Trastuzumab

曲妥珠单抗
  • DOI:
    文章类型: Journal Article
    HER2受体酪氨酸激酶是表皮生长因子受体家族成员之一,包括EGFR,HER3和HER4已知它们在正常发育和癌症中起关键作用。乳腺癌的一个子集与HER2基因有关,在20-25%的浸润性乳腺癌中扩增和/或过表达,并与肿瘤对化疗的耐药性相关,转移性乳腺癌(MBC)和患者生存率低。受体酪氨酸激酶抑制剂的出现改善了HER2阳性乳腺癌的预后;然而,HER2+MBC总是进展(获得性抗性或从头抗性)。基于单克隆抗体的药物(大分子TKIs)靶向HER2的细胞外结合结构域;而小分子TKIs在细胞内作用以抑制增殖和存活信号。我们回顾了TKIs的作用方式,以期显示哪些TKIs可以结合在纳米颗粒中,以受益于纳米技术的力量(降低毒性,改善疏水性药物的溶解度,循环半衰期长,绕过外排泵并防止网状内皮系统(单核吞噬细胞系统)捕获。纳米治疗剂还介导掺入纳米颗粒中的多种药物的药代动力学和生物分布的同步。提到了目前正在研究的有或没有纳米颗粒递送的新型TKIs,并提出了基于纳米的策略来改善其交付。目前在临床实践中的免疫疗法,讨论了临床试验或临床前阶段。然而,免疫疗法仅在相对较小的患者亚群中有效。将纳米医学与免疫疗法相结合可以提高治疗效果,通过将“冷”非免疫反应性肿瘤和转移转化为“热”免疫反应性病变。
    The HER2 receptor tyrosine kinase is a member of the epidermal growth factor receptor family which includes EGFR, HER3 and HER4. They are known to play critical roles in both normal development and cancer. A subset of breast cancers is associated with the HER2 gene, which is amplified and/or overexpressed in 20-25% of invasive breast cancers and is correlated with tumor resistance to chemotherapy, Metastatic Breast Cancer (MBC) and poor patient survival. The advent of receptor tyrosine kinase inhibitors has improved the prognosis of HER2-postive breast cancers; however, HER2+MBC invariably progresses (acquired resistance or de novo resistance). The monoclonal antibody-based drugs (large molecule TKIs) target the extracellular binding domain of HER2; while the small molecule TKIs act intracellularly to inhibit proliferation and survival signals. We reviewed the modes of action of the TKIs with a view to showing which of the TKIs could be combined in nanoparticles to benefit from the power of nanotechnology (reduced toxicity, improved solubility of hydrophobic drugs, long circulation half-lives, circumventing efflux pumps and preventing capture by the reticuloendothelial system (mononuclear phagocyte system). Nanotherapeutics also mediate the synchronization of the pharmacokinetics and biodistribution of multiple drugs incorporated in the nanoparticles. Novel TKIs that are currently under investigation with or without nanoparticle delivery are mentioned, and nano-based strategies to improve their delivery are suggested. Immunotherapies currently in clinical practice, clinical trials or at the preclinical stage are discussed. However, immunotherapy only works well in relatively small subsets of patients. Combining nanomedicine with immunotherapy can boost therapeutic outcomes, by turning \"cold\" non-immunoresponsive tumors and metastases into \"hot\" immunoresponsive 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
    背景:在靶向治疗期间,HER2阳性乳腺癌总是失去HER2DNA扩增。相比之下,有趣的是,HER2蛋白可能丢失或获得。为了纵向和系统地了解HER2DNA/蛋白质化学计量的复杂/不一致变化,HER2DNA拷贝数和可溶性血液蛋白(aHER2/sHER2)平行测试,非侵入性(通过液体活检),在二维中,因此HER2-2D。
    方法:在使用抗体-药物偶联物(ADC)曲妥珠单抗-emtansine(T-DM1)对晚期HER2阳性乳腺癌患者(n=37)进行标准治疗之前和之后,通过数字PCR和ELISA评估aHER2和sHER2。
    结果:如预期,aHER2总是被T-DM1抑制,但这种损失令人惊讶地反映了sHER2增益,有时是相当大的实体,在大多数(30/37;81%)患者中。HER2致癌剂量的这种非正统分裂在两个代表性病例中得到了aHER2/sHER2动力学的支持。和免疫组织化学高状态,尽管4/5的T-DM1肿瘤后可再活检sHER2患者的拷贝数中性。此外,通过T-DM1体外处理的垂死的乳腺癌细胞系优先释放sHER2。最后,sHER2增加与比sHER2损失更长的PFS相关(平均PFS282天vs133天,95%CI[210-354]对[56-209],对数秩检验p=0.047),特别是当排除在T-DM1治疗期间出现循环HER2旁路改变的病例(n=11)时(平均PFS349vs139天,95%CI[255-444]vs[45-232],对数秩检验p=0.009)。
    结论:HER2增益是在肿瘤组织中适应性选择的,并通过sHER2增益在血液中概括。可能,在用裸抗体抗HER2治疗期间,增加致癌剂量对肿瘤有益,但在用强细胞毒性ADC如T-DM1治疗期间对宿主有利。在后一种情况下,HER2-gain肿瘤可以暂时保持在检查,直到替代致癌驱动因素,液体活检显示,绕过HER2。无论哪种解释,HER2-2D可能有助于定制/优先考虑抗HER2治疗,尤其是对低aHER2/低sHER2肿瘤有活性的ADC。
    背景:NCT05735392于2023年1月31日回顾性注册https://www。
    结果:gov/search?term=NCT05735392。
    BACKGROUND: During targeted treatment, HER2-positive breast cancers invariably lose HER2 DNA amplification. In contrast, and interestingly, HER2 proteins may be either lost or gained. To longitudinally and systematically appreciate complex/discordant changes in HER2 DNA/protein stoichiometry, HER2 DNA copy numbers and soluble blood proteins (aHER2/sHER2) were tested in parallel, non-invasively (by liquid biopsy), and in two-dimensions, hence HER2-2D.
    METHODS: aHER2 and sHER2 were assessed by digital PCR and ELISA before and after standard-of-care treatment of advanced HER2-positive breast cancer patients (n=37) with the antibody-drug conjugate (ADC) Trastuzumab-emtansine (T-DM1).
    RESULTS: As expected, aHER2 was invariably suppressed by T-DM1, but this loss was surprisingly mirrored by sHER2 gain, sometimes of considerable entity, in most (30/37; 81%) patients. This unorthodox split in HER2 oncogenic dosage was supported by reciprocal aHER2/sHER2 kinetics in two representative cases, and an immunohistochemistry-high status despite copy-number-neutrality in 4/5 available post-T-DM1 tumor re-biopsies from sHER2-gain patients. Moreover, sHER2 was preferentially released by dying breast cancer cell lines treated in vitro by T-DM1. Finally, sHER2 gain was associated with a longer PFS than sHER2 loss (mean PFS 282 vs 133 days, 95% CI [210-354] vs [56-209], log-rank test p=0.047), particularly when cases (n=11) developing circulating HER2-bypass alterations during T-DM1 treatment were excluded (mean PFS 349 vs 139 days, 95% CI [255-444] vs [45-232], log-rank test p=0.009).
    CONCLUSIONS: HER2 gain is adaptively selected in tumor tissues and recapitulated in blood by sHER2 gain. Possibly, an increased oncogenic dosage is beneficial to the tumor during anti-HER2 treatment with naked antibodies, but favorable to the host during treatment with a strongly cytotoxic ADC such as T-DM1. In the latter case, HER2-gain tumors may be kept transiently in check until alternative oncogenic drivers, revealed by liquid biopsy, bypass HER2. Whichever the interpretation, HER2-2D might help to tailor/prioritize anti-HER2 treatments, particularly ADCs active on aHER2-low/sHER2-low tumors.
    BACKGROUND: NCT05735392 retrospectively registered on January 31, 2023 https://www.
    RESULTS: gov/search?term=NCT05735392.
    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
    炎症性乳腺癌(IBC)是一种罕见的,侵袭性乳腺癌的特点是预后不良。治疗需要多学科的方法,新辅助化疗,手术,和放射治疗(RT)。特别是,历史上,在化疗和乳房切除术后或作为不符合手术条件的患者的根治性治疗的辅助治疗中,高剂量的常规RT被给予.这里,我们报告了一例49岁女性患者的IBC不适合手术,并采用晶格RT和分割的外部束RT联合曲妥珠单抗治疗,有治愈的目的。RT后一年,患者出现完全缓解和可耐受的毒性反应.这是用这种特殊类型的RT治疗的不可手术的IBC患者的首例报道病例。
    Inflammatory breast cancer (IBC) is a rare, aggressive form of breast cancer characterized by poor prognosis. The treatment requires a multidisciplinary approach, with neoadjuvant chemotherapy, surgery, and radiation therapy (RT). Particularly, high doses of conventional RT have been historically delivered in the adjuvant setting after chemotherapy and mastectomy or as radical treatment in patients ineligible for surgery. Here, we report the case of a 49-year-old woman patient with IBC unsuitable for surgery and treated with a combination of lattice RT and fractionated external beam RT concurrent with trastuzumab, with a curative aim. One year after RT, the patient showed a complete response and tolerable toxicities. This is the first reported case of a not-operable IBC patient treated with this particular kind of RT.
    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:乳腺癌(BC)是全球最常见的恶性疾病。局部阶段的BC可以通过手术成功治疗。然而,局部复发发生在约4-10%的患者,需要系统性治疗,损害患者的生活质量,缩短预期寿命。因此,需要新的治疗选择,可以在术中使用,并有助于完全去除手术区域的残留肿瘤细胞。在本研究中,我们描述了抗HER2抗体曲妥珠单抗的半胱氨酸修饰变体,与硅酞菁光敏剂染料WB692-CB1偶联,用于BC的光免疫疗法(PIT)。
    方法:克隆半胱氨酸修饰的曲妥珠单抗变体并在Expi293F细胞中表达。通过固定化亲和层析纯化后,抗体与染料偶联。通过流式细胞术测量抗体和抗体染料缀合物的细胞结合。将BC细胞与缀合物孵育并通过红光照射激活染料后,确定细胞活力。
    结果:抗体和缀合物显示与表达HER2的BC细胞的特异性结合。用缀合物处理HER2highBC细胞系SK-BR-3,然后用32J/cm2的红光剂量照射导致24小时内完全杀死细胞。
    结论:我们的新型抗体染料缀合物代表了局部BC术中治疗的有希望的候选者,旨在消除手术区域残留的肿瘤细胞,并有可能减少局部复发,从而改善BC患者的康复前景。
    OBJECTIVE: Breast cancer (BC) is the most common malignant disease worldwide. Localized stages of BC can be successfully treated by surgery. However, local recurrence occurs in about 4-10% of patients, requiring systemic treatments that impair the patients\' quality of life and shortens life expectancy. Therefore, new therapeutic options are needed, which can be used intraoperatively and contribute to the complete removal of residual tumor cells in the surgical area. In the present study, we describe a cysteine-modified variant of the anti-HER2 antibody trastuzumab, that was coupled to the silicon phthalocyanine photosensitizer dye WB692-CB1 for the photoimmunotherapy (PIT) of BC.
    METHODS: The cysteine modified trastuzumab variant was cloned and expressed in Expi293F cells. After purification via immobilized affinity chromatography, the antibody was coupled to the dye. Cell binding of the antibody and the antibody dye conjugate was measured by flow cytometry. After incubation of BC cells with the conjugate and activation of the dye by irradiation with red light, cell viability was determined.
    RESULTS: The antibody and the conjugate showed specific binding to HER2-expressing BC cells. Treatment of the HER2high BC cell line SK-BR-3 with the conjugate followed by irradiation with a red light dose of 32 J/cm2 led to complete cell killing within 24 h.
    CONCLUSIONS: Our novel antibody dye conjugate represents a promising candidate for intraoperative treatment of localized BC, aiming to eliminate residual tumor cells in the surgical area and potentially reduce local recurrence, thereby improving recovery prospects for BC 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
    在过去的十年中,HER2靶向疗法在乳腺癌治疗方面取得了进步。临床上,通过免疫组织化学或通过荧光原位杂交的基因调控评估肿瘤细胞表面的HER2水平来确定HER2治疗的合格性.HER2治疗在HER2水平升高的患者中并不总是有效,这质疑HER2的量是否足以预测患者的预后。此外,HER2靶向抗体-药物偶联物(ADC)Enhertu®最近被批准用于转移性低HER2癌症,证实HER2治疗对低HER2水平患者的益处。为了评估HER2水平与治疗效果之间的相关性,我们使用流式细胞术定量了8种细胞系的HER2,同时测定了2种HER2靶向ADC的毒性.高HER2细胞系和低HER2细胞系均对ADC具有显著的毒性反应。我们量化了HER2内在化,发现HER2水平和内在化百分比之间没有相关性。我们发现了一个有用的指标,表明运输到溶酶体的HER2受体的最小数量足以提供有效的治疗。我们的结果表明,目前确定HER2治疗资格的标准可能会限制患者获得有效治疗。总之,HER2水平并不完全足以确定对ADC治疗的反应。
    HER2-targeting therapies have advanced breast cancer treatment over the past decade. Clinically, eligibility for HER2 therapies is determined by assessing HER2 levels on tumor cell surfaces through immunohistochemistry or by gene regulation through fluorescence in situ hybridization. HER2 therapies are not always effective in patients with elevated levels of HER2, questioning whether the amount of HER2 is sufficiently predictive of patient outcomes. Additionally, the HER2-targeting antibody-drug conjugate (ADC) Enhertu® was recently approved for metastasized HER2-low cancers, confirming the benefits of HER2 treatment for patients with low HER2 levels. To evaluate the correlation between HER2 levels and treatment efficacy, we quantified HER2 on eight cell lines using flow cytometry while simultaneously determining the toxicity of two HER2-targeting ADCs. Both HER2-high cell lines and HER2-low cell lines had significant toxicity responses to ADCs. We quantified HER2 internalization and found no correlation between HER2 levels and the percentage of internalization. We found a useful metric suggesting that a minimum number of HER2 receptors trafficked to lysosomes is sufficient to provide effective treatment. Our results indicate that the current standards of determining eligibility for HER2 therapy could limit patients\' access to effective treatment. In conclusion, HER2 levels are not wholly adequate to determine the response to ADC treatment.
    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
    背景和目的:人表皮生长因子受体2(HER2)阳性,激素受体阳性(HR阳性)转移性乳腺癌(MBC)通常在后续系列中接受曲妥珠单抗emtansine(T-DM1)治疗.将内分泌治疗(ET)与T-DM1结合可以改善该亚型的治疗结果。因此,本研究旨在探讨T-DM1联合ET在HER2阳性和HR阳性MBC中的获益.这项研究是第一个研究ET与T-DM1结合的益处。材料和方法:本研究分析了2010年6月至2021年12月接受T-DM1治疗的HER2阳性和HR阳性MBC患者的医疗记录。根据患者是否同时接受ET和T-DM1分为两组。主要终点是确定无进展生存期(PFS),而次要终点是总生存期(OS),客观反应率,和治疗的安全性。结果:我们的分析检查了88例患者,其中32例(36.4%)接受T-DM1联合ET治疗。联合治疗显示中位数PFS显着改善(15.4vs.6.4个月;p=0.00004)和中位OS(35.0与23.1个月;p=0.026)与单独的T-DM1相比。组合组的ORR也较高(65.6%vs.29.3%;p=0.026)。与未接受帕妥珠单抗治疗的患者相比,先前接受帕妥珠单抗治疗的患者在T-DM1上的中位PFS降低(11.7vs.5.4个月,分别为;p<0.01)。与HER2+患者相比,T-DM1在HER23+患者中显示出更好的中位PFS。扩增率>2.0(10.8vs5.8个月,分别为;p=0.049)。安全性与以前的T-DM1研究一致。结论:T-DM1联合ET可显著改善HER2阳性和HR阳性MBC患者的PFS和OS。我们的研究表明,先前的帕妥珠单抗治疗加曲妥珠单抗治疗可能会降低T-DM1疗效。
    Background and Objectives: Patients with human epidermal growth factor receptor 2 (HER2) -positive, hormone receptor-positive (HR-positive) metastatic breast cancer (MBC) usually undergo trastuzumab emtansine (T-DM1) therapy in subsequent lines. Combining endocrine therapy (ET) with T-DM1 can improve treatment outcomes in this subtype. Therefore, this study aimed to investigate the benefits of using T-DM1 with ET in HER2-positive and HR-positive MBC. This study was the first to investigate the benefits of combining ET with T-DM1. Material and Methods: This study analyzed the medical records of patients with HER2-positive and HR-positive MBC who were treated with T-DM1 from June 2010 to December 2021. The patients were divided into groups based on whether they received concomitant ET with T-DM1. The primary endpoint was to determine the progression-free survival (PFS), while the secondary endpoints were overall survival (OS), objective response rate, and safety of the treatment. Results: Our analysis examined 88 patients, of whom 32 (36.4%) were treated with T-DM1 in combination with ET. The combination therapy showed a significant improvement in median PFS (15.4 vs. 6.4 months; p = 0.00004) and median OS (35.0 vs. 23.1 months; p = 0.026) compared to T-DM1 alone. The ORR was also higher in the combination group (65.6% vs. 29.3%; p = 0.026). Patients treated with pertuzumab priorly had reduced median PFS on T-DM1 compared to those who were not treated with pertuzumab (11.7 vs. 5.4 months, respectively; p < 0.01). T-DM1 demonstrated better median PFS in HER2 3+ patients compared to HER2 2+ patients, with an amplification ratio of >2.0 (10.8 vs 5.8 months, respectively; p = 0.049). The safety profiles were consistent with previous T-DM1 studies. Conclusions: The combination of T-DM1 with ET can significantly improve PFS and OS in patients with HER2-positive and HR-positive MBC. Our study suggests that prior pertuzumab treatment plus trastuzumab treatment might decrease T-DM1 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
    Vault是由78个主要的Vault蛋白(MVP)拷贝组成的真核核糖核蛋白,组装成基于体积的尺寸约为60纳米的纳米粒子,封闭其他蛋白质和RNA。不管他们的生理作用,金库代表理想,天然中空纳米粒子,由唯一的MVP组装而成。这里,我们已经在Komagataellaphaffi中表达并纯化了携带C末端Z肽(vault-Z)的MVP变体,可以紧密结合抗体的Fc部分,鉴于有针对性的交付。通过表面等离子体共振分析,我们可以确定单克隆抗体曲妥珠单抗(Tz)/vault-Z1:1相互作用的亲和力为2.5nM.然后,我们通过共孵育表征了溶液中的相互作用,超速离心,和沉淀蛋白质的分析。这显示了高达至少10:1的Tz/vault-Z比率的几乎不可逆的结合。作为概念的证明,我们用荧光团标记了Tz的Fc部分,并将其与纳米颗粒缀合,以及Tz或西妥昔单抗,另一种单克隆抗体。因此,我们可以证明抗体依赖,SKBR3和MDA-MB231乳腺癌细胞系的选择性摄取。这些调查提供了一部小说,灵活的技术平台,极大地扩展了Vault-Z\的应用程序,因为它可以与精细调节量的抗体以及其他分子稳定缀合,如荧光团,细胞靶向肽,或药物,使用Fc部分作为支架。
    Vaults are eukaryotic ribonucleoproteins consisting of 78 copies of the major vault protein (MVP), which assemble into a nanoparticle with an about 60 nm volume-based size, enclosing other proteins and RNAs. Regardless of their physiological role(s), vaults represent ideal, natural hollow nanoparticles, which are produced by the assembly of the sole MVP. Here, we have expressed in Komagataella phaffi and purified an MVP variant carrying a C-terminal Z peptide (vault-Z), which can tightly bind an antibody\'s Fc portion, in view of targeted delivery. Via surface plasmon resonance analysis, we could determine a 2.5 nM affinity to the monoclonal antibody Trastuzumab (Tz)/vault-Z 1:1 interaction. Then, we characterized the in-solution interaction via co-incubation, ultracentrifugation, and analysis of the pelleted proteins. This showed virtually irreversible binding up to an at least 10:1 Tz/vault-Z ratio. As a proof of concept, we labeled the Fc portion of Tz with a fluorophore and conjugated it with the nanoparticle, along with either Tz or Cetuximab, another monoclonal antibody. Thus, we could demonstrate antibody-dependent, selective uptake by the SKBR3 and MDA-MB 231 breast cancer cell lines. These investigations provide a novel, flexible technological platform that significantly extends vault-Z\'s applications, in that it can be stably conjugated with finely adjusted amounts of antibodies as well as of other molecules, such as fluorophores, cell-targeting peptides, or drugs, using the Fc portion as a scaffold.
    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
    嵌合抗原受体(CAR)T细胞疗法在治疗恶性肿瘤方面显示出希望。然而,使用人表皮生长因子受体2(HER2)CAR-T细胞具有严重毒性的风险,包括细胞因子释放综合征,由于他们对HER2的“上靶肿瘤外”识别。增强HER2CAR的质量和功能可以大大提高CAR-T细胞的治疗潜力。在这项研究中,我们开发了一种新型的抗HER2单克隆抗体,Ab8,其靶向HER2的结构域III,不同于曲妥珠单抗的结构域IV识别。尽管两种抗HER2mAb诱导了相似水平的抗体依赖性细胞毒性,基于曲妥珠单抗的CAR-T细胞对HER2阳性癌细胞表现出有效的抗肿瘤活性。总之,我们的研究结果提供了科学证据,即抗体对近侧膜结构域的识别可促进HER2特异性CAR-T细胞的抗肿瘤反应.
    Chimeric antigen receptor (CAR) T cell therapy shows promise in treating malignant tumors. However, the use of human epidermal growth factor receptor-2 (HER2) CAR-T cells carries the risk of severe toxicity, including cytokine release syndrome, due to their \"on-target off-tumor\" recognition of HER2. Enhancing the quality and functionality of HER2 CARs could greatly improve the therapeutic potential of CAR-T cells. In this study, we developed a novel anti-HER2 monoclonal antibody, Ab8, which targets domain III of HER2, distinct from the domain IV recognition of trastuzumab. Although two anti-HER2 mAbs induced similar levels of antibody-dependent cellular cytotoxicity, trastuzumab-based CAR-T cells exhibited potent antitumor activity against HER2-positive cancer cells. In conclusion, our findings provide scientific evidence that antibody recognition of the membrane-proximal domain promotes the anti-tumor response of HER2-specific CAR-T cells.
    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
    毒性表皮坏死松解症(TEN)是一种罕见的,但可能致命的皮肤黏膜反应,这可能是由于对某些药物的免疫反应而发生的。然而,由曲妥珠单抗触发的TEN极为罕见。早期诊断,认可,及时停止不良药物和开始类固醇治疗以及支持性管理是管理TEN的最重要措施。虽然罕见,重要的是要警惕与曲妥珠单抗相关的这种潜在不良反应,以确保患者安全并有助于改善结局.
    Toxic Epidermal Necrolysis (TEN) is a rare, but potentially fatal mucocutaneous reaction, that may occur due to an immunologic response to certain medications. However, TEN triggered by Trastuzumab is extremely rare. Early diagnosis, recognition, and prompt cessation of the offending drugs and initiation of steroid therapy with supportive management are the most important actions for managing TEN. Although rare, it is important to be vigilant about this potential adverse reactions associated with trastuzumab to ensure patient safety and contribute to better outcomes.
    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
    曲妥珠单抗的开发是20世纪最重要的癌症药物开发项目之一。曲妥珠单抗成为治疗人类表皮生长受体2(HER2)阳性乳腺癌的游戏者,对疾病复发和生存率有显著的积极影响。曲妥珠单抗的开发是癌症药物开发新时代的开始,这表明了我们了解分子病理生理学和药物作用机制的重要性。药物-诊断共同发展模式,其中该药物与预测性生物标志物测定平行开发,对今天的癌症药物开发产生了重大影响,当涉及到临床富集试验设计时,我们感谢曲妥珠单抗.曲妥珠单抗并不是唯一针对HER2蛋白开发的药物。在过去的几十年里,已经开发了几种新的HER2靶向疗法,包括小分子酪氨酸激酶抑制剂(TKI),单克隆抗体,和抗体-药物缀合物(ADC)。特别是,ADC曲妥珠单抗deruxtecan似乎为HER2靶向治疗提供了新的途径,不仅针对乳腺癌,也适用于胃癌和非小细胞肺癌。随着曲妥珠单抗作为参考点的发展,本文将对HER2靶向治疗的疗效进行简要总结,包括HER2阳性测试,因为它在过去25年中不断发展。
    The development of trastuzumab is among the most significant cancer drug development projects in the 20th century. Trastuzumab became a gamechanger for the treatment of human epidermal growth receptor 2 (HER2) positive breast cancer, with a significant positive impact on disease recurrence and survival. The development of trastuzumab was the beginning of a new era of cancer drug development, which showed us the importance of understanding the molecular pathophysiology and drug mechanism of action. The drug-diagnostic codevelopment model, in which the drug is developed in parallel with a predictive biomarker assay, has had a significant impact on today\'s cancer drug development, and we are indebted to trastuzumab when it comes to the clinical enrichment trial design. Trastuzumab is not the only drug developed to target the HER2 protein. Over the past few decades, several new HER2 targeted therapies have been developed, including small-molecule tyrosine kinase inhibitors (TKI), monoclonal antibodies, and antibody-drug conjugates (ADC). In particular, the ADC trastuzumab deruxtecan seems to pave new avenues when it comes to HER2 targeted treatment not only for breast cancer, but also for gastric cancer and non-small cell lung cancer. With the development of trastuzumab as a reference point, this article will provide a brief summary of the efficacy of HER2 targeted therapy, including testing for HER2 positivity, as it has evolved over the past 25 years.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号