Pancreatic Cancer

胰腺癌
  • 文章类型: Journal Article
    这是急性和慢性胰腺炎之间以及急性胰腺炎与胰腺癌之间关系的概述。急性胰腺炎和复发性急性胰腺炎是慢性胰腺炎的病因。基于人群的研究计算出急性胰腺炎首次发作后发生急性复发性胰腺炎的风险为20%,而急性胰腺炎首次发作后发生慢性胰腺炎的风险为10%。一个重要的风险因素是吸烟。急性和慢性胰腺炎是胰腺癌的危险因素。急性胰腺炎的风险与急性胰腺炎的复发次数有关,但不是急性胰腺炎的病因。急性胰腺炎,以及慢性胰腺炎,是胰腺癌的危险因素。急性胰腺炎或复发性急性胰腺炎发作后,应将患者视为高风险。
    This is an overview of relation between acute and chronic pancreatitis and between acute pancreatitis and pancreatic cancer. Acute pancreatitis and recurrent acute pancreatitis are an etiological factor of chronic pancreatitis. Population-based studies have calculated the risk of acute recurrent pancreatitis after the first attack of acute pancreatitis to be 20% and development of chronic pancreatitis after first attack of acute pancreatitis is 10%. An important risk factor is tobacco smoking. Acute and chronic pancreatitis are risk factors for pancreatic cancer. The risk of acute pancreatitis is related to the number of recurrences of acute pancreatitis, but not the etiology of acute pancreatitis. Acute pancreatitis, as well as chronic pancreatitis, are risk factors for pancreatic cancer. After an attack of acute pancreatitis or recurrent acute pancreatitis a patient should be regarded as a high risk.
    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
    胰腺导管腺癌(PDAC)的治疗选择有限,强调迫切需要有效的治疗方法。PDAC的主要驱动因素是突变的KRAS原癌基因,KRA,90%的患者。直接KRAS抑制剂的出现为治疗提供了一个有希望的途径,特别是那些针对KRASG12C突变的等位基因,在临床试验中显示出令人鼓舞的结果。然而,耐药性的发展需要探索有效的联合疗法。我们的目标是通过无偏倚的药物筛选来确定有效的KRASG12C抑制剂联合疗法。结果显示与七种同源物1(SOS1)抑制剂的协同作用,酪氨酸蛋白磷酸酶非受体11型(PTPN11)/Src同源区2域含磷酸酶2(SHP2)抑制剂,和广谱多激酶抑制剂。在新颖且独特的KRASG12C突变的患者来源的类器官模型中的验证证实了来自筛选实验的所述命中。我们的发现提出了增强KRASG12C抑制剂疗效的策略,指导临床试验设计和分子肿瘤委员会。
    Pancreatic ductal adenocarcinoma (PDAC) has limited treatment options, emphasizing the urgent need for effective therapies. The predominant driver in PDAC is mutated KRAS proto-oncogene, KRA, present in 90% of patients. The emergence of direct KRAS inhibitors presents a promising avenue for treatment, particularly those targeting the KRASG12C mutated allele, which show encouraging results in clinical trials. However, the development of resistance necessitates exploring potent combination therapies. Our objective was to identify effective KRASG12C-inhibitor combination therapies through unbiased drug screening. Results revealed synergistic effects with son of sevenless homolog 1 (SOS1) inhibitors, tyrosine-protein phosphatase non-receptor type 11 (PTPN11)/Src homology region 2 domain-containing phosphatase-2 (SHP2) inhibitors, and broad-spectrum multi-kinase inhibitors. Validation in a novel and unique KRASG12C-mutated patient-derived organoid model confirmed the described hits from the screening experiment. Our findings propose strategies to enhance KRASG12C-inhibitor efficacy, guiding clinical trial design and molecular tumor boards.
    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
    胰腺癌,一种高度致命的恶性肿瘤,预计未来十年将成为癌症相关死亡的第二大原因。这凸显了对个性化诊断和治疗的新见解的迫切需要。尽管胰腺癌的分子亚型在基因组学和转录组学中已经建立,很少有已知的分子分类被翻译来指导临床策略,需要进行范式转换.值得注意的是,长期发展和持续改进的高通量技术和系统是进一步从表观基因组学角度描绘胰腺癌分子格局的重要动力,蛋白质组学,代谢组学,和宏基因组学。因此,使用整合的多组学方法在多个水平上更全面地理解分子分类,对于开发更多潜在的治疗选择具有巨大的前景.在这次审查中,我们从不同的组学水平概括了分子谱,讨论了多组学手段的各种亚型,朝着具有临床影响的胰腺癌的平台到旁边的翻译前进了一步,并提出了一些方法论和科学挑战。
    Pancreatic cancer, a highly fatal malignancy, is predicted to rank as the second leading cause of cancer-related death in the next decade. This highlights the urgent need for new insights into personalized diagnosis and treatment. Although molecular subtypes of pancreatic cancer were well established in genomics and transcriptomics, few known molecular classifications are translated to guide clinical strategies and require a paradigm shift. Notably, chronically developing and continuously improving high-throughput technologies and systems serve as an important driving force to further portray the molecular landscape of pancreatic cancer in terms of epigenomics, proteomics, metabonomics, and metagenomics. Therefore, a more comprehensive understanding of molecular classifications at multiple levels using an integrated multi-omics approach holds great promise to exploit more potential therapeutic options. In this review, we recapitulated the molecular spectrum from different omics levels, discussed various subtypes on multi-omics means to move one step forward towards bench-to-beside translation of pancreatic cancer with clinical impact, and proposed some methodological and scientific challenges in store.
    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
    胰腺癌(PC),其特点是其侵略性和低患者生存率,仍然是一个具有挑战性的恶性肿瘤。Anoikis,抑制转移性癌细胞扩散的过程,通过失巢凋亡相关基因与癌症进展和转移密切相关。尽管如此,这些基因在PC中的确切作用机制尚不清楚。
    研究数据来自癌症基因组图谱(TCGA)数据库,在基因表达综合(GEO)数据库中访问验证数据。进行差异表达分析和单变量Cox分析以确定与失巢凋亡相关的预后相关差异表达基因(DEGs)。然后采用无监督聚类分析对癌症样品进行分类。随后,对确定的DEGs进行了最小绝对收缩和选择算子(LASSO)Cox回归分析,以建立临床预后基因标签.使用此签名得出的风险评分,癌症患者被分为高风险和低风险组,通过生存分析进行进一步评估,免疫浸润分析,和突变分析。外部验证数据用于确认结果,Westernblot和免疫组织化学用于验证临床预后基因标记的风险基因。
    共获得20个与失巢凋亡相关的预后相关的DEGs。TCGA数据集揭示了两个不同的子组:簇1和簇2。利用20个DEG,构建了包含两个风险基因(CDKN3和LAMA3)的临床预后基因标签.胰腺腺癌(PAAD)患者根据其风险评分分为高风险和低风险组,后者表现出优越的存活率。在两组之间的免疫浸润和突变水平之间注意到统计学上的显着差异。验证队列结果与最初的发现一致。此外,实验验证证实了CDKN3和LAMA3在肿瘤样品中的高表达。
    我们的研究解决了在理解与失巢凋亡相关的基因在PAAD中的参与方面的差距。本文开发的临床预后基因标签准确地对PAAD患者进行分层,有助于为这些患者提供精准医疗。
    UNASSIGNED: Pancreatic cancer (PC), characterized by its aggressive nature and low patient survival rate, remains a challenging malignancy. Anoikis, a process inhibiting the spread of metastatic cancer cells, is closely linked to cancer progression and metastasis through anoikis-related genes. Nonetheless, the precise mechanism of action of these genes in PC remains unclear.
    UNASSIGNED: Study data were acquired from the Cancer Genome Atlas (TCGA) database, with validation data accessed at the Gene Expression Omnibus (GEO) database. Differential expression analysis and univariate Cox analysis were performed to determine prognostically relevant differentially expressed genes (DEGs) associated with anoikis. Unsupervised cluster analysis was then employed to categorize cancer samples. Subsequently, a least absolute shrinkage and selection operator (LASSO) Cox regression analysis was conducted on the identified DEGs to establish a clinical prognostic gene signature. Using risk scores derived from this signature, patients with cancer were stratified into high-risk and low-risk groups, with further assessment conducted via survival analysis, immune infiltration analysis, and mutation analysis. External validation data were employed to confirm the findings, and Western blot and immunohistochemistry were utilized to validate risk genes for the clinical prognostic gene signature.
    UNASSIGNED: A total of 20 prognostic-related DEGs associated with anoikis were obtained. The TCGA dataset revealed two distinct subgroups: cluster 1 and cluster 2. Utilizing the 20 DEGs, a clinical prognostic gene signature comprising two risk genes (CDKN3 and LAMA3) was constructed. Patients with pancreatic adenocarcinoma (PAAD) were classified into high-risk and low-risk groups per their risk scores, with the latter exhibiting a superior survival rate. Statistically significant variation was noted across immune infiltration and mutation levels between the two groups. Validation cohort results were consistent with the initial findings. Additionally, experimental verification confirmed the high expression of CDKN3 and LAMA3 in tumor samples.
    UNASSIGNED: Our study addresses the gap in understanding the involvement of genes linked to anoikis in PAAD. The clinical prognostic gene signature developed herein accurately stratifies patients with PAAD, contributing to the advancement of precision medicine for these 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
    由于对吉西他滨(GEM)的耐药性,胰腺癌(PC)患者通常预后差,生存率低。我们研究的目的是探索外泌体PPP3CB对PC中GEM抗性的影响,同时分析miR-298/STAT3信号通路的调控作用。
    通过透射电子显微镜(TEM)验证从PC细胞分离的外泌体,纳米粒子跟踪分析(NTA)和蛋白质印迹(WB)。PPP3CB和miR-298之间的相互作用使用双荧光素酶报告基因检测进行验证,然后使用CCK8测定法评估细胞生长和死亡,EdU染色,和流式细胞术。
    在GEM抗性PC细胞中观察到增加的PPP3CB表达。通过超高速离心成功提取PC细胞和GEM抗性PC细胞的外泌体。共聚焦显微镜显示PC细胞内化荧光素酰胺(FAM)标记的GEM抗性外泌体。PPP3CB增强GEM耐药PC细胞的增殖,抑制其凋亡,而下调PPP3CB促进PC细胞的死亡并抑制GEM抗性PC细胞的增殖,并增强PC细胞对GEM的敏感性。此外,PPP3CB通过下调miR-298正向调节PC细胞中STAT3的表达,从而促进PC细胞的生长并抑制其死亡。
    PC细胞衍生的外泌体PPP3CB通过下调miR-298,刺激细胞生长,增强STAT3表达,抑制细胞死亡,从而增加PC电池对GEM的电阻。
    UNASSIGNED: Due to resistance to gemcitabine (GEM), patients with pancreatic cancer (PC) usually have poor prognosis and low survival rate. The purpose of our research was to explore the impact of exosome PPP3CB on GEM resistance in PC, and concurrently analyze the regulatory role of the miR-298/STAT3 signaling pathway.
    UNASSIGNED: Exosomes isolated from PC cells were verified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blotting (WB). The interaction between PPP3CB and miR-298 was verified using dual-luciferase reporter gene assay, followed by evaluation of cell growth and death using CCK8 assay, EdU staining, and flow cytometry.
    UNASSIGNED: Increased PPP3CB expression was observed in GEM-resistant PC cells. Exosomes from PC cells and GEM-resistant PC cells were successfully extracted by ultra-high speed centrifugation. Confocal microscopy showed internalization of fluorescein amide (FAM)-labeled GEM-resistant exosomes by PC cells. PPP3CB enhanced the proliferation of GEM-resistant PC cells and inhibited their apoptosis, whereas down-regulation of PPP3CB promoted the death of PC cells and inhibited the proliferation of GEM-resistant PC cells, and enhance the susceptibility of PC cells to GEM. Additionally, PPP3CB positively regulated STAT3 expression in PC cells by down-regulating miR-298, thus promoting the growth and inhibiting the death of PC cells.
    UNASSIGNED: PC cell-derived exosome PPP3CB enhances STAT3 expression by downregulating miR-298, stimulating cell growth, and suppressing cell death, thereby increasing the resistance of PC cells to GEM.
    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:尽管呼吁将胰腺癌(PC)护理区域化到高容量中心(HVC),许多PC患者选择在离家较近或多个机构接受治疗.在跨机构PC护理的背景下,与协调护理相关的挑战知之甚少.
    方法:在这项定性研究中,我们对HVC(n=9)和社区医院(n=11)的肿瘤临床医生进行了半结构化访谈,以评估他们在各自机构中协调治疗和治疗PC患者的观点。采访被转录,编码,并使用演绎和归纳方法进行分析,以确定与跨机构协调挑战相关的主题,并注意到改进机会。
    结果:临床医生发现了与闭环通信相关的挑战,在某种程度上,无法访问共享的电子健康记录。患者共同管理的挑战归因于患者接受不同临床医生的不一致建议。为了应对这些挑战,参与者提出了一些改进的机会,例如与各机构的临床医生建立融洽的关系,并更新肿瘤委员会的流程.据报道,更新肿瘤委员会流程的机会是多维的,可能涉及:(1)指定肿瘤委员会协调员;(2)记录和传播肿瘤委员会的建议;(3)在肿瘤委员会会议期间使用电话会议促进基于社区的临床医生参与。
    结论:鉴于与患者共同管理相关的沟通障碍和挑战,促进PC临床医生之间的关系发展和改善多学科肿瘤委员会的实践可能会促进跨机构的协调.有必要研究多学科肿瘤委员会协调员和电话会议平台如何加强跨机构沟通,从而改善患者预后。
    BACKGROUND: Despite calls for regionalizing pancreatic cancer (PC) care to high-volume centers (HVCs), many patients with PC elect to receive therapy closer to their home or at multiple institutions. In the context of cross-institutional PC care, the challenges associated with coordinating care are poorly understood.
    METHODS: In this qualitative study we conducted semi-structured interviews with oncology clinicians from a HVC (n = 9) and community-based hospitals (n = 11) to assess their perspectives related to coordinating the care of and treating PC patients across their respective institutions. Interviews were transcribed, coded, and analyzed using deductive and inductive approaches to identify themes related to cross-institutional coordination challenges and to note improvement opportunities.
    RESULTS: Clinicians identified challenges associated with closed-loop communication due, in part, to not having access to a shared electronic health record. Challenges with patient co-management were attributed to patients receiving inconsistent recommendations from different clinicians. To address these challenges, participants suggested several improvement opportunities such as building rapport with clinicians across institutions and updating tumor board processes. The opportunity to update tumor board processes was reportedly multi-dimensional and could involve: (1) designating a tumor board coordinator; (2) documenting and disseminating tumor board recommendations; and (3) using teleconferencing to facilitate community-based clinician engagement during tumor board meetings.
    CONCLUSIONS: In light of communication barriers and challenges associated with patient co-management, enabling the development of relationships among PC clinicians and improving the practices of multidisciplinary tumor boards could potentially foster cross-institutional coordination. Research examining how multidisciplinary tumor board coordinators and teleconferencing platforms could enhance cross-institutional communication and thereby improve patient outcomes is warranted.
    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:由于边际疗效,免疫检查点抑制剂(ICIs)很少用于治疗晚期胰腺导管腺癌(PDAC)。
    方法:本研究包括92例诊断为晚期或复发性PDAC的连续患者,这些患者接受了基于nivolumab的治疗。使用单变量和多变量分析来确定预后因素。选择301例PDAC患者的对照组,这些患者通过姑息性化疗实现了疾病控制,但没有ICI,并使用倾向评分匹配(PSM)进行比较。
    结果:纳武单抗治疗后的中位总生存期(OS)为15.8(95%置信区间[CI],12.5–19.0),2.4(95%CI1.2-3.6),和1.1(95%CI1.0-1.2)个月的患者在化疗实现疾病控制后接受附加纳武单抗,在没有预先确认疾病控制的情况下,同时接受纳武单抗和化疗的患者,在那些接受nivolumab而没有伴随化疗的人中,分别(P<0.001)。PSM之后,与接受单独化疗的患者(13.8个月;95%CI10.8~16.9)相比,接受纳武单抗(19.8个月;95%CI14.5~25.1)的患者自同时化疗开始后实现疾病控制的中位总生存期(OS)显著更长(P=0.026).与单独接受化疗的患者相比,切除患者中肿瘤的免疫谱分析显示,添加nivolumab的患者对Tregs的CD8T细胞得分更高。
    结论:在化疗后获得疾病控制的晚期PDAC患者中,添加纳武单抗与OS改善相关。
    BACKGROUND: Immune checkpoint inhibitors (ICIs) are rarely used to treat advanced pancreatic ductal adenocarcinoma (PDAC) due to marginal efficacy.
    METHODS: This study included 92 consecutive patients diagnosed with advanced or recurrent PDAC who received nivolumab-based treatment. Univariate and multivariate analyses were used to identify prognostic factors. A control group of 301 patients with PDAC who achieved disease control with palliative chemotherapy but without ICIs was selected for comparison using propensity score matching (PSM).
    RESULTS: The median overall survival (OS) since nivolumab treatment was 15.8 (95% confidence interval [CI], 12.5-19.0), 2.4 (95% CI 1.2-3.6), and 1.1 (95% CI 1.0-1.2) months in patients who received add-on nivolumab after achieving disease control with chemotherapy, in those who received concomitant nivolumab and chemotherapy without prerequisite confirmation of disease control, and in those who received nivolumab without concomitant chemotherapy, respectively (P < 0.001). After PSM, the median overall survival (OS) since initiation of the concomitant chemotherapy that achieved disease control was significantly longer (P = 0.026) in patients who received add-on nivolumab (19.8 months; 95% CI 14.5-25.1) than in those who received chemotherapy alone (13.8 months; 95% CI 10.8-16.9). The immune profiling of the tumors in resected patients revealed higher scores of CD8+ T cells to Tregs in patients with add-on nivolumab comparing to those who received chemotherapy alone.
    CONCLUSIONS: Adding-on nivolumab was associated with improved OS in patients with advanced PDAC who achieved disease control following chemotherapy.
    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: Editorial
    透明细胞肉瘤(CCS)是一种恶性肿瘤,可由肌腱和腱膜引起。这种具有黑素细胞谱系的细胞的恶性增殖通常发生在年轻患者中,它通常在四肢被识别。然而,还描述了包括胃肠器官在内的不同部位。由于分子和组织病理学评估的困难,诊断常与恶性黑色素瘤混淆。大多数病例采用手术切除治疗,但总的来说,预后较差。在这篇社论中,我们将讨论一个在胰腺中发现的非常有趣的CCS病例。我们将讨论这类癌症治疗的文献和争议。此外,我们将研究纳入这些病例的分子策略,以更好地了解肿瘤的原发位置.最后,将描述该领域的未来前景和新的治疗策略。
    Clear cell sarcoma (CCS) is a type of malignant tumor that can arise from tendons and aponeuroses. This malignant proliferation of cells with melanocytic lineage normally occurs in young patients, and it is normally identified in extremities. However, different sites including gastrointestinal organs are also described. Due difficulties in the molecular and histopathology evaluation, the diagnosis is often confused with malignant melanoma. Most cases are treated with surgical resection, but overall, the prognosis is poor. In this editorial, we will discuss a very interesting case of CCS identified in the pancreas. We will discuss the literature and controversies in the management of this type of cancer. Furthermore, we will address molecular strategies to be incorporated in those cases to better understand the primary location of the tumor. Finally, future perspectives of the field and new strategies of treatment will be described.
    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
    比较高强度聚焦超声(HIFU)和根治性手术对非转移性胰腺癌(PC)的疗效和安全性。
    我们回顾性分析了2020年1月至2021年12月在湘雅三医院接受HIFU(n=43)或手术(n=46)的89例I/II/III期PC患者。疼痛缓解,Karnofsky绩效量表(KPS),总生存期(OS),评估与治疗相关的并发症和OS的危险因素.
    两组治疗后30天的疼痛缓解率没有显着差异。然而,与手术组相比,HIFU组治疗后VAS评分显著降低(p=0.019).在手术组,治疗后30天的KPS低于治疗前的KPS(70vs80;p=0.015)。这种关系在HIFU组中逆转(80vs70;p=0.024)。中位OS优于HIFU(23vs10个月;p<0.001),1年OS率较高(69.57%vs32.6%;p<0.001)。然而,III期患者的OS在两组之间没有显着差异(p=0.177)。HIFU组的并发症评分≥III级为2.33%,手术组为32.6%。多变量分析表明,年龄,KPS,治疗方法是OS的独立预后因素。
    HIFU在早期KPS方面显示出优于手术的优势,VAS改进,和胰腺癌的安全性;然而,长期结果有利于手术。对于III期疾病,HIFU在总生存率方面不劣于手术。
    UNASSIGNED: To compare the efficacy and safety of high-intensity focused ultrasound (HIFU) and radical surgery for non-metastatic pancreatic cancer (PC).
    UNASSIGNED: We retrospectively analyzed 89 stage I/II/III PC patients who underwent HIFU (n = 43) or surgery (n = 46) at the Third Xiangya Hospital from January 2020 to December 2021. Pain relief, Karnofsky Performance Scale (KPS), overall survival (OS), treatment-related complications and risk factors for OS were assessed.
    UNASSIGNED: There was no significant difference in the pain relief rate at 30 days post-treatment between the two groups. However, compared with the surgery group, the HIFU group showed significantly lower post-treatment VAS scores (p = 0.019). In the surgery group, the KPS at 30 days post-treatment was lower than pretreatment KPS (70 vs 80; p = 0.015). This relationship was reversed in the HIFU group (80 vs 70; p = 0.024). Median OS favored surgery over HIFU (23 vs 10 months; p < 0.001), with a higher 1-year OS rate (69.57% vs 32.6%; p < 0.001). However, there was no significant difference in OS between the two groups for stage III patients (p = 0.177). Complications rated ≥ grade III were 2.33% in the HIFU group and 32.6% in the surgery group. Multivariate analyses showed that age, KPS, and treatment methods were independent prognostic factors for OS.
    UNASSIGNED: HIFU demonstrates advantages over surgery in terms of early KPS, VAS improvements, and safety for pancreatic cancer; however, long-term outcomes favor surgery. For III-stage disease, HIFU was noninferior to surgery in overall survival.
    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
    在当前的医学时代,人类健康面临各种挑战,癌症是一个突出的问题。因此,通过不断涌入选择性靶向肿瘤细胞的新型分子,同时对正常细胞显示最小的毒性,增强癌症的治疗武器库是必要的。这项研究探讨了新报道的螺羟吲哚-吡唑并[3,4-b]吡啶衍生物8a-h和10a-h的抗增殖和EGFR激酶抑制活性。对人癌细胞系A-549(肺癌)生长的抑制作用,Panc-1(胰腺癌),和A-431(皮肤表皮样癌)进行了评估,通过分析明确了SAR。IC50值在个位数微摩尔范围内,化合物8b,8d,10a-b,和10d被证明是针对所研究的癌细胞系的最有效的抗增殖候选物。它们还对人肺MRC5细胞系产生可忽略的细胞毒性(选择性评分在8.63和30.02之间)。此外,我们研究了化合物8b的潜在抑制作用,8d,10a-b,和10d对EGFR和VEGFR-2。10a是本次研究最有效的EGFR抑制剂,IC50值为0.54μM。最终,与厄洛替尼相比,同源物10a的分子对接分析通过检查其结合模式和对接评分,强调了其对EGFR的有效抑制.这些发现强调了螺羟吲哚-吡唑并[3,4-b]吡啶衍生物作为靶向EGFR激酶的有希望的抗癌剂的潜力。
    In the current medical era, human health is confronted with various challenges, with cancer being a prominent concern. Therefore, enhancing the therapeutic arsenal for cancer with a constant influx of novel molecules that selectively target tumor cells while displaying minimal toxicity toward normal cells is imperative. This study delves into the antiproliferative and EGFR kinase inhibitory activities of newly reported spirooxindole-pyrazolo[3,4-b]pyridine derivatives 8a-h and 10a-h. The inhibitory effects on the growth of human cancer cell lines A-549 (lung carcinoma), Panc-1 (pancreatic carcinoma), and A-431 (skin epidermoid carcinoma) were evaluated, and the SAR has been clarified through analysis. With IC50 values in the single-digit micromolar range, compounds 8b, 8d, 10a-b, and 10d were shown to be the most effective antiproliferative candidates against the studied cancer cell lines. They also exerted negligible cytotoxicity (with selectivity scores between 8.63 and 30.02) against the human lung MRC5 cell line. Additionally, we investigated the potential inhibitory action of compounds 8b, 8d, 10a-b, and 10d on EGFR and VEGFR-2. 10a was this investigation\'s most effective EGFR inhibitor, with an IC50 value of 0.54 μM. Ultimately, the molecular docking analysis of congener 10a highlighted its effective suppression of EGFR by examining its binding mode and docking score compared to Erlotinib. These findings underscore the potential of spirooxindole-pyrazolo[3,4-b]pyridine derivatives as promising anticancer agents targeting EGFR kinase.
    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