hu3F8

hu3F8
  • 文章类型: Journal Article
    目的:目前治疗骨肉瘤(OS)的预后较差,特别是对于有转移和复发的患者,强调迫切需要新的靶向治疗来提高生存率。靶向α-粒子疗法选择性地将细胞毒性有效载荷递送至具有识别肿瘤相关抗原的放射性标记的分子的肿瘤。我们最近证明了FDA批准的潜力,人源化抗GD2抗体,hu3F8,作为OS放射性药物成像的靶向递送载体。目前的研究旨在推进该系统用于OS的α粒子治疗。
    方法:hu3F8抗体用actinium-225进行放射性标记,并在OS和自发发生的OS的原位鼠异种移植物中评估[225Ac]Ac-DOTA-hu3F8的安全性和治疗效果。
    结果:在两种情况下都证明了显着的抗肿瘤活性,提高总体生存率。在鼠异种移植的情况下,生物发光成像显示,与未治疗组相比,肿瘤生长延迟了16-18天.结果在治疗后33天用磁共振成像进一步验证,验尸后显微计算机断层扫描和平面显微放射成像。组织学评估显示辐射诱导的肾毒性,表现为肾脏上皮细胞核肿大和暗示多倍体,提示放射性损伤后肾功能迅速恢复。两名犬科患者的治疗延迟了转移扩散的进展,总生存时间为211天和437天,超过记录转移的生存时间为111天和84天,分别。
    结论:这项研究强调了基于hu3F8的α-粒子治疗作为OS的一种有希望的治疗策略的潜力。
    OBJECTIVE: Current treatments for osteosarcoma (OS) have a poor prognosis, particularly for patients with metastasis and recurrence, underscoring an urgent need for new targeted therapies to improve survival. Targeted alpha-particle therapy selectively delivers cytotoxic payloads to tumors with radiolabeled molecules that recognize tumor-associated antigens. We have recently demonstrated the potential of an FDA approved, humanized anti-GD2 antibody, hu3F8, as a targeted delivery vector for radiopharmaceutical imaging of OS. The current study aims to advance this system for alpha-particle therapy of OS.
    METHODS: The hu3F8 antibody was radiolabeled with actinium-225, and the safety and therapeutic efficacy of the [225Ac]Ac-DOTA-hu3F8 were evaluated in both orthotopic murine xenografts of OS and spontaneously occurring OS in canines.
    RESULTS: Significant antitumor activity was proven in both cases, leading to improved overall survival. In the murine xenograft\'s case, tumor growth was delayed by 16-18 days compared to the untreated cohort as demonstrated by bioluminescence imaging. The results were further validated with magnetic resonance imaging at 33 days after treatment, and microcomputed tomography and planar microradiography post-mortem. Histological evaluations revealed radiation-induced renal toxicity, manifested as epithelial cell karyomegaly and suggestive polyploidy in the kidneys, suggesting rapid recovery of renal function after radiation damage. Treatment of the two canine patients delayed the progression of metastatic spread, with an overall survival time of 211 and 437 days and survival beyond documented metastasis of 111 and 84 days, respectively.
    CONCLUSIONS: This study highlights the potential of hu3F8-based alpha-particle therapy as a promising treatment strategy for OS.
    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: Preprint
    背景:由于用抗GD2单克隆抗体治疗神经母细胞瘤(NB)可在患有微小残留病的儿童中提供生存益处,并且我们的临床前研究表明,抗CD3x抗GD2双特异性抗体(GD2Bi)武装的T细胞(GD2BAT)对GD2细胞系具有高度细胞毒性,我们在复发/难治性患者中进行了I/II期研究,以确定GD2BAT的安全性并探讨其临床获益.方法:3+3剂量递增研究(NCT02173093)I期涉及9名可评估的NB患者(n=5),骨肉瘤(OST)(n=3),和促纤维增生性小圆细胞肿瘤(DSRCT)(n=1),每周两次输注GD2BAT,以40、80或160x106GD2BAT/kg/输注每日白细胞介素2(300,000IU/m2)和每周两次粒细胞-巨噬细胞集落刺激因子(250µg/m2)。试验的II期部分在160×106GD2BAT/kg/输注的剂量3水平的NB患者中进行,但未能招募计划数量的患者。结果:12例患者中的9例完成了治疗。没有剂量限制性毒性(DLT)。所有患者均出现轻度和可控制的细胞因子释放综合征(CRS),伴有2-3级发烧/寒战,头痛,在输注GD2BAT后72小时内偶尔出现低血压。GD2抗体相关疼痛在这项研究中并不显著。I期和有限II期患者的中位OS分别为18.0和31.2个月,分别,而合并OS为21.1个月.在患有NB的I期患者之一中,存在完全的骨髓应答,并且疾病总体稳定。12例II期患者中有10例可以评估反应:1例部分反应。另外三名患者被认为具有长期稳定疾病的临床益处。通过干扰素-γ(IFN-γ)Elispots测量,超过50%的可评估患者在GD2BAT后对GD2靶标显示出增强的免疫反应,Th1细胞因子,和/或趋化因子。结论:我们的研究证明了高达160×106个细胞/kg/输注GD2BAT的安全性。结合治疗后内源性免疫反应发展的证据,该数据支持在更大的II期临床试验中对GD2BAT的进一步研究.
    UNASSIGNED: Since treatment of neuroblastoma (NB) with anti-GD2 monoclonal antibodies provides a survival benefit in children with minimal residual disease and our preclinical study shows that anti-CD3 x anti-GD2 bispecific antibody (GD2Bi) armed T cells (GD2BATs) were highly cytotoxic to GD2+ cell lines, we conducted a phase I/II study in recurrent/refractory patients to establish safety and explore the clinical benefit of GD2BATs.
    UNASSIGNED: The 3+3 dose escalation study (NCT02173093) phase I involved 9 evaluable patients with NB (n=5), osteosarcoma (OST) (n=3), and desmoplastic small round cell tumors (DSRCT) (n=1) with twice weekly infusions of GD2BATs at 40, 80, or 160 x 106 GD2BATs/kg/infusion with daily interleukin 2 (300,000 IU/m2) and twice weekly granulocyte-macrophage colony stimulating factor (250 μg/m2). Phase II portion of the trial was conducted in patients with NB at the dose 3 level of 160 x 106 GD2BATs/kg/infusion but failed to enroll the planned number of patients.
    UNASSIGNED: Nine of 12 patients in the phase I completed therapy. There were no dose limiting toxicities (DLTs). All patients developed mild and manageable cytokine release syndrome (CRS) with grade 2-3 fevers/chills, headaches, and occasional hypotension up to 72 hours after GD2BAT infusions. GD2-antibody associated pain was not significant in this study. The median OS for patients in the Phase I and limited Phase II was 18.0 and 31.2 months, respectively, whereas the combined OS was 21.1 months. There was a complete bone marrow response with overall stable disease in one of the phase I patients with NB. Ten of 12 phase II patients were evaluable for response: 1 had partial response. Three additional patients were deemed to have clinical benefit with prolonged stable disease. More than 50% of evaluable patients showed augmented immune responses to GD2+ targets after GD2BATs as measured by interferon-gamma (IFN-γ) EliSpots, Th1 cytokines, and/or chemokines.
    UNASSIGNED: Our study demonstrated safety of up to 160 x 106 cells/kg/infusion of GD2BATs. Combined with evidence for the development of post treatment endogenous immune responses, this data supports further investigation of GD2 BATs in larger Phase II clinical 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
    骨肉瘤(OS)是儿童中最常诊断的骨癌,在过去的几十年中总体生存率几乎没有改善。二唾液酸神经节苷脂GD2在OS肿瘤上的高表面表达和在正常组织中的限制性表达使其成为抗OS放射性药物的理想靶标。由于人类和犬OS具有许多生物学和分子特征,犬科动物自发发生的OS一直是测试人类翻译的新成像和治疗方式的理想模型。在这项研究中,我们评估了人源化抗GD2抗体,hu3F8,作为一种潜在的递送载体,用于人和犬OS的靶向放射性药物成像。
    通过免疫组织化学和流式细胞术检查hu3F8与人和犬OS细胞和肿瘤的交叉反应性。hu3F8用铟-111放射性标记,并在荷瘤异种移植物小鼠中评估[111In]In-hu3F8的生物分布。在自发性OS犬科动物中测试了[111In]In-hu3F8对转移性OS的靶向能力。
    hu3F8以高结合亲和力与人和犬OS细胞以及犬OS肿瘤交叉反应。生物分布研究揭示[111In]In-hu3F8在肿瘤组织中的选择性摄取。自发性OS犬的SPECT/CT成像显示所有转移性病变中[111In]In-hu3F8的强烈摄取。免疫组织化学证实了放射性标记的hu3F8在骨和软病变中的广泛结合。
    这项研究证明了使用基于hu3F8的放射性药物成像在OS细胞和自发性OS犬肿瘤上靶向GD2的可行性。它以有针对性的方式提供成像有效载荷的能力支持hu3F8用于OS的精确成像和放射性药物治疗的潜在未来用途。
    Osteosarcoma (OS) is the most frequently diagnosed bone cancer in children with little improvement in overall survival in the past decades. The high surface expression of disialoganglioside GD2 on OS tumors and restricted expression in normal tissues makes it an ideal target for anti-OS radiopharmaceuticals. Since human and canine OS share many biological and molecular features, spontaneously occurring OS in canines has been an ideal model for testing new imaging and treatment modalities for human translation. In this study, we evaluated a humanized anti-GD2 antibody, hu3F8, as a potential delivery vector for targeted radiopharmaceutical imaging of human and canine OS.
    The cross-reactivity of hu3F8 with human and canine OS cells and tumors was examined by immunohistochemistry and flow cytometry. The hu3F8 was radiolabeled with indium-111, and the biodistribution of [111In]In-hu3F8 was assessed in tumor xenograft-bearing mice. The targeting ability of [111In]In-hu3F8 to metastatic OS was tested in spontaneous OS canines.
    The hu3F8 cross reacts with human and canine OS cells and canine OS tumors with high binding affinity. Biodistribution studies revealed selective uptake of [111In]In-hu3F8 in tumor tissue. SPECT/CT imaging of spontaneous OS canines demonstrated avid uptake of [111In]In-hu3F8 in all metastatic lesions. Immunohistochemistry confirmed the extensive binding of radiolabeled hu3F8 within both osseous and soft lesions.
    This study demonstrates the feasibility of targeting GD2 on OS cells and spontaneous OS canine tumors using hu3F8-based radiopharmaceutical imaging. Its ability to deliver an imaging payload in a targeted manner supports the utility of hu3F8 for precision imaging of OS and potential future use in radiopharmaceutical therapy.
    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
    高危神经母细胞瘤(HRNBL)的治疗包括多模式治疗,包括化疗,手术,放射治疗,清髓治疗,然后自体造血干细胞移植,和免疫疗法。GD2是一种二唾液酸神经节苷脂,在神经母细胞瘤细胞表面高表达,在正常组织上表达有限,这使得它成为免疫疗法的一个有吸引力的靶点。免疫疗法与鼠和嵌合抗GD2抗体制剂的组合在HRNBL患者中与标准疗法相比具有改善的结果。Naxitamab(Danyelza),完全人源化的抗GD2抗体,是在纪念斯隆·凯特琳癌症中心(MSKCC)开发的,以减轻与外来鼠和嵌合抗原不耐受有关的不良反应。I期和II期研究证明了纳西他单抗在复发/难治性(r/r)HRNBL患者中的耐受性和有效性,促使美国食品和药物管理局(FDA)于2020年批准了具有骨或骨髓受累的HRNBL。纳西他单抗的初步结果令人鼓舞;然而,需要进行未来的试验,以最大限度地提高药物耐受性,并阐明其在神经母细胞瘤治疗中的最佳作用,以及其他治疗策略.本文讨论了纳西他单抗联合粒细胞-巨噬细胞集落刺激因子(GM-CSF)治疗r/rHRNBL的应用。
    Therapy for high-risk neuroblastoma (HR NBL) is comprised of multimodal therapy including chemotherapy, surgery, radiation therapy, myeloablative therapy followed by autologous hematopoietic stem cell transplant, and immunotherapy. GD2 is a disialoganglioside that is highly expressed on the surface of neuroblastoma cells, with limited expression on normal tissues, which makes it an attractive target for immunologic therapy. The combination of immunotherapy with murine and chimeric anti-GD2 antibody formulations has improved outcomes compared with standard therapy in HR NBL patients. Naxitamab (Danyelza), a fully humanized anti-GD2 antibody, was developed at Memorial Sloan Kettering Cancer Center (MSKCC) to mitigate adverse reactions related to intolerance of foreign murine and chimeric antigens. Phase I and II studies demonstrating the tolerability and efficacy of naxitamab in patients with relapsed/refractory (r/r) HR NBL prompted its approval by the U.S. Food and Drug Administration (FDA) in 2020 for HR NBL with bone or bone marrow involvement. Initial outcomes with naxitamab are encouraging; however, future trials to maximize drug tolerance and elucidate its optimal role in neuroblastoma therapy in conjunction with other treatment strategies are needed. This review discusses the use of naxitamab in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment of r/r HR NBL.
    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
    The role of T cells in controlling human cancers is well known. Their success requires continued persistence in vivo and efficient trafficking to tumor sites, requirements shared by other effectors such as Natural Killer (NK) cells. To date, cytokine IL2 remains the only clinically approved cytokine therapy available to expand, maintain, and activate these effector lymphoid cells, but toxicities can be severe. Cytokine IL15 offers similar T cell proliferation and activation properties, but without the unwanted side-effects seen with IL2. Several IL15-cytokine fusion proteins have been developed to improve their in vivo function, typically exploiting the IL15Rα to complex with IL15, to extend serum half-life and increase affinity for IL15β receptor on immune cells. Here we describe a novel IL15 complex incorporating the full-length IL15Rα to complex with wild type IL15 to form spontaneous trimers of dimers (6 IL15 + 6 IL15Rα) during co-expression, resulting in a substantial increase in serum half-life and enhancement of in vivo cytokine effect on IgG or T cell engaging antibody-dependent cell-mediated cytotoxicities, when compared to alternative strategies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Fifty-seven stage 4 patients with refractory/relapsed neuroblastoma were enrolled in a phase I trial (Clinicaltrials.gov NCT01757626) using humanized anti-GD2 monoclonal antibody hu3F8 in combination with granulocyte-macrophage colony-stimulating factor. The influence of body weight and human anti-human antibody (HAHA) on the pharmacokinetics (PK) of hu3F8, and the effect of de novo anti-GD2 response on patient outcome were explored. Serum samples before hu3F8 infusion, and serially up to day 12 during treatment cycle #1, and at 5 min after each hu3F8 infusion for all subsequent cycles were collected. PK was analyzed using non-compartmental modeling. Immunogenicity was assayed by HAHA response, and vaccination effect by induced host anti-GD2 response measured periodically until disease progression or last followup. Progression-free and overall survival was estimated by the Kaplan-Meier method. Despite dosing being based on body weight, smaller patients had consistently lower area-under-the-curve and faster clearance over the 15 dose levels (0.9 to 9.6 mg/kg per treatment cycle) in this trial. Positive HAHA, defined by the upper limit of normal, when measured within 10 days from the last hu3F8 dose received, was associated with significantly lower serum hu3F8. Despite prior sensitization to other anti-GD2 antibody, e.g. mouse 3F8 or ch14.18, 75% of the patients never developed HAHA response even after getting more treatment cycles. Hu3F8 induced a de novo anti-GD2 response in patients, which was prognostic of progression-free survival. We conclude that hu3F8 had low immunogenicity. During treatment, positive HAHA and low body weight affected PK adversely, whereas induced anti-GD2 response was an outcome predictor.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号