gastrin-releasing peptide receptor

  • 文章类型: Journal Article
    前列腺癌(PC)的高发病率和沉重的疾病负担需要准确而全面的评估以进行适当的疾病管理。前列腺特异性膜抗原(PSMA)正电子发射断层扫描(PET)不能检测PSMA阴性病变,尽管它在PC疾病管理中起着关键作用。据报道,PC病变中胃泌素释放肽受体(GRPR)的过表达是PC诊断和治疗的补充靶标。已经开发了衍生自GRPR的天然配体的放射性药物。这些放射性药物能够在体内可视化和量化GRPR,可用于疾病评估和治疗指导。最近开发的放射性药物表现出改善的药代动力学参数,而亲和力没有恶化。已经构建了几种靶向GRPR的异二聚体作为替代方案,因为它们具有以单靶标检测的低诊断效率检测肿瘤病变的潜力。此外,一些针对GRPR的放射性药物已进入临床试验,用于PC的初始分期或生化复发检测,以指导疾病分层和治疗,表明在PC疾病管理方面具有相当大的潜力。在这里,我们全面总结了针对GRPR的放射性药物的研究进展。特别是,我们讨论了配体的影响,螯合剂,和连接剂对放射性药物的分布。此外,我们总结了一个潜在的设计方案,以促进放射性药物的发展,因此,提示临床翻译。
    The high incidence and heavy disease burden of prostate cancer (PC) require accurate and comprehensive assessment for appropriate disease management. Prostate-specific membrane antigen (PSMA) positron emission tomography (PET) cannot detect PSMA-negative lesions, despite its key role in PC disease management. The overexpression of gastrin-releasing peptide receptor (GRPR) in PC lesions reportedly performs as a complementary target for the diagnosis and therapy of PC. Radiopharmaceuticals derived from the natural ligands of GRPR have been developed. These radiopharmaceuticals enable the visualization and quantification of GRPR within the body, which can be used for disease assessment and therapeutic guidance. Recently developed radiopharmaceuticals exhibit improved pharmacokinetic parameters without deterioration in affinity. Several heterodimers targeting GRPR have been constructed as alternatives because of their potential to detect tumor lesions with a low diagnostic efficiency of single target detection. Moreover, some GRPR-targeted radiopharmaceuticals have entered clinical trials for the initial staging or biochemical recurrence detection of PC to guide disease stratification and therapy, indicating considerable potential in PC disease management. Herein, we comprehensively summarize the progress of radiopharmaceuticals targeting GRPR. In particular, we discuss the impact of ligands, chelators, and linkers on the distribution of radiopharmaceuticals. Furthermore, we summarize a potential design scheme to facilitate the advancement of radiopharmaceuticals and, thus, prompt clinical translation.
    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
    目标:牙龈卟啉单胞菌(P.牙龈),一种主要的牙周病原体,与动脉粥样硬化的进展和恶化有关。在这项研究中,我们的目的是研究胃泌素释放肽受体拮抗剂,RC-3095可以减弱牙龈卟啉单胞菌LPS诱导的内皮细胞和巨噬细胞的炎症反应,以及用牙龈卟啉单胞菌LPS处理的ApoE-/-小鼠模型中的动脉粥样硬化。
    方法:使用HUVECs和大鼠主动脉内皮检查RC-3095对牙龈卟啉单胞菌LPS诱导的内皮炎症的影响。通过暴露于牙龈卟啉单胞菌LPS,将THP-1细胞极化为M1巨噬细胞,有或没有RC-3095。通过注射牙龈卟啉单胞菌LPS,在高脂喂养的雄性ApoE-/-小鼠中评估RC-3095对动脉粥样硬化进展的影响,RC-3095,或两者的组合。
    结果:RC-3095通过抑制NF-κB依赖性的ICAM-1和VCAM-1的表达,显著降低牙龈卟啉单胞菌LPS诱导的白细胞与内皮细胞和主动脉内皮的粘附。此外,RC-3095通过阻断MAPK和NF-κB信号通路抑制牙龈卟啉单胞菌LPS诱导的M1巨噬细胞极化。此外,RC-3095减少ApoE-/-小鼠动脉粥样硬化病变面积,通过牙龈卟啉单胞菌LPS注射加速,降低动脉粥样硬化小鼠主动脉组织中ICAM-1和VCAM-1的表达。
    结论:RC-3095可以减轻牙龈卟啉单胞菌LPS诱导的内皮细胞炎症,巨噬细胞极化,和动脉粥样硬化进展,提示其作为牙周病原体相关动脉粥样硬化的治疗方法的潜力。
    OBJECTIVE: Porphyromonas gingivalis (P. gingivalis), one of the major periodontopathogens, is associated with the progression and exacerbation of atherosclerosis. In this study, we aimed to investigate whether the gastrin-releasing peptide receptor antagonist, RC-3095, could attenuate P. gingivalis LPS-induced inflammatory responses in endothelial cells and macrophages, as well as atherosclerosis in an ApoE-/- mouse model treated with P. gingivalis LPS.
    METHODS: The effect of RC-3095 on P. gingivalis LPS-induced endothelial inflammation was examined using HUVECs and rat aortic endothelium. THP-1 cells were polarized into M1 macrophages by exposure to P. gingivalis LPS, with or without RC-3095. The effect of RC-3095 on atherosclerosis progression was assessed in high-fat-fed male ApoE-/- mice through injections of P. gingivalis LPS, RC-3095, or a combination of both.
    RESULTS: RC-3095 significantly reduced P. gingivalis LPS-induced leukocyte adhesion to endothelial cells and aortic endothelium by suppressing NF-κB-dependent expressions of ICAM-1 and VCAM-1. In addition, RC-3095 inhibited the P. gingivalis LPS-induced polarization of M1 macrophages by blocking the MAPK and NF-κB signaling pathways. Moreover, RC-3095 decreased the area of atherosclerotic lesions in ApoE-/- mice, which was accelerated by P. gingivalis LPS injection, and lowered the expressions of ICAM-1 and VCAM-1 in the aortic tissue of mice with atherosclerosis.
    CONCLUSIONS: RC-3095 can alleviate P. gingivalis LPS-induced endothelial inflammation, macrophage polarization, and atherosclerosis progression, suggesting its potential as a therapeutic approach for periodontal pathogen-associated atherosclerosis.
    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
    我们开发了一种针对胃泌素释放肽受体(GRPrs)的新型位点特异性双峰MRI/荧光纳米颗粒造影剂,在侵袭性前列腺癌中过度表达。使用葡萄糖和酪蛋白涂层合成了生物相容性超小超顺磁性氧化铁(USPIO)纳米颗粒,然后与Cy7.5-K-8AOC-BBN[7-14]肽缀合物缀合。所得USPIO(Cy7.5)-BBN纳米颗粒通过100kDa膜透析纯化,并使用透射电子显微镜(TEM)充分表征。动态光散射(DLS),傅里叶变换红外(FTIR)光谱,和磁共振成像(MRI)弛豫率,以及评估PC-3前列腺癌细胞和异种移植荷瘤小鼠的体外和体内结合特异性和成像功效。USPIO(Cy7.5)-BBN纳米颗粒具有4.93±0.31nm的核直径和35.56±0.58nm的流体动力学直径。在7TMRI下测得的r2弛豫率为70.2±2.5s-1mM-1。Cy7.5-K-8AOC-BBN[7-14]肽与纳米颗粒的比率被确定为21:1。体外GRPr抑制结合(IC50)值为2.5±0.7nM,表明USPIO(Cy7.5)-BBN对PC-3细胞上的GRPr具有非常高的结合亲和力。体内MRI显示,与阻断组(4h:15.3±2.0%和24h:-2.8±6.8%;p<0.005)相比,注射后4h(31.1±3.4%)和24h(25.7±2.1%)摄取组的肿瘤肌肉对比增强。体内和离体近红外荧光(NIRF)成像显示,与阻断组相比,摄取组的肿瘤荧光显着增加。这些发现证明了双峰USPIO(Cy7.5)-BBN纳米颗粒对表达GRPr的PC-3细胞的高特异性,表明它们在侵袭性前列腺癌中具有靶向成像的潜力。
    We developed a novel site-specific bimodal MRI/fluorescence nanoparticle contrast agent targeting gastrin-releasing peptide receptors (GRPrs), which are overexpressed in aggressive prostate cancers. Biocompatible ultra-small superparamagnetic iron oxide (USPIO) nanoparticles were synthesized using glucose and casein coatings, followed by conjugation with a Cy7.5-K-8AOC-BBN [7-14] peptide conjugate. The resulting USPIO(Cy7.5)-BBN nanoparticles were purified by 100 kDa membrane dialysis and fully characterized using transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier transform infrared (FTIR) spectroscopy, and magnetic resonance imaging (MRI) relaxivity, as well as evaluated for in vitro and in vivo binding specificity and imaging efficacy in PC-3 prostate cancer cells and xenografted tumor-bearing mice. The USPIO(Cy7.5)-BBN nanoparticles had a core diameter of 4.93 ± 0.31 nm and a hydrodynamic diameter of 35.56 ± 0.58 nm. The r2 relaxivity was measured to be 70.2 ± 2.5 s-1 mM-1 at 7T MRI. The Cy7.5-K-8AOC-BBN [7-14] peptide-to-nanoparticle ratio was determined to be 21:1. The in vitro GRPr inhibitory binding (IC50) value was 2.5 ± 0.7 nM, indicating a very high binding affinity of USPIO(Cy7.5)-BBN to the GRPr on PC-3 cells. In vivo MRI showed significant tumor-to-muscle contrast enhancement in the uptake group at 4 h (31.1 ± 3.4%) and 24 h (25.7 ± 2.1%) post-injection compared to the blocking group (4 h: 15.3 ± 2.0% and 24 h: -2.8 ± 6.8%; p < 0.005). In vivo and ex vivo near-infrared fluorescence (NIRF) imaging revealed significantly increased fluorescence in tumors in the uptake group compared to the blocking group. These findings demonstrate the high specificity of bimodal USPIO(Cy7.5)-BBN nanoparticles towards GRPr-expressing PC-3 cells, suggesting their potential for targeted imaging in aggressive prostate cancer.
    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
    胃泌素释放肽受体(GRPR),在许多实体瘤中过度表达,是一个很有前途的成像标记和治疗靶点。大多数报道的GRPR靶向放射性配体含有C末端酰胺。根据报道的强效拮抗剂D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH,我们合成了C末端异羟肟酸盐衍生的[68Ga]Ga-LW02075([68Ga]Ga-DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH)和[68Ga]Ga-LW02050([68Ga]-DOTA-Pip-D-NHal-Phe-并将它们与密切相关和临床验证的[68Ga]Ga-SB3([68Ga]Ga-DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHEt)进行了比较。Ga-SB3、Ga-LW02075和Ga-LW02050的结合亲和力(Ki)分别为1.20±0.31、1.39±0.54和8.53±1.52nM,分别。通过钙释放测定证实Ga-LW02075和Ga-LW02050都是GRPR拮抗剂。影像学研究表明,在PET图像中,[68Ga]Ga-SB3和[68Ga]Ga-LW02050注射后1小时,PC-3前列腺癌肿瘤异种移植物清晰可见。但不是[68Ga]Ga-LW02075。注射后1小时进行的离体生物分布研究表明,[68Ga]Ga-LW02050的肿瘤摄取与[68Ga]Ga-SB3相当(5.38±1.00vs.6.98±1.36%内径/g),然后是[68Ga]Ga-LW02075(3.97±1.71%ID/g)。[68Ga]Ga-SB3的胰腺摄取最高(37.3±6.90%ID/g),其次是[68Ga]Ga-LW02075(17.8±5.24%ID/g),而[68Ga]Ga-LW02050的胰腺摄取仅为0.53±0.11%ID/g。我们的数据表明[68Ga]Ga-LW02050是一种有前途的PET示踪剂,可用于检测表达GRPR的癌症病变。
    Gastrin-releasing peptide receptor (GRPR), overexpressed in many solid tumors, is a promising imaging marker and therapeutic target. Most reported GRPR-targeted radioligands contain a C-terminal amide. Based on the reported potent antagonist D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH, we synthesized C-terminal hydroxamate-derived [68Ga]Ga-LW02075 ([68Ga]Ga-DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH) and [68Ga]Ga-LW02050 ([68Ga]Ga-DOTA-Pip-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHOH), and compared them with the closely related and clinically validated [68Ga]Ga-SB3 ([68Ga]Ga-DOTA-pABzA-DIG-D-Phe-Gln-Trp-Ala-Val-Gly-His-Leu-NHEt). Binding affinities (Ki) of Ga-SB3, Ga-LW02075, and Ga-LW02050 were 1.20 ± 0.31, 1.39 ± 0.54, and 8.53 ± 1.52 nM, respectively. Both Ga-LW02075 and Ga-LW02050 were confirmed to be GRPR antagonists by calcium release assay. Imaging studies showed that PC-3 prostate cancer tumor xenografts were clearly visualized at 1 h post injection by [68Ga]Ga-SB3 and [68Ga]Ga-LW02050 in PET images, but not by [68Ga]Ga-LW02075. Ex vivo biodistribution studies conducted at 1 h post injection showed that the tumor uptake of [68Ga]Ga-LW02050 was comparable to that of [68Ga]Ga-SB3 (5.38 ± 1.00 vs. 6.98 ± 1.36 %ID/g), followed by [68Ga]Ga-LW02075 (3.97 ± 1.71 %ID/g). [68Ga]Ga-SB3 had the highest pancreas uptake (37.3 ± 6.90 %ID/g) followed by [68Ga]Ga-LW02075 (17.8 ± 5.24 %ID/g), while the pancreas uptake of [68Ga]Ga-LW02050 was only 0.53 ± 0.11 %ID/g. Our data suggest that [68Ga]Ga-LW02050 is a promising PET tracer for detecting GRPR-expressing cancer lesions.
    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
    胃泌素释放肽受体(GRPR)在各种癌症中过度表达,是癌症诊断和治疗的有希望的靶标。然而,大多数报道的GRPR靶向放射性配体观察到的高胰腺摄取和/或代谢不稳定性可能限制了其临床应用.我们小组最近报道了一种GRPR靶向拮抗剂示踪剂,[68Ga]Ga-TacsBOMB2([68Ga]Ga-DOTA-Pip-D-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Leu13ivthz14-NH2),在临床前小鼠模型中显示最小的胰腺摄取。在这项研究中,我们合成了四种具有非天然氨基酸取代的衍生物(Tle10衍生的Ga-LW01158,NMe-His12衍生的Ga-LW01160,α-Me-Trp8-和Tle10衍生的Ga-LW01186以及Tle10-和N-Me-Gly11-Ga衍生的-LW02002),并评估了它们的潜力。Ga-LW01158、Ga-LW01160、Ga-LW01186和Ga-LW02002的结合亲和力(Ki(GRPR))分别为5.11±0.47、187±17.8、6.94±0.95和11.0±0.39nM,分别。[68Ga]Ga-LW01158、[68Ga]Ga-LW01186和[68Ga]Ga-LW02002使得能够在PET图像中清楚地可视化小鼠中皮下植入的人前列腺癌PC-3肿瘤异种移植物。离体生物分布研究显示[68Ga]Ga-LW01158在注射后1小时具有最高的肿瘤摄取(11.2±0.65%ID/g)和良好的肿瘤-背景摄取比。[68Ga]Ga-LW01158、[68Ga]Ga-LW01186和[68Ga]Ga-LW02002观察到相当的体内稳定性(76.5-80.7%在注射后15分钟在小鼠血浆中保持完整)。总之,Tle10替换,单独或与α-Me-Trp8或NMe-Gly11取代组合,在Ga-TacsBOMB2中产生的衍生物保留了良好的GRPR结合亲和力和体内稳定性。具有良好的肿瘤摄取和肿瘤背景成像对比度,[68Ga]Ga-LW01158有望用PET检测表达GRPR的病变。
    Gastrin-releasing peptide receptor (GRPR) is overexpressed in various cancers and is a promising target for cancer diagnosis and therapy. However, the high pancreas uptake and/or metabolic instability observed for most reported GRPR-targeted radioligands might limit their clinical applications. Our group recently reported a GRPR-targeted antagonist tracer, [68Ga]Ga-TacsBOMB2 ([68Ga]Ga-DOTA-Pip-D-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Leu13ψThz14-NH2), which showed a minimal pancreas uptake in a preclinical mouse model. In this study, we synthesized four derivatives with unnatural amino acid substitutions (Tle10-derived Ga-LW01158, NMe-His12-derived Ga-LW01160, α-Me-Trp8- and Tle10-derived Ga-LW01186, and Tle10- and N-Me-Gly11-derived Ga-LW02002) and evaluated their potential for detecting GRPR-expressing tumors with positron emission tomography (PET). The binding affinities (Ki(GRPR)) of Ga-LW01158, Ga-LW01160, Ga-LW01186, and Ga-LW02002 were 5.11 ± 0.47, 187 ± 17.8, 6.94 ± 0.95, and 11.0 ± 0.39 nM, respectively. [68Ga]Ga-LW01158, [68Ga]Ga-LW01186, and [68Ga]Ga-LW02002 enabled clear visualization of subcutaneously implanted human prostate cancer PC-3 tumor xenografts in mice in PET images. Ex vivo biodistribution studies showed that [68Ga]Ga-LW01158 had the highest tumor uptake (11.2 ± 0.65 %ID/g) and good tumor-to-background uptake ratios at 1 h post-injection. Comparable in vivo stabilities were observed for [68Ga]Ga-LW01158, [68Ga]Ga-LW01186, and [68Ga]Ga-LW02002 (76.5-80.7% remaining intact in mouse plasma at 15 min post-injection). In summary, the Tle10 substitution, either alone or combined with α-Me-Trp8 or NMe-Gly11 substitution, in Ga-TacsBOMB2 generates derivatives that retained good GRPR binding affinity and in vivo stability. With good tumor uptake and tumor-to-background imaging contrast, [68Ga]Ga-LW01158 is promising for detecting GRPR-expressing lesions with PET.
    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
    每个肿瘤都有自己独特的分子身份。治疗,因此,应该针对这种独特的癌症表型。Theragnostics使用相同的化合物进行靶向成像和治疗,放射性标记到适当的放射性核素,分别。胃泌素释放肽受体(GRPR)在前列腺癌中过度表达,和放射性标记的GRPR拮抗剂在分期和生化复发方面显示出很高的诊断性能。已经在临床前开发了几种针对GRPR的治疗无关化合物。他们正在转化为诊所,有4项临床试验招募GRPR表达肿瘤的参与者。
    Each tumor has its own distinctive molecular identity. Treatment, therefore, should be tailored to this unique cancer phenotype. Theragnostics uses the same compound for targeted imaging and treatment, radiolabeled to an appropriate radionuclide, respectively. Gastrin-releasing peptide receptors (GRPRs) are overexpressed in prostate cancer, and radiolabeled GRPR antagonists have shown high diagnostic performance at staging and biochemical recurrence. Several GRPR-targeting theragnostic compounds have been developed preclinically. Their translation into clinics is underway with 4 clinical trials recruiting participants with GRPR-expressing 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:在各种实体瘤中过表达,胃泌素释放肽受体(GRPR)是一个很有前途的癌症成像标记和治疗靶点。尽管由于潜在的副作用较少,拮抗剂对于GRPR靶向的放射性药物的开发是优选的,激动剂的内化可能导致更长的肿瘤保留和更好的治疗效果。在这项研究中,我们系统地研究了非天然氨基酸取代,以提高体内稳定性和肿瘤摄取以前报道的GRPR靶向激动剂示踪剂,[68Ga]Ga-TacBOMB2(68Ga-DOTA-Pip-D-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Leu13-Thz14-NH2)。
    结果:对Gln7、Trp8、Ala9、Val10、Gly11和His12进行单独或组合的非天然氨基酸置换。在25个非天然氨基酸取代中,tert-Leu10(Tle10)和NMe-His12取代被鉴定为优选的修饰,尤其是在组合中。与先前报道的[68Ga]Ga-TacBOMB2相比,Tle10和NMe-His12衍生的[68Ga]Ga-LW01110显示出保留的激动剂特性和改善的GRPR结合亲和力(Ki=7.62vs1.39nM),体内稳定性(注射后15分钟,小鼠血浆中12.7%完整示踪剂为89.0%)和肿瘤摄取(注射后1小时,5.95vs16.6%ID/g)。
    结论:非天然氨基酸取代是提高基于肽的放射性药物的体内稳定性和肿瘤摄取的有效策略。具有出色的肿瘤摄取和肿瘤背景对比,[68Ga]Ga-LW01110有望用PET检测表达GRPR的癌症病变。由于激动剂可导致与受体结合后的内化和可预见的长期肿瘤滞留,我们优化的GRPR靶向序列,[Tle10,NMe-His12,Thz14]孟买(7-14),是用于设计GRPR靶向放射治疗剂的有前途的模板。
    BACKGROUND: Overexpressed in various solid tumors, gastrin-releasing peptide receptor (GRPR) is a promising cancer imaging marker and therapeutic target. Although antagonists are preferable for the development of GRPR-targeted radiopharmaceuticals due to potentially fewer side effects, internalization of agonists may lead to longer tumor retention and better treatment efficacy. In this study, we systematically investigated unnatural amino acid substitutions to improve in vivo stability and tumor uptake of a previously reported GRPR-targeted agonist tracer, [68Ga]Ga-TacBOMB2 (68Ga-DOTA-Pip-D-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Leu13-Thz14-NH2).
    RESULTS: Unnatural amino acid substitutions were conducted for Gln7, Trp8, Ala9, Val10, Gly11 and His12, either alone or in combination. Out of 25 unnatural amino acid substitutions, tert-Leu10 (Tle10) and NMe-His12 substitutions were identified to be preferable modifications especially in combination. Compared with the previously reported [68Ga]Ga-TacBOMB2, the Tle10 and NMe-His12 derived [68Ga]Ga-LW01110 showed retained agonist characteristics and improved GRPR binding affinity (Ki = 7.62 vs 1.39 nM), in vivo stability (12.7 vs 89.0% intact tracer in mouse plasma at 15 min post-injection) and tumor uptake (5.95 vs 16.6 %ID/g at 1 h post-injection).
    CONCLUSIONS: Unnatural amino acid substitution is an effective strategy to improve in vivo stability and tumor uptake of peptide-based radiopharmaceuticals. With excellent tumor uptake and tumor-to-background contrast, [68Ga]Ga-LW01110 is promising for detecting GRPR-expressing cancer lesions with PET. Since agonists can lead to internalization upon binding to receptors and foreseeable long tumor retention, our optimized GRPR-targeted sequence, [Tle10,NMe-His12,Thz14]Bombesin(7-14), is a promising template for use for the design of GRPR-targeted radiotherapeutic agents.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial
    目的:低级别神经胶质瘤(LGG)是一组浸润性生长的神经胶质脑肿瘤,其特征是复杂的肿瘤内异质性和与非肿瘤组织的细微视觉外观差异,这可能导致病理组织采样错误。尽管5-ALA荧光一直是在手术过程中观察胶质瘤的重要方法,由于灵敏度低,其有效性在LGG的情况下是有限的。因此,我们开发了一种针对神经胶质瘤细胞中胃泌素释放肽受体(GRPR)的新型PET/NIR双模态图像探针,以增强肿瘤可视化并提高采样的准确性。方法:前瞻性,非随机化,单中心可行性临床试验(NCT03407781)于2016年10月21日至2018年8月17日在转诊中心进行.连续招募包括怀疑患有LGGs的患者,并认为适合手术切除的候选人。第1组包括10名患者,他们接受了术前68Ga-IRDye800CW-BBNPET/MRI评估,然后进行了术中荧光引导手术。第2组包括42例接受IRDye800CW-BBN荧光引导手术的患者。主要终点是术前PET成像对术中荧光的预测价值以及荧光引导采样的敏感性和特异性。结果:39例患者被纳入终点的深入分析,有25个(64.1%)表现出可见的荧光,14人(35.9%)没有。与MRI增强相比,术前PET阳性摄取在预测术中荧光方面表现出更高的准确性(100%[10/10]与87.2%[34/39])。在手术期间收集总共125个样品。与病理学相比,主观荧光强度在鉴定WHOIII级样本方面的敏感性为88.6%,特异性为88.2%.对于WHOII级样本,荧光检测的灵敏度和特异度分别为54.7%和88.2%,分别。结论:这项研究证明了新型双模态成像技术在LGG手术中通过相同的分子受体进行术前和术中靶向成像的可行性。与LGG患者的5-ALA相比,PET/NIR双模态探针有望在荧光引导手术中进行术前手术计划,并在引导肿瘤采样方面提供更高的准确性。
    Purpose: Lower-grade gliomas (LGGs) are a group of infiltrative growing glial brain tumors characterized by intricate intratumoral heterogeneity and subtle visual appearance differences from non-tumor tissue, which can lead to errors in pathologic tissue sampling. Although 5-ALA fluorescence has been an essential method for visualizing gliomas during surgery, its effectiveness is limited in the case of LGGs due to low sensitivity. Therefore, we developed a novel PET/NIR dual-modality image probe targeting gastrin-releasing peptide receptor (GRPR) in glioma cells to enhance tumor visualization and improve the accuracy of sampling. Methods: A prospective, non-randomized, single-center feasibility clinical trial (NCT03407781) was conducted in the referral center from October 21, 2016, to August 17, 2018. Consecutive enrollment included patients suspected of having LGGs and considered suitable candidates for surgical removal. Group 1 comprised ten patients who underwent preoperative 68Ga-IRDye800CW-BBN PET/MRI assessment followed by intraoperative fluorescence-guided surgery. Group 2 included 42 patients who underwent IRDye800CW-BBN fluorescence-guided surgery. The primary endpoints were the predictive value of preoperative PET imaging for intraoperative fluorescence and the sensitivity and specificity of fluorescence-guided sampling. Results: Thirty-nine patients were included in the in-depth analysis of endpoints, with 25 (64.1%) exhibiting visible fluorescence, while 14 (35.9%) did not. The preoperative positive PET uptake exhibited a greater accuracy in predicting intraoperative fluorescence compared to MRI enhancement (100% [10/10] vs. 87.2% [34/39]). A total of 125 samples were harvested during surgery. Compared with pathology, subjective fluorescence intensity showed a sensitivity of 88.6% and a specificity of 88.2% in identifying WHO grade III samples. For WHO grade II samples, the sensitivity and specificity of fluorescence were 54.7% and 88.2%, respectively. Conclusion: This study has demonstrated the feasibility of the novel dual-modality imaging technique for integrated pre- and intraoperative targeted imaging via the same molecular receptor in surgeries for LGGs. The PET/NIR dual-modality probe exhibits promise for preoperative surgical planning in fluorescence-guided surgery and provides greater accuracy in guiding tumor sampling compared to 5-ALA in patients with LGGs.
    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
    尽管肿瘤学取得了巨大的发展,前列腺癌仍然是一种使人衰弱的恶性肿瘤.解决这一问题的最有希望的方法之一是利用纳米医学的进步与完善的核医学和放射疗法相结合。按照这个想法,我们已经开发了基于聚丙烯酸的电子束合成纳米凝胶的放射性同位素纳米载体平台。我们开发了一个功能化协议,显示缀合在靶向配体-蛙皮素衍生物中非常高(>97%)的效率。这种工程肽可以结合在前列腺癌细胞中过度表达的胃泌素释放肽受体;此外,它带有放射性同位素螯合部分。我们的纳米平台在体外表现出非常有希望的性能;放射性标记的纳米载体在标记缓冲液和人血清中保持>90%的高放射化学纯度长达14天。靶向纳米载体的应用也允许PC-3前列腺癌细胞的有效和特异性摄取,4小时后高达近30%,与无载体的放射性标记的肽相比,这是统计学上显著的改进。尽管我们的系统需要进一步研究才能在体内获得更有希望的结果,我们的研究代表了放射医学领域的重要进展,这是导致去势抵抗性前列腺癌有效治疗的众多步骤之一.
    Despite the tremendous development of oncology, prostate cancer remains a debilitating malignancy. One of the most promising approaches to addressing this issue is to exploit the advancements of nanomedicine in combination with well-established nuclear medicine and radiotherapy. Following this idea, we have developed a radioisotope nanocarrier platform of electron-beam-synthesized nanogels based on poly(acrylic acid). We have developed a functionalization protocol, showing the very high (>97%) efficiency of the conjugation in targeting a ligand-bombesin derivative. This engineered peptide can bind gastrin-releasing peptide receptors overexpressed in prostate cancer cells; moreover, it bears a radioisotope-chelating moiety. Our nanoplatform exhibits very promising performance in vitro; the radiolabeled nanocarriers maintained high radiochemical purity of >90% in both the labeling buffer and human serum for up to 14 days. The application of the targeted nanocarrier allowed also effective and specific uptake in PC-3 prostate cancer cells, up to almost 30% after 4 h, which is a statistically significant improvement in comparison to carrier-free radiolabeled peptides. Although our system requires further studies for more promising results in vivo, our study represents a vital advancement in radionanomedicine-one of many steps that will lead to effective therapy for castration-resistant prostate cancer.
    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
    胃泌素释放肽受体(GRPr)在各种癌症类型中过度表达,包括前列腺癌和乳腺癌,使其成为分子成像和治疗的有吸引力的目标。在这项工作中,我们设计了一种包含金属螯合剂NODIA-Me的新型GRPr拮抗探针。这种基于1,4,7-三氮杂环壬烷的螯合剂由于其中性甲基咪唑臂而形成带正电荷的金属络合物。因为GRPr缀合物的N-末端处的正电荷负责高受体亲和力,如当前的金标准DOTA-RM2所例示的,所以我们研究了带正电荷的放射性金属络合物是否可以用作药代动力学修饰剂以也产生高亲和力GRPr缀合物。在这方面,生物缀合物NODIA-Me-Ahx-JMV594通过固相肽合成和基于溶液的反应的组合以94%的产率制备。放射性标记提供了68Ga标记的缀合物,其放射化学产率>95%,放射化学纯度>98%,平均摩尔活性为Am〜17MBqnmol-1。无金属和69/71Ga标记的缀合物的竞争性GRPr亲和力被确定为IC50=0.41±0.06和1.45±0.06nM,分别。无金属GRPr拮抗剂DOTA-RM2及其相应的69/71Ga复合物的IC50值为1.42±0.07和0.98±0.19nM,分别。携带GRPr()PC-3肿瘤的小鼠的小动物PET成像显示,肿瘤和胰腺中作为GRPr表达水平高的器官的放射性积累。这些发现得到了相应的离体生物分布数据的证实,其中肿瘤和胰腺表现出最高的放射性积累。通过共同注射过量的NODIA-Me-Ahx-JMV594,肿瘤和GRPr(+)器官的摄取显着降低,确认特异性受体介导的摄取。使用68Ga标记的NODIA-Me-Ahx-JMV594通过PET成像清楚地显示了女性乳腺癌患者的雌激素受体阳性肿瘤。总结一下,Ga(NODIA-Me)复合物诱导的缀合物N末端的正电荷导致与有效拮抗剂DOTA-RM2相当的高GRPr亲和力。缀合NODIA-Me-Ahx-JMV594是用于GRPr肿瘤成像的有前途的探针,值得在更大的患者队列中以及与其他放射性金属的组合进行进一步评估。
    The gastrin-releasing peptide receptor (GRPr) is overexpressed in various cancer types including prostate and breast carcinomas, making it an attractive target for molecular imaging and therapy. In this work, we designed a novel GRPr antagonistic probe comprising metal chelator NODIA-Me. This 1,4,7-triazacyclononane-based chelator forms positively charged metal complexes due to its neutral methylimidazole arms. Because a positive charge at the N-terminus of GRPr conjugates is responsible for high receptor affinity as exemplified by the current gold standard DOTA-RM2, we investigated if a positively charged radiometal complex can be used as a pharmacokinetic modifier to also produce high-affinity GRPr conjugates. In this respect, the bioconjugate NODIA-Me-Ahx-JMV594 was prepared by a combination of solid-phase peptide synthesis and solution-based reactions in a 94% yield. Radiolabeling provided the 68Ga-labeled conjugate in radiochemical yields of >95% and radiochemical purities of >98% with mean molar activities of Am ∼17 MBq nmol-1. The competitive GRPr affinity of the metal-free and 69/71Ga-labeled conjugate was determined to be IC50 = 0.41 ± 0.06 and 1.45 ± 0.06 nM, respectively. The metal-free GRPr antagonist DOTA-RM2 and its corresponding 69/71Ga complex had IC50 values of 1.42 ± 0.07 and 0.98 ± 0.19 nM, respectively. Small-animal PET imaging of mice bearing GRPr(+) PC-3 tumors revealed high radioactivity accumulation in the tumors and in the pancreas as an organ with high levels of GRPr expression. These findings were corroborated by the corresponding ex vivo biodistribution data, in which the tumors and the pancreas exhibited the highest radioactivity accumulation. By coinjection of an excess of NODIA-Me-Ahx-JMV594, uptake in the tumors and GRPr(+) organs was significantly reduced, confirming specific receptor-mediated uptake. The estrogen receptor-positive tumor of a female breast cancer patient was clearly visualized by PET imaging using 68Ga-labeled NODIA-Me-Ahx-JMV594. To summarize, the positive charge at the N-terminus of the conjugate induced by the Ga(NODIA-Me) complex resulted in high GRPr affinity comparable to that of the potent antagonist DOTA-RM2. The conjugate NODIA-Me-Ahx-JMV594 is a promising probe for imaging of GRPr tumors that warrants further evaluation in larger patient cohorts as well as in combination with other radiometals.
    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