Endocrine Resistance

内分泌抵抗
  • 文章类型: Journal Article
    乳腺癌是女性死亡的主要原因,尽管个性化治疗取得了进展,由于耐药性,转移性疾病在很大程度上仍然无法治愈。雌激素受体(ER,ESR1)在所有乳腺癌的三分之二中表达,在内分泌压力下,体细胞ESR1突变出现在30%的导致内分泌抵抗的病例中。我们和其他人报道了ESR1融合是ER介导的内分泌抵抗的机制。ER融合,保留AF1和DNA结合域,携带ESR1外显子1-6与框内基因配偶体融合,导致ER配体结合结构域(LBD)的丢失。我们证明,在非特殊类型(浸润性导管癌(IDC)-NST)和浸润性小叶癌(ILC)细胞系中,ER融合表现出与雌二醇或抗内分泌疗法无关的经典ER信号通路的强大过度激活。我们采用稳定过表达ER融合的细胞系模型,同时内源性ER敲低,以最大程度地减少内源性ER的影响。细胞系在已知是转移性疾病驱动因素的途径中表现出共同的转录组富集,特别是MYC信号。表达3'融合配偶体SOX9和YAP1的细胞一致地表现出增强的生长和细胞存活。表达DAB2融合的ILC细胞导致生长增强,生存,和迁移;在IDC-NSTDAB2模型中不认识的表型。在这里,我们报告说细胞系活动是亚型-,fusion-,和试验特异性表明LBD损失,聚变伙伴,和细胞景观都影响融合活动。因此,评估融合频率至关重要,在肿瘤临床病理的背景下。
    Breast cancer is a leading cause of female mortality and despite advancements in personalized therapeutics, metastatic disease largely remains incurable due to drug resistance. The estrogen receptor (ER, ESR1) is expressed in two-thirds of all breast cancer, and under endocrine stress, somatic ESR1 mutations arise in approximately 30% of cases that result in endocrine resistance. We and others reported ESR1 fusions as a mechanism of ER-mediated endocrine resistance. ER fusions, which retain the activation function 1- and DNA-binding domains, harbor ESR1 exons 1 to 6 fused to an in-frame gene partner resulting in loss of the ER ligand-binding domain (LBD). We demonstrate that in a no-special type (invasive ductal carcinoma [IDC]-NST) and an invasive lobular carcinoma (ILC) cell line, ER fusions exhibit robust hyperactivation of canonical ER signaling pathways independent of estradiol or antiendocrine therapies. We employ cell line models stably overexpressing ER fusions with concurrent endogenous ER knockdown to minimize endogenous ER influence. Cell lines exhibited shared transcriptomic enrichment in pathways known to be drivers of metastatic disease, notably MYC signaling. Cells expressing the 3\' fusion partners SOX9 and YAP1 consistently demonstrated enhanced growth and cell survival. ILC cells expressing the DAB2 fusion led to enhanced growth, survival, and migration, phenotypes not appreciated in the IDC-NST DAB2 model. Herein, we report that cell line activity is subtype-, fusion-, and assay-specific, suggesting that LBD loss, the fusion partner, and the cellular landscape all influence fusion activities. Therefore, it will be critical to assess fusion frequency in the context of the clinicopathology.
    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
    对内分泌治疗的获得性抗性仍然是主要的临床挑战。在这项研究中,我们发现desmoglein-2(DSG2)在ER+乳腺癌(BC)的获得性内分泌抵抗和细胞可塑性中起主要作用。通过使用单细胞RNA-seq分析完善的氟维司群抗性ER+BC模型,我们发现,ER抑制导致癌细胞群中DSG2的特异性增加,这反过来又增强了桥粒在体外和体内的形成和细胞表型可塑性,促进对治疗的抗性。DSG2耗竭减少氟维司群耐药异种移植模型中的肿瘤发生和转移,并提高氟维司群效率。机械上,DSG2与JUP和波形蛋白形成桥粒复合体并触发Wnt/PCP信号传导。我们发现DSG2水平升高,随着ER水平的降低和Wnt/PCP通路的激活,预测生存不佳,这表明DSG2high签名可用于治疗干预。我们的分析强调了抗雌激素治疗后DSG2介导的桥粒连接的关键作用。
    Acquired resistance to endocrine treatments remains a major clinical challenge. In this study, we found that desmoglein-2 (DSG2) plays a major role in acquired endocrine resistance and cellular plasticity in ER+ breast cancer (BC). By analysing the well-established fulvestrant-resistant ER+ BC model using single-cell RNA-seq, we revealed that ER inhibition leads to a specific increase in DSG2 in cancer cell populations, which in turn enhances desmosome formation in vitro and in vivo and cell phenotypic plasticity that promotes resistance to treatment. DSG2 depletion reduced tumorigenesis and metastasis in fulvestrant-resistant xenograft models and promoted fulvestrant efficiency. Mechanistically, DSG2 forms a desmosome complex with JUP and Vimentin and triggers Wnt/PCP signalling. We showed that elevated DSG2 levels, along with reduced ER levels and an activated Wnt/PCP pathway, predicted poor survival, suggesting that a DSG2high signature could be exploited for therapeutic interventions. Our analysis highlighted the critical role of DSG2-mediated desmosomal junctions following antiestrogen treatment.
    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
    乳腺癌(BC)是一种高度异质性的肿瘤,已超过肺癌,成为女性最常诊断的癌症。在临床实践中,治疗雌激素受体α(ERα)阳性BC的主要方法是通过内分泌治疗,这涉及使用他莫昔芬和氟维司群等药物靶向ERα。然而,从头或获得性耐药的问题提出了重大的临床挑战,强调开发新的治疗策略的迫切需要。在这方面,我们已经成功地设计和开发了一种名为OBHSA的新型选择性雌激素受体降解剂(SERD),特异性靶向并降解ERα,表现出显著的疗效。我们的发现揭示了OBHSA抑制各种BC细胞增殖的有效性,包括他莫昔芬敏感和他莫昔芬耐药的BC细胞,表明其克服内分泌抵抗的巨大潜力。在机制方面,我们发现OBHSA通过两种不同的途径克服了他莫昔芬耐药性.首先,OBHSA以ERα依赖性方式降解细胞周期蛋白D1,从而阻断细胞周期。其次,OBHSA诱导细胞内活性氧的升高,引发未折叠蛋白反应(UPR)的过度激活,并最终导致细胞凋亡。总之,我们的发现表明OBHSA通过诱导细胞周期阻滞和UPR介导的细胞凋亡发挥抗肿瘤作用.这些发现为开发针对内分泌抗性BC的新型治疗药物提供了希望。
    Breast cancer (BC) is a highly heterogeneous tumor that has surpassed lung cancer as the most frequently diagnosed cancer in women. In clinical practice,the primary approach for treating estrogen receptor alpha (ERα)-positive BC is through endocrine therapy, which involves targeting the ERα using medications like tamoxifen and fulvestrant. However, the problem of de novo or acquired resistance poses a significant clinical challenge, emphasizing the critical need for the development of novel therapeutic strategies. In this regard, we have successfully designed and developed a novel selective estrogen receptor degrader (SERD) called OBHSA, which specifically targets and degrades ERα, demonstrating remarkable efficacy. Our findings revealed the effectiveness of OBHSA in inhibiting the proliferation of various BC cells, including both tamoxifen-sensitive and tamoxifen-resistant BC cells, indicating its great potential to overcome endocrine resistance. In terms of mechanism, we discovered that OBHSA overcame tamoxifen resistance through two distinct pathways. Firstly, OBHSA degraded cyclin D1 in an ERα-dependent manner, thereby blocking the cell cycle. Secondly, OBHSA induced an elevation in intracellular reactive oxygen species, triggering an excessive activation of the unfolded protein response (UPR) and ultimately leading to apoptotic cell death. In summary, our finding demonstrated that OBHSA exerts anti-tumor effects by inducing cell cycle arrest and UPR-mediated apoptosis. These findings hold promise for the development of novel therapeutic drugs targeting endocrine-resistant BC.
    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
    成纤维细胞生长因子受体(FGFR)在雌激素受体阳性(ER+)乳腺癌中通过促进肿瘤发生和内分泌抵抗发挥重要作用。这篇评论探讨了结构,信号通路,以及FGFRs的含义,特别是ER+乳腺癌中的FGFR1、FGFR2、FGFR3和FGFR4。FGFR1经常被扩增,尤其是侵袭性管腔B样肿瘤,其扩增与不良预后和治疗耐药性有关。FGFR1与癌基因如EIF4EBP1和NSD3的共扩增使其作为独立的致癌驱动因子的作用复杂化。FGFR2扩增,虽然不太常见,在激素受体调节中至关重要,驱动增殖和治疗抗性。FGFR3和FGFR4也通过各种机制促进内分泌抵抗,包括PI3K/AKT/mTOR和RAS/RAF/MEK/ERK等替代信号通路的激活。内分泌耐药仍然是一个主要的临床挑战,大约70%的乳腺癌最初激素受体阳性。尽管CDK4/6抑制剂联合内分泌治疗(ET)取得了成功,抵抗力经常发展,需要新的治疗策略。FGFR抑制剂在临床前研究中显示出潜力,但由于脱靶毒性和缺乏预测性生物标志物,临床试验取得的成功有限.目前的临床试验,包括那些评估FGFR抑制剂如erdafitinib的人,lucitanib,还有多替尼,表现出混合的结果,强调乳腺癌中FGFR信号的复杂性。FGFR和其他信号通路之间的相互作用凸显了对全面分子谱分析和个性化治疗方法的需求。未来的研究应该集中在识别强大的生物标志物和开发联合疗法,以提高FGFR靶向治疗的疗效。总之,在ER+乳腺癌中靶向FGFR信号既是挑战也是机遇.更深入地了解分子机制和耐药途径对于FGFR抑制剂成功融入临床实践至关重要。旨在改善内分泌耐药乳腺癌患者的预后。
    Fibroblast Growth Factor Receptors (FGFRs) play a significant role in Estrogen Receptor-positive (ER+) breast cancer by contributing to tumorigenesis and endocrine resistance. This review explores the structure, signaling pathways, and implications of FGFRs, particularly FGFR1, FGFR2, FGFR3, and FGFR4, in ER+ breast cancer. FGFR1 is frequently amplified, especially in aggressive Luminal B-like tumors, and its amplification is associated with poor prognosis and treatment resistance. The co-amplification of FGFR1 with oncogenes like EIF4EBP1 and NSD3 complicates its role as a standalone oncogenic driver. FGFR2 amplification, though less common, is critical in hormone receptor regulation, driving proliferation and treatment resistance. FGFR3 and FGFR4 also contribute to endocrine resistance through various mechanisms, including the activation of alternate signaling pathways like PI3K/AKT/mTOR and RAS/RAF/MEK/ERK. Endocrine resistance remains a major clinical challenge, with around 70% of breast cancers initially hormone receptor positive. Despite the success of CDK 4/6 inhibitors in combination with endocrine therapy (ET), resistance often develops, necessitating new treatment strategies. FGFR inhibitors have shown potential in preclinical studies, but clinical trials have yielded limited success due to off-target toxicities and lack of predictive biomarkers. Current clinical trials, including those evaluating FGFR inhibitors like erdafitinib, lucitanib, and dovitinib, have demonstrated mixed outcomes, underscoring the complexity of FGFR signaling in breast cancer. The interplay between FGFR and other signaling pathways highlights the need for comprehensive molecular profiling and personalized treatment approaches. Future research should focus on identifying robust biomarkers and developing combination therapies to enhance the efficacy of FGFR-targeted treatments. In conclusion, targeting FGFR signaling in ER+ breast cancer presents both challenges and opportunities. A deeper understanding of the molecular mechanisms and resistance pathways is crucial for the successful integration of FGFR inhibitors into clinical practice, aiming to improve outcomes for patients with endocrine-resistant breast cancer.
    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
    针对雌激素受体(ER/ESR1)的内分泌疗法是治疗ER阳性乳腺癌患者的基石。但是阻力往往限制了它们的有效性。因此,了解分子机制是优化现有药物和开发新的ER调节剂的关键。尽管报告数据的分散方式减少了其潜在影响,但已经取得了显著进展。这里,我们介绍EstroGene2.0,它的前身1.0版本的扩展数据库。EstroGene2.0专注于乳腺癌模型中对内分泌疗法的反应和抗性。整合了来自28个细胞系的212项研究的361个实验的多维分析,用户友好的浏览器提供全面的数据可视化和元数据挖掘功能(https://estrogeneii。Web。app/)。利用统一的数据收集,我们的后续荟萃分析显示,对不同类型的ER调节剂(包括SERM)的反应存在很大差异,SERD,SERCA和LDD/PROTAC。值得注意的是,内分泌抗性模型表现出一系列转录组改变,包括ER和干扰素信号的反向移位,这是临床上概括的。此外,解剖多个ESR1突变细胞模型揭示了基因组编辑与异位过表达模型工程的不同临床相关性,并确定了高置信度突变ER靶标,例如NPY1R。这些例子证明了EstroGene2.0如何帮助研究乳腺癌对内分泌疗法的反应并探索耐药机制。
    Endocrine therapies targeting the estrogen receptor (ER/ESR1) are the cornerstone to treat ER-positive breast cancers patients, but resistance often limits their effectiveness. Understanding the molecular mechanisms is thus key to optimize the existing drugs and to develop new ER-modulators. Notable progress has been made although the fragmented way data is reported has reduced their potential impact. Here, we introduce EstroGene2.0, an expanded database of its precursor 1.0 version. EstroGene2.0 focusses on response and resistance to endocrine therapies in breast cancer models. Incorporating multi-omic profiling of 361 experiments from 212 studies across 28 cell lines, a user-friendly browser offers comprehensive data visualization and metadata mining capabilities (https://estrogeneii.web.app/). Taking advantage of the harmonized data collection, our follow-up meta-analysis revealed substantial diversity in response to different classes of ER-modulators including SERMs, SERDs, SERCA and LDD/PROTAC. Notably, endocrine resistant models exhibit a spectrum of transcriptomic alterations including a contra-directional shift in ER and interferon signaling, which is recapitulated clinically. Furthermore, dissecting multiple ESR1-mutant cell models revealed the different clinical relevance of genome-edited versus ectopic overexpression model engineering and identified high-confidence mutant-ER targets, such as NPY1R. These examples demonstrate how EstroGene2.0 helps investigate breast cancer\'s response to endocrine therapies and explore resistance mechanisms.
    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
    目的本研究旨在确定在国家癌症研究所(INCan)接受治疗的西班牙裔墨西哥乳腺癌(BC)患者队列中内分泌抵抗的患病率。此外,确定了与内分泌抵抗相关的临床病理因素,并探讨了它们对患者生存的影响。方法回顾性分析2012年至2016年间200例接受INCan治疗的雌激素受体(ER)和孕激素受体(PR)阳性肿瘤患者的临床资料。根据《乳腺癌进展国际共识指南2》的定义对内分泌耐药进行了定义。分析其临床病理特征,以确定与内分泌抵抗的存在。我们使用了敏感性分析和多变量调整逻辑回归,卡普兰-迈耶曲线,和多变量调整后的Cox回归。P值<0.05被认为具有统计学意义。结果32.5%的患者出现内分泌耐药。激素抵抗和敏感性之间的区别受肿瘤大小和淋巴结状态的影响。内分泌抵抗病例的平均直径为7.15cm,而非内分泌病例的平均直径为5.71cm,在20%的内分泌抵抗病例中存在N3状态,而在非内分泌病例中只有2.2%(p值<0.001)。临床分期与内分泌抵抗密切相关(风险比[RR]4.39,95%置信区间[95CI]1.50,11.43)。此外,内分泌抵抗显著影响随访期间的死亡率,在多变量调整模型中,危险比[HR]为23.7(95CI5.20,108.42)。然而,一个完整的病理反应降低了内分泌抵抗的风险,风险比(RR)为0.15(95%CI0.03,0.75)。结论西班牙裔墨西哥BC患者诊断为晚期临床分期可预测内分泌抵抗。局部晚期疾病患者的完全病理反应也是内分泌抵抗的关键预测因子。这些结果表明,内分泌抵抗是随访期间BC的关键因素。
    UNASSIGNED: This study aimed to determine the prevalence of endocrine resistance in a cohort of Hispanic Mexican breast cancer (BC) patients receiving care at Instituto Nacional de Cancerología (INCan). Additionally, the clinical-pathological factors associated with endocrine resistance were identified, and their impact on patient survival was explored.
    UNASSIGNED: A retrospective analysis of 200 BC patients who attended INCan between 2012 and 2016 with estrogen receptor (ER) and progesterone receptor (PR) positive tumors was made. Endocrine resistance was defined according to the International Consensus Guidelines for Advance Breast Cancer 2 definition. Their clinicopathological characteristics were analyzed to determine the association with endocrine resistance presence. We used sensitivity analyses and multivariate-adjusted logistic regressions, Kaplan-Meier curves, and multivariate-adjusted Cox regressions. P-value < 0.05 was considered as statistically significant.
    UNASSIGNED: Endocrine resistance was observed in 32.5% of patients included in this study. The distinction between hormone resistance and sensitivity was influenced by tumor size and node status. It had a mean diameter of 7.15 cm in endocrine resistance cases compared to 5.71 cm in non-endocrine, with N3 status present in 20% of endocrine resistance cases versus only 2.2% in non-endocrine (p-value < 0.001). The clinical stage exhibited a strong association with endocrine resistance (Risk Ratio [RR] 4.39, 95% confidence interval [95%CI] 1.50, 11.43). Furthermore, endocrine resistance significantly impacted mortality during the follow-up, with a Hazard Ratio [HR] of 23.7 (95%CI 5.20, 108.42) in multivariable-adjusted models. However, a complete pathological response reduced the endocrine resistance risk, as demonstrated by a Risk Ratio (RR) of 0.15 (95% CI 0.03, 0.75).
    UNASSIGNED: Advanced clinical stage at diagnosis predicted endocrine resistance in Hispanic Mexican BC patients. Complete pathologic response in locally advanced disease patients was also a key predictor of endocrine resistance. These results indicated that endocrine resistance was a critical factor in BC during follow-up.
    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
    低级别浆液性卵巢癌(LGSOC)是一种罕见的卵巢恶性肿瘤,主要影响年轻女性,其特征是缓慢的生长模式。它表现出惰性生长和高雌激素/孕激素受体表达,提示对内分泌治疗的潜在反应。然而,由于内分泌抵抗的发展,治疗效果仍然有限。抵抗的机制,无论是初级的还是后天的,在很大程度上仍然未知,并且在这些患者中通过内分泌治疗实现良好的治疗结果存在重大障碍。在雌激素受体阳性乳腺癌中,内分泌抵抗的机制已经得到了广泛的探索,并且已经出现了克服抵抗的新治疗策略。考虑到LGSOC和乳腺癌共同的雌激素受体阳性,我们想探讨是否存在任何平行的耐药机制,以及我们是否可以将内分泌乳腺癌治疗扩展到LGSOC.这篇综述旨在强调可能驱动卵巢癌内分泌抵抗的潜在分子机制。同时还探索克服这种耐药性的可用治疗机会。通过解开所涉及的潜在途径并检查新兴战略,这篇综述探讨了在LGSOC中推进治疗方案和改善患者预后的宝贵见解,有限的治疗选择。
    Low-grade serous ovarian cancer (LGSOC) is a rare ovarian malignancy primarily affecting younger women and is characterized by an indolent growth pattern. It exhibits indolent growth and high estrogen/progesterone receptor expression, suggesting potential responsiveness to endocrine therapy. However, treatment efficacy remains limited due to the development of endocrine resistance. The mechanisms of resistance, whether primary or acquired, are still largely unknown and present a significant hurdle in achieving favorable treatment outcomes with endocrine therapy in these patients. In estrogen receptor-positive breast cancer, mechanisms of endocrine resistance have been largely explored and novel treatment strategies to overcome resistance have emerged. Considering the shared estrogen receptor positivity in LGSOC and breast cancer, we wanted to explore whether there are any parallel mechanisms of resistance and whether we can extend endocrine breast cancer treatments to LGSOC. This review aims to highlight the underlying molecular mechanisms possibly driving endocrine resistance in ovarian cancer, while also exploring the available therapeutic opportunities to overcome this resistance. By unraveling the potential pathways involved and examining emerging strategies, this review explores valuable insights for advancing treatment options and improving patient outcomes in LGSOC, which has limited therapeutic options available.
    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
    乳腺癌是女性最常见的癌症类型。绝大多数乳腺癌患者具有激素受体阳性(HR+)肿瘤。在晚期HR+乳腺癌中,内分泌治疗联合细胞周期蛋白依赖性激酶4/6(CDK4/6)抑制剂被认为是一线治疗的标准.然而,最终会出现对激素治疗和CDK4/6抑制剂的抵抗,导致疾病的进展。抗体-药物缀合物(ADC)是一种有希望的治疗选择,对HR+乳腺癌患者具有显著疗效。对内分泌治疗有抵抗力。ADC通常由通过接头连接到靶向特定肿瘤相关抗原的单克隆抗体的细胞毒性有效载荷组成。提供了更有选择性地将化疗药物递送到癌细胞的优势。在这次审查中,我们专注于ADC的作用机制,它们的毒性特征和治疗用途以及相关的生物标志物和晚期HR+乳腺癌的未来前景。
    Breast cancer is the most common cancer type in women. The vast majority of breast cancer patients have hormone receptor-positive (HR+) tumors. In advanced HR+ breast cancer, the combination of endocrine therapy with cyclin-dependent kinase 4/6 (CDK4/6) inhibitors is considered the standard of care in the front-line setting. Nevertheless, resistance to hormonal therapy and CDK4/6 inhibitors eventually occurs, leading to progression of the disease. Antibody-drug conjugates (ADCs) comprise a promising therapeutic choice with significant efficacy in patients with HR+ breast cancer, which is resistant to endocrine treatment. ADCs typically consist of a cytotoxic payload attached by a linker to a monoclonal antibody that targets a specific tumor-associated antigen, offering the advantage of a more selective delivery of chemotherapy to cancer cells. In this review, we focus on the ADC mechanisms of action, their toxicity profile and therapeutic uses as well as on related biomarkers and future perspectives in advanced HR+ breast cancer.
    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:更新CDK4/6抑制剂(CDK4/6i)治疗激素受体(HR)阳性的最新临床证据,人表皮生长因子受体(HER)2阴性乳腺癌。
    结果:在过去的十年中,CDK4/6i已成为转移性和高风险早期HR+/HER2-乳腺癌患者护理治疗标准的一部分。三个可用的CDK4/6i(palbociclib,ribociclib和abemaciclib)与内分泌治疗(ET)联合治疗转移性乳腺癌(mBC)已得到广泛研究,可持续延长无进展生存期;然而,瑞博西尼已成为mBC的首选一线药物,其总体生存获益优于内分泌单一疗法.在公元前早期,abemaciclib是目前唯一被批准的药物,而ribociclib具有早期阳性的临床试验数据。毒性和经济负担限制了CDK4/6i在所有患者和资源贫乏环境中的使用,在mBC中使用它们的最佳时机仍不清楚。有相当多的证据表明CDK4/6i在转移性和早期HR+/HER2-乳腺癌中的应用,但是知识差距仍然存在,需要进一步的研究来更好地定义它们的最佳使用。
    OBJECTIVE: Update on the most recent clinical evidence on CDK4/6 inhibitors (CDK4/6i) in the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor (HER)2-negative breast cancer.
    RESULTS: Over the past decade, CDK4/6i have become part of the standard of care treatment of patients with both metastatic and high-risk early HR + /HER2- breast cancers. The three available CDK4/6i (palbociclib, ribociclib and abemaciclib) have been extensively studied in combination with endocrine therapy (ET) in metastatic breast cancer (mBC) with consistent prolongation of progression free survival; however, ribociclib has emerged as the preferred first line agent in mBC given overall survival benefit over endocrine monotherapy. In early BC, abemaciclib is the only currently approved agent while ribociclib has early positive clinical trial data. Toxicities and financial burden limit the use of CDK4/6i in all patients and resource-poor settings, and optimal timing of their use in mBC remains unclear. There is considerable evidence for the use of CDK4/6i in metastatic and early HR + /HER2- breast cancer, but knowledge gaps remain, and further research is necessary to better define their optimal use.
    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
    使用细胞周期蛋白依赖性激酶4/6抑制剂(CDK4/6i)的内分泌疗法(ET)目前是大多数激素受体阳性(HR)和人表皮生长受体2阴性(HER2-)患者的一线标准治疗方法转移性或晚期乳腺癌。然而,大多数肿瘤对CDK4/6is产生反应并最终产生耐药性。抗药性的机制知之甚少,在快速变化的治疗环境中,最佳的进展后治疗方案及其顺序继续发展。在这次审查中,我们总结了CDK4/6is和ET的耐药机制,并描述使用小分子抑制剂的临床试验结果,抗体-药物偶联物和免疫治疗,提供这些新策略如何逆转治疗耐药性的见解,并讨论一些没有转化为临床益处。最后,我们根据当前新出现的证据提供合理的治疗策略.
    Endocrine therapy (ET) with a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) is currently the first-line standard treatment for most patients with hormone receptor-positive (HR+) and human epidermal growth receptor 2-negative (HER2-) metastatic or advanced breast cancer. However, the majority of tumors response to and eventually develop resistance to CDK4/6is. The mechanisms of resistance are poorly understood, and the optimal postprogression treatment regimens and their sequences continue to evolve in the rapidly changing treatment landscape. In this review, we generally summarize the mechanisms of resistance to CDK4/6is and ET, and describe the findings from clinical trials using small molecule inhibitors, antibody-drug conjugates and immunotherapy, providing insights into how these novel strategies may reverse treatment resistance, and discussing how some have not translated into clinical benefit. Finally, we provide rational treatment strategies based on the current emerging evidence.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号