humanized tumor mice (HTM)

  • 文章类型: Journal Article
    HER4受体对雌激素受体阳性乳腺癌的生长和治疗的影响尚不确定。使用CRISPR/Cas9技术,我们产生了来自HER4阳性MCF-7,T-47D的稳定HER4敲除变体,和ZR-75-1乳腺癌细胞系。我们研究了肿瘤细胞增殖以及他莫昔芬的细胞和分子机制,abemaciclib,AMG232和NRG1处理作为体外HER4的功能。HER4差异影响对他莫昔芬和abemaciclib治疗的细胞反应。最显著的是MCF-7在HER4敲除后的体外敏感性增加和NRG1对细胞增殖的抑制。此外,我们基于MCF-7HER4-野生型和相应的HER4-敲除细胞,评估了人源化肿瘤小鼠(HTM)对他莫昔芬和abemaciclib治疗的反应和免疫效应.没有任何治疗,HER4基因敲除后HTM中MCF-7肿瘤生长增强,提示HER4在临床前但类似人的条件下具有肿瘤抑制作用.这种现象与体内MCF-7中HER2表达增加有关。与HER4无关,abemaciclib和他莫昔芬治疗可显著抑制这些小鼠的肿瘤生长。然而,经abemaciclib治疗的激素受体阳性乳腺癌患者的肿瘤相关mdm2基因拷贝增加或HER4表达显著,无事件生存率降低.显然,HER4的存在影响他莫昔芬和abemaciclib治疗不同雌激素受体阳性乳腺癌细胞的疗效,甚至在不同程度上,并且与接受abemaciclib治疗的患者的不良结局相关。
    The impact of the HER4 receptor on the growth and treatment of estrogen receptor-positive breast cancer is widely uncertain. Using CRISPR/Cas9 technology, we generated stable HER4 knockout variants derived from the HER4-positive MCF-7, T-47D, and ZR-75-1 breast cancer cell lines. We investigated tumor cell proliferation as well as the cellular and molecular mechanisms of tamoxifen, abemaciclib, AMG232, and NRG1 treatments as a function of HER4 in vitro. HER4 differentially affects the cellular response to tamoxifen and abemaciclib treatment. Most conspicuous is the increased sensitivity of MCF-7 in vitro upon HER4 knockout and the inhibition of cell proliferation by NRG1. Additionally, we assessed tumor growth and immunological effects as responses to tamoxifen and abemaciclib therapy in humanized tumor mice (HTM) based on MCF-7 HER4-wildtype and the corresponding HER4-knockout cells. Without any treatment, the enhanced MCF-7 tumor growth in HTM upon HER4 knockout suggests a tumor-suppressive effect of HER4 under preclinical but human-like conditions. This phenomenon is associated with an increased HER2 expression in MCF-7 in vivo. Independent of HER4, abemaciclib and tamoxifen treatment considerably inhibited tumor growth in these mice. However, abemaciclib-treated hormone receptor-positive breast cancer patients with tumor-associated mdm2 gene copy gains or pronounced HER4 expression showed a reduced event-free survival. Evidently, the presence of HER4 affects the efficacy of tamoxifen and abemaciclib treatment in different estrogen receptor-positive breast cancer cells, even to different extents, and is associated with unfavorable outcomes in abemaciclib-treated 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
    术前放射治疗目前尚未整合到乳腺癌的治疗方案中。然而,通过新辅助照射将免疫“冷”乳腺癌转化为其“热”变体,应该引起内源性肿瘤免疫防御,因此,提高免疫治疗效率。我们研究了亚致死的细胞和免疫效应,ERpos的新辅助照射。,HER2位置。,人源化肿瘤小鼠(HTM)的体外和体内三阴性乳腺癌亚型。这种小鼠模型的特征是类似人的免疫系统,因此有助于详细分析新佐剂的机制和效率。辐照诱导的“原位疫苗接种”,特别是在同时应用检查点治疗的情况下。类似于临床表现,我们观察到HER2-pos管腔的免疫原性逐渐增加。到HTM的三阴性亚型,表明免疫细胞浸润到肿瘤组织中。抗PD-L1治疗将HER2-pos分开。将阴性HTM分为响应者和非响应者,而管腔HTM基本上没有反应。在HER2-pos中单独照射是有效的。和腔亚型特异性HTM,并支持克服单一抗PD-L1治疗的无反应性。治疗成功与脾脏中T细胞比例和PD-1表达的显著增加相关。在所有亚型特异性HTM联合治疗中,被证明对减少肿瘤生长最有效,增强免疫反应,并在抗PD-L1治疗期间将无应答者转化为应答者。在HTM中,新辅助放疗以亚型特异性方式增强抗PD-L1检查点治疗乳腺癌.根据“板凳到床边”的原则,这项研究为在检查点治疗中临床应用新辅助放疗提供了重要的基础.
    Pre-operative radiation therapy is not currently integrated into the treatment protocols for breast cancer. However, transforming immunological \"cold\" breast cancers by neoadjuvant irradiation into their \"hot\" variants is supposed to elicit an endogenous tumor immune defense and, thus, enhance immunotherapy efficiency. We investigated cellular and immunological effects of sub-lethal, neoadjuvant irradiation of ER pos., HER2 pos., and triple-negative breast cancer subtypes in-vitro and in-vivo in humanized tumor mice (HTM). This mouse model is characterized by a human-like immune system and therefore facilitates detailed analysis of the mechanisms and efficiency of neoadjuvant, irradiation-induced \"in-situ vaccination\", especially in the context of concurrently applied checkpoint therapy. Similar to clinical appearances, we observed a gradually increased immunogenicity from the luminal over the HER2-pos. to the triple negative subtype in HTM indicated by an increasing immune cell infiltration into the tumor tissue. Anti-PD-L1 therapy divided the HER2-pos. and triple negative HTM groups into responder and non-responder, while the luminal HTMs were basically irresponsive. Irradiation alone was effective in the HER2-pos. and luminal subtype-specific HTM and was supportive for overcoming irresponsiveness to single anti-PD-L1 treatment. The treatment success correlated with a significantly increased T cell proportion and PD-1 expression in the spleen. In all subtype-specific HTM combination therapy proved most effective in diminishing tumor growth, enhancing the immune response, and converted non-responder into responder during anti-PD-L1 therapy. In HTM, neoadjuvant irradiation reinforced anti-PD-L1 checkpoint treatment of breast cancer in a subtype -specific manner. According to the \"bench to bedside\" principle, this study offers a vital foundation for clinical translating the use of neoadjuvant irradiation in the context of checkpoint therapy.
    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
    检查点阻断特别基于PD-1/PD-L1抑制性抗体。然而,有效的免疫肿瘤防御不仅可以被PD-(L)1阻断,而且还可以被其他免疫检查点分子的存在阻断。这里,我们研究了几种免疫检查点蛋白及其可溶性形式的共表达(例如,PD-1,TIM-3,LAG-3,PD-L1,PD-L2等)在同时携带细胞系来源(JIMT-1,MDA-MB-231,MCF-7)或患者来源的乳腺癌和功能性人类免疫系统的人源化肿瘤小鼠(HTM)中。我们鉴定了具有三阳性PD-1、LAG-3和TIM-3表型的肿瘤浸润性T细胞。虽然PD-1在CD4和CD8T细胞中的表达增加,在基于MDA-MB-231的HTM模型中,发现TIM-3尤其在细胞毒性T细胞中上调。在血清中检测到高水平的可溶性TIM-3和半乳糖凝集素-9(TIM-3配体)。令人惊讶的是,在携带PD-L1阳性肿瘤的小鼠中发现可溶性PD-L2,但仅低水平的sPD-L1。在R2基因组学分析平台上对包含3039个原发性乳腺癌样品的数据集的分析显示,TIM-3,半乳糖凝集素-9和LAG-3表达增加,不仅在三阴性乳腺癌中,而且在HER2+和激素受体阳性乳腺癌亚型中也是如此。这些数据表明LAG-3和TIM-3代表乳腺癌抗免疫领域内的其他关键分子。
    Checkpoint blockade is particularly based on PD-1/PD-L1-inhibiting antibodies. However, an efficient immunological tumor defense can be blocked not only by PD-(L)1 but also by the presence of additional immune checkpoint molecules. Here, we investigated the co-expression of several immune checkpoint proteins and the soluble forms thereof (e.g., PD-1, TIM-3, LAG-3, PD-L1, PD-L2 and others) in humanized tumor mice (HTM) simultaneously harboring cell line-derived (JIMT-1, MDA-MB-231, MCF-7) or patient-derived breast cancer and a functional human immune system. We identified tumor-infiltrating T cells with a triple-positive PD-1, LAG-3 and TIM-3 phenotype. While PD-1 expression was increased in both the CD4 and CD8 T cells, TIM-3 was found to be upregulated particularly in the cytotoxic T cells in the MDA-MB-231-based HTM model. High levels of soluble TIM-3 and galectin-9 (a TIM-3 ligand) were detected in the serum. Surprisingly, soluble PD-L2, but only low levels of sPD-L1, were found in mice harboring PD-L1-positive tumors. Analysis of a dataset containing 3039 primary breast cancer samples on the R2 Genomics Analysis Platform revealed increased TIM-3, galectin-9 and LAG-3 expression, not only in triple-negative breast cancer but also in the HER2+ and hormone receptor-positive breast cancer subtypes. These data indicate that LAG-3 and TIM-3 represent additional key molecules within the breast cancer anti-immunity landscape.
    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
    “人源化”小鼠已广泛用于人类癌症进展的表征,并作为强大的临床前模型。多色表型的标准化可以帮助识别与检查点相关的并发症有关的免疫细胞模式。因此,我们将已建立的免疫细胞谱分析方案应用于我们的人源化患者来源异种移植(hPDX)模型.hPDX的特征在于人类免疫系统和患者来源的肿瘤移植的共存。这些小鼠在CD34干细胞移植后具有类似人的免疫系统,而免疫系统的重建水平与移植的CD34细胞的数量无关。造血干细胞(HSC)移植中CD3细胞的污染≤1.2%,不会引发异常的T细胞成熟。确定了不同的B和T细胞分化阶段,以及表达TIGIT的调节性T细胞(Tregs)和耗尽的T细胞,PD-1或KLRG1。总的来说,使用流式细胞术表征免疫细胞的标准化方案的应用将有助于更好地理解免疫肿瘤过程.
    \"Humanized\" mice have been widely used for the characterization of human cancer progression and as a powerful preclinical model. Standardization of multicolor phenotyping could help to identify immune cell patterns involved in checkpoint-related complications. Therefore, we applied established protocols for immune cell profiling to our humanized Patient-Derived Xenograft (hPDX) model. hPDX are characterized by the co-existence of a human immune system and a patient-derived tumor transplant. These mice possess a human-like immune system after CD34+ stem cell transplantation while the reconstitution level of the immune system was not related to the quantity of transplanted CD34+ cells. Contamination ≤ 1.2% by CD3+ cells in the hematopoietic stem cell (HSC) transplant did not trigger abnormal T cell maturation. Different B and T cell differentiation stages were identified, as well as regulatory T cells (Tregs) and exhausted T cells that expressed TIGIT, PD-1, or KLRG1. Overall, the application of standardized protocols for the characterization of immune cells using flow cytometry will contribute to a better understanding of immune-oncologic processes.
    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
    CX3CL1是一种多功能趋化因子,参与许多生物过程,如免疫细胞吸引和增强肿瘤免疫细胞相互作用,还能增强肿瘤细胞的增殖和转移。多种活性部分由两种CX3CL1亚型决定,通过蛋白酶的蛋白水解切割产生的膜结合和可溶性形式。这里,我们研究了CX3CL1过表达对MDA-MB-453和SK-BR-3乳腺癌细胞的影响。此外,我们评估了基质金属蛋白酶抑制剂TMI-1和GI254023X联合抗HER2抗体曲妥珠单抗在体外和体内的治疗能力.TMI-1和GI254023X在体外引起CX3CL1和HER2的脱落减少,但对肿瘤细胞增殖或活力没有影响。此外,除非CX3CL1在转染时过表达(MDA-MB-453CX3CL1),否则曲妥珠单抗治疗不会延迟MDA-MB-453细胞体外扩增。在人源化肿瘤小鼠中,这表明人类肿瘤和人类免疫系统共存,CX3CL1过表达导致肿瘤生长略微增强。然而,曲妥珠单抗治疗可减弱MDA-MB-453CX3CL1和空载体转染的MDA-MB-453移植小鼠的肿瘤生长,但显示出增强的效率,尤其是在预防CX3CL1过表达癌细胞的肺转移方面。然而,TMI-1没有进一步增强曲妥珠单抗治疗功效。
    CX3CL1 is a multifunctional chemokine that is involved in numerous biological processes, such as immune cell attraction and enhanced tumor immune cell interaction, but also in enhancing tumor cell proliferation and metastasis. The multifarious activity is partially determined by two CX3CL1 isoforms, a membrane-bound and a soluble version generated by proteolytic cleavage through proteases. Here, we investigated the impact of CX3CL1 overexpression in MDA-MB-453 and SK-BR-3 breast cancer cells. Moreover, we evaluated the therapeutic capacity of Matrix-Metalloproteinases-inhibitors TMI-1 and GI254023X in combination with the anti-HER2 antibody trastuzumab in vitro and in vivo. TMI-1 and GI254023X caused a reduced shedding of CX3CL1 and of HER2 in vitro but without effects on tumor cell proliferation or viability. In addition, trastuzumab treatment did not retard MDA-MB-453 cell expansion in vitro unless CX3CL1 was overexpressed upon transfection (MDA-MB-453CX3CL1). In humanized tumor mice, which show a coexistence of human tumor and human immune system, CX3CL1 overexpression resulted in a slightly enhanced tumor growth. However, trastuzumab treatment attenuated tumor growth of both MDA-MB-453CX3CL1 and empty vector transfected MDA-MB-453 transplanted mice but showed enhanced efficiency especially in preventing lung metastases in CX3CL1 overexpressing cancer cells. However, TMI-1 did not further enhance the trastuzumab treatment efficacy.
    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
    Cancer immunotherapy has been shown to enhance established treatment regimens. We evaluated the potential reinforcing effect of IL-15 in trastuzumab treated humanized tumor mice (HTM) which were generated by concurrent transplantation of neonatal NOD-scid IL2Rγnull mice with human hematopoietic stem cells (HSC) and HER2 positive breast cancer cells (metastasizing SK-BR-3, solid tumor forming BT474).We found that trastuzumab treatment efficacy mainly depends on the immediate anti-tumorigenic cellular effect which is significantly enhanced by tumor interacting immune cells upon cotransplantion of HSC. However, trastuzumab treatment caused elevated CD44 expression on tumor cells that metastasized into the lung and liver but did not hinder tumor cell dissemination into the bone marrow. Moreover, in a number of SK-BR-3-transplanted animals disseminated CD44high/CD24low tumor cells lost trastuzumab sensitivity. Concerning the FcγRIIIa polymorphism, trastuzumab treatment efficiency in HTM was higher in mice with NK-cells harboring the high affinity FcγRIIIa compared to those with low affinity FcγRIIIa. In contrast, IL-15 caused the strongest NK-cell activation in heterozygous low affinity FcγRIIIa animals. Although IL-15 enhanced the trastuzumab mediated tumor defense, an unspecific immune stimulation resulted in preterm animal death due to systemic inflammation. Overall, treatment studies based on \"patient-like\" HTM revealed critical and adverse immune-related mechanisms which must be managed prior to clinical testing.
    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
    通过将人造血干细胞和过表达HER2的人乳腺癌细胞共移植到新生NOD-scidIL2Rγ(null)(NSG)小鼠中来产生人源化肿瘤小鼠(HTM)。这些小鼠的特征在于人类免疫系统的发育与人类乳腺癌的生长相结合。由于同时移植到新生小鼠中,MHC不匹配的肿瘤细胞的转移导致固体共存和免疫细胞激活(CD4(+)T细胞,自然杀伤细胞,和骨髓细胞),但没有拒绝的证据.HTM脾脏的组织学染色显示人抗原呈递细胞与人T和B细胞共同定位,从而允许MHC依赖性相互作用。从而产生T细胞依赖性抗体。这里,我们研究了这些小鼠产生人类肿瘤特异性抗体的能力,以及与肿瘤生长相关的免疫球蛋白滴度.我们在HTM的血清中发现了可检测的IgM和IgG,当IgG血清浓度高于10µg/ml时,它显然控制了肿瘤的发展。Westernblot分析显示,HTM中产生的肿瘤特异性抗体不识别HER2/neu抗原,但不同,可能与乳腺癌治疗相关的抗原。总之,HTM提供了一种新的方法来生成完全的人单克隆抗体,该抗体不需要进一步的遗传操作(例如g.,人性化)在人类中的潜在应用。此外,所产生的抗体的功效和安全性可以在类似人的条件下在相同的小鼠模型中进行测试。这对于目前没有可用的抗体疗法的癌症亚型可能特别感兴趣。
    Humanized tumor mice (HTM) were generated by the co-transplantation of human hematopoietic stem cells and human breast cancer cells overexpressing HER2 into neonatal NOD-scid IL2Rγ(null) (NSG) mice. These mice are characterized by the development of a human immune system in combination with human breast cancer growth. Due to concurrent transplantation into newborn mice, transfer of MHC-mismatched tumor cells resulted in solid coexistence and immune cell activation (CD4(+) T cells, natural killer cells, and myeloid cells), but without evidence for rejection. Histological staining of the spleen of HTM revealed co-localization of human antigen-presenting cells together with human T and B cells allowing MHC-dependent interaction, and thereby the generation of T cell-dependent antibody production. Here, we investigated the capability of these mice to generate human tumor-specific antibodies and correlated immunoglobulin titers with tumor outgrowth. We found detectable IgM and also IgG amounts in the serum of HTM, which apparently controlled tumor development when IgG serum concentrations were above 10 µg/ml. Western blot analyses revealed that the tumor-specific antibodies generated in HTM did not recognize HER2/neu antigens, but different, possibly relevant antigens for breast cancer therapy. In conclusion, HTM offer a novel approach to generate complete human monoclonal antibodies that do not require further genetic manipulation (e. g., humanization) for a potential application in humans. In addition, efficacy and safety of the generated antibodies can be tested in the same mouse model under human-like conditions. This might be of particular interest for cancer subtypes with no currently available antibody therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号