Tipifarnib

Tipifarnib
  • 文章类型: Journal Article
    Tipifarnib是HRAS突变(HRASmt)复发性或转移性头颈部鳞状细胞癌(HNSCC)的唯一靶向治疗突破。鉴于HRASmt的低频率,HRASmt癌症的分子谱难以探索。本研究旨在了解分子共变,免疫谱,524例HRASmt实体瘤包括尿路上皮癌(UC)的临床结果,乳腺癌(BC),非小细胞肺癌(NSCLC),黑色素瘤,和HNSCC。HRASmt在UC中最常见(3.0%),其次是HNSCC(2.82%),黑色素瘤(1.05%),BC(0.45%),非小细胞肺癌(0.44%)。无论激素受体状态如何,Her2BC中都不存在HRASmt。与非鳞状细胞肺癌相比,HRASmt与鳞状细胞肺癌的相关性更高(60%与40%在HRASwt,p=0.002)。HRASmt的肿瘤微环境(TME)显示三阴性BC(TNBC)中M1巨噬细胞增加,HNSCC,鳞状NSCLC,和UC;在TNBC中增加M2巨噬细胞;并且在HNSCC中增加CD8+T细胞(所有p<0.05)。最后,HRASmt与HNSCC的总生存期较短相关(HR:1.564,CI:1.16-2.11,p=0.003),但与其他检查的癌症类型无关。总之,这项研究为HRASmt肿瘤的独特分子谱提供了新的见解,这可能有助于确定新的靶标并指导未来的临床试验设计.
    Tipifarnib is the only targeted therapy breakthrough for HRAS-mutant (HRASmt) recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). The molecular profiles of HRASmt cancers are difficult to explore given the low frequency of HRASmt. This study aims to understand the molecular co-alterations, immune profiles, and clinical outcomes of 524 HRASmt solid tumors including urothelial carcinoma (UC), breast cancer (BC), non-small-cell lung cancer (NSCLC), melanoma, and HNSCC. HRASmt was most common in UC (3.0%), followed by HNSCC (2.82%), melanoma (1.05%), BC (0.45%), and NSCLC (0.44%). HRASmt was absent in Her2+ BC regardless of hormone receptor status. HRASmt was more frequently associated with squamous compared to non-squamous NSCLC (60% vs. 40% in HRASwt, p = 0.002). The tumor microenvironment (TME) of HRASmt demonstrated increased M1 macrophages in triple-negative BC (TNBC), HNSCC, squamous NSCLC, and UC; increased M2 macrophages in TNBC; and increased CD8+ T-cells in HNSCC (all p < 0.05). Finally, HRASmt was associated with shorter overall survival in HNSCC (HR: 1.564, CI: 1.16-2.11, p = 0.003) but not in the other cancer types examined. In conclusion, this study provides new insights into the unique molecular profiles of HRASmt tumors that may help to identify new targets and guide future clinical trial design.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Systematic Review
    背景:口腔鳞状细胞癌(OSCC)是一种常见的恶性肿瘤,其复杂的遗传改变有助于其发展。H-Ras,一个原癌基因,当突变时成为癌基因,并与各种癌症有关。这篇系统的综述旨在研究H-Ras的表达和突变在多大程度上有助于OSCC的发生和发展。以及这种分子改变如何影响OSCC患者的临床特征和预后。
    方法:在PUBMED进行了全面的电子科学文献检索,Scopus,和2007年至2021年的GOOGLESCHOLAR数据库。搜索策略产生了120篇文章。通过我们的纳入和排除标准汇总和过滤所有结果后,共有9篇文章被纳入我们的文献综述。它也已在PROSPERO(CRD42023485202)注册。
    结果:发现Ras基因的突变通常在密码子12、13和61的热点中报道,导致下游信号通路的激活,导致异常和不受控制的细胞生长。这项系统评价显示,在分化良好的OSCC中,H-Ras突变的患病率增加,在参与多种危险行为的个体中,H-Ras突变的患病率也增加。特别是咀嚼烟草,证明与H-Ras阳性患病率较高存在显著关联。
    结论:这篇综述揭示了H-Ras突变的患病率,它们与临床特征的关联,以及它们对OSCC预后的潜在影响。它还增强了我们对OSCC基础的分子机制的理解,并为进一步研究基于H-Ras改变的靶向治疗铺平了道路。
    BACKGROUND: Oral squamous cell carcinoma (OSCC) is a commonly occurring malignancy with complex genetic alterations contributing to its development. The H-Ras, a proto-oncogene, becomes an oncogene when mutated and has been implicated in various cancers. This systematic review aims to research to what extent H-Ras expression and mutation contribute to the development and progression of OSCC, and how does this molecular alteration impacts the clinical characteristics and prognosis in patients with OSCC.
    METHODS: A thorough electronic scientific literature search was carried out in PUBMED, SCOPUS, and GOOGLE SCHOLAR databases from 2007 to 2021. The search strategy yielded 120 articles. Following aggregation and filtering all results through our inclusion and exclusion criteria total 9 articles were included in our literature review. It has also been registered with PROSPERO (CRD42023485202).
    RESULTS: It was found that mutations in the Ras gene commonly reported in hotspots at codons 12, 13, and 61 resulting in the activation of downstream signaling pathways causing abnormal and uncontrolled cell growth. This systematic review has shown an increased prevalence of H-Ras mutation in well-differentiated OSCC and also the prevalence of H-Ras mutation in individuals engaging in multiple risk behaviors, particularly chewing tobacco, demonstrated a significant association with a higher prevalence of H-Ras positivity.
    CONCLUSIONS: This review sheds light on the prevalence of H-Ras mutations, their association with clinical characteristics, and their potential implications for OSCC prognosis. It also enhances our comprehension of the molecular mechanisms that underlie OSCC and paves the way for further research into targeted treatments based on H-Ras alterations.
    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
    头颈部鳞状细胞癌(HNSCC)是全球范围内高度流行的恶性肿瘤,相当比例的患者发展为复发性和/或转移性(R/M)疾病。尽管最近在治疗方面取得了进展,晚期HNSCC患者的预后仍然较差.这里,我们介绍了一例复发的转移性HNSCC患者,该患者具有HRASG12S突变,对替比法尼治疗有持久的反应,法尼基转移酶的选择性抑制剂。该患者是一名48岁的女性,以前接受过多种治疗,没有明显的临床反应。然而,替比法尼治疗导致持续8个月的持久部分反应。连续基因组和转录组学分析显示,与原发肿瘤相比,转移性病变中YAP1和AXL上调,肿瘤微环境从免疫富集到血管生成增加的纤维化亚型的演变,和PI3K/AKT/mTOR通路在替比法尼治疗中的激活。最后,在HRAS突变的PDX和同基因HRAS模型中,我们证明了替比法尼的疗效受到AKT通路激活的限制,替比法尼和PI3K抑制剂双重治疗,BYL719导致增强的抗肿瘤功效。我们的案例研究强调了在R/MHNSCC中用替比法尼靶向HRAS突变的潜力,并确定了对替比法尼的获得性抗性的潜在机制。随着免疫-,化学-,和放射治疗。临床前结果为进一步研究R/MHRAS驱动的HNSCC中HRAS和PI3K靶向疗法的药物组合提供了坚实的基础。
    Head and neck squamous cell carcinoma (HNSCC) is a highly prevalent malignancy worldwide, with a significant proportion of patients developing recurrent and/or metastatic (R/M) disease. Despite recent advances in therapy, the prognosis for patients with advanced HNSCC remains poor. Here, we present the case of a patient with recurrent metastatic HNSCC harboring an HRAS G12S mutation who achieved a durable response to treatment with tipifarnib, a selective inhibitor of farnesyltransferase. The patient was a 48-year-old woman who had previously received multiple lines of therapy with no significant clinical response. However, treatment with tipifarnib resulted in a durable partial response that lasted 8 months. Serial genomic and transcriptomic analyses demonstrated upregulation of YAP1 and AXL in metastatic lesions compared with the primary tumor, the evolution of the tumor microenvironment from an immune-enriched to a fibrotic subtype with increased angiogenesis, and activation of the PI3K/AKT/mTOR pathway in tipifarnib treatment. Lastly, in HRAS-mutated PDXs and in the syngeneic HRAS model, we demonstrated that tipifarnib efficacy is limited by activation of the AKT pathway, and dual treatment with tipifarnib and the PI3K inhibitor, BYL719, resulted in enhanced anti-tumor efficacy. Our case study highlights the potential of targeting HRAS mutations with tipifarnib in R/M HNSCC and identifies potential mechanisms of acquired resistance to tipifarnib, along with immuno-, chemo-, and radiation therapy. Preclinical results provide a firm foundation for further investigation of drug combinations of HRAS-and PI3K -targeting therapeutics in R/M HRAS-driven HNSCC.
    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
    可操作的基因突变和癌基因融合的评估已使非小细胞肺癌(NSCLC)的治疗策略发生了范式转变。HRAS突变涉及约0.2-0.8%的NSCLC患者,大部分在61号密码子上.对于这些患者来说,关于临床特征和治疗反应的数据很少.
    在南特大学医院常规进行的下一代测序(NGS)用于鉴定NSCLC患者的HRAS分子改变。我们鉴定并描述了4例HRASp.GlnQ61Leu突变患者。回顾了以前HRAS突变的NSCLC病例的文献,以及使用最先进的H-Ras特异性抑制剂的实体瘤的可用数据,替比法尼,被介绍了。
    在2018年1月至2020年12月诊断为晚期NSCLC的1614例患者中,有4例(0.25%)有HRASp.Gln61Leu突变。其中三人在一线全身治疗期间死亡。此外,文献中发现另外3例病例.所有病例均为当前或以前的吸烟者,大多数患者在诊断时出现胸膜或心包积液。
    HRAS突变型NSCLC患者的临床病程尚不清楚。应确定其他病例,以明确预后和对治疗的反应。Tipifarnib,一种法尼基转移酶抑制剂,是靶向HRAS突变肿瘤的有希望的候选者,应该在NSCLC患者中进行探索。
    Assessment of actionable gene mutations and oncogene fusions have made a paradigm shift in treatment strategies of non-small cell lung cancer (NSCLC). HRAS mutations involved around 0.2-0.8% of NSCLC patients, mostly on codon 61. For these patients, few data are available regarding clinical characteristics and response to therapies.
    Next-Generation Sequencing (NGS) done routinely at Nantes University Hospital was used to identify HRAS molecular alterations in NSCLC patients. We identified and described four HRAS p.GlnQ61Leu mutated patients. Literature of previously HRAS-mutant NSCLC cases was reviewed, and available data in solid tumour with the most advanced H-Ras specific inhibitor, tipifarnib, were presented.
    Of 1614 patients diagnosed with advanced NSCLC from January 2018 to December 2020, four (0.25%) had HRAS p.Gln61Leu mutation. Three of them died during the first-line systemic therapy. Furthermore, three additional cases were identified in literature. All cases were current or former smokers, most of them had pleural or pericardial effusion at diagnosis.
    The clinical course of patients with HRAS-mutant NSCLC remains unclear. Furthers cases should be identified in order to clarify prognosis and response to therapies. Tipifarnib, a farnesyl transferase inhibitor, is a promising candidate to target HRAS-mutant tumours and should be explored in NSCLC patients.
    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
    BACKGROUND: This article summarizes new findings on chemo- and targeted therapy of head and neck squamous cell carcinoma presented at the 2021 American Society of Clinical Oncology (ASCO) conference.
    OBJECTIVE: Relevant publications were evaluated and summarized.
    METHODS: The abstract database of the 2021 ASCO conference was searched. Publications were selected if they focused on chemo- or targeted therapies in head and neck squamous cell carcinoma. Publications covered in other articles of this issue were omitted, as were study proposals without results.
    RESULTS: The authors present six articles. Dosage and novel forms of application are shown for cisplatin. Cetuximab is used in the neoadjuvant setting and for progressive disease following immune therapy. Lastly, two new drugs in the form of liposomal irinotecan and the mHRAS inhibitor tipifarnib are discussed.
    CONCLUSIONS: Patient-specific and possibly local application of cisplatin might be considered in the future. Moreover, mHRAS inhibition might also be useful for selected patients after sequencing.
    UNASSIGNED: HINTERGRUND: Bei der Konferenz der American Society of Clinical Oncology (ASCO) 2021 wurde Neues rund um Chemotherapien und zielgerichtete Therapien vorgestellt. im vorliegenden Beitrag werden einige dieser Publikationen zusammengefasst.
    UNASSIGNED: Relevante Publikationen werden zusammenfasst dargestellt und bewertet.
    METHODS: Über eine Datenbankabfrage wurden Abstracts des diesjährigen ASCO-Kongresses gesucht, die sich mit Chemotherapie oder zielgerichteten Therapien bei Kopf-Hals-Plattenepithelkarzinomen beschäftigten. Forschungsankündigungen ohne Daten sowie Publikationen mit Überschneidung zu den weiteren Artikeln dieser Ausgabe wurden aussortiert.
    UNASSIGNED: Die Autor*innen stellen 6 Artikel weiterführend vor. Hierbei geht es um neue Applikationsformen sowie die Dosisfindung bei Cisplatin. Zudem geht es um Neoadjuvanzien sowie Cetuximab nach Immuntherapie. Neu untersucht werden liposomales Irinotecan und der mHRAS-Inhibitor Tipifarnib.
    UNASSIGNED: Patientenspezifischere Dosierungen von Cisplatin sowie die lokale Applikation könnten in Zukunft von Bedeutung sein. Die Blockade von mHRAS wird im Einzelfall nach Sequenzierung Nutzen finden.
    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
    背景:酪氨酸激酶抑制剂(TKI)最初被证明是肾细胞癌(RCC)的有效治疗方法。然而,在中位治疗时间为14个月后,绝大多数患者产生耐药性。这项研究分析了针对外泌体赋予耐药性的替比法尼(Tipi)+舒尼替尼的联合治疗。
    方法:786-O,786-O-SR(舒尼替尼耐药),A498,A498-SR,Caki-2,Caki-2-SR,培养293T细胞。使用差异超速离心收集外来体。细胞增殖,JurkatT细胞免疫测定,和免疫印迹分析用于下游分析。
    结果:SR外泌体处理对免疫细胞显示出细胞毒性作用。与舒尼替尼敏感(SS)外泌体相比,这种细胞毒性作用与SR外泌体上PD-L1的表达增加有关。此外,Tipi处理下调源自SR细胞系的外泌体上的PD-L1表达。Tipi下调外泌体中PD-L1的能力在患者中具有重要应用。从患有RCC的患者收集的外泌体显示比没有RCC的受试者增加的PD-L1表达。接下来,然后在Tipi治疗后比较外泌体浓度,所有SS细胞系显示更大的减少。在免疫印迹分析中,293T细胞显示Alix的剂量依赖性增加,nSMase或Rab27a均无变化。相反,所有SS和SR细胞系在所有三种标志物中显示出减少。在对所有SS和SR细胞系进行48小时处理的细胞增殖后,药物组合显示出降低肿瘤生长的协同能力。
    结论:Tipifarnib减弱了转运(ESCRT)依赖性和ESCRT非依赖性途径所需的内体分选复合物所需的外泌体内体分选复合物,从而阻断外泌体生物发生和分泌以及下调SS和SR细胞上的PD-L1。
    BACKGROUND: Tyrosine kinase inhibitors (TKI) were initially demonstrated as an efficacious treatment for renal cell carcinoma (RCC). However, after a median treatment length of 14 months, a vast majority of patients develop resistance. This study analyzed a combination therapy of tipifarnib (Tipi) + sunitinib that targeted exosome-conferred drug resistance.
    METHODS: 786-O, 786-O-SR (sunitinib resistant), A498, A498-SR, Caki-2, Caki-2-SR, and 293T cells were cultured. Exosomes were collected using differential ultracentrifugation. Cell proliferation, Jurkat T cell immune assay, and immunoblot analysis were used for downstream analysis.
    RESULTS: SR exosomes treatment displayed a cytotoxic effect on immune cells. This cytotoxic effect was associated with increased expression of PD-L1 on SR exosomes when compared to sunitinib-sensitive (SS) exosomes. Additionally, Tipi treatment downregulated PD-L1 expression on exosomes derived from SR cell lines. Tipi\'s ability to downregulate PD-L1 in exosomes has a significant application within patients. Exosomes collected from patients with RCC showed increased PD-L1 expression over subjects without RCC. Next, exosome concentrations were then compared after Tipi treatment, with all SS cell lines displaying an even greater reduction. On immunoblot assay, 293T cells showed a dose-dependent increase in Alix with no change in either nSMase or Rab27a. Conversely, all the SS and SR cell lines displayed a decrease in all three markers. After a cell proliferation employed a 48-h treatment on all SS and SR cell lines, the drug combination displayed synergistic ability to decrease tumor growth.
    CONCLUSIONS: Tipifarnib attenuates both the exosome endosomal sorting complex required for endosomal sorting complex required for transport (ESCRT)-dependent and ESCRT-independent pathways, thereby blocking exosome biogenesis and secretion as well as downregulating PD-L1 on SS and SR cells.
    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
    Farnesyltransferase inhibitors (FTIs) are focus for the treatment of several diseases, particularly in the field of cancer therapy. Their potential, however, goes even further, as a number of studies have evaluated FTIs for the treatment of infectious diseases such as malaria, African sleeping sickness, leishmaniosis, and hepatitis D virus infection. Little is known about protein prenylation mechanisms in human pathogens. However, disruption of IspA, a gene encoding the geranyltranstransferase of Staphylococcus aureus (S. aureus) leads to reprogramming of cellular behavior as well as impaired growth and decreased resistance to cell wall-targeting antibiotics. We used an agar well diffusion assay and a time kill assay and determined the minimum inhibitory concentrations of the FTIs lonafarnib and tipifarnib. Additionally, we conducted cell viability assays. We aimed to characterize the effect of these FTIs on S. aureus, methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus epidermidis (S. epidermidis), Escherichia coli (E. coli), Enterococcus faecium (E. faecium), Klebsiella pneumoniae (K. pneumoniae), Pseudomonas aeruginosa (P. aeruginosa), and Streptococcus pneumoniae (S. pneumoniae). Both the FTIs lonafarnib and tipifarnib were capable of inhibiting the growth of the Gram-positive bacteria S. aureus, MRSA, S. epidermidis, and S. pneumoniae, whereas no effect was observed on Gram-negative bacteria. The analysis of the impact of lonafarnib and tipifarnib on common human pathogens might lead to novel insights into their defense mechanisms and therefore provide new therapeutic targets for antibiotic-resistant bacterial infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase III
    Despite the achievement of complete remission with chemotherapy in patients with acute myeloid leukemia (AML), relapse is common and the majority of patients will die of their disease. Patients who achieve a remission after refractory or relapsed disease as well as elderly patients have a very high rate of relapse even if they achieve a complete remission. A phase 3 randomized ECOG-ACRIN-led intergroup study was conducted to determine whether post-remission therapy with the farnesyl transferase inhibitor, tipifarnib (R115777), improved the disease-free survival (DFS) of adult patients with AML in complete remission (CR), at high risk for relapse.
    Adult patients with AML in remission after salvage therapy and/or over age 60 in first remission were enrolled in this study. They were randomly assigned to treatment with tipifarnib or observation (control). The primary objective was to compare the disease-free survival (DFS) between the two arms based on intention to treat, which includes all randomized patients.
    One hundred and forty-four patients were enrolled on the study. Median DFS was 8.9 vs 5.3 months, for tipifarnib vs observation (one-sided p = 0.026) and did not cross the pre-specified boundary to call the study positive. For the 134 eligible patients, median DFS was 10.8 vs 5.3 months for those randomized to tipifarnib vs observation (one-sided p = 0.008). Moreover in an ad hoc evaluation of all women (n = 71) median DFS was 12.1 vs 3.9 months for tipifarnib vs observation (one-sided p = 0.0004) while median OS was 26.5 vs 8.4 months respectively (one-sided p = 0.001).
    This study was not able to demonstrate a benefit to tipifarnib as maintenance therapy in patients with AML in remission. While subsets of patients may indeed benefit, additional studies would be needed to elucidate that benefit which is unlikely given that other seemingly better options have since become 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
    Current therapies for recurrent and metastatic SCC are associated with poor outcomes, and options for later lines of treatment are limited. Insights into potential therapeutic targets, as well as mechanisms of resistance to available therapies, have begun to be elucidated, creating the basis for exploration of combination approaches to drive better patient outcomes. Tipifarnib, a farnesyl transferase inhibitor (FTI), is a small molecule drug that has demonstrated encouraging clinical activity in a genetically-defined subset of head and neck squamous cell carcinoma (HNSCC)-specifically, tumors that express a mutation in the HRAS protooncogene. More recently, bioinformatic analyses and results from patient-derived xenograft modeling indicate that HRAS pathway dependency may extend to a broader subpopulation of SCCs beyond HRAS mutants in the context of combination with agents such as cisplatin, cetuximab, or alpelisib. In addition, tipifarnib can also inactivate additional farnesylated proteins implicated in resistance to approved therapies, including immunotherapies, through a variety of distinct mechanisms, suggesting that tipifarnib could serve as an anchor for combination regimens in SCCs and other tumor types.
    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
    To test the potential ability of tipifarnib to impair proliferation and to enhance the activity of the EGFR inhibitor cetuximab in wild-type H-Ras HNSCC, which accounts for the majority of HNSCC.
    Cell growth, apoptosis and signaling changes in HNSCC cells following tipifarnib exposure in vitro were assessed by SRB, colony formation assay, annexin V staining and Western blot, respectively. A patient-derived xenograft (PDX) animal model was adopted to evaluate the efficacy of tipifarnib in vivo with and without cetuximab.
    Treatment of wild-type H-Ras HNSCC cell lines in vitro with tipifarnib reduced cell growth and increased levels of defarnesylated H-Ras in a dose-dependent manner. In a PDX mouse model, treatment with single-agent tipifarnib led to only near-significant growth inhibition. The addition of cetuximab resulted in increased anti-proliferative effect both in culture and in PDX models, which was also mirrored by Western blot and apoptosis assay results.
    Tipifarnib has only a moderate ability to slow tumor growth as a single agent in HNSCC with wild type H-Ras, despite specifically inhibiting the farnesyltransferase upon which the function of H-Ras depends. The combination of cetuximab and tipifarnib appears to enhance the anti-proliferative effect of single-agent tipifarnib and marginally enhance that of single agent cetuximab. These findings deserve further evaluation.
    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