Solid tumor therapy

实体瘤治疗
  • 文章类型: Journal Article
    在术语“精确肿瘤学”下实施抗癌策略的许多局限性已被广泛讨论。虽然一些作者提出了有希望的未来方向,其他人不那么乐观,使用错觉等短语,炒作,和错误的假设。通过在各种在线出版物上执业的临床医生和癌症患者,其中一篇文章指出,“在寻求下一次癌症治疗的过程中,很少有研究人员费心回头看看失败药物的墓地,以找出哪里出了问题。"信息很明确:具有吸引人的名字的新型治疗策略(例如,尽管进行了数十年的广泛研究和临床试验,但合成的“杀伤力”)仍未兑现其承诺。这篇综述的主要目的是讨论实体瘤治疗中的关键挑战,这些挑战令人惊讶地继续被细胞死亡命名委员会(NCCD)和许多其他作者忽视。这些挑战包括:化疗诱导的基因组混乱的影响(例如,多核化)对抗性和复发,caspase3的致癌功能,癌细胞转移(从细胞凋亡的晚期恢复),以及普遍使用的临床前化学敏感性测定的陷阱(例如,在活体动物中进行的细胞“生存能力”和肿瘤生长延迟研究)将这种促生存反应评分为“致死”事件。本文概述的研究强调了在实体瘤的管理中需要新的方向。
    The many limitations of implementing anticancer strategies under the term \"precision oncology\" have been extensively discussed. While some authors propose promising future directions, others are less optimistic and use phrases such as illusion, hype, and false hypotheses. The reality is revealed by practicing clinicians and cancer patients in various online publications, one of which has stated that \"in the quest for the next cancer cure, few researchers bother to look back at the graveyard of failed medicines to figure out what went wrong\". The message is clear: Novel therapeutic strategies with catchy names (e.g., synthetic \"lethality\") have not fulfilled their promises despite decades of extensive research and clinical trials. The main purpose of this review is to discuss key challenges in solid tumor therapy that surprisingly continue to be overlooked by the Nomenclature Committee on Cell Death (NCCD) and numerous other authors. These challenges include: The impact of chemotherapy-induced genome chaos (e.g., multinucleation) on resistance and relapse, oncogenic function of caspase 3, cancer cell anastasis (recovery from late stages of apoptosis), and pitfalls of ubiquitously used preclinical chemosensitivity assays (e.g., cell \"viability\" and tumor growth delay studies in live animals) that score such pro-survival responses as \"lethal\" events. The studies outlined herein underscore the need for new directions in the management of solid tumors.
    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:HER3(ErbB3),人类表皮生长因子受体家族的成员,在各种癌症中经常过度表达。多种HER3靶向抗体和抗体-药物缀合物(ADC)被开发用于实体瘤治疗。然而,目前还没有一种HER3靶向药物被批准用于肿瘤治疗.我们开发了DB-1310,一种HER3ADC,由一种新型人源化抗HER3单克隆抗体与专有DNA拓扑异构酶I抑制剂有效载荷(P1021)共价连接组成,并评估DB-1310在临床前模型中的疗效和安全性。
    方法:通过ELISA和SPR测量DB-1310与Her3和其他HER家族的结合。通过FACS测试DB-1310和帕特单抗的结合表位的竞争。乳房的敏感性,肺,通过体外细胞杀伤试验评估了DB-1310的前列腺癌和结肠癌细胞系。体内生长抑制研究评估了DB-1310对Her3+乳腺的敏感性,肺,结肠癌和前列腺癌异种移植模型。还在食蟹猴中测量了安全性特征。
    结果:DB-1310通过具有高亲和力和内化能力的新型表位结合HER3。体外,DB-1310在许多HER3+乳腺中表现出细胞毒性,肺,前列腺癌和结肠癌细胞系。HER3+HCC1569乳腺癌的体内研究,NCI-H441肺癌和Colo205结肠癌异种移植模型显示DB-1310具有剂量依赖性杀肿瘤活性。在HER3+非小细胞肺癌(NSCLC)和前列腺癌患者来源的异种移植物(PDX)模型中也观察到肿瘤抑制。此外,DB-1310显示出比派妥克替康(HER3-DXd)更强的肿瘤生长抑制活性,这是在相同剂量下临床开发中的另一种HER3ADC。DB-1310的抑瘤活性与EGFR酪氨酸激酶抑制剂协同作用,奥希替尼,并在奥希替尼耐药的PDX模型中发挥作用。食蟹猴的安全性的临床前评估进一步显示,DB-1310具有良好的安全性,最高非严重毒性剂量(HNSTD)为45mg/kg。
    结论:这些发现表明,DB-1310在体外和体内模型中对HER3+肿瘤具有有效的抗肿瘤活性,并在非临床物种中显示出可接受的安全性。因此,DB-1310可有效用于HER3+实体瘤的临床治疗。
    BACKGROUND: HER3 (ErbB3), a member of the human epidermal growth factor receptor family, is frequently overexpressed in various cancers. Multiple HER3-targeting antibodies and antibody-drug conjugates (ADCs) were developed for the solid tumor treatment, however none of HER3-targeting agent has been approved for tumor therapy yet. We developed DB-1310, a HER3 ADC composed of a novel humanized anti-HER3 monoclonal antibody covalently linked to a proprietary DNA topoisomerase I inhibitor payload (P1021), and evaluate the efficacy and safety of DB-1310 in preclinical models.
    METHODS: The binding of DB-1310 to Her3 and other HER families were measured by ELISA and SPR. The competition of binding epitope for DB-1310 and patritumab was tested by FACS. The sensitivity of breast, lung, prostate and colon cancer cell lines to DB-1310 was evaluated by in vitro cell killing assay. In vivo growth inhibition study evaluated the sensitivity of DB-1310 to Her3 + breast, lung, colon and prostate cancer xenograft models. The safety profile was also measured in cynomolgus monkey.
    RESULTS: DB-1310 binds HER3 via a novel epitope with high affinity and internalization capacity. In vitro, DB-1310 exhibited cytotoxicity in numerous HER3 + breast, lung, prostate and colon cancer cell lines. In vivo studies in HER3 + HCC1569 breast cancer, NCI-H441 lung cancer and Colo205 colon cancer xenograft models showed DB-1310 to have dose-dependent tumoricidal activity. Tumor suppression was also observed in HER3 + non-small cell lung cancer (NSCLC) and prostate cancer patient-derived xenograft (PDX) models. Moreover, DB-1310 showed stronger tumor growth-inhibitory activity than patritumab deruxtecan (HER3-DXd), which is another HER3 ADC in clinical development at the same dose. The tumor-suppressive activity of DB-1310 synergized with that of EGFR tyrosine kinase inhibitor, osimertinib, and exerted efficacy also in osimertinib-resistant PDX model. The preclinical assessment of safety in cynomolgus monkeys further revealed DB-1310 to have a good safety profile with a highest non severely toxic dose (HNSTD) of 45 mg/kg.
    CONCLUSIONS: These finding demonstrated that DB-1310 exerted potent antitumor activities against HER3 + tumors in in vitro and in vivo models, and showed acceptable safety profiles in nonclinical species. Therefore, DB-1310 may be effective for the clinical treatment of HER3 + solid tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    酪氨酸激酶抑制剂(TKIs)为EGFR突变的非小细胞肺癌(NSCLC)患者提供了巨大的益处。虽然以前的TKIs对EGFR外显子20插入突变(EGFREx20Ins)的疗效有限,莫博替尼(TAK-788)旨在特异性抑制这些Ex20Ins突变。在I/II期临床试验中,在一组接受铂类预处理的EGFREx20Ins突变型NSCLC患者中,莫博替尼显示出有意义的获益.对于这个队列,客观缓解率为28%(95%置信区间[CI],20%-37%)。中位无进展生存期和缓解持续时间分别为7.3个月(95%CI,5.5-9.2)和17.5个月(95%CI,7.4-20.3),分别,两者都由独立审查委员会评估。根据这些结果,mobocertinib于2021年被美国食品和药物管理局(FDA)批准为第一个用于治疗该适应症的TKI。这篇综述总结了莫博塞替尼的临床前发展和导致其批准的早期临床数据,并讨论了莫博塞替尼发展的潜在方向。
    Tyrosine kinase inhibitors (TKIs) have provided great benefit for patients with EGFR-mutant non-small cell lung cancer (NSCLC). While prior TKIs have demonstrated limited efficacy against exon 20 insertion mutations of EGFR (EGFR Ex20Ins), mobocertinib (TAK-788) is designed to specifically inhibit these Ex20Ins mutations. In a phase I/II clinical trial, mobocertinib demonstrated meaningful benefits among a cohort of platinum-pretreated patients with EGFR Ex20Ins mutant NSCLC. For this cohort, the objective response rate was 28% (95% confidence interval [CI], 20%-37%). The median progression-free survival and duration of response were 7.3 months (95% CI, 5.5-9.2) and 17.5 months (95% CI, 7.4-20.3), respectively, both by independent review committee assessment. On the basis of these results, mobocertinib was granted accelerated approval as the first TKI for treatment of this indication by the U.S. Food and Drug Administration (FDA) in 2021. This review summarizes the preclinical development of mobocertinib and the early-phase clinical data leading to its approval and discusses potential directions for mobocertinib\'s development.
    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
    Sotorasib,具有G12C突变的Kirsten大鼠肉瘤病毒癌基因(KRAS)的直接抑制剂,获得美国食品和药物管理局(FDA)的批准,作为含有KRASG12C突变的局部晚期或转移性非小细胞肺癌(NSCLC)的二线治疗,根据II期临床试验(Code-BreaK100)的结果。在这篇文章中,我们回顾了KRASG12C抑制剂的作用机制和索托拉西布的最新临床试验,以全面了解其疗效和毒性。我们还回顾了对KRASG12C抑制剂产生抗性的机制以及与KRASG12C突变肿瘤的联合治疗相关的临床前研究。目前,临床数据表明,索托拉西单药治疗对KRASG12C突变和可耐受毒性的NSCLC患者有显著疗效,它可以代表一种新的靶向疗法。需要进一步的研究来描述索托拉西的耐药机制,并确定联合疗法治疗含有KRASG12C突变的NSCLC的疗效和安全性。
    Sotorasib, a direct inhibitor of the enzyme Kirsten rat sarcoma viral oncogene (KRAS) with the G12C mutation, was approved by the U.S. Food and Drug Administration (FDA), as a second-line treatment for locally advanced or metastatic non-small cell lung cancer (NSCLC) containing the KRAS G12C mutation, on the basis of results of a phase II clinical trial (Code- BreaK100). In this article, we review the mechanism of action of KRAS G12C inhibitors and the latest clinical trials with sotorasib to provide a comprehensive understanding of its efficacy and toxicity. We also review the mechanisms that produce resistance to the KRAS G12C inhibitors and the preclinical research related to combination treatments for KRAS G12C-mutated tumors. Currently, clinical data suggests that sotorasib monotherapy has significant efficacy in NSCLC patients with the KRAS G12C mutation and tolerable toxicity, and it could represent a novel targeted therapy. Additional research will be required to delineate the mechanisms of resistance to sotorasib and determine the efficacy and safety of combination therapy for the treatment of NSCLC containing the KRAS G12C mutation.
    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
    组蛋白脱乙酰酶(HDAC)是表观遗传学中的关键酶,是抗癌治疗的有希望的靶标。尽管几种靶向HDAC的药物已被批准用于治疗肿瘤,它们的临床使用受到有害副作用和不良疗效的限制。在这里,我们通过药效团模型筛选和分子对接发现了4种有效的HDAC抑制剂。这些化合物能够以纳摩尔亲和力结合HDAC1、3和6。其中,与伏立诺他(SAHA)相比,化合物3对HDAC1、3和6的抑制作用更大。抗癌活性的评估表明,化合物3显着抑制实体癌细胞的生长,包括HGC-27,AGS,MDA-MB-231、A549、MCF-7和H460细胞。体内抗癌研究表明,化合物3还可以显着抑制HGC-27细胞来源的异种移植物的生长,没有可观察到的毒性。这些发现表明化合物3可作为实体瘤治疗的潜在HDAC靶向抑制剂。
    Histone deacetylases (HDACs) are key enzymes in epigenetics and promising targets for anticancer therapy. Although several drugs targeting HDAC have been approved for the treatment of tumors, their clinical use has been limited by their deleterious side effects and poor efficacy. Herein, we discover four potent HDAC inhibitors through pharmacophore model screening and molecular docking. These compounds are able to bind HDACs 1, 3, and 6 with nanomolar affinity. Among them, compound 3 shows greater inhibitory effect on HDACs 1, 3, and 6 than that of vorinostat (SAHA). Evaluation of anticancer activity indicates that compound 3 significantly inhibits the growth of solid cancer cells including HGC-27, AGS, MDA-MB-231, A549, MCF-7, and H460 cells. In vivo anticancer study suggests that compound 3 can also markedly inhibit the growth of HGC-27 cells-derived xenograft, with no observable toxicity. These findings suggest that compound 3 may be as a potential HDAC-targeting inhibitor for solid tumor therapy.
    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
    转移性环境中的免疫治疗极大地改变了各种类型恶性肿瘤的治疗前景,包括结直肠癌.免疫检查点抑制剂的类别尤其是作为一类基于对免疫细胞-癌细胞调节和肿瘤微环境随时间的演变的更全面理解的疗法而出现。策略包括过继性细胞疗法,肿瘤疫苗,和抗体也证明了增强抗肿瘤免疫力的能力。在这篇文章中,我们全面回顾了结直肠癌免疫治疗策略的现状,并深入了解了这些策略在未来10年可能如何演变,并适应更多局部形式的结肠癌和直肠癌.我们特别关注正在研究的各种组合方法,以逆转癌症诱导的免疫抑制,特别是在错配修复精通/微卫星稳定的结直肠肿瘤中。最后,我们总结了目前对局部免疫调节中一个最近确定的整体因素的理解,结肠微生物组。本文的目的是确定当前的挑战和改进的障碍,并指定将免疫治疗领域的知识应用于旨在改善新辅助治疗患者生存率和相关结局的临床试验的合理设计的机会。
    Immunotherapy in the metastatic setting has drastically altered the landscape of treatment for various types of malignancy, including colorectal cancer. The category of immune checkpoint inhibitors has especially emerged as a class of therapy predicated on a more comprehensive understanding of immune cell-cancer cell regulation and evolution of the tumor microenvironment over time. Strategies including adoptive cellular therapies, tumor vaccines, and antibodies have also demonstrated the ability to enhance antitumor immunity. In this article, we provide a comprehensive review of the current landscape of immunotherapeutic strategies in colorectal cancer and provide insight into how these strategies may evolve in the next decade and be adapted to more localized forms of cancers of the colon and rectum. We provide particular focus on various combination approaches under investigation for reversing cancer-induced immunosuppression, especially in mismatch repair-proficient/microsatellite-stable colorectal tumors. Finally, we summarize current understanding on a recently identified integral factor in local immune regulation, the colonic microbiome. The aim of this article is to identify current challenges and barriers to improvement and to specify opportunities for applying knowledge in the immunotherapy sphere to rational design of clinical trials intended to improve survival and related outcomes in patients treated in the neoadjuvant setting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    最近,在不使用常规毒性化学药物的情况下,通过纳米催化药物提高肿瘤内高毒性活性氧(ROS)的肿瘤内水平,用于肿瘤特异性治疗,引起了人们的极大兴趣。which,然而,由于纳米药物在肿瘤部位的积累相对较差,并且在很大程度上阻止了肿瘤内浸润,因此治疗效果仍然不令人满意。在这里,一种超声(US)触发的双尺寸/电荷可切换纳米催化药物,指定为Cu-LDH/HMME@Lips,通过催化ROS生成构建用于深部实体瘤治疗。纳米药物的带负电荷的脂质体外层可以延长血液循环,从而显着增强肿瘤的积累,而在US刺激下从脂质体释放的带正电荷的Fenton样催化剂Cu-LDH显示出通过胞吞作用的肿瘤内渗透增强。同时,共释放的声敏剂血卟啉单甲醚(HMME)在US辐照下催化单线态氧(1O2)的产生,和深部肿瘤浸润的Cu-LDH催化H2O2分解,特别是在轻度酸性肿瘤微环境(TME)内产生高毒性的羟基自由基(·OH)。通过双重大小/电荷切换机制有效的肿瘤内积累和渗透,以及通过声敏化和Fenton样反应产生的ROS,保证了纳米催化药物对肿瘤深部治疗的高疗效。
    Elevating intratumoral levels of highly toxic reactive oxygen species (ROS) by nanocatalytic medicine for tumor-specific therapy without using conventional toxic chemodrugs is recently of considerable interest, which, however, still suffers from less satisfactory therapeutic efficacy due to the relatively poor accumulation at the tumor site and largely blocked intratumoral infiltration of nanomedicines. Herein, an ultrasound (US)-triggered dual size/charge-switchable nanocatalytic medicine, designated as Cu-LDH/HMME@Lips, is constructed for deep solid tumor therapy via catalytic ROS generations. The negatively charged liposome outer-layer of the nanomedicine enables much-prolonged blood circulation for significantly enhanced tumoral accumulation, while the positively charged Fenton-like catalyst Cu-LDH released from the liposome under the US stimulation demonstrates much enhanced intratumoral penetration via transcytosis. In the meantime, the co-released sonosensitizer hematoporphyrin monomethyl ether (HMME) catalyze the singlet oxygen (1O2) generation upon the US irradiation, and deep-tumoral infiltrated Cu-LDH catalyzes the H2O2 decomposition to produce highly toxic hydroxyl radical (·OH) specifically within the mildly acidic tumor microenvironment (TME). The efficient intratumoral accumulation and penetration via the dual size/charge switching mechanism, and the ROS generations by both sonosensitization and Fenton-like reactions, ensures the high therapeutic efficacy for the deep tumor therapy by the nanocatalytic medicine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    常规化疗仍是癌症治疗的重要选择,但是它的细胞选择性差,严重的副作用,和抗药性。近年来,利用纳米颗粒(NPs)改善化疗药物的治疗效果已成为人们关注的焦点。纳米技术通过高负载能力极大地改变了肿瘤学的面貌,毒性较小,药物的靶向递送,增加对目标部位的吸收,和优化传统药物的药代动力学模式。目前,正在设想在新型纳米药物设计领域进行研究,如脂质体,聚合物NP,生物NP,和无机NP,从而使化疗有效和持久。到现在为止,已经使用广泛的纳米载体治疗包括肺在内的实体瘤进行了许多研究,乳房,胰腺,大脑,还有肝脏.为纳米载药制剂的进一步应用提供参考,这篇综述概述了纳米载体的最新发展,以及它们在几种实体瘤治疗中的最新应用情况。
    Conventional chemotherapy is still an important option of cancer treatment, but it has poor cell selectivity, severe side effects, and drug resistance. Utilizing nanoparticles (NPs) to improve the therapeutic effect of chemotherapeutic drugs has been highlighted in recent years. Nanotechnology dramatically changed the face of oncology by high loading capacity, less toxicity, targeted delivery of drugs, increased uptake to target sites, and optimized pharmacokinetic patterns of traditional drugs. At present, research is being envisaged in the field of novel nano-pharmaceutical design, such as liposome, polymer NPs, bio-NPs, and inorganic NPs, so as to make chemotherapy effective and long-lasting. Till now, a number of studies have been conducted using a wide range of nanocarriers for the treatment of solid tumors including lung, breast, pancreas, brain, and liver. To provide a reference for the further application of chemodrug-loaded nanoformulations, this review gives an overview of the recent development of nanocarriers, and the updated status of their use in the treatment of several solid tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Conventional systemic chemotherapeutic regimens suffer from challenges such as nonspecificity, shorter half-life, clearance of drugs, and dose-limiting toxicity. Localized delivery of chemotherapeutic drugs through noninvasive spatiotemporally controllable stimuli-responsive drug delivery systems could overcome these drawbacks while utilizing drugs approved for cancer treatment. In this regard, we developed photoelectro active nanocomposite silk-based drug delivery systems (DDS) exhibiting on-demand drug release in vivo. A functionally modified single-walled carbon nanotube loaded with doxorubicin (DOX) was embedded within a cross-linker free silk hydrogel. The resultant nanocomposite silk hydrogel showed electrical field responsiveness and near-infrared (NIR) laser-induced hyperthermal effect. The remote application of these stimuli in tandem or independent manner led to the increased thermal and electrical conductivity of nanocomposite hydrogel, which effectively triggered the intermittent on-demand drug release. In a proof-of-concept in vivo tumor regression study, the nanocomposite hydrogel was administered in a minimally invasive way at the periphery of the tumor by covering most of it. During the 21-day study, drastic tumor regression was recorded upon regular stimulation of nanocomposite hydrogel with simultaneous or individual external application of an electric field and NIR laser. Tumor cell death marker expression analysis uncovered the induction of apoptosis in tumor cells leading to its shrinkage. Heart ultrasound and histology revealed no cardiotoxicity associated with localized DOX treatment. To our knowledge, this is also the first report to show the simultaneous application of electric field and NIR laser in vivo for localized tumor therapy, and our results suggested that such strategy might have high clinical translational potential.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Necrosis targeting radiopharmaceutical (131)I-hypericin ((131)I-Hyp) has been studied for the therapy of solid malignancies. However, serious side effects may be caused by its unwanted radioactivity after being metabolized by the liver and excreted via bile in the digestive tract. Thus the aim of this study was to investigate two kinds of bile draining for reducing them. Thirty-eight normal rats were intravenously injected with (131)I-Hyp, 24 of which were subjected to the common bile duct (CBD) drainage for gamma counting of collected bile and tissues during 1-6, 7-12, 13-18, and 19-24 h (n = 6 each group), 12 of which were divided into two groups (n = 6 each group) for comparison of the drainage efficiency between CBD catheterization and duodenum intubation by collecting their bile at the first 4 h. Afterwards the 12 rats together with the last two rats which were not drained were scanned via single-photon emission computerized tomography/computed tomography (SPECT/CT) to check the differences. The images showed that almost no intestinal radioactivity can be found in those 12 drained rats while discernible radioactivity in the two undrained rats. The results also indicated that the most of the radioactivity was excreted from the bile within the first 12 h, accounting to 92% within 24 h. The radioactive metabolites in the small and large intestines peaked at 12 h and 18 h, respectively. No differences were found in those two ways of drainages. Thus bile drainage is highly recommended for the patients who were treated by (131)I-Hyp if human being and rats have a similar excretion pattern. This strategy can be clinically achieved by using a nasobiliary or nasoduodenal drainage catheter.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号