Radioimmunotherapy

放射免疫疗法
  • 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
    免疫PET的组合-其中抗体(Ab)用同位素标记,用于PET成像和放射免疫治疗(RIT),使用相同的抗体和治疗同位素,在癌症管理方面具有显著优势。ImmunoPET允许抗原表达的非侵入性成像,这有助于患者选择后续的放射免疫治疗。它还有助于评估肿瘤对治疗的反应,允许必要时进行治疗调整。此外,免疫PET提供了关键的药代动力学数据,包括抗体生物分布和清除率,这对于剂量测定计算和治疗方案优化至关重要。仍有挑战需要克服。鉴定在癌细胞上选择性表达而在正常组织上最低限度表达的适当靶抗原仍然是降低脱靶毒性的主要障碍。此外,优化放射性标记抗体的药代动力学以最大化肿瘤摄取和最小化正常组织摄取是至关重要的,尤其是肝脏和肾脏等重要器官。这种方法通过利用单克隆抗体的特异性和放射的细胞毒性作用,提供了靶向和个性化癌症治疗的潜力,具有降低的全身毒性。然而,需要进一步的研究来应对剩余的挑战,并优化这些技术的临床应用.
    The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
    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
    细胞背包在治疗和诊断应用中都具有巨大的潜力,这使得进一步探索它们与宿主细胞的相互作用变得至关重要。目前的证据表明,背包可以诱导持续的免疫反应。我们最初的目标是在背包中加入模型抗原,以促进树突状细胞成熟并促进抗原呈递,从而诱导免疫反应。然而,我们意外地发现,装载抗原的背包和空背包都表现出相当的诱导树突状细胞成熟的能力,导致T细胞增殖几乎相同的效力。我们的机械研究表明,背包的附着通过打开PIEZO1机械离子通道在树突状细胞上诱导机械力。这种相互作用导致细胞内细胞骨架的重塑,并促进树突状细胞产生I型干扰素。因此,机械免疫驱动的树突状细胞背包,当与放射治疗相结合时,诱导强大的抗肿瘤作用。这项研究提出了一种利用机械转导来增强联合免疫治疗策略的途径。可能导致该领域的突破性进展。
    Cell backpacks present significant potential in both therapeutic and diagnostic applications, making it essential to further explore their interactions with host cells. Current evidence indicates that backpacks can induce sustained immune responses. Our original objective was to incorporate a model antigen into the backpacks to promote dendritic cell maturation and facilitate antigen presentation, thereby inducing immune responses. However, we unexpectedly discovered that both antigen-loaded backpacks and empty backpacks demonstrated comparable abilities to induce dendritic cell maturation, resulting in nearly identical potency in T-cell proliferation. Our mechanistic studies suggest that the attachment of backpacks induces mechanical forces on dendritic cells via opening the PIEZO1 mechanical ion channel. This interaction leads to the remodeling of the intracellular cytoskeleton and facilitates the production of type I interferons by dendritic cells. Consequently, the mechano-immune-driven dendritic cell backpacks, when combined with radiotherapy, induce a robust antitumor effect. This research presents an avenue for leveraging mechanotransduction to enhance combination immunotherapeutic strategies, potentially leading to groundbreaking advancements in the field.
    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
    免疫抑制细胞浸润到乳腺肿瘤微环境(TME)与抑制的效应T细胞(Teff)反应有关,加速肿瘤生长,和不良的临床结果。我们小组和其他人的先前研究发现,免疫抑制性骨髓来源的抑制细胞(MDSC)和调节性T细胞(Tregs)的浸润是原位claudin低肿瘤模型中免疫功能障碍的关键贡献者,限制过继性细胞疗法的功效。然而,目前缺乏在TME中靶向这些细胞的方法。为了克服这个障碍,聚合物胶束纳米颗粒(PMNP)用于共同递送激活Toll样受体7和8(TLR7/8)并抑制PI3Kδ(PI3Kδ)的小分子药物。TLR7/8激动剂和PI3K抑制剂对TME的免疫调节导致1型巨噬细胞极化,减少了MDSC的积累,选择性地减少了TME中的组织驻留Treg,同时增强T细胞和B细胞的适应性免疫反应。与单独的局部放射疗法相比,PMNP显着增强了带有原位claudin低肿瘤的小鼠的局部放射疗法(RT)的抗肿瘤活性。一起来看,这些数据表明,RT联合纳米配制的免疫刺激剂减少了免疫抑制性TME,导致肿瘤消退.这些发现为这种方法的临床研究奠定了基础。
    Infiltration of immunosuppressive cells into the breast tumor microenvironment (TME) is associated with suppressed effector T cell (Teff) responses, accelerated tumor growth, and poor clinical outcomes. Previous studies from our group and others identified infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) as critical contributors to immune dysfunction in the orthotopic claudin-low tumor model, limiting the efficacy of adoptive cellular therapy. However, approaches to target these cells in the TME are currently lacking. To overcome this barrier, polymeric micellular nanoparticles (PMNPs) were used for the co-delivery of small molecule drugs activating Toll-like receptors 7 and 8 (TLR7/8) and inhibiting PI3K delta (PI3Kδ). The immunomodulation of the TME by TLR7/8 agonist and PI3K inhibitor led to type 1 macrophage polarization, decreased MDSC accumulation and selectively decreased tissue-resident Tregs in the TME, while enhancing the T and B cell adaptive immune responses. PMNPs significantly enhanced the anti-tumor activity of local radiation therapy (RT) in mice bearing orthotopic claudin-low tumors compared to RT alone. Taken together, these data demonstrate that RT combined with a nanoformulated immunostimulant diminished the immunosuppressive TME resulting in tumor regression. These findings set the stage for clinical studies of this approach.
    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
    放射疗法发挥免疫刺激和免疫抑制作用,都是当地的,在辐射的肿瘤微环境中,系统的,在辐射场之外。受临床前数据的启发,显示放疗和免疫检查点抑制剂之间的协同作用,基于以下假设启动了多项临床试验:放疗和免疫检查点抑制剂联合治疗可刺激强大的全身免疫反应并改善临床结局.然而,尽管早期乐观,在治疗和转移环境中的放射免疫疗法试验收效甚微.在这篇评论中,我们总结了放疗的免疫刺激作用,为放疗和免疫检查点抑制剂的联合试验提供了理论基础。我们还讨论了纳入放疗和免疫检查点抑制剂的临床试验的结果,并在放疗的免疫抑制作用的背景下检查这些试验的成功。最后,我们强调了缓解放疗诱导的免疫抑制的目标,目的是增强放疗和免疫检查点抑制剂的联合作用。
    Radiotherapy exerts immunostimulatory and immunosuppressive effects, both locally, within the irradiated tumour microenvironment, and systemically, outside the radiation field. Inspired by preclinical data that showed synergy between radiotherapy and immune checkpoint inhibitors, multiple clinical trials were initiated with the hypothesis that combined treatment with radiotherapy and immune checkpoint inhibitors could stimulate a robust systemic immune response and improve clinical outcomes. However, despite early optimism, radioimmunotherapy trials in the curative and metastatic settings have met with little success. In this Review, we summarise the immunostimulatory effects of radiotherapy that provided the theoretical basis for trials of combination radiotherapy and immune checkpoint inhibitors. We also discuss findings from clinical trials incorporating radiotherapy and immune checkpoint inhibitors and examine the success of these trials in the context of the immunosuppressive effects of radiotherapy. We conclude by highlighting targets for relieving radiotherapy-induced immunosuppression with the goal of enhancing the combined effects of radiotherapy and immune checkpoint inhibitors.
    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
    放射免疫疗法(RIT)是一种新颖且有前景的癌症治疗方法,随着对RIT抗体选择的持续研究,放射性核素,治疗方案,并使患者群体受益。当我们深入研究肿瘤生物学机制时,需要更深入地探索RIT如何影响肿瘤组织,以提供改善临床治疗结果和患者预后的新途径.我们用碘131(131I)标记抗PD-L1单克隆抗体阿替珠单抗,用SephadexG-25介质凝胶过滤树脂分离纯化标记的mAb,并测试了产品的稳定性。我们通过分析其体内生物分布并进行SPECT成像来检测131I-PD-L1mAb的体内活性,然后设置不同的治疗组以研究131I-atezolizumab对荷瘤小鼠存活的影响。蛋白质印迹,实时定量PCR,免疫组化检测Caspase8和Nlrp3在肿瘤中的表达水平。TUNEL荧光染色检测肿瘤细胞凋亡。放射性药物分子探针131I-atezolizumab显示出高稳定性和体内生物活性。所采用的治疗方案对荷瘤小鼠的存活有积极影响。131I内照射上调肿瘤中的Caspase8,并最终通过激活凋亡途径抑制实体瘤生长。我们还发现NLRP3的表达显着增加,在焦凋亡途径中起重要作用,在肿瘤中。总之,我们的数据表明,放射性药物联合免疫治疗通过调节相关的生物学途径影响肿瘤组织,从而达到更好的抗肿瘤效果相比单一疗法,并提供新的见解,促进更好的患者预后和联合治疗策略。
    Radioimmunotherapy (RIT) is a novel and promising cancer treatment method, with ongoing research focusing on RIT antibody selection, radionuclides, treatment options, and benefited patient groups. As we dive into the mechanisms of tumor biology, a deeper exploration of how RIT affects tumor tissue is needed to provide new ways to improve clinical treatment outcome and patient prognosis. We labeled the anti-PD-L1 monoclonal antibody atezolizumab with iodine-131 (131I), separated and purified the labeled mAb with Sephadex G-25 medium gel filtration resin, and tested product stability. We detected the in vivo activity of 131I-PD-L1 mAb by analyzing its in vivo biodistribution and performing SPECT imaging and then set different treatment groups to study the effect of 131I-atezolizumab on the survival of tumor-bearing mice. Western blot, real-time quantitative PCR, and immunohistochemistry were used to detect the expression level of Caspase8 and Nlrp3 in tumor. TUNEL fluorescence staining was used to detect the apoptosis in the tumor. The radiopharmaceutical molecular probe 131I-atezolizumab showed high stability and in vivo biological activity. The treatment regimen adopted had a positive effect on the survival of tumor-bearing mice. 131I internal irradiation upregulated Caspase8 in tumor and ultimately inhibited solid tumor growth by activating apoptosis pathways. We also found a significant increase in the expression of NLRP3, which plays an important role in the pyroptosis pathway, in tumor. In summary, our data demonstrated that radiopharmaceuticals combined with immunotherapy affected tumor tissue by modulating relevant biological pathways, thereby achieving better antitumor effects compared with single therapy and providing new insights for promoting better patient prognosis and combination treatment strategies.
    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
    大多数癌症免疫治疗的实验方法都是针对荷瘤小鼠中相对较小的肿瘤进行测试的。因为在大多数情况下,晚期癌症对治疗有抵抗力。在这项研究中,我们询问是否通过合理设计的联合放射免疫疗法(CRI)方案可以根除晚期小鼠肿瘤.
    CRI包括局部放疗,肿瘤内IL-12,缓释全身IL-2和抗CTLA-4抗体。CRI对几种弱免疫原性和免疫原性小鼠肿瘤,包括B78黑色素瘤的治疗作用,评估了MC38和CT26结肠癌和9464D神经母细胞瘤。进行免疫细胞消耗和流式细胞术分析以确定抗肿瘤作用的机制。
    体积为2,000mm3或更大的肿瘤被CRI根除。肿瘤的流动分析显示CRI后T调节(Treg)细胞减少和CD8/Treg比率增加。治疗的肿瘤快速缩小不需要T细胞,而T细胞参与对抗远处肿瘤的全身效应。治愈的小鼠产生了免疫记忆。
    这些发现强调了合理设计的联合免疫疗法方案甚至可以有效对抗大,晚期肿瘤。
    UNASSIGNED: The majority of experimental approaches for cancer immunotherapy are tested against relatively small tumors in tumor-bearing mice, because in most cases advanced cancers are resistant to the treatments. In this study, we asked if even late-stage mouse tumors can be eradicated by a rationally designed combined radio-immunotherapy (CRI) regimen.
    UNASSIGNED: CRI consisted of local radiotherapy, intratumoral IL-12, slow-release systemic IL-2 and anti- CTLA-4 antibody. Therapeutic effects of CRI against several weakly immunogenic and immunogenic mouse tumors including B78 melanoma, MC38 and CT26 colon carcinomas and 9464D neuroblastoma were evaluated. Immune cell depletion and flow cytometric analysis were performed to determine the mechanisms of the antitumor effects.
    UNASSIGNED: Tumors with volumes of 2,000 mm3 or larger were eradicated by CRI. Flow analyses of the tumors revealed reduction of T regulatory (Treg) cells and increase of CD8/Treg ratios following CRI. Rapid shrinkage of the treated tumors did not require T cells, whereas T cells were involved in the systemic effect against the distant tumors. Cured mice developed immunological memory.
    UNASSIGNED: These findings underscore that rationally designed combination immunotherapy regimens can be effective even against large, late-stage 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
    表皮生长因子受体(EGFR)蛋白在一系列恶性肿瘤中高度表达。尽管针对EGFR的治疗干预措施已经在癌症患者中产生了治疗反应,副作用是常见的,因为野生型EGFR的正常组织表达。我们开发了一种用225Ac标记的新型肿瘤特异性抗EGFR嵌合抗体ch806,并在胶质母细胞瘤和结直肠癌的鼠模型中评估了其体外特性和治疗效果。方法:使用不同的螯合剂制备225Ac-ch806,产生[225Ac]Ac-macropa-tzPEG3Sq-ch806和[225Ac]Ac-DOTA-dhPzPEG4-ch806。放射化学产率,纯度,表观比活度,定量测定225Ac-ch806的血清稳定性。检查了体外细胞杀伤作用。在具有U87MG的小鼠中研究了225Ac-ch806的生物分布和治疗功效。de2-7和DiFi肿瘤。治疗后肿瘤的药效学分析,包括γH2AX的DNA双链断裂免疫荧光,以及增殖的免疫组织化学,细胞周期停滞,和凋亡。结果:[225Ac]Ac-macropa-tzPEG3Sq-ch806的放射化学产率超过[225Ac]Ac-DOTA-dhPzPEG4-ch806,纯度,表观比活度,和血清稳定性。[225Ac]Ac-macropa-tzPEG3Sq-ch806因此用于体外和体内研究。它显示了一个重要的,具体,和剂量依赖性的体外细胞杀伤作用U87MG。de2-7细胞。225Ac-ch806在正常组织中也显示出高肿瘤摄取和最小摄取。225Ac-ch806在两种U87MG中均显著抑制肿瘤生长并延长存活。de2-7和DiFi模型。与对照相比,在225Ac-ch806处理的肿瘤中观察到增强的γH2AX染色。Ki-67表达降低在所有225Ac-ch806处理的肿瘤中是明显的。在U87MG中显示p21和裂解的半胱天冬酶3的表达增加。de2-7和DiFi肿瘤用225Ac-ch806治疗。结论:在胶质母细胞瘤和结直肠肿瘤模型中,225Ac-ch806通过诱导双链断裂显著抑制肿瘤生长,从而限制癌细胞增殖,同时诱导细胞周期停滞和凋亡。这些发现强调了225Ac-ch806作为表达EGFR的实体瘤的潜在疗法的潜在临床适用性。
    The epidermal growth factor receptor (EGFR) protein is highly expressed in a range of malignancies. Although therapeutic interventions directed toward EGFR have yielded therapeutic responses in cancer patients, side effects are common because of normal-tissue expression of wild-type EGFR. We developed a novel tumor-specific anti-EGFR chimeric antibody ch806 labeled with 225Ac and evaluated its in vitro properties and therapeutic efficacy in murine models of glioblastoma and colorectal cancer. Methods: 225Ac-ch806 was prepared using different chelators, yielding [225Ac]Ac-macropa-tzPEG3Sq-ch806 and [225Ac]Ac-DOTA-dhPzPEG4-ch806. Radiochemical yield, purity, apparent specific activity, and serum stability of 225Ac-ch806 were quantified. In vitro cell killing effect was examined. The biodistribution and therapeutic efficacy of 225Ac-ch806 were investigated in mice with U87MG.de2-7 and DiFi tumors. Pharmacodynamic analysis of tumors after therapy was performed, including DNA double-strand break immunofluorescence of γH2AX, as well as immunohistochemistry for proliferation, cell cycle arrest, and apoptosis. Results: [225Ac]Ac-macropa-tzPEG3Sq-ch806 surpassed [225Ac]Ac-DOTA-dhPzPEG4-ch806 in radiochemical yield, purity, apparent specific activity, and serum stability. [225Ac]Ac-macropa-tzPEG3Sq-ch806 was therefore used for both in vitro and in vivo studies. It displayed a significant, specific, and dose-dependent in vitro cell-killing effect in U87MG.de2-7 cells. 225Ac-ch806 also displayed high tumor uptake and minimal uptake in normal tissues. 225Ac-ch806 significantly inhibited tumor growth and prolonged survival in both U87MG.de2-7 and DiFi models. Enhanced γH2AX staining was observed in 225Ac-ch806-treated tumors compared with controls. Reduced Ki-67 expression was evident in all 225Ac-ch806-treated tumors. Increased expression of p21 and cleaved caspase 3 was shown in U87MG.de2-7 and DiFi tumors treated with 225Ac-ch806. Conclusion: In glioblastoma and colorectal tumor models, 225Ac-ch806 significantly inhibited tumor growth via induction of double-strand breaks, thereby constraining cancer cell proliferation while inducing cell cycle arrest and apoptosis. These findings underscore the potential clinical applicability of 225Ac-ch806 as a potential therapy for EGFR-expressing solid 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
    这项研究调查了一种靶向肿瘤血管生成的新型放射免疫疗法策略。我们通过使用DOTA作为螯合剂,用放射性同位素177Lu标记抗三磷酸腺苷合酶(ATPS)单克隆抗体(mAb),开发了一种放射性药物复合物。177Lu-DOTA-ATPSmAb在血清中表现出高标记效率(99.0%)和稳定性。MKN-45癌细胞表现出最高的细胞摄取,可以被未标记的ATPSmAb特异性阻断。在老鼠身上,177Lu-DOTA-ATPSmAb在肿瘤中显著积累,第7天的肿瘤摄取为16.0±1.5%ID/g。177Lu-DOTA-ATPSmAb处理以剂量依赖性方式显著降低MKN-45细胞的活力。在异种移植肿瘤模型中,这种放射免疫治疗策略导致显著的肿瘤生长抑制(82.8%).此外,177Lu-DOTA-ATPS单克隆抗体与舒尼替尼相结合,一种抗血管生成药物,增强舒尼替尼在小鼠模型中的治疗效果。我们的研究成功开发了177Lu-DOTA-ATPSmAb,靶向肿瘤血管的放射免疫治疗药物。这种方法证明了抑制肿瘤生长的重要前景,既可以作为单一疗法,也可以与其他抗癌药物联合使用。
    This study investigated a novel radioimmunotherapy strategy for targeting tumor angiogenesis. We developed a radiopharmaceutical complex by labeling an anti-adenosine triphosphate synthase (ATPS) monoclonal antibody (mAb) with the radioisotope 177Lu using DOTA as a chelating agent. 177Lu-DOTA-ATPS mAb demonstrated high labeling efficiency (99.0%) and stability in serum. MKN-45 cancer cells exhibited the highest cellular uptake, which could be specifically blocked by unlabeled ATPS mAb. In mice, 177Lu-DOTA-ATPS mAb accumulated significantly in tumors, with a tumor uptake of 16.0 ± 1.5%ID/g on day 7. 177Lu-DOTA-ATPS mAb treatment significantly reduced the viability of MKN-45 cells in a dose-dependent manner. In a xenograft tumor model, this radioimmunotherapy strategy led to substantial tumor growth inhibition (82.8%). Furthermore, combining 177Lu-DOTA-ATPS mAb with sunitinib, an anti-angiogenic drug, enhanced the therapeutic efficacy of sunitinib in the mouse model. Our study successfully developed 177Lu-DOTA-ATPS mAb, a radioimmunotherapy agent targeting tumor blood vessels. This approach demonstrates significant promise for inhibiting tumor growth, both as a single therapy and in combination with other anti-cancer drugs.
    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
    α-发射器211At在几个细胞直径内沉积大量能量,导致不可修复的DNA双链断裂,同时最大限度地减少脱靶毒性。我们研究了使用211At标记的抗CD45单克隆抗体(mAb)211At-CD45-B10作为犬白细胞抗原单倍体造血细胞移植的非清髓性预处理方案。方法:在第-3天,给17只健康犬注射0.50(n=14)或0.75(n=3)mg/kg剂量的用211At(8.436-23.199MBq[0.228-0.627mCi/kg])标记的抗CD45mAb。在第0天给予来自狗-白细胞-抗原-单倍体供体的外周血干细胞。通过聚合酶链反应测定法计算外周血嵌合状态,使用酶联免疫吸附测定和系列血液样品的放射性测量研究了放射免疫缀合物的血液清除率。结果:所有狗在第28天达到供体嵌合状态(范围,27%-100%)。造血植入率为100%,虽然雕刻耐久性是可变的。对于所研究的2mAb给药水平,观察到对血液的吸收剂量没有差异。中性粒细胞减少症(0-29细胞/μL),淋巴细胞减少症(36-130细胞/μL),观察到血小板减少(1.5-9×103/μL)并迅速恢复。与211At-CD45-B10相关的主要不良非血液学事件是轻度可逆性转氨酶。未见移植物抗宿主病。17只狗中有12只活了30天,供体嵌合体在3%至99%的范围内。结论:结果表明,211At-CD45-B10的非清髓性预处理可用于单倍体相合造血细胞移植,但植入量不同。
    The α-emitter 211At deposits a high amount of energy within a few cell diameters, resulting in irreparable DNA double-strand breaks while minimizing off-target toxicity. We investigated the use of the 211At-labeled anti-CD45 monoclonal antibody (mAb) 211At-CD45-B10 as a nonmyeloablative conditioning regimen for dog-leukocyte-antigen-haploidentical hematopoietic cell transplantation. Methods: Seventeen healthy dogs were injected with either a 0.50 (n = 14) or 0.75 (n = 3) mg/kg dose of anti-CD45 mAb labeled with 211At (8.436-23.199 MBq [0.228-0.627 mCi/kg]) on day -3. Peripheral blood stem cells from dog-leukocyte-antigen-haploidentical donors were given on day 0. Peripheral blood chimerism was calculated by polymerase chain reaction assays, and blood clearance of the radioimmunoconjugate was studied using enzyme-linked immunosorbent assay and radioactivity measurements of serial blood samples. Results: All dogs achieved donor chimerism by day 28 (range, 27%-100%). The hematopoietic engraftment rate was 100%, though engraftment durability was variable. No difference in absorbed dose to blood was seen for the 2 mAb dosing levels studied. Neutropenia (0-29 cells/μL), lymphocytopenia (36-130 cells/μL), and thrombocytopenia (1.5-9 × 103/μL) with prompt recovery were observed. The main adverse nonhematologic event related to 211At-CD45-B10 was mild reversible transaminitis. Graft-versus-host disease was not seen. Twelve of the 17 dogs survived over 30 d, with donor chimerism ranging from 3% to 99%. Conclusion: The results suggest that nonmyeloablative conditioning with 211At-CD45-B10 could be used in haploidentical hematopoietic cell transplantation though with variable engraftment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号