Antibody–drug conjugates

抗体 - 药物偶联物
  • 文章类型: Journal Article
    在过去的几年中,通过使用尖端的递送方法或生物制剂,这给生物分析带来了困难。每年,新的生物制药产品创新问世,但是这些产品的分析开发具有挑战性。定量的产品和组分的共轭分子结构是必要的临床前和临床研究,以指导治疗的发展,考虑到它们内在的复杂性。此外,通过LC-MS技术测量这些独特的模态需要大量的信息。已经开发了许多基于LC-MS的方法,包括AEX-HPLC-MS,RP-IP-LCMS,HILIC-MS,LCHRMS,微流-LC-MS,ASMS,混合LBA/LC-MS,还有更多.然而,这些方法继续面临问题,促使替代方法的发展。因此,开发如此复杂的生物分子,低浓度需要熟练的基于LC-MS的方法和知识渊博的人员。这篇综述涵盖了一般的新模式分类,样品制备技术,分析各种新模式的现状和生物分析策略,包括基因生物疗法,寡核苷酸、抗体-药物缀合物,单克隆抗体和PROTACs。它还涵盖了过去如何使用这些策略,以及现在如何使用它们来应对基于LC-MS的方法开发中的挑战。以及改进策略,当前的进步和最近开发的方法。我们还介绍了基于不同LC-MS技术的优点和缺点,用于检查生物制药产品和未来前景。
    The last few years have seen a rise in the identification and development of bio-therapeutics through the use of cutting-edge delivery methods or bio-formulations, which has created bio-analytical difficulties. Every year, new bio-pharmaceutical product innovations come out, but the analytical development of these products is challenging. Quantifying the products and components of conjugated molecular structures is essential for preclinical and clinical research in order to guide therapeutic development, given their intrinsic complexity. Furthermore, a significant amount of information is needed for the measurement of these unique modalities by LC-MS techniques. Numerous LC-MS based methods have been developed, including AEX-HPLC-MS, RP-IP-LCMS, HILIC-MS, LCHRMS, Microflow-LC-MS, ASMS, Hybrid LBA/LC-MS, and more. However, these methods continue to face problems, prompting the development of alternative approaches. Therefore, developing bio-molecules that are this complicated and, low in concentration requires a skilled LC-MS based approach and knowledgeable personnel. This review covers general novel modalities classifications, sample preparation techniques, current status and bio-analytical strategies for analyzing various novel modalities, including gene bio-therapeutics, oligonucleotides, antibody-drug conjugates, monoclonal antibodies and PROTACs. It also covers how these strategies have been used in the past and how they are being used now to address challenges in the development of LC-MS based methods, as well as improvement strategies, current advancements and recent developed methods. We additionally covered on the benefits and drawbacks of different LC-MS based techniques for the examination of bio-pharmaceutical products and the future perspectives.
    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
    抗体-药物缀合物(ADC),它结合了抗体的精确靶向能力和小分子药物的强大细胞毒性,已经发展成为一种有希望的肿瘤治疗方法。然而,传统的共价偶联方法需要根据小分子药物的特性设计特定的接头,这极大地限制了ADC的发展和可以使用的药物范围。在这里,提出了一种新型的抗体-杯芳烃药物缀合物(ACDC),其通过使用抗体将药物递送到其靶标而功能类似于ADC,但不需要药物与抗体的共价缀合。通过用超分子接头代替常规接头,ACDC可以通过主客体相互作用负载各种化疗药物。此外,ACDC在到达低氧微环境时容易减少,导致药物的快速释放。凭借这种精确的药物封装和控释机制,ACDC将药物有效地递送至肿瘤组织并实现显著增强的抗肿瘤效果。考虑到ACDC可以通过将来自肿瘤靶向抗体的抗体-杯芳烃缀合物与各种小分子药物组合来容易地制备,ACDCs可能提供一种有前途的平台技术来加速ADC的发展,从而提高化疗的疗效。
    Antibody-drug conjugates (ADCs), which combine the precise targeting capabilities of antibodies with the powerful cytotoxicity of small-molecule drugs, have evolved into a promising approach for tumor treatment. However, the traditional covalent coupling method requires the design of a specific linker tailored to the properties of the small-molecule drugs, which greatly limits the development of ADCs and the range of drugs that can be used. Herein, a novel type of antibody-calixarene drug conjugates (ACDCs) that function similarly to ADCs by delivering drugs to their targets using antibodies but without the requirement of covalent conjugation of the drugs with antibodies is presented. By replacement of conventional linkers with supramolecular linkers, the ACDCs can load various chemotherapeutic drugs through host-guest interactions. Furthermore, ACDCs are readily reduced upon reaching the hypoxic microenvironment, resulting in rapid release of the drugs. With this precise drug encapsulation and controlled release mechanism, ACDCs deliver drugs to tumor tissues effectively and achieve a significantly enhanced antitumor effect. Considering that the ACDCs can be easily prepared by combining antibody-calixarene conjugates derived from tumor-targeting antibodies with various small-molecule drugs, ACDCs may provide a promising platform technology to accelerate ADC development and thus improve the therapeutic efficacy of chemotherapy.
    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
    抗体-药物缀合物(ADC)由靶向肿瘤细胞的单克隆抗体和通过接头连接的细胞毒性药物组成。通过利用抗体的靶向特性,在鉴定肿瘤抗原后,ADC通过内吞作用将细胞毒性药物递送到肿瘤细胞中。这种精确的方法旨在选择性地杀死肿瘤细胞,同时最大限度地减少对正常细胞的伤害。提供安全有效的治疗益处。近年来,随着ADC的发展,抗肿瘤治疗取得了重大进展,为患者提供新的和有效的治疗选择。有300多个ADC探索各种肿瘤适应症,其中一些已经批准用于临床,诸如抗原表达等因素引起的耐药性等挑战,ADC处理,有效载荷已经出现。这篇综述旨在概述ADC的发展历史,他们的结构,作用机制,最近的构图进步,目标选择,已完成和正在进行的临床试验,抵抗机制,和干预策略。此外,它将深入研究带有新标记的ADC的潜力,连接子,有效载荷,和创新的行动机制,以加强癌症治疗选择。ADC的发展也导致了联合治疗作为提高药物疗效的新治疗方法的出现。
    Antibody-drug conjugates (ADCs) consist of monoclonal antibodies that target tumor cells and cytotoxic drugs linked through linkers. By leveraging antibodies\' targeting properties, ADCs deliver cytotoxic drugs into tumor cells via endocytosis after identifying the tumor antigen. This precise method aims to kill tumor cells selectively while minimizing harm to normal cells, offering safe and effective therapeutic benefits. Recent years have seen significant progress in antitumor treatment with ADC development, providing patients with new and potent treatment options. With over 300 ADCs explored for various tumor indications and some already approved for clinical use, challenges such as resistance due to factors like antigen expression, ADC processing, and payload have emerged. This review aims to outline the history of ADC development, their structure, mechanism of action, recent composition advancements, target selection, completed and ongoing clinical trials, resistance mechanisms, and intervention strategies. Additionally, it will delve into the potential of ADCs with novel markers, linkers, payloads, and innovative action mechanisms to enhance cancer treatment options. The evolution of ADCs has also led to the emergence of combination therapy as a new therapeutic approach to improve drug efficacy.
    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
    目前针对人类表皮生长因子受体(HER)家族的疗法,包括单克隆抗体(mAb)和酪氨酸激酶抑制剂(TKIs),受到耐药性和全身毒性的限制。抗体-药物缀合物(ADC)是最快速扩展的抗癌治疗剂类别之一,目前已获得FDA批准。重要的是,ADC代表了一种有希望的治疗选择,具有通过向HER过表达的癌细胞特异性递送高效细胞毒素并发挥mAb和有效载荷介导的抗肿瘤功效来克服传统HER靶向治疗抗性的潜力。HER靶向ADC的临床效用通过HER2靶向ADC(包括曲妥珠单抗依坦素和曲妥珠单抗deruxtecan)的巨大成功来举例说明。尽管如此,改善现有HER2靶向ADC以及开发针对其他HER家族成员的ADC的策略,特别是EGFR和HER3,引起了极大的兴趣。迄今为止,没有HER4靶向ADC的报道。在这次审查中,我们广泛详述临床阶段EGFR-,HER2-,和HER3靶向单特异性ADC以及针对该受体家族的新型临床和临床前双特异性ADC(bsADC)。最后,我们讨论了针对HER的ADC发展的新趋势,包括新型ADC有效载荷和HER配体靶向ADC。
    Current therapies targeting the human epidermal growth factor receptor (HER) family, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs), are limited by drug resistance and systemic toxicities. Antibody-drug conjugates (ADCs) are one of the most rapidly expanding classes of anti-cancer therapeutics with 13 presently approved by the FDA. Importantly, ADCs represent a promising therapeutic option with the potential to overcome traditional HER-targeted therapy resistance by delivering highly potent cytotoxins specifically to HER-overexpressing cancer cells and exerting both mAb- and payload-mediated antitumor efficacy. The clinical utility of HER-targeted ADCs is exemplified by the immense success of HER2-targeted ADCs including trastuzumab emtansine and trastuzumab deruxtecan. Still, strategies to improve upon existing HER2-targeted ADCs as well as the development of ADCs against other HER family members, particularly EGFR and HER3, are of great interest. To date, no HER4-targeting ADCs have been reported. In this review, we extensively detail clinical-stage EGFR-, HER2-, and HER3-targeting monospecific ADCs as well as novel clinical and pre-clinical bispecific ADCs (bsADCs) directed against this receptor family. We close by discussing nascent trends in the development of HER-targeting ADCs, including novel ADC payloads and HER ligand-targeted ADCs.
    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
    背景:三阴性乳腺癌(TNBC)由于其侵袭性和新辅助失败后有限的治疗选择,仍然是一种具有临床挑战性的亚型。历史上,卡培他滨一直是未达到病理完全缓解(pCR)的TNBC患者的辅助治疗的基石。然而,免疫治疗和PARP抑制剂等新方法的整合促使人们对传统的新辅助治疗后方法进行了重新评估.
    方法:这篇综述综合了来自关键临床试验和荟萃分析的数据,以评估新辅助治疗后的疗效。我们专注于免疫检查点抑制剂(ICIs)的作用,PARP抑制剂(PARPis),在TNBC治疗范例中,抗体-药物缀合物(ADC)与卡培他滨一起或代替卡培他滨。
    结果:在新辅助治疗方案中加入pembrolizumab等ICIs已显示出pCR率增加和无事件生存率改善,提出了关于新辅助治疗后最佳治疗的新问题。同样,PARPis已证明在BRCA突变的TNBC群体中有效,无病生存期(DFS)和总生存期(OS)显着改善。对ADC的新兴研究进一步使佐剂景观复杂化,提供卡培他滨的潜在有效替代品,特别是在新辅助治疗后残留疾病的患者中。
    结论:将这些新的治疗方法有效地融入临床实践仍然是一个挑战。考虑到耐药性等因素,患者特有的特征,和社会经济障碍。这篇综述讨论了这些疗法的含义,并提出了TNBC个性化医疗方法的未来方向。
    结论:随着TNBC治疗前景的发展,卡培他滨的作用正在严格检查中。虽然它仍然是某些患者群体的可行选择,ICIs的推出,PARPis,ADC提供了有希望的替代方案,可以重新定义辅助治疗标准。正在进行和未来的试验对于确定新辅助治疗后残留疾病的TNBC患者的最佳治疗策略至关重要。
    BACKGROUND: Triple-negative breast cancer (TNBC) remains a clinically challenging subtype due to its aggressive nature and limited treatment options post-neoadjuvant failure. Historically, capecitabine has been the cornerstone of adjuvant therapy for TNBC patients not achieving a pathological complete response (pCR). However, the integration of new modalities such as immunotherapy and PARP inhibitors has prompted a re-evaluation of traditional post-neoadjuvant approaches.
    METHODS: This review synthesizes data from pivotal clinical trials and meta-analyses to evaluate the efficacy of emerging therapies in the post-neoadjuvant setting. We focus on the role of immune checkpoint inhibitors (ICIs), PARP inhibitors (PARPis), and antibody-drug conjugates (ADCs) alongside or in place of capecitabine in TNBC treatment paradigms.
    RESULTS: The addition of ICIs like pembrolizumab to neoadjuvant regimens has shown increased pCR rates and improved event-free survival, posing new questions about optimal post-neoadjuvant therapies. Similarly, PARPis have demonstrated efficacy in BRCA-mutated TNBC populations, with significant improvements in disease-free survival (DFS) and overall survival (OS). Emerging studies on ADCs further complicate the adjuvant landscape, offering potentially efficacious alternatives to capecitabine, especially in patients with residual disease after neoadjuvant therapy.
    CONCLUSIONS: The challenge remains to integrate these new treatments into clinical practice effectively, considering factors such as drug resistance, patient-specific characteristics, and socio-economic barriers. This review discusses the implications of these therapies and suggests a future direction focused on personalized medicine approaches in TNBC.
    CONCLUSIONS: As the treatment landscape for TNBC evolves, the role of capecitabine is being critically examined. While it remains a viable option for certain patient groups, the introduction of ICIs, PARPis, and ADCs offers promising alternatives that could redefine adjuvant therapy standards. Ongoing and future trials will be pivotal in determining the optimal therapeutic strategies for TNBC patients with residual disease post-neoadjuvant 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: Journal Article
    几十年来,以铂为基础的化疗一直是晚期尿路上皮癌一线治疗的基石,基于其已证明的疗效和良好的安全性。尽管enfortumabvedotin(EV)联合pembrolizumab在EV-3023期试验中显示出优于铂类化疗的疗效,与EV+pembrolizumab相关的常见和潜在累积毒性可能使这种组合不太适合某些患者,比如那些预先存在神经病变的人,高血糖症,或肝功能损害,或患者可能有良好的结果与铂类药物为基础的化疗。EV+pembrolizumab在不同国家的可用性也可能受到财务考虑的限制。因此,铂类化疗可能仍然是晚期尿路上皮癌的一个有价值的选择.以顺铂或卡铂为基础的方案的资格可以通过评估肾功能来确定,性能状态,和特定的合并症。在铂类化疗后无疾病进展的顺铂患者中,基于JAVELIN膀胱1003期试验的结果,阿维鲁单抗一线维持是标准的治疗方法,这表明,在临床相关亚组中,与单用最佳支持治疗相比,阿维鲁单抗一线维持治疗加最佳支持治疗延长了总生存期和无进展生存期.与avelumab相关的不良事件通常与其他免疫检查点抑制剂观察到的不良事件一致。长期随访显示,长期治疗没有新的安全性问题.在基于顺铂或卡铂的化疗后接受阿维鲁单抗一线维持治疗的患者中,疗效获益和安全性相似。阿维鲁单抗一线维持的有效性和安全性已经在一些现实世界的研究中得到证实。总的来说,这些数据支持对所有接受铂类治疗且无疾病进展的患者使用阿维鲁单抗一线治疗.在这个播客中,我们在EV-302试验结果的背景下讨论了铂类化疗在这一疾病背景下的演变作用,并描述了在接受基于顺铂或卡铂的一线化疗后接受阿维鲁单抗维持治疗的患者中的实际考虑因素.
    Platinum-based chemotherapy has been the cornerstone of first-line treatment for advanced urothelial carcinoma for decades, based on its proven efficacy and well-characterized safety profile. Although enfortumab vedotin (EV) plus pembrolizumab showed superior efficacy versus platinum-based chemotherapy in the EV-302 phase 3 trial, common and potentially cumulative toxicities associated with EV plus pembrolizumab may make this combination less suitable for some patients, such as those with pre-existing neuropathy, hyperglycemia, or hepatic impairment, or patients likely to have favorable outcomes with platinum-based chemotherapy. The availability of EV plus pembrolizumab in various countries may also be limited by financial considerations. Thus, platinum-based chemotherapy is likely to remain a valuable option for advanced urothelial carcinoma. Eligibility for cisplatin- or carboplatin-based regimens can be determined by assessing renal function, performance status, and specific comorbidities. In cisplatin-eligible and -ineligible patients without disease progression following platinum-based chemotherapy, avelumab first-line maintenance is standard of care based on findings from the JAVELIN Bladder 100 phase 3 trial, which showed that avelumab first-line maintenance plus best supportive care prolonged overall survival and progression-free survival compared with best supportive care alone across clinically relevant subgroups. Adverse events associated with avelumab were generally consistent with those observed with other immune checkpoint inhibitors, and long-term follow-up showed no new safety concerns with prolonged treatment. Efficacy benefits and safety profiles were similar in patients who received avelumab first-line maintenance after cisplatin- or carboplatin-based chemotherapy. The effectiveness and safety of avelumab first-line maintenance have been confirmed in several real-world studies. Overall, these data support the use of avelumab first-line maintenance for all platinum-treated patients without disease progression. In this podcast, we discuss the evolving role of platinum-based chemotherapy in this disease setting in the context of EV-302 trial results and describe practical considerations in patients receiving first-line cisplatin- or carboplatin-based chemotherapy followed by avelumab maintenance 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
    抗肿瘤疗法是本世纪的主要研究主题之一。已经实施了现代方法来靶向和增强细胞抑制药物对肿瘤的作用并减少其一般/非特异性毒性。在这种情况下,抗体-药物缀合物(ADC)代表了一种有希望和成功的策略。本次审查的目的是评估有关ADC的不同方面。它们是从化学和药理学的角度以及结构等方面提出的,共轭和发展特殊性以及效应,临床试验,讨论了安全性问题以及未来使用这些药物的前景和挑战.代表性实例包括但不限于以下ADC的主要结构成分:单克隆抗体(曲妥珠单抗,brentuximab),接头(pH敏感,还原敏感,基于肽的,磷酸盐基,和其他),和有效载荷(多柔比星,emtansine,ravtansine,卡利车霉素)。关于药物治疗的成功,大量正在进行的临床试验支持了与ADC治疗相关的高疗效预期.首先讨论了发展战略等主要方面,优点和缺点,安全性和有效性,提供了对这个问题的回顾性见解。第二部分是前瞻性回顾,专注于克服先前确定的困难的各种计划。
    Antineoplastic therapy is one of the main research themes of this century. Modern approaches have been implemented to target and heighten the effect of cytostatic drugs on tumors and diminish their general/unspecific toxicity. In this context, antibody-drug conjugates (ADCs) represent a promising and successful strategy. The aim of this review was to assess different aspects regarding ADCs. They were presented from a chemical and a pharmacological perspective and aspects like structure, conjugation and development particularities alongside effects, clinical trials, safety issues and perspectives and challenges for future use of these drugs were discussed. Representative examples include but are not limited to the following main structural components of ADCs: monoclonal antibodies (trastuzumab, brentuximab), linkers (pH-sensitive, reduction-sensitive, peptide-based, phosphate-based, and others), and payloads (doxorubicin, emtansine, ravtansine, calicheamicin). Regarding pharmacotherapy success, the high effectiveness expectation associated with ADC treatment is supported by the large number of ongoing clinical trials. Major aspects such as development strategies are first discussed, advantages and disadvantages, safety and efficacy, offering a retrospective insight on the subject. The second part of the review is prospective, focusing on various plans to overcome the previously identified difficulties.
    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
    抗体-药物缀合物(ADC)代表实体瘤和血液肿瘤的系统治疗中的革命性方法。由抗体构成,细胞毒性有效载荷,和一个链接器,ADC旨在选择性地将细胞毒性剂递送至肿瘤,同时保留正常组织。到目前为止,已经测试并批准了多种ADC用于多种实体瘤,但是如果有一个对临床实践有重大影响,这就是曲妥珠单抗-deruxtecan(T-DXd).值得注意的是,T-DXd被批准用于HER2阳性和HER2低转移性乳腺癌(MBC),HER2阳性胃癌(GC),HER2突变型非小细胞肺癌(NSCLC)和HER23+实体瘤。此外,它接受了HER2阳性结直肠癌(CRC)的突破性治疗.
    我们回顾了T-DXd的临床前和临床数据,重点是正在进行的早期试验,探索联合疗法以增强表达HER2的实体瘤中T-DXd的活性。
    T-DXd的临床使用仍然引起患者选择的问题,治疗持续时间,优先于其他批准的ADC,和管理抵抗。对T-DXd毒性的担忧仍然存在,特别是涉及潜在毒性药物的组合。生物标志物识别和联合疗法的进步为提高疗效和克服对T-DXd的耐药性提供了有希望的途径。最终改善癌症患者的预后。
    UNASSIGNED: Antibody-drug conjugates (ADCs) represent a revolutionary approach in the systemic treatment for both solid and hematologic tumors. Constituted by an antibody, a cytotoxic payload, and a linker, ADCs aim to selectively deliver cytotoxic agents to tumors while sparing normal tissues. Various ADCs have been tested and approved for multiple solid tumors so far, but if there is one that had a major impact on clinical practice, this is Trastuzumab-deruxtecan (T-DXd). Notably, T-DXd was approved for HER2-positive and HER2-low metastatic breast cancer (MBC), HER2-positive gastric cancer (GC), HER2-mutant non-small cell lung cancer (NSCLC) and HER2 3+ solid tumors. Moreover, it received Breakthrough Therapy Designation for HER2-positive colorectal cancer (CRC).
    UNASSIGNED: We review preclinical and clinical data of T-DXd, focusing on early-phase ongoing trials exploring combination therapies to enhance the activity of T-DXd in HER2-expressing solid tumors.
    UNASSIGNED: The clinical use of T-DXd still raises questions about selection of patients, treatment duration, prioritization over other approved ADCs, and management of resistance. Concerns regarding the toxicity of T-DXd remain, particularly with combinations involving potentially toxic drugs. Advancements in biomarker identification and combination therapies offer promising avenues to enhance efficacy and overcome resistance to T-DXd, ultimately improving outcomes for patients with cancer.
    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
    抗体-药物偶联物(ADC)是胃癌靶向治疗的重要组成部分,可能改变传统的治疗模式。许多ADC已经进入基于生物学理论和临床前实验的严格临床试验。还与单克隆抗体疗法结合进行了模态试验,化疗,免疫疗法,和其他治疗方法的疗效增强药物的协调作用。然而,ADC在治疗胃癌方面表现出局限性,包括由其结构或其他因素引发的阻力。正在进行的深入研究和临床前实验正在产生改进,虽然在治疗期间药物开发过程和伴随诊断的增强积极地提高ADC功效。胃癌患者的最佳治疗策略正在不断发展。本文就ADCs治疗胃癌的临床进展作一综述,分析ADC联合治疗的机制,讨论阻力模式,并为ADC药物开发和伴随诊断的未来应用提供了有希望的前景。
    Antibody-drug conjugates (ADCs) represent a crucial component of targeted therapies in gastric cancer, potentially altering traditional treatment paradigms. Many ADCs have entered rigorous clinical trials based on biological theories and preclinical experiments. Modality trials have also been conducted in combination with monoclonal antibody therapies, chemotherapies, immunotherapies, and other treatments to enhance the efficacy of drug coordination effects. However, ADCs exhibit limitations in treating gastric cancer, including resistance triggered by their structure or other factors. Ongoing intensive researches and preclinical experiments are yielding improvements, while enhancements in drug development processes and concomitant diagnostics during the therapeutic period actively boost ADC efficacy. The optimal treatment strategy for gastric cancer patients is continually evolving. This review summarizes the clinical progress of ADCs in treating gastric cancer, analyzes the mechanisms of ADC combination therapies, discusses resistance patterns, and offers a promising outlook for future applications in ADC drug development and companion diagnostics.
    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
    在非小细胞肺癌(NSCLC)患者中,<5%的病例中存在的致癌变异被认为是罕见的,其中主要包括人类表皮生长因子受体2(HER2)突变,间充质-上皮转化(MET)改变,c-ros癌基因1(ROS1)重排,转染(RET)融合过程中的重排,v-raf小鼠肉瘤病毒癌基因同源物B1(BRAF)突变,和神经营养肌钙蛋白受体激酶(NTRK)融合。脑转移(BMs)发生在大约10%-50%的具有罕见遗传变异的NSCLC患者中。最近小分子酪氨酸激酶抑制剂和大分子抗体-药物缀合物(ADC)的出现为具有罕见驱动改变的NSCLC患者赋予了显著的生存益处。尽管大多数靶向药物都能有效控制脑部病变,并且有希望报道与新型ADC相关的颅内缓解,BM仍然是一个主要的治疗挑战。本文综述了近年来罕见遗传变异和BM,特别关注颅内功效,并探讨如何最好地治疗这些患者的未来观点。
    In patients with non-small cell lung cancer (NSCLC), oncogenic variants present in <5% of cases are considered rare, the predominant of which include human epidermal growth factor receptor 2 (HER2) mutations, mesenchymal-epithelial transition (MET) alterations, c-ros oncogene 1 (ROS1) rearrangements, rearrangement during transfection (RET) fusions, v-raf mouse sarcoma virus oncogene homolog B1 (BRAF) mutations, and neurotrophic troponin receptor kinase (NTRK) fusions. Brain metastases (BMs) occur in approximately 10%-50% of patients with NSCLC harboring rare genetic variants. The recent advent of small-molecule tyrosine kinase inhibitors and macromolecular antibody-drug conjugates (ADCs) has conferred marked survival benefits to patients with NSCLC harboring rare driver alterations. Despite effective brain lesion control for most targeted agents and promising reports of intracranial remission associated with novel ADCs, BM continues to be a major therapeutic challenge. This review discusses the recent advances in the treatment of NSCLC with rare genetic variants and BM, with a particular focus on intracranial efficacy, and explores future perspectives on how best to treat these patients.
    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