Antibody-drug conjugate

抗体 - 药物偶联物
  • 文章类型: Journal Article
    后期临床试验失败会增加新药上市的总体成本和风险。在临床前研究和早期临床试验中确定毒性和功效的药代动力学(PK)驱动因素支持通过计算模型定量优化药物方案和剂量。此外,这种方法允许在开发早期对具有更好PK特性的先导候选物进行优先排序.Mylotarg是一种抗体-药物偶联物(ADC),经过17年的临床试验,在分次给药方案下获得了美国食品和药物管理局(FDA)的批准。包括市场上的10年期限,导致数百起致命的不良事件。尽管ADC由于其靶向性质而通常被认为具有较低的毒性风险,脱靶活性和释放的有效载荷仍然可以限制给药和驱动临床失败。根据其最初的时间表,Mylotarg的剂量很少高,这是典型的ADC,因为它们的半衰期长。然而,我们的PK模型表明,这些方案增加了与最大血浆浓度(Cmax)相关的毒性,同时产生对靶受体的次优暴露.我们的分析表明,剂量分割对Mylotarg耐受性的益处应该在药物临床开发的早期就很明显,并且可能会减少无效的III期研究的增殖。我们还确定了在不损害耐受性的情况下可能更有效的时间表。或者,循环时间较长的Mylotarg制剂可以避免剂量分级的需要,允许优越的病人方便。通过定量建模方法进行早期PK优化可以加速临床发展并防止后期失败。
    Late-stage clinical trial failures increase the overall cost and risk of bringing new drugs to market. Determining the pharmacokinetic (PK) drivers of toxicity and efficacy in preclinical studies and early clinical trials supports quantitative optimization of drug schedule and dose through computational modeling. Additionally, this approach permits prioritization of lead candidates with better PK properties early in development. Mylotarg is an antibody-drug conjugate (ADC) that attained U.S. Food and Drug Administration (FDA) approval under a fractionated dosing schedule after 17 years of clinical trials, including a 10-year period on the market resulting in hundreds of fatal adverse events. Although ADCs are often considered lower risk for toxicity due to their targeted nature, off-target activity and liberated payload can still constrain dosing and drive clinical failure. Under its original schedule, Mylotarg was dosed infrequently at high levels, which is typical for ADCs because of their long half-lives. However, our PK modeling suggests that these regimens increase maximum plasma concentration (Cmax)-related toxicities while producing suboptimal exposures to the target receptor. Our analysis demonstrates that the benefits of dose fractionation for Mylotarg tolerability should have been obvious early in the drug\'s clinical development and could have curtailed the proliferation of ineffective Phase III studies. We also identify schedules likely to be even more efficacious without compromising on tolerability. Alternatively, a longer-circulating Mylotarg formulation could obviate the need for dose fractionation, allowing superior patient convenience. Early-stage PK optimization through quantitative modeling methods can accelerate clinical development and prevent late-stage failures.
    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
    三阴性乳腺癌(TNBC)是一种高度异质性的乳腺癌亚型,其特征还在于攻击性表型,复发率高,预后不良。抗体-药物缀合物(ADC)是具有通过接头连接的细胞毒性有效载荷的单克隆抗体。ADC作为靶向抗癌剂越来越受到关注。新兴ADC药物如沙妥珠单抗戈维替康和曲妥珠单抗deruxtecan在转移性乳腺癌(包括TNBC)患者中的临床研究进展迅速。鉴于其优异的临床疗效和良好的耐受性,Sacituzumabgovitecan在2020年获得FDA加速批准用于治疗晚期转移性TNBC。这篇综述讨论了TNBC的治疗现状和挑战,重点介绍了TNBC和转移性乳腺癌的ADC开发和临床试验的现状。我们还总结了TNBC患者ADC开发的临床经验和未来的探索方向。
    Triple-negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype, which is also characterized by the aggressive phenotype, high recurrence rate, and poor prognosis. Antibody-drug conjugate (ADC) is a monoclonal antibody with a cytotoxic payload connected by a linker. ADC is gaining more and more attention as a targeted anti-cancer agent. Clinical studies of emerging ADC drugs such as sacituzumab govitecan and trastuzumab deruxtecan in patients with metastatic breast cancer (including TNBC) are progressing rapidly. In view of its excellent clinical efficacy and good tolerability, Sacituzumab govitecan gained accelerated approval by the FDA for the treatment of advanced metastatic TNBC in 2020. This review discusses the treatment status and challenges in TNBC, with an emphasis on the current status of ADC development and clinical trials in TNBC and metastatic breast cancer. We also summarize the clinical experience and future exploration directions of ADC development for TNBC patients.
    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
    Polatuzumabvedotin,第一个FDA批准的抗体-药物偶联物(ADC)靶向CD79b,用于治疗先前未治疗的弥漫性大B细胞淋巴瘤(DLBCL)或高级B细胞淋巴瘤(HGBL),以及复发或难治性(R/R)DLBCL。尽管得到了批准,关于polatuzumabvedotin的长期安全性的担忧仍然存在.这项研究旨在评估自2019年批准以来与polatuzumabvedotin相关的不良事件(AE),利用适用于FDA不良事件报告系统(FAERS)的数据挖掘策略。
    信号检测采用四种方法,包括报告赔率比(ROR),比例报告比率(PRR),贝叶斯置信传播神经网络(BCPNN),和多项目gammapoissonshrinker(MGPS),评估和量化polatuzumabvedotin相关AE的信号。此外,根据患者年龄进行亚组分析,性别,和致命病例进行调查,以调查特定亚群中的AE发生情况。
    从FAERS数据库中收集了总共1,521份报告,将polatuzumabvedotin列为“主要嫌疑人(PS)”药物。通过同时遵守四种算法,检测到19个显著的标准化MedDRA查询(SMQ)AE和92个显著的优选术语(PT)AE。亚组分析显示,男性患者的PT发生率高于女性患者,老年患者polatuzumabvedotin相关AE的可能性增加(>65岁),具有高致死性病例风险的不良事件包括:血乳酸脱氢酶升高,血细胞减少,和肾积水.polatuzumabvedotin开始后发生不良事件的中位时间为18.5(5~57.75)天,95%的AE发生在162天内。
    这项研究确定了与polatuzumabvedotin相关的各种不良事件,为接受polatuzumabvedotin治疗的患者的临床监测和风险识别提供重要见解。
    UNASSIGNED: Polatuzumab vedotin, the first FDA-approved antibody-drug conjugate (ADC) targeting CD79b, is utilized in the treatment of previously untreated diffuse large B-cell lymphoma (DLBCL) or high-grade B-cell lymphoma (HGBL), as well as relapsed or refractory (R/R) DLBCL. Despite its approval, concerns persist regarding the long-term safety profile of polatuzumab vedotin. This study aims to evaluate the adverse events (AEs) associated with polatuzumab vedotin since its approval in 2019, utilizing data mining strategies applied to the FDA Adverse Event Reporting System (FAERS).
    UNASSIGNED: Signal detection employed four methodologies, including reporting odds ratio (ROR), proportional reporting ratio (PRR), bayesian confidence propagation neural network (BCPNN), and multi-item gamma poisson shrinker (MGPS), to evaluate and quantify the signals of polatuzumab vedotin-associated AEs. Additionally, subgroup analyses based on patients age, gender, and fatal cases were conducted to investigate AEs occurrences in specific subpopulations.
    UNASSIGNED: A total of 1,521 reports listing polatuzumab vedotin as a \"principal suspect (PS)\" drug were collected from the FAERS database. Through concurrent compliance with four algorithms, 19 significant Standardized MedDRA Query (SMQ) AEs and 92 significant Preferred Term (PT) AEs were detected. Subgroup analyses revealed a higher incidence of PTs in male patients compared to female patients, increased likelihood of polatuzumab vedotin-associated AEs in elder patients (>65 years), and AEs with a high risk of fatal cases include: blood lactate dehydrogenase increased, cytopenia, and hydronephrosis. The median time to AEs occurrence following polatuzumab vedotin initiation was 18.5 (5∼57.75) days, with 95% of AEs occurred within 162 days.
    UNASSIGNED: This study identified various AEs associated with polatuzumab vedotin, offering critical insights for clinical monitoring and risk identification in patients receiving polatuzumab vedotin therapy.
    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
    乳腺癌仍然是世界范围内主要的恶性肿瘤之一。在无法手术的晚期或转移性人表皮生长因子受体2(HER2)阳性乳腺癌的情况下,全身管理主要依赖于HER2靶向单克隆抗体.随着抗HER2抗体-药物偶联物(ADC)的成功开发,这些药物已被越来越多地纳入转移性乳腺癌的治疗方案.这里,我们介绍了一例42岁的HER2阳性肺转移性乳腺癌女性患者,该患者接受了广泛的治疗方案.这个方案包括化疗,放射治疗,激素治疗,乳房手术干预,和抗HER2治疗。抗HER2治疗涉及使用曲妥珠单抗和ADCdiitamabvedotin(RC48)的单一和双重靶向策略,为期8年。在使用RC48进行HER2靶向治疗后经历疾病进展后,患者通过联合曲妥珠单抗(DS8201)和tislelizumab的治疗方案实现了明显的部分缓解。数据表明,DS8201在治疗HER2扩增的转移性乳腺癌的晚期中具有有希望的作用。特别是在使用ADC的初始HER2定向治疗后显示进展的病例。此外,其与抗PD-1药物的组合通过增强抗肿瘤免疫应答而增强治疗功效.
    Breast cancer remains one of the predominant malignancies worldwide. In the context of inoperable advanced or metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer, systemic management primarily relies on HER2-targeting monoclonal antibodies. With the successful development of anti-HER2 antibody-drug conjugates (ADCs), these agents have been increasingly integrated into therapeutic regimens for metastatic breast cancer. Here, we present the case of a 42-year-old female patient with HER2-positive pulmonary metastatic breast cancer who underwent an extensive treatment protocol. This protocol included chemotherapy, radiation therapy, hormonal therapy, surgical intervention on the breast, and anti-HER2 therapies. The anti-HER2 therapies involved both singular and dual targeting strategies using trastuzumab and the ADC disitamab vedotin (RC48) over an 8-year period. After experiencing disease progression following HER2-targeted therapy with RC48, the patient achieved noticeable partial remission through a therapeutic regimen that combined trastuzumab deruxtecan (DS8201) and tislelizumab. The data suggest a promising role for DS8201 in managing advanced stages of HER2-amplified metastatic breast cancer, especially in cases that demonstrate progression after initial HER2-directed therapies using ADCs. Furthermore, its combination with anti-PD-1 agents enhances therapeutic efficacy by augmenting the anti-tumoral immune response.
    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
    目的:Mirvetuximabsoravantine可能是卵巢低级别浆液性癌(LGSC)的潜在有效治疗选择,但叶酸受体α(FRα)过度表达在这种肿瘤类型中的患病率尚不清楚.我们试图表征LGSC中FRα的表达及其与临床和分子特征的关联。
    方法:在由89个LGSCs和42个卵巢浆液性交界性肿瘤(SBT)组成的组织微阵列上进行FRα免疫组织化学。对78个LGSC进行基于临床肿瘤-正常组的测序。检查了FRα高状态与临床病理特征和生存结果之间的关联。
    结果:在89个LGSC中,36(40%)为高FRα(≥75%的活肿瘤细胞表现出中至强膜表达)。在9名LGSC患者和来自不同时间点的样本中,4人(44%)的结果不一致,在3例(33%)中从FRα阴性转化为FRα高。FRα高状态与年龄之间没有关联,种族,或无进展/总生存期。MAPK通路遗传改变,最常见的是KRAS(n=23),存在于45(58%)LGSC中。与MAPK改变的LGSCs相比,缺乏MAPK通路改变的人更可能是FRα高(61%vs20%,p<0.001)。在SBT中,与没有恶性复发的常规SBT相比,FRα高表达与高风险(微乳头状)组织学和/或随后的LGSC复发相关(53%vs9%,p=0.008)。
    结论:有必要对LGSC患者进行FRα定向治疗的未来研究。复发时FRα状态不一致提示复检的潜在益处。建议采用生物标志物驱动的方法来指导LGSC中的治疗选择。由于高FRα表达在缺乏MAPK通路遗传改变的肿瘤中更常见,FRa测试以确定米维妥昔单抗soravantine治疗的资格特别推荐用于此亚组。
    OBJECTIVE: Mirvetuximab soravtansine may be a potentially effective therapeutic option for ovarian low-grade serous carcinoma (LGSC), but the prevalence of folate receptor alpha (FRα) overexpression in this tumor type is unknown. We sought to characterize FRα expression in LGSC and its association with clinical and molecular features.
    METHODS: FRα immunohistochemistry was performed on a tissue microarray comprised of 89 LGSCs and 42 ovarian serous borderline tumors (SBTs). Clinical tumor-normal panel-based sequencing was performed on 78 LGSCs. Associations between FRα-high status and clinicopathologic characteristics and survival outcomes were examined.
    RESULTS: Of 89 LGSCs, 36 (40%) were FRα-high (≥75% of viable tumor cells exhibiting moderate-to-strong membranous expression). Of 9 patients with LGSC and samples from different timepoints, 4 (44%) had discordant results, with conversion from FRα-negative to FRα-high in 3 (33%) cases. There was no association between FRα-high status with age, race, or progression-free/overall survival. A MAPK pathway genetic alteration, most commonly involving KRAS (n = 23), was present in 45 (58%) LGSCs. Those lacking MAPK pathway alterations were more likely to be FRα-high compared to MAPK-altered LGSCs (61% vs 20%, p < 0.001). In SBTs, FRα-high expression was associated with high-risk (micropapillary) histology and/or subsequent LGSC recurrence compared to conventional SBTs without malignant recurrence (53% vs 9%, p = 0.008).
    CONCLUSIONS: Future studies of FRα-directed therapy in patients with LGSC are warranted. Discordant FRα status at recurrence suggests potential benefit for retesting. A biomarker-driven approach to direct treatment selection in LGSC is recommended. As high FRα expression is more common amongst tumors lacking MAPK pathway genetic alterations, FRα testing to determine eligibility for mirvetuximab soravtansine therapy is particularly recommended for this subgroup.
    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:抗体-药物缀合物(ADC)是化疗药物递送方面相对较新的进展,可改善细胞毒性剂的靶向性。然而,尽管它们具有抗肿瘤活性,严重的眼部不良反应,包括视力丧失,已经报道了几个ADC。已提出通过大细胞胞吞作用将ADC非特异性摄取到人角膜上皮细胞(HCECs)及其前体中是眼部毒性的主要机制。在这项研究中,我们评估了一种新型聚合物的能力,聚(l-赖氨酸)-接枝-聚(乙二醇)(PLL-g-PEG),降低ADC利妥昔单抗-mc单甲基立他汀F(MMAF)(RIX)对人角膜上皮(HCE-T)细胞的摄取。方法:将HCE-T细胞暴露于增加浓度的RIX以确定细胞增殖的抑制作用。HCE-T细胞用PLL-g-PEG处理,巨噬细胞吞噬抑制剂5-(N-乙基-N-异丙基)阿米洛利(EIPA),或车辆。孵育30分钟后,添加了RIX。如通过显微镜观察,通过荧光抗人免疫球蛋白G和荧光缀合的葡聚糖检测ADC。结果:RIX引起HCE-T细胞增殖的剂量依赖性抑制。如通过使用荧光缀合的抗人抗体对RIX的直接定量以及使用荧光缀合的葡聚糖对巨成细胞作用的定量所评估的,EIPA显著降低RIX摄取和降低巨成细胞作用。PLL-g-PEG导致RIX摄取的剂量依赖性抑制,其最大抑制浓度为0.022%-0.023%PLL-g-PEG。结论:数据显示PLL-g-PEG是角膜上皮细胞摄取RIX的有效抑制剂,并支持将其用作预防与ADC治疗相关的眼部不良事件的新型治疗方法。
    Purpose: Antibody-drug conjugates (ADCs) are a relatively recent advance in the delivery of chemotherapeutics that improve targeting of cytotoxic agents. However, despite their antitumor activity, severe ocular adverse effects, including vision loss, have been reported for several ADCs. The nonspecific uptake of ADCs into human corneal epithelial cells (HCECs) and their precursors via macropinocytosis has been proposed to be the primary mechanism of ocular toxicity. In this study, we evaluated the ability of a novel polymer, poly(l-lysine)-graft-poly(ethylene glycol) (PLL-g-PEG), to decrease the ADC rituximab-mc monomethylauristatin F (MMAF) (RIX) uptake into human corneal epithelial (HCE-T) cells. Methods: HCE-T cells were exposed to increasing concentrations of RIX to determine inhibition of cell proliferation. HCE-T cells were treated with PLL-g-PEG, the macropinocytosis inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA), or vehicle. After 30 min of incubation, RIX was added. ADC was detected by fluorescent anti-human immunoglobulin G and fluorescently conjugated dextran as viewed by microscopy. Results: RIX caused dose-dependent inhibition of HCE-T cell proliferation. EIPA significantly reduced RIX uptake and decreased macropinocytosis as assessed by direct quantification of RIX using a fluorescently conjugated anti-human antibody as well as quantification of macropinocytosis using fluorescently conjugated dextran. PLL-g-PEG resulted in a dose-dependent inhibition of RIX uptake with half-maximal inhibitory concentrations of 0.022%-0.023% PLL-g-PEG. Conclusion: The data show PLL-g-PEG to be a potent inhibitor of RIX uptake by corneal epithelial cells and support its use as a novel therapeutic approach for the prevention of ocular adverse events associated with ADC 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
    近年来,免疫疗法行业的到来带来了提供变革的可能性,耐用,以及各种恶性肿瘤的潜在治愈结果。然而,进一步的研究表明,有许多问题显着降低了免疫疗法的有效性,尤其是实体瘤。首先,这些问题与肿瘤及其微环境的保护机制有关。目前,主要努力集中在通过使用不同的过继细胞疗法变体和基因工程构建体的修饰来克服保护机制。此外,需要复杂的劳动力来开发和实施这些治疗方法。为了克服这些重大挑战,创新的策略和方法是必要的工程更强大的免疫疗法的变化,提高抗肿瘤活性和降低毒性.在这次审查中,我们讨论了最近的免疫疗法创新,旨在提高实体瘤的临床疗效,以及克服各种免疫疗法局限性的策略。
    In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
    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
    调节性T细胞(Tregs)在介导肿瘤微环境中的免疫抑制中起着至关重要的作用。此外,Tregs导致程序性细胞死亡蛋白1(PD-1)阻断免疫疗法缺乏疗效和过度进行性疾病。因此,Tregs被认为是一个有希望的治疗靶点,特别是与PD-1阻断结合时。然而,Tregs的全身消耗会导致严重的自身免疫不良事件,这对Treg定向治疗提出了严峻挑战。这里,我们开发了一种新的治疗方法,通过近红外多卡霉素光释放(NIR-DPR)局部和主要损害Tregs。在这项技术中,我们制备了抗CD25F(ab')2偶联物,暴露于NIR光后,在表达CD25的细胞中位点特异性地捕获多卡霉素。体外,CD25靶向的NIR-DPR显著增加表达CD25的HT2-A5E细胞的凋亡。当肿瘤在体内用NIR光照射时,肿瘤内CD25+Treg群体减少,Ki-67和白细胞介素-10表达受到抑制,提示肿瘤内CD25+Tregs功能受损。CD25靶向的NIR-DPR抑制肿瘤生长并改善同系小鼠肿瘤模型的存活率。值得注意的是,CD25靶向NIR-DPR协同增强PD-1阻断的疗效,尤其是在CD8+/TregPD-1比例较高的肿瘤中。此外,联合疗法诱导显著的抗癌免疫,包括树突状细胞的成熟,细胞毒性CD8+T细胞的广泛肿瘤内浸润,和增加分化为CD8+记忆T细胞。总之,CD25靶向的NIR-DPR局部和主要靶向肿瘤微环境中的Treg,并协同提高PD-1阻断的疗效,表明这种联合疗法可以是一种合理的抗癌联合免疫疗法。
    Regulatory T cells (Tregs) play a crucial role in mediating immunosuppression in the tumor microenvironment. Furthermore, Tregs contribute to the lack of efficacy and hyperprogressive disease upon Programmed cell death protein 1 (PD-1) blockade immunotherapy. Thus, Tregs are considered a promising therapeutic target, especially when combined with PD-1 blockade. However, systemic depletion of Tregs causes severe autoimmune adverse events, which poses a serious challenge to Treg-directed therapy. Here, we developed a novel treatment to locally and predominantly damage Tregs by near-infrared duocarmycin photorelease (NIR-DPR). In this technology, we prepared anti-CD25 F(ab\')2 conjugates, which site-specifically uncage duocarmycin in CD25-expressing cells upon exposure to NIR light. In vitro, CD25-targeted NIR-DPR significantly increased apoptosis of CD25-expressing HT2-A5E cells. When tumors were irradiated with NIR light in vivo, intratumoral CD25+ Treg populations decreased and Ki-67 and Interleukin-10 expression was suppressed, indicating impaired functioning of intratumoral CD25+ Tregs. CD25-targeted NIR-DPR suppressed tumor growth and improved survival in syngeneic murine tumor models. Of note, CD25-targeted NIR-DPR synergistically enhanced the efficacy of PD-1 blockade, especially in tumors with higher CD8+/Treg PD-1 ratios. Furthermore, the combination therapy induced significant anti-cancer immunity including maturation of dendritic cells, extensive intratumoral infiltration of cytotoxic CD8+ T cells, and increased differentiation into CD8+ memory T cells. Altogether, CD25-targeted NIR-DPR locally and predominantly targets Tregs in the tumor microenvironment and synergistically improves the efficacy of PD-1 blockade, suggesting that this combination therapy can be a rational anti-cancer combination immunotherapy.
    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
    高级别浆液性卵巢癌(HGSOC)和卵巢透明细胞癌(CC),具有生物学侵袭性的肿瘤具有快速转移到腹腔和远处器官的能力。约10%的HGSOC和30%的CC证明HER2IHC3+受体过表达。我们评估曲妥珠单抗deruxtecan(T-DXd;DS-8201a)的疗效,针对多种HGSOC和CC肿瘤模型的新型HER2靶向抗体-药物缀合物(ADC)与ADC同种型对照(CTLADC)。11种卵巢癌细胞系,包括从同一患者建立的匹配的原发性和转移性细胞系,通过免疫组织化学和流式细胞术评估HER2表达,和通过荧光原位杂交分析进行基因扩增。体外实验证明,与CTLADC相比,T-DXd对HER23+HGSOC和CC细胞系显著更有效(p<0.0001)。当与HER23+细胞混合时,T-DXd诱导HER2非表达肿瘤细胞的有效旁观者杀伤。在HER2IHC3+HGSOC和CC小鼠异种移植模型中研究了T-DXd的体内活性。我们发现T-DXd比CTLADC对HER23+HGSOC(KR(CH)31)和CC(OVA10)异种移植物显著更有效,从第8天开始肿瘤生长显著差异(KR(CH)31p=0.0003,OVA10p<0.0001)。T-DXd在两种异种移植模型中也赋予存活优势。T-DXd可以代表针对原发性和转移性HER2过表达HGSOC和CC的有效ADC。
    High-grade serous ovarian cancer (HGSOC) and ovarian clear cell carcinoma (CC), are biologically aggressive tumors endowed with the ability to rapidly metastasize to the abdominal cavity and distant organs. About 10% of HGSOC and 30% of CC demonstrate HER2 IHC 3 + receptor over-expression. We evaluated the efficacy of trastuzumab deruxtecan (T-DXd; DS-8201a), a novel HER2-targeting antibody-drug conjugate (ADC) to an ADC isotype control (CTL ADC) against multiple HGSOC and CC tumor models. Eleven ovarian cancer cell lines including a matched primary and metastatic cell line established from the same patient, were evaluated for HER2 expression by immunohistochemistry and flow cytometry, and gene amplification by fluorescence in situ hybridization assays. In vitro experiments demonstrated T-DXd to be significantly more effective against HER2 3 + HGSOC and CC cell lines when compared to CTL ADC (p < 0.0001). T-DXd induced efficient bystander killing of HER2 non-expressing tumor cells when admixed with HER2 3 + cells. In vivo activity of T-DXd was studied in HER2 IHC 3 + HGSOC and CC mouse xenograft models. We found T-DXd to be significantly more effective than CTL ADC against HER2 3 + HGSOC (KR(CH)31) and CC (OVA10) xenografts with a significant difference in tumor growth starting at day 8 (p = 0.0003 for KR(CH)31, p < 0.0001 for OVA10). T-DXd also conferred a survival advantage in both xenograft models. T-DXd may represent an effective ADC against primary and metastatic HER2-overexpressing HGSOC and CC.
    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
    肺癌是癌症死亡的主要原因,非小细胞肺癌(NSCLC)约占所有肺癌的85%,治疗和预后不良。常规临床化疗和免疫治疗受到全身毒性和耐药性的挑战,因此,研究人员越来越关注抗体-药物偶联物(ADC),结合化疗和靶向治疗的创新概念,其中药物通过抗体选择性地与肿瘤细胞表面的抗原结合,将ADC内在化,然后通过内体将ADC转移到溶酶体以降解药物并杀死肿瘤细胞。尽管ADC很有前途,迄今为止,FDA尚未批准用于任何适应症(包括NSCLC)的ADC产品上市.在这次审查中,我们总结了ADC的主要优势,并深入探讨了NSCLC治疗最理想的ADC的设计。除了临床前研究,我们通过总结ClinicalTrials的实时临床试验数据,重点关注ADC作为NSCLC治疗干预措施的临床研究现状。并合理推测未来几代ADC的设计方向。
    Lung cancer is the leading cause of cancer death, with non-small cell lung cancer (NSCLC) accounting for approximately 85 % of all lung cancers and having a poor treatment and prognosis. Conventional clinical chemotherapy and immunotherapy are challenged by systemic toxicity and drug resistance, so researchers are increasingly focusing on antibody-drug conjugate (ADC), an innovative concept combining chemotherapy and targeted therapy, in which a drug selectively binds to antigens on the surface of a tumor cell via antibodies, which internalize the ADC, and then transfers the ADC to the lysosome via the endosomes to degrade the drug and kill the tumor cell. Despite the promising nature of ADCs, no ADC product for any indication including NSCLC has been approved for marketing by the FDA to date. In this review, we summarize the main advantages of ADCs and discuss in depth the design of the most desirable ADCs for NSCLC therapy. In addition to preclinical studies, we focus on the current state of clinical research on ADCs as interventions for the treatment of NSCLC by summarizing real-time clinical trial data from ClinicalTrials.gov, and reasonably speculate on the direction of the design of future generations of ADCs.
    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