Novel combination therapy

  • 文章类型: Case Reports
    背景:胰腺导管腺癌(PDAC)是一种高度致命的疾病,有效治疗有限,尤其是在一线化疗后。人表皮生长因子受体2(HER-2)免疫组织化学(IHC)阳性与PDAC中更具侵略性的临床行为和较短的总生存期相关。
    方法:我们介绍了1例多发性转移性PDAC,IHC错配修复有效,但HER-2IHC弱阳性诊断,一线nab-紫杉醇联合吉西他滨和PD-1抑制剂治疗后肿瘤没有消退。RC48(HER2-抗体-药物偶联物)的新型联合治疗PRaG3.0,放射治疗,PD-1抑制剂,然后应用粒细胞-巨噬细胞集落刺激因子和白细胞介素-2作为二线治疗,患者确认部分缓解良好,无进展生存期为6.5个月,总生存期为14.2个月.她在任何时候都没有出现任何2级或以上的治疗相关不良事件。在包含放疗的PRaG3.0治疗的前两个激活周期中,外周CD8Temra和CD4Temra的百分比增加,但在不包含放疗的维持周期中降低至基线。
    结论:PRaG3.0可能是HER2阳性转移性PDAC患者的一种新策略,这些患者以前的一线方法甚至PD-1免疫治疗均失败,但在前瞻性试验中需要更多数据。
    BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal disease with limited effective treatment especially after first-line chemotherapy. The human epidermal growth factor receptor 2 (HER-2) immunohistochemistry (IHC) positive is associated with more aggressive clinical behavior and shorter overall survival in PDAC.
    METHODS: We present a case of multiple metastatic PDAC with IHC mismatch repair proficient but HER-2 IHC weakly positive at diagnosis that didn\'t have tumor regression after first-line nab-paclitaxel plus gemcitabine and PD-1 inhibitor treatment. A novel combination therapy PRaG 3.0 of RC48 (HER2-antibody-drug conjugate), radiotherapy, PD-1 inhibitor, granulocyte-macrophage colony-stimulating factor and interleukin-2 was then applied as second-line therapy and the patient had confirmed good partial response with progress-free-survival of 6.5 months and overall survival of 14.2 month. She had not developed any grade 2 or above treatment-related adverse events at any point. Percentage of peripheral CD8+Temra and CD4+Temra were increased during first two activation cycles of PRaG 3.0 treatment containing radiotherapy but deceased to the baseline during the maintenance cycles containing no radiotherapy.
    CONCLUSIONS: PRaG 3.0 might be a novel strategy for HER2-positive metastatic PDAC patients who failed from previous first-line approach and even PD-1 immunotherapy but needs more data in prospective trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    BACKGROUND: The diversity in the genomic landscape of advanced and metastatic tumors calls for combination therapies based on the genomic signature associated with each tumor. Determining safe and tolerable doses for novel combinations of oncology drugs is essential for a precision medicine approach, but can also require dose reductions. Trametinib, palbociclib, and everolimus are among the targeted therapies most often used in novel combinations at our precision medicine clinic.
    OBJECTIVE: To evaluate the safe, tolerable dosing of trametinib, palbociclib, and everolimus when used as part of novel combinations with other agents for the treatment of advanced or metastatic solid tumors.
    METHODS: This retrospective study included adult patients with advanced or metastatic solid tumors who received trametinib, everolimus, or palbociclib plus other therapies as a part of novel combinations between December 2011 and July 2018 at the University of California San Diego. Patients were excluded if they received trametinib, everolimus, or palbociclib in standard combinations, such as dabrafenib plus trametinib, everolimus plus fulvestrant, everolimus plus letrozole, and palbociclib plus letrozole. Dosing and adverse events were determined through a review of the electronic medical records. A safe, tolerable drug combination dose was defined as being tolerated for at least 1 month, with no clinically significant serious adverse events.
    RESULTS: A safe, tolerable dose was determined for 76% of the 71 patients who received trametinib, 88% of the 48 patients who received everolimus, and 73% of the 41 patients receiving palbociclib when used in combination with other therapies. For patients with clinically significant adverse events, dose reductions were attempted in 30% of the trametinib recipients, in 17% of everolimus recipients, and in 45% of palbociclib recipients. When used in combination with other therapies, the optimal dosing of trametinib, palbociclib, and everolimus was lower than the standard single-agent dosing: it was 1 mg daily for trametinib; 5 mg daily for everolimus; and 75 mg daily, for 3 weeks on and 1 week off for palbociclib. Of note, everolimus could not be given concomitantly with trametinib at these doses.
    CONCLUSIONS: Safe and tolerable dosing of novel combination therapies that includes trametinib, everolimus, or palbociclib is feasible for a precision medicine approach. However, neither results from this study nor results from previous studies could support the use of everolimus in combination with trametinib, even at reduced doses.
    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
    尽管存在明显的肿瘤内和肿瘤间异质性,但小细胞肺癌(SCLC)仍被视为整体疾病。非特异性DNA损伤剂几十年来一直是一线治疗。最近,新出现的SCLC肿瘤的转录组和基因组分析确定了不同的SCLC亚型和对靶向治疗的脆弱性,包括核酶聚(ADP-核糖)聚合酶(PARPi)的抑制剂。与健康肺组织和肺肿瘤的其他亚型相比,SCLC细胞系和肿瘤显示PARP1蛋白和mRNA水平升高。在临床前SCLC模型中也观察到对PARPi的显著反应。临床上,PARPi单药治疗对SCLC患者具有不同的益处。迄今为止,正在积极开展研究,以检查PARPi反应的预测性生物标志物和各种PARPi组合策略,以最大限度地提高PARPi的临床效用.这篇叙述性综述总结了支持PARPi单药治疗的现有临床前证据,联合治疗,和各自的翻译到诊所。具体来说,我们涵盖了PARPi与DNA损伤化疗(顺铂,依托泊苷,替莫唑胺),胸部放疗,免疫疗法(免疫检查点抑制剂),和许多其他靶向DNA损伤反应的新型治疗剂,肿瘤微环境,表观遗传调制,血管生成,泛素-蛋白酶体系统,或自噬。推定的生物标志物,如SLFN11表达式,MGMT甲基化,E2F1表达,和铂灵敏度,可以预测对不同治疗组合的反应,也进行了讨论。SCLC治疗的未来正在经历快速变化,重点是量身定制和个性化的治疗策略。使用PARPi的癌症治疗的进一步发展将极大地受益于至少一部分生物标志物定义的SCLC患者。
    Small cell lung cancer (SCLC) is treated as a monolithic disease despite the evident intra- and intertumoral heterogeneity. Non-specific DNA-damaging agents have remained the first-line treatment for decades. Recently, emerging transcriptomic and genomic profiling of SCLC tumors identified distinct SCLC subtypes and vulnerabilities towards targeted therapeutics, including inhibitors of the nuclear enzyme poly (ADP-ribose) polymerase (PARPi). SCLC cell lines and tumors exhibited an elevated level of PARP1 protein and mRNA compared to healthy lung tissues and other subtypes of lung tumors. Notable responses to PARPi were also observed in preclinical SCLC models. Clinically, PARPi monotherapy exerted variable benefits for SCLC patients. To date, research is being vigorously conducted to examine predictive biomarkers of PARPi response and various PARPi combination strategies to maximize the clinical utility of PARPi. This narrative review summarizes existing preclinical evidence supporting PARPi monotherapy, combination therapy, and respective translation to the clinic. Specifically, we covered the combination of PARPi with DNA-damaging chemotherapy (cisplatin, etoposide, temozolomide), thoracic radiotherapy, immunotherapy (immune checkpoint inhibitors), and many other novel therapeutic agents that target DNA damage response, tumor microenvironment, epigenetic modulation, angiogenesis, the ubiquitin-proteasome system, or autophagy. Putative biomarkers, such as SLFN11 expression, MGMT methylation, E2F1 expression, and platinum sensitivity, which may be predictive of response to distinct therapeutic combinations, were also discussed. The future of SCLC treatment is undergoing rapid change with a focus on tailored and personalized treatment strategies. Further development of cancer therapy with PARPi will immensely benefit at least a subset of biomarker-defined SCLC 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
    Mycobacterium abscessus is a difficult respiratory pathogen to treat, when compared to other nontuberculus mycobacteria (NTM), due to its drug resistance. In this study, we aimed to find a new clarithromycin partner that potentiated strong, positive, synergy against M. abscessus among current anti-M. abscessus drugs, including omadacycline, amikacin, rifabutin, bedaquiline, and cefoxitine. First, we determined the minimum inhibitory concentrations required of all the drugs tested for M. abscessus subsp. abscessus CIP104536T treatment using a resazurin microplate assay. Next, the best synergistic partner for clarithromycin against M. abscessus was determined using an in vitro checkerboard combination assay. Among the drug combinations evaluated, omadacycline showed the best synergistic effect with clarithromycin, with a fractional inhibitory concentration index of 0.4. This positive effect was also observed against M. abscessus clinical isolates and anti-M. abscessus drug resistant strains. Lastly, this combination was further validated using a M. abscessus infected zebrafish model. In this model, the clarithromycin-omadacyline regimen was found to inhibit the dissemination of M. abscessus, and it significantly extended the lifespan of the M. abscessus infected zebrafish. In summation, the synergy between two anti-M. abscessus compounds, clarithromycin and omadacycline, provides an attractive foundation for a new M. abscessus treatment regimen.
    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
    临床医生的传统癌症治疗选择是手术,化疗,辐射和免疫疗法,可单独使用或以各种组合使用。除电离辐射之外的其他物理模态包括光动力疗法和加热以及称为肿瘤治疗场(TTFields)的最近的方法。TTFelds是中频,低强度,使用非侵入性阵列施加于肿瘤区域和细胞的交变电场。TTField彻底改变了新诊断和复发性胶质母细胞瘤(GBM)以及不可切除和局部晚期恶性胸膜间皮瘤(MPM)的治疗。TTField被认为主要通过破坏有丝分裂来杀死肿瘤细胞;然而,已经表明,TTField可以提高不同类药物的疗效。直接靶向有丝分裂,复制应激和DNA损伤途径。因此,需要详细了解TTFields的作用机制才能在临床上有效使用该疗法。最近的发现暗示了TTFields在不同的重要途径中的作用,如DNA损伤反应和复制应激,ER压力,膜渗透性,自噬,和免疫反应。这篇综述集中于TTFelds抗肿瘤作用的潜在新机制及其在已完成和正在进行的临床试验和临床前研究中的意义。此外,这篇综述讨论了化疗药物和放疗联合TTField治疗未来临床应用的优势和策略.
    Traditional cancer therapy choices for clinicians are surgery, chemotherapy, radiation and immune therapy which are used either standalone therapies or in various combinations. Other physical modalities beyond ionizing radiation include photodynamic therapy and heating and the more recent approach referred to as Tumor Treating Fields (TTFields). TTFields are intermediate frequency, low-intensity, alternating electric fields that are applied to tumor regions and cells using noninvasive arrays. TTFields have revolutionized the treatment of newly diagnosed and recurrent glioblastoma (GBM) and unresectable and locally advanced malignant pleural mesothelioma (MPM). TTFields are thought to kill tumor cells predominantly by disrupting mitosis; however it has been shown that TTFields increase efficacy of different classes of drugs, which directly target mitosis, replication stress and DNA damage pathways. Hence, a detailed understanding of TTFields\' mechanisms of action is needed to use this therapy effectively in the clinic. Recent findings implicate TTFields\' role in different important pathways such as DNA damage response and replication stress, ER stress, membrane permeability, autophagy, and immune response. This review focuses on potentially novel mechanisms of TTFields anti-tumor action and their implications in completed and ongoing clinical trials and pre-clinical studies. Moreover, the review discusses advantages and strategies using chemotherapy agents and radiation therapy in combination with TTFields for future clinical use.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号