gilteritinib

吉特替尼
  • 文章类型: Journal Article
    一名72岁女性复发性FLT3-ITD阳性急性髓系白血病患者接受gilteritinib治疗,并获得完全缓解,血液学恢复不完全。然而,两个月后,她的右眼出现视神经浸润和视力丧失,同时维持gilteritinib的血液学缓解.鞘内注射细胞毒性药物减少了脑脊液(CSF)中的母细胞数,但她的视力没有恢复.视神经浸润开始时,Gilteritinib的剂量为80毫克/天,gilteritinib的血浆谷和CSF水平分别为151.9ng/ml和1.9ng/ml,分别,中枢神经系统(CNS)穿透率为1.3%。40天后检测到血液学进行性疾病(PD),病人一个月后死亡。血液PD时的目标测序显示FLT3F691L突变,这是已知的赋予耐药gilteritinib。在这个病人身上,药代动力学(gilteritinib的低CNS渗透)和药效学(获得耐药突变)机制被认为是CNS复发和血液学PD的原因,分别。考虑到文献中关于FLT3抑制剂的中枢神经系统渗透及其对中枢神经系统疾病的影响的数据的缺乏,我们认为这是一个有价值的案例。
    A 72-year-old woman with relapsed FLT3-ITD-positive acute myeloid leukemia was treated with gilteritinib and achieved complete remission with incomplete hematological recovery. However, two months later, she developed optic nerve infiltration and lost vision in her right eye while maintaining hematological remission on gilteritinib. Intrathecal injection of cytotoxic drugs reduced the number of blasts in the cerebrospinal fluid (CSF), but her vision did not recover. At the onset of optic nerve infiltration, at a dose of 80 mg/day gilteritinib, the plasma trough and CSF levels of gilteritinib were 151.9 ng/ml and 1.9 ng/ml, respectively, with a central nervous system (CNS) penetration rate of 1.3%. Hematologic progressive disease (PD) was detected after 40 days, and the patient died one month later. Target sequencing at the time of hematologic PD revealed the FLT3 F691L mutation, which is known to confer resistance to gilteritinib. In this patient, pharmacokinetic (low CNS penetration of gilteritinib) and pharmacodynamic (acquisition of a drug resistance mutation) mechanisms were thought to be responsible for the CNS relapse and hematologic PD, respectively. We believe this is a valuable case to report considering the scarcity of data on CNS penetration of FLT3 inhibitors and their effects on CNS disease in the literature.
    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: Case Reports
    维奈托克,选择性BCL-2抑制剂,在AML的治疗中显示出优越的疗效。然而,一些患有AML1-ETO的AML患者对venetoclax治疗反应较差.在这份报告中,1例复发/难治性(R/R)venetoclax耐药AML1-ETO阳性AML患者在接受吉利替尼和维奈托克联合治疗后,肿瘤快速消退.其他实验室发现表明,两种药物的综合作用可能与综合应激反应的诱导有关。本病例为FLT3野生型RR的治疗提供了一种新的治疗方法,对venetoclax有原发性耐药性的AML1-ETOAML患者。
    Venetoclax, a selective BCL-2 inhibitor, has shown superior efficacy in the treatment of AML. Nevertheless, some AML patients with AML1-ETO respond poorly to venetoclax treatment. In this report, a relapsed/refractory (R/R) venetoclax resistant AML1-ETO positive AML patient showed rapid tumor regression after combination therapy with gilteritinib and venetoclax. Additional laboratory findings indicated that the combined impact of the two drugs may be associated with the induction of the integrated stress response. This case presents a novel therapeutic approach for the treatment of FLT3 wild-type RR, AML1-ETO AML patients who have primary resistance to venetoclax.
    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
    FMS相关的酪氨酸激酶3(FLT3)抑制剂gilteritinib是复发性/难治性(R/R)FLT3突变(FLT3mut)急性髓性白血病(AML)的标准疗法,但总生存率(OS)仅为大约20%,很少患者达到深度和/或持久的反应。我们回顾性分析了29例接受三联疗法(gilteritinib+venetoclax[VEN]+阿扎胞苷[AZA])治疗的R/RFLT3mutAML患者。19例患者(65.5%)曾接受过FLT3抑制剂治疗。改良复合完全缓解(mCRc)率为62.1%(n=18;CR,4/29,13.8%;CRi,6/29,20.7%;MLFS,8/29,27.6%)。在获得mCRc的18例患者中,FLT3-PCR阴性为94.4%(n=17),流式细胞术阴性为77.7%(n=14)。10例无FLT3TKI暴露的患者的mCRc率为70%(n=7),19例有FLT3TKI暴露的患者的mCRc率为57.8%(n=11)(P=0.52)。在第一个周期结束时,在反应者中,ANC>0.5×109/L的中位时间为38天,血小板>50×109/L的中位时间为31天,但60天死亡率为0%。所有R/RFLT3mut患者的2年OS估计为60.9%。1年OS为80%和58.8%的患者没有和先前的FLT3TKI暴露,分别为(P=0.79)。在三联疗法后接受allo-HSCT的19例患者(65.5%)中,估计的2年OS为62%,在未接受allo-HSCT的10例患者中为37%(P=0.03)。总之,gilteritinib三联疗法,VEN,AZA是有效和安全的,是R/RFLT3mutAML的优秀前线选择。
    The FMS-related tyrosine kinase 3 (FLT3) inhibitor gilteritinib is standard therapy for relapsed/refractory (R/R) FLT3-mutated (FLT3mut) acute myeloid leukemia (AML) but the overall survival (OS) is only approximately 20 % and few patients achieve deep and/ or durable response. We retrospectively analyzed 29 R/R FLT3mut AML patients treated on triplet regimens (gilteritinib+ venetoclax[VEN] +azacitidine[AZA]). Nineteen patients (65.5 %) had received prior FLT3 inhibitor therapy. The modified composite complete remission (mCRc) rate was 62.1 % (n = 18; CR, 4/29,13.8 %; CRi, 6/29, 20.7 %; MLFS, 8/29, 27.6 %). Among 18 patients achieved mCRc, FLT3-PCR negativity was 94.4 % (n=17), and flow-cytometry negativity was 77.7 % (n=14). The mCRc rate was 70 % (n=7) in 10 patients without prior FLT3 TKI exposure and 57.8 % (n=11) in 19 patients with prior FLT3 TKI exposure (P=0.52). At the end of the first cycle, the median time to ANC > 0.5× 109/L was 38 days and platelet > 50× 109/L was 31 days among responders, but 60-day mortality was 0 %. The estimated 2-year OS was 60.9 % for all R/R FLT3mut patients. The 1-year OS was 80 % and 58.8 % in patients without and with prior FLT3 TKI exposure, respectively (P=0.79). The estimated 2-year OS was 62 % in 19 (65.5 %) patients who received allo-HSCT after triplet therapy and 37 % in 10 patients who did not receive allo-HSCT (P=0.03). In conclusion, triplet therapy with gilteritinib, VEN, and AZA is effective and safe and an excellent frontline option for R/R FLT3mut AML.
    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
    急性髓性白血病(AML)由于其频繁复发,儿科和成人患者的生存率均较低。为了阐明AML复发的生物能量原理,我们研究了线粒体-核双基因组的转录调控,这些基因组负责治疗抗性母细胞的代谢可塑性.功能的获得和丧失结果表明,NFκB2是NFκB(活化B细胞的核因子κ-轻链增强子)家族的非经典转录因子(TF),可以控制TFAM(线粒体转录因子A)的表达,已知这对于代谢生物发生至关重要。此外,遗传追踪和启动子分析显示,NFκB2在线粒体中,可以结合调节D-loop域的特定“TTGGGGGGTG”区域,以激活轻链启动子(LSP)和重链启动子1(HSP1),线粒体基因组的启动子。基于我们发现NFκB2调节线粒体-核双重基因组的新功能,我们探索了一种新的三联疗法,包括NFκB2,酪氨酸激酶的抑制剂,和线粒体ATP合酶可有效消除具有FMS相关受体酪氨酸激酶3(FLT3)突变的原发性AML母细胞,并对离体对照细胞表现出最小的毒性。因此,AML的有效治疗必须包括对介导代谢可塑性的双基因组的强抑制作用,以改善白血病预后.
    Acute myeloid leukemia (AML) has a poor survival rate for both pediatric and adult patients due to its frequent relapse. To elucidate the bioenergetic principle underlying AML relapse, we investigated the transcriptional regulation of mitochondrial-nuclear dual genomes responsible for metabolic plasticity in treatment-resistant blasts. Both the gain and loss of function results demonstrated that NFκB2, a noncanonical transcription factor (TF) of the NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) family, can control the expression of TFAM (mitochondrial transcription factor A), which is known to be essential for metabolic biogenesis. Furthermore, genetic tracking and promoter assays revealed that NFκB2 is in the mitochondria and can bind the specific \"TTGGGGGGTG\" region of the regulatory D-loop domain to activate the light-strand promoter (LSP) and heavy-strand promoter 1 (HSP1), promoters of the mitochondrial genome. Based on our discovery of NFκB2\'s novel function of regulating mitochondrial-nuclear dual genomes, we explored a novel triplet therapy including inhibitors of NFκB2, tyrosine kinase, and mitochondrial ATP synthase that effectively eliminated primary AML blasts with mutations of the FMS-related receptor tyrosine kinase 3 (FLT3) and displayed minimum toxicity to control cells ex vivo. As such, effective treatments for AML must include strong inhibitory actions on the dual genomes mediating metabolic plasticity to improve leukemia prognosis.
    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
    急性髓细胞性白血病(AML)是一种遗传多样性和挑战性的恶性肿瘤,FLT3基因突变是特别常见和有害的。Gilteritinib,一种有效的FLT3抑制剂,已被美国食品和药物管理局(FDA)批准用于治疗具有FLT3突变的复发/难治性AML。尽管gilteritinib是基于其对FLT3激酶的抑制活性而开发的,了解其抗白血病活性的确切机制对于管理耐药性和发现生物标志物非常重要.本研究旨在阐明gilteritinib对FLT3表达水平的影响。结果显示gilteritinib诱导FLT3磷酸化和表达的剂量依赖性降低。治疗48小时后,这种减少尤其明显。发现FLT3表达的减少与FLT3mRNA转录的变化无关,提示转录后调控机制。在各种AML细胞系和具有FLT3野生型和FLT3突变体两者的细胞中进行进一步的研究显示通过吉特替尼治疗的FLT3减少。此外,评价了其他FLT3抑制剂降低FLT3表达的能力.其他FLT3抑制剂,Midostaurin,Crenolanib,还有Quizartinib,也降低了FLT3的表达,与gilteritinib的效果一致。这些发现为优化AML患者的gilteritinib治疗提供了巨大的希望。然而,重要的是要认识到,需要进一步的研究以充分了解这些机制及其在FLT3减少中的临床意义.
    Acute myeloid leukemia (AML) is a genetically diverse and challenging malignancy, with mutations in the FLT3 gene being particularly common and deleterious. Gilteritinib, a potent FLT3 inhibitor, has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of relapsed/refractory AML with FLT3 mutations. Although gilteritinib was developed based on its inhibitory activity against FLT3 kinase, it is important to understand the precise mechanisms of its antileukemic activity in managing drug resistance and discovering biomarkers. This study was designed to elucidate the effect of gilteritinib on the FLT3 expression level. The results showed that gilteritinib induced a dose-dependent decrease in both FLT3 phosphorylation and expression. This reduction was particularly pronounced after 48 h of treatment. The decrease in FLT3 expression was found to be independent of changes in FLT3 mRNA transcription, suggesting post-transcriptional regulatory mechanisms. Further studies were performed in various AML cell lines and cells with both FLT3 wild-type and FLT3 mutant exhibited FLT3 reduction by gilteritinib treatment. In addition, other FLT3 inhibitors were evaluated for their ability to reduce FLT3 expression. Other FLT3 inhibitors, midostaurin, crenolanib, and quizartinib, also reduced FLT3 expression, consistent with the effect of gilteritinib. These findings hold great promise for optimizing gilteritinib treatment in AML patients. However, it is important to recognize that further research is warranted to gain a full understanding of these mechanisms and their clinical implications in the context of FLT3 reduction.
    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
    Fms样酪氨酸激酶3(FLT3)突变,存在于超过30%的急性髓性白血病(AML)病例中,并以FLT3内部串联重复(FLT3-ITD)为主,与AML患者的不良预后相关。而酪氨酸激酶抑制剂(TKIs;例如,gilteritinib)是有效的,他们面临诸如抗药性等挑战,复发,和高成本。这里,我们报告二甲双胍,一个便宜的,安全,和广泛使用的抗糖尿病药,与gilteritinib在治疗FLT3-ITDAML中表现出惊人的协同作用。二甲双胍显著使FLT3-ITDAML细胞(包括TKI耐药细胞)对gilteritinib敏感。在FLT3-ITDAML小鼠模型中,二甲双胍+吉特替尼(低剂量)显著抑制白血病进展并延长生存期。机械上,联合治疗共同抑制polo样激酶1(PLK1)的表达和FLT3/STAT5/ERK/mTOR的磷酸化.临床分析还显示服用二甲双胍的FLT3-ITDAML患者的生存率提高。因此,二甲双胍/gilteritinib联合治疗FLT3突变的AML患者是一种有希望且具有成本效益的治疗方法,特别是对于那些收入/负担能力低的人。
    Fms-like tyrosine kinase 3 (FLT3) mutations, present in over 30% of acute myeloid leukemia (AML) cases and dominated by FLT3-internal tandem duplication (FLT3-ITD), are associated with poor outcomes in patients with AML. While tyrosine kinase inhibitors (TKIs; e.g., gilteritinib) are effective, they face challenges such as drug resistance, relapse, and high costs. Here, we report that metformin, a cheap, safe, and widely used anti-diabetic agent, exhibits a striking synergistic effect with gilteritinib in treating FLT3-ITD AML. Metformin significantly sensitizes FLT3-ITD AML cells (including TKI-resistant ones) to gilteritinib. Metformin plus gilteritinib (low dose) dramatically suppresses leukemia progression and prolongs survival in FLT3-ITD AML mouse models. Mechanistically, the combinational treatment cooperatively suppresses polo-like kinase 1 (PLK1) expression and phosphorylation of FLT3/STAT5/ERK/mTOR. Clinical analysis also shows improved survival rates in patients with FLT3-ITD AML taking metformin. Thus, the metformin/gilteritinib combination represents a promising and cost-effective treatment for patients with FLT3-mutated AML, particularly for those with low income/affordability.
    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
    FLT3-ITD是急性髓性白血病(AML)疾病的一种不良预后因素。Gilteritinib,第二代FLT3酪氨酸激酶抑制剂,在ADMIRALIII期试验中,复发/难治性FLT3突变型AML患者的总生存期得到改善.然而,关于Gilteritinib为基础的治疗FLT3突变的AML伴中枢神经系统(CNS)受累的疗效和安全性的数据很少.我们使用gilteritinib治疗1例异基因造血干细胞移植后中枢神经系统复发AML患者。gilteritinib的积极抗白血病作用可能为FLT3突变的AML伴CNS复发的治疗带来新的希望。
    FLT3-ITD is a type of poor prognostic factors in acute myeloid leukemia (AML) disease. Gilteritinib, the second-generation FLT3 tyrosine kinase inhibitor, improved the overall survival of patients with relapsed/refractory FLT3-mutated AML in the ADMIRAL phase III trial. However, few data are available on the efficacy and safety of gilteritinib-based therapy for FLT3-mutated AML with central nervous system (CNS) involvement. We performed gilteritinib to treat a patient with CNS relapsed AML after allogeneic hematopoietic stem cell transplantation. The positive antileukemic effect of gilteritinib may bring new hope for the treatment of FLT3-mutated AML with CNS relapse.
    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:FMS样酪氨酸激酶3内部串联重复(FLT3-ITD)是急性髓性白血病(AML)中常见的突变类型,通常与患者预后不良有关。随着分子诊断的进步和酪氨酸激酶抑制剂(TKI)的发展,FLT3-ITD突变的AML患者的总生存期(OS)在一定程度上延长,但是复发和耐药性仍然是巨大的挑战。宁替尼是一种新型的TKI,可抵抗与肿瘤发病机理有关的各种激酶,目前正在进行肺癌的临床试验。在这项研究中,我们在体内和体外研究了宁替尼对FLT3突变的AML的抗肿瘤活性.
    方法:在AML细胞系和表达各种FLT3突变的Ba/F3细胞中进行细胞增殖测定,以验证宁吉替尼的体外抗白血病活性。免疫印迹试验用于验证宁替尼对FLT3蛋白和下游途径的影响。分子对接和CETSA用于验证宁替尼与靶蛋白的相互作用。在Ba/F3-FLT3-ITD-,MOLM13,Ba/F3-FLT3-ITD-F691L-,MOLM13-FLT3-ITD-F691L诱导的白血病小鼠模型。我们还使用患者来源的原代细胞来确定宁替尼的疗效。
    结果:宁替尼抑制细胞增殖,阻断了细胞周期,诱导细胞凋亡并结合FLT3抑制其下游信号通路,包括STAT5、AKT和ERK通路,在FLT3-ITDAML细胞系中。在FLT3-ITD和FLT3-ITD-F691L突变的小鼠模型中,宁替尼表现出优于现有临床药物gilteritinib和quizartinib的抗白血病活性,显著延长小鼠的生存期。此外,宁替尼对患者来源的携带FLT3-ITD突变的原代细胞表现出活性.
    结论:总体而言,我们的研究证实了宁替尼在具有FLT3-ITD突变的AML中的治疗作用,为临床耐药患者提供了一个潜在的新选择。
    BACKGROUND: FMS-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) is a common mutation type in acute myeloid leukemia (AML) and is usually associated with poor patient prognosis. With advancements in molecular diagnostics and the development of tyrosine kinase inhibitors (TKI), the overall survival (OS) of AML patients with FLT3-ITD mutations has been prolonged to some extent, but relapse and drug resistance are still substantial challenges. Ningetinib is a novel TKI against various kinases in relation to tumour pathogenesis and is undergoing clinical trials of lung cancer. In this study, we explored the antitumor activity of ningetinib against AML with FLT3 mutations both in vivo and in vitro.
    METHODS: Cell proliferation assays were performed in AML cell lines and Ba/F3 cells expressing various FLT3 mutations to validate the antileukemic activity of ningetinib in vitro. Immunoblot assays were used to verify the effect of ningetinib on the FLT3 protein and downstream pathways. Molecular docking and CETSA were used to validate the interaction of ningetinib with target proteins. The survival benefit of ningetinib in vivo was assessed in Ba/F3-FLT3-ITD-, MOLM13, Ba/F3-FLT3-ITD-F691L-, MOLM13-FLT3-ITD-F691L-induced leukemia mouse models. We also used patient-derived primary cells to determine the efficacy of ningetinib.
    RESULTS: Ningetinib inhibited cell proliferation, blocked the cell cycle, induced apoptosis and bound FLT3 to inhibit its downstream signaling pathways, including the STAT5, AKT and ERK pathways, in FLT3-ITD AML cell lines. In the mouse models with FLT3-ITD and FLT3-ITD-F691L mutation, ningetinib showed superior anti-leukemia activity to existing clinical drugs gilteritinib and quizartinib, significantly prolongating the survival of mice. In addition, ningetinib exhibited activity against patient-derived primary cells harboring FLT3-ITD mutations.
    CONCLUSIONS: Overall, our study confirmed the therapeutic role of ningetinib in AML with FLT3-ITD mutations, providing a potential new option for clinically resistant 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: English Abstract
    Objective: This study aims to evaluate the safety and effectiveness of gilteritinib (Gilt) -based combination therapy bridging allo-HSCT for FLT3-ITD(+) R/R AML. Additionally, it aims to assess the impact of Gilt maintenance therapy on the prognosis of patients after allo-HSCT. Methods: The clinical data of 26 patients with FLT3-ITD(+) R/R AML treated at the First Affiliated Hospital of Soochow University from August 2019 to January 2023 were retrospectively analyzed. The analysis included an assessment of the composite complete remission rate (CRc), overall survival (OS) time, disease-free survival (DFS) time, and adverse events experienced by all enrolled patients. Results: A total of 26 patients with FLT3-ITD(+) R/R AML were enrolled, including 14 men and 12 women with a median age of 38 (18-65) years. A total of 18 cases were refractory, and eight cases were relapsed. The curative effect evaluation conducted between 14 and 21 days showed that the complete remission (CR) rate was 26.9% (7/26), the CR with hematology incomplete recovery was 57.7% (15/26), and the partial response (PR) rate was 7.7% (2/26). The CRc was 84.6% (22/26), and the minimal residual disease (MRD) negativity rate was 65.4%. The 12 month cumulative OS rate for all patients was 79.0%, and the 24 month cumulative OS rate was 72.0%. The median OS time was not determined. The median follow-up time was 16.0 months. Among the patients who responded to treatment, the 12 month cumulative DFS rate was 78.0%, and the 24 month cumulative DFS rate was 71.0%. The median DFS time was not determined. Patients who received allo-HSCT had a median OS time that was significantly longer than those who did not receive allo-HSCT (3.3 months, 95%CI 2.2-4.3 months, P=0.005). The median OS time of patients with or without Gilt maintenance therapy after allo-HSCT was not determined, but the OS time of patients with Gilt maintenance therapy after allo-HSCT treatment was longer than that of patients without Gilt maintenance therapy after allo-HSCT treatment (P=0.019). The FLT3-ITD mutation clearance rate in this study was 38.5%, and the median OS time of patients with FLT3-ITD mutation clearance was not determined but was significantly longer than the median OS of patients without FLT3-ITD mutation clearance (15.0 months; P=0.018). The most common grade 3 and above hematological adverse events of Gilt-based combination therapy included leukopenia (76.9%), neutropenia (76.9%), febrile neutropenia (61.5%), thrombocytopenia (69.2%), and anemia (57.7%). One patient developed differentiation syndrome during oral Gilt maintenance therapy after allo-HSCT treatment, but his condition improved after treatment. Conclusion: The Gilt-based combination therapy is highly effective in treating FLT3-ITD(+) R/R AML. It demonstrates a high CRc, MRD negativity rate, and rapid onset, leading to a significant improvement in patients\' survival. Furthermore, the clearance rate of FLT3-ITD mutation is notably high. Additionally, implementing bridging allo-HSCT and Gilt maintenance therapy after allo-HSCT treatment has considerably enhances patients\' survival. Closely monitoring and managing any adverse event that may occur during treatment are crucial.
    目的: 探讨Gilteritinib(Gilt)为基础的方案桥接异基因造血干细胞移植(allo-HSCT)对伴FMS样酪氨酸激酶3(FLT3)基因内部串联重复(ITD)突变的复发难治性急性髓系白血病(R/R AML)患者的安全性、有效性,以及移植后Gilt维持治疗对FLT3-ITD阳性R/R AML患者预后的影响。 方法: 回顾性分析2019年8月至2023年1月苏州大学附属第一医院收治的26例伴FLT3-ITD突变的R/R AML患者。统计所有纳入患者的复合完全缓解(CRc)率、总生存(OS)期、无病生存(DFS)期和不良反应。 结果: 26例FLT3-ITD突变阳性R/R AML患者中,男14例,女12例,中位年龄38(18~65)岁。难治18例,复发8例。用药第14~21天疗效:完全缓解(CR)率为26.9%(7/26),CR伴血液学不完全恢复(CRi)率为57.7%(15/26),部分缓解(PR)率为7.7%(2/26),CRc率为84.6%(22/26),微小残留病(MRD)转阴率为65.4%。所有患者12、24个月累计OS率分别为79.0%和72.0%。中位OS期未达到。中位随访时间为16.0个月。全部治疗有效患者12、24个月累计DFS率分别为78.0%和71.0%。中位DFS期未达到。接受allo-HSCT的患者中位OS期未达到,较未接受allo-HSCT的患者(3.3个月,95%CI 2.2~4.3个月)显著延长(P=0.005)。移植后是否应用Gilt维持治疗的患者中位OS期均未达到,且移植后维持治疗的患者OS明显优于移植后未进行维持治疗的患者(P=0.019)。本研究中FLT3-ITD基因突变清除率为38.5%,且FLT3-ITD基因突变转阴的患者中位OS期未达到,明显长于未转阴的患者(15.0个月)(P=0.018)。Gilt为基础的方案最常见的3级及以上血液学不良反应包括白细胞减少(76.9%)、中性粒细胞减少(76.9%)、中性粒细胞减少性发热(61.5%)、血小板减少(69.2%)和贫血(57.7%)。其中1例患者在移植后口服Gilt维持治疗时出现分化综合征,治疗后好转。 结论: Gilt为基础的方案桥接allo-HSCT治疗FLT3-ITD突变阳性R/R AML患者的CRc率较高,MRD转阴率也较高,起效迅速,有效延长患者生存期。此外,FLT3-ITD基因突变清除率较高,且桥接移植和移植后Gilt维持治疗明显改善患者生存。治疗过程中不良事件的监测和管理至关重要。.
    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
    FLT3突变的急性髓性白血病(AML)患者复发或难以强化诱导(R/R),预后较差。Gilteritinib最近已成为R/RFLT3突变AML患者的标准治疗方法。我们调查了与gilteritinib单一疗法相比,在gilteritinib(gilt-ven)中添加venetoclax是否可以改善预后。我们纳入了强化化疗后接受gilteritinib(n=19)和gilt-ven(n=17)治疗R/RAML的患者。Gilteritinib和gilt-ven组在mCRc率方面没有差异(53%和65%,p=0.51)和实现异基因造血干细胞移植(HSCT,47%和35%,p=0.5)。两组总生存期(OS)相当,尽管gilt-ven(12个月OS58.8%[95%CI39.5%-87.6%])与gilteritinib(gilteritinib的42.1%[95%CI24.9%-71.3%])相比有更好的OS趋势.与任何其他基于gilteritinib的方法相比,早期挽救与最佳结果相关(p=0.031)。联合治疗与血液学毒性增加有关。总之,与gilteritinib相比,gilt-ven并未改善R/RFLT3突变AML患者的缓解率或HSCT实现率,并且与血液学毒性增加相关.虽然操作系统没有区别,gilt-ven提出了更好的生存趋势,并且在早期进行抢救排序时,gilt-ven方法显示了生存益处。
    Patients with FLT3-mutated acute myeloid leukaemia (AML) that relapse or are refractory (R/R) to intensive induction have poor outcomes. Gilteritinib has recently become standard-of-care for patients with R/R FLT3-mutated AML. We investigated whether adding venetoclax to gilteritinib (gilt-ven) improves outcomes as compared with gilteritinib monotherapy. We included patients treated with gilteritinib (n = 19) and gilt-ven (n = 17) for R/R AML after intensive chemotherapy. Gilteritinib and gilt-ven groups did not differ in terms of mCRc rates (53% and 65%, p = 0.51) and realization of allogeneic haematopoietic stem-cell transplantation (HSCT, 47% and 35%, p = 0.5). Overall survival (OS) was comparable between groups, although a trend towards better OS was seen with gilt-ven (12-month OS 58.8% [95% CI 39.5%-87.6%]) versus gilteritinib (42.1% [95% CI 24.9%-71.3%] for gilteritinib). Early salvage with gilt-ven versus any other gilteritinib-based approach was associated with the best outcome (p = 0.031). Combination therapy was associated with increased haematological toxicity. In summary, gilt-ven did not improve remissions or HSCT-realization rates in patients with R/R FLT3-mutated AML as compared with gilteritinib and was associated with increased haematological toxicity. Although OS did not differ, a trend towards better survival was suggested with gilt-ven and a survival benefit was shown for gilt-ven approach when sequenced early for salvage.
    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