义:这项工作证明了使用辐射变色胶片
对β发射放射性药物疗法
跨材料界面进行水吸收剂量测量的可行性。
    Objective.In this work, we present and evaluate a technique for performing interface measurements of beta particle-emitting radiopharmaceutical therapy agents in solution.Approach.Unlaminated EBT3 film was calibrated for absorbed dose to water using a NIST matched x-ray beam. Custom acrylic source phantoms were constructed and placed above interfaces comprised of bone, lung, and water-equivalent materials. The film was placed perpendicular to these interfaces and measurements for absorbed dose to water using solutions of90Y and177Lu were performed and compared to Monte Carlo absorbed dose to water estimates simulated with EGSnrc. Surface and depth dose profile measurements were also performed.Main results.Surface absorbed dose to water measurements agreed with predicted results within 3.6% for177Lu and 2.2% for90Y. The agreement between predicted and measured absorbed dose to water was better for90Y than177Lu for depth dose and interface profiles. In general, agreement withink= 1 uncertainty bounds was observed for both radionuclides and all interfaces. An exception to this was found for the bone-to-water interface for177Lu due to the increased sensitivity of the measurements to imperfections in the material surfaces.Significance. This work demonstrates the feasibility and limitations of using radiochromic film for performing absorbed dose to water measurements on beta particle-emitting radiopharmaceutical therapy agents across material interfaces.
    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
    脑转移瘤(BrM)是中枢神经系统常见的恶性病变,由于诊断延迟和治疗选择有限,对晚期恶性肿瘤构成重大威胁。它们独特的基因组谱强调了分子谱以定制有效治疗的需要。癌症生物学的最新进展揭示了肿瘤起始的分子驱动因素,programming,和转移。这个,加上分子成像技术和放射性示踪剂合成的进步,为开发对BrM特异性靶标具有增强特异性和亲和力的创新放射性药物铺平了道路。尽管血脑屏障对有效的药物输送提出了挑战,几种放射性标记的化合物在检测和靶向BrM方面显示出希望。本手稿概述了临床和临床前环境中用于核成像和靶向放射性核素治疗的分子生物标志物的最新进展。此外,它探讨了解决BrM带来的独特挑战的潜在疗法应用。
    Brain metastases (BrM) are common malignant lesions in the central nervous system, and pose a significant threat in advanced-stage malignancies due to delayed diagnosis and limited therapeutic options. Their distinct genomic profiles underscore the need for molecular profiling to tailor effective treatments. Recent advances in cancer biology have uncovered molecular drivers underlying tumor initiation, progression, and metastasis. This, coupled with the advances in molecular imaging technology and radiotracer synthesis, has paved the way for the development of innovative radiopharmaceuticals with enhanced specificity and affinity for BrM specific targets. Despite the challenges posed by the blood-brain barrier to effective drug delivery, several radiolabeled compounds have shown promise in detecting and targeting BrM. This manuscript provides an overview of the recent advances in molecular biomarkers used in nuclear imaging and targeted radionuclide therapy in both clinical and preclinical settings. Additionally, it explores potential theranostic applications addressing the unique challenges posed by BrM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号