NK cell therapy

NK 细胞治疗
  • 文章类型: Journal Article
    mRNA的应用经历了前所未有的应用-从疫苗接种到细胞治疗。自然杀伤(NK)细胞被认为在免疫疗法中具有显著的潜力。基于NK的细胞疗法已引起人们的注意,因为同种异体移植物具有最小的移植物抗宿主风险,从而导致更容易的现成生产。NK细胞可以用病毒载体或电穿孔进行工程改造,涉及高成本,风险,和毒性,强调需要替代方式作为mRNA技术。我们成功开发,筛选,并优化了基于咪唑脂质的新型脂质平台。制剂通过微流体混合制备,并表现出约100nm的尺寸,多分散指数小于0.2。他们能够转染NK-92细胞,KHYG-1细胞,和高效无细胞毒性的原代NK细胞,而LipofectamineMessengerMax和D-Lin-MC3基于脂质纳米颗粒的制剂则没有。此外,与修饰的mRNA相比,未修饰的mRNA的翻译更高,时间更稳定。值得注意的是,治疗相关的白介素2mRNA的递送导致NK细胞系和原代NK细胞的生存力以及保留的活化标记和细胞毒性。总之,我们的平台具备成功部署基于NK的治疗策略所需的所有先决条件.
    mRNA applications have undergone unprecedented applications-from vaccination to cell therapy. Natural killer (NK) cells are recognized to have a significant potential in immunotherapy. NK-based cell therapy has drawn attention as allogenic graft with a minimal graft-versus-host risk leading to easier off-the-shelf production. NK cells can be engineered with either viral vectors or electroporation, involving high costs, risks, and toxicity, emphasizing the need for alternative way as mRNA technology. We successfully developed, screened, and optimized novel lipid-based platforms based on imidazole lipids. Formulations are produced by microfluidic mixing and exhibit a size of approximately 100 nm with a polydispersity index of less than 0.2. They are able to transfect NK-92 cells, KHYG-1 cells, and primary NK cells with high efficiency without cytotoxicity, while Lipofectamine Messenger Max and D-Lin-MC3 lipid nanoparticle-based formulations do not. Moreover, the translation of non-modified mRNA was higher and more stable in time compared with a modified one. Remarkably, the delivery of therapeutically relevant interleukin 2 mRNA resulted in extended viability together with preserved activation markers and cytotoxic ability of both NK cell lines and primary NK cells. Altogether, our platforms feature all prerequisites needed for the successful deployment of NK-based therapeutic strategies.
    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
    衰老过程错综复杂地涉及免疫系统动力学,在管理衰老细胞(SNCs)及其衰老相关分泌表型(SASPs)方面具有关键作用。不幸的是,免疫衰老,随着年龄的增长,免疫力逐渐失调,阻碍有效的SNC消除,导致积累,再加上SASPs的发布,which,反过来,抑制免疫力和增加对衰老相关疾病(AAD)的易感性。自然杀伤(NK)细胞,先天免疫系统的组成部分,在迅速解决SNCs方面发挥着关键作用。这些细胞还与先天免疫和适应性免疫的其他成分协调以监视和消除这些细胞。因此,在衰老过程中保持NK细胞功能对于逃避AAD和促进健康衰老至关重要。或者,基于NK细胞的疗法为解决与衰老相关的挑战提供了有希望的途径。值得注意的,最近在过继NK细胞疗法的研究显示了恢复免疫衰老的前景,消除SNCs,减轻SASPs。这一进展为过继NK细胞疗法提供了证据概念,并有望成为长寿疗法的新兴革命。
    The aging process intricately involves immune system dynamics, with a crucial role in managing senescent cells (SNCs) and their senescence-associated secretory phenotypes (SASPs). Unfortunately, immunosenescence, a progressively dysregulated immunity with age, hampers effective SNC elimination, leading to accumulation, coupled with the release of SASPs, which, in turn, inhibits immunity and heightened susceptibility to aging-associated diseases (AADs). Natural killer (NK) cells, integral to the innate immune system, play a pivotal role in addressing SNCs swiftly. These cells also coordinate with other components of both innate and adaptive immunity to surveil and eliminate these cells. Accordingly, preserving NK cell function during aging is crucial for evading AADs and promoting healthy aging. Alternatively, NK-cell-based therapies present promising avenues for addressing the challenges associated with aging. Notable, recent studies in adoptive NK cell therapy have shown promise in rejuvenating immunosenescence, eliminating SNCs, and alleviating SASPs. This progress provides the proof-concept of adoptive NK cell therapy for senotherapy and holds promise as an emerging revolution in longevity therapeutics.
    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
    NK细胞疗法作为一种潜在的癌症治疗方法已经获得了极大的关注。走向治疗用途,NK细胞需要被激活和扩增以获得高效和大量的有效剂量。这通常通过用细胞因子离体刺激以增强功能或扩增10-14天以增加其活性和数量来完成。对于现成的产品,获得产生大剂量的有效NK细胞的稳健方法是非常期望的。值得注意的是,过去的报道表明,用IL-12,IL-15和IL-18刺激NK细胞赋予它们类似记忆的特性,更好的抗肿瘤活性,和坚持。虽然这种方法产生的NK细胞具有临床上有利的特征,并得到了令人鼓舞的早期血液恶性肿瘤治疗结果的支持。其有限的可扩展性,初始剂量的可变性,以及患者特定生产的必要性阻碍了其更广泛的应用。在这项研究中,用来自K562-41BBL-mbIL21细胞的PM21颗粒刺激NK细胞与使用细胞因子IL-12,IL-15和IL-18的记忆样诱导相结合,以产生具有增强的抗肿瘤功能的NK细胞。使用细胞因子与PM21颗粒(细胞因子和颗粒,CAP)显着增强NK细胞扩增,在14天内实现了惊人的8200倍。机械上,与单独的PM21颗粒相比,这种显着改善是由于关键刺激配体(4-1BBL和IL-2)的受体上调,导致驱动NK细胞大量生长的协同作用,展示了更有效的治疗应用的潜力。CAP-NK细胞的治疗潜力由增强的代谢适应性证明。持久性,和抗肿瘤功能在体外和体内。最后,CAP-NK细胞适用于目前用于开发治疗性NK细胞产品的技术,包括基于CRISPR/Cas9的技术来产生三基因敲除或基因敲入。一起来看,这些数据表明,细胞因子的添加增强了已经有效的体外产生治疗性NK细胞与PM21颗粒的方法,产生优异的NK细胞产品的制造效率和潜在的治疗应用。
    NK cell therapeutics have gained significant attention as a potential cancer treatment. Towards therapeutic use, NK cells need to be activated and expanded to attain high potency and large quantities for an effective dosage. This is typically done by ex vivo stimulation with cytokines to enhance functionality or expansion for 10-14 days to increase both their activity and quantity. Attaining a robust methodology to produce large doses of potent NK cells for an off-the-shelf product is highly desirable. Notably, past reports have shown that stimulating NK cells with IL-12, IL-15, and IL-18 endows them with memory-like properties, better anti-tumor activity, and persistence. While this approach produces NK cells with clinically favorable characteristics supported by encouraging early results for the treatment of hematological malignancies, its limited scalability, variability in initial doses, and the necessity for patient-specific production hinder its broader application. In this study, stimulation of NK cells with PM21-particles derived from K562-41BBL-mbIL21 cells was combined with memory-like induction using cytokines IL-12, IL-15, and IL-18 to produce NK cells with enhanced anti-tumor function. The use of cytokines combined with PM21-particles (cytokine and particle, CAP) significantly enhanced NK cell expansion, achieving a remarkable 8,200-fold in 14 days. Mechanistically, this significant improvement over expansion with PM21-particles alone was due to the upregulation of receptors for key stimulating ligands (4-1BBL and IL-2), resulting in a synergy that drives substantial NK cell growth, showcasing the potential for more effective therapeutic applications. The therapeutic potential of CAP-NK cells was demonstrated by the enhanced metabolic fitness, persistence, and anti-tumor function both in vitro and in vivo. Finally, CAP-NK cells were amenable to current technologies used in developing therapeutic NK cell products, including CRISPR/Cas9-based techniques to generate a triple-gene knockout or a gene knock-in. Taken together, these data demonstrate that the addition of cytokines enhanced the already effective method of ex vivo generation of therapeutic NK cells with PM21-particles, yielding a superior NK cell product for manufacturing efficiency and potential therapeutic applications.
    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
    难治性急性髓系白血病(AML),定义为诊断时两个周期的诱导治疗或复发时一个周期的失败,代表一个结果不佳的子组。单倍体相同的自然杀伤细胞(NK)治疗是一种在难治性恶性肿瘤中正在探索的策略。历史上,在我们的中心,难治性AML患者接受了细胞减灭性治疗(氟达拉滨+胞嘧啶+粒细胞集落刺激因子±伊达比星或米托蒽醌+依托泊苷),随后休息1周,然后接受氟达拉滨+美法仑减量移植.我们在这项试验中使用了相同的骨架(CTRI/2019/02/017505),并在化疗完成后1天添加了来自家庭供体的CD56阳性细胞。CD56阳性选择是使用CliniMACSProdigy系统(MiltenyiBiotec,BergischGladbach,德国),然后在2微摩尔三氧化二砷和500U/mL白介素2的自体血浆中过夜孵育。从2019年2月开始,本试验纳入了14名中位年龄为29岁(四分位距[IQR]:16.5-38.5)的患者。六个是女性。六个患有原发性难治性AML,而八个患有复发性难治性AML。CD56细胞输注剂量中位数为46.16×106/kg(IQR:25.06-70.36)。一名患者在NK细胞输注后撤回同意。在进行移植的13名患者中,5人死于移植后即刻并发症,2人未移植,但处于无形态白血病状态(2人随后均在第二次移植后死于感染性并发症).其余6名移植后存活超过1个月的患者,两人出现疾病复发并死亡。其余四名患者在最后一次随访时仍存活且无复发(存活患者的平均随访时间为24个月)。该队列的2年估计总生存率为28.6%±12.1%,而采用该方法的治疗相关死亡率(TRM)为38.5%±13.5%。单倍型NK细胞治疗作为移植的辅助手段是安全的,在AML患者中需要进一步探索。对于难治性AML,移植后NK输注和减少TRM同时使用移植前NK输注的策略值得探索。
    Refractory acute myeloid leukemia (AML), defined as failure of two cycles of induction therapy at diagnosis or of one cycle at relapse, represents a subgroup with poor outcomes. Haploidentical natural killer cell (NK) therapy is a strategy that is being explored in refractory malignancies. Historically, at our center, patients with refractory AML have been treated with cytoreductive therapy (fludarabine + cytosine + granulocyte colony-stimulating factor ± idarubicin or mitoxantrone + etoposide) followed by 1-week rest and then reduced-intensity transplant with fludarabine + melphalan. We used the same backbone for this trial (CTRI/2019/02/017505) with the addition of CD56-positive cells from a family donor infused 1 day after the completion of chemotherapy. CD56-positive selection was done using a CliniMACS Prodigy system (Miltenyi Biotec, Bergisch Gladbach, Germany) followed by overnight incubation in autologous plasma with 2 micromolar arsenic trioxide and 500 U/mL of interleukin-2. From February 2019, 14 patients with a median age of 29 years (interquartile range [IQR]: 16.5-38.5) were enrolled in this trial. Six were females. Six had primary refractory AML while eight had relapsed refractory AML. The median CD56-cell dose infused was 46.16 × 106/kg (IQR: 25.06-70.36). One patient withdrew consent after NK cell infusion. Of the 13 patients who proceeded to transplant, five died of immediate post-transplant complications while two did not engraft but were in morphologic leukemia-free state (both subsequently died of infective complications after the second transplant). Of the remaining six patients who engrafted and survived beyond 1 month of the transplant, two developed disease relapse and died. The remaining four patients are alive and relapse free at the last follow-up (mean follow-up duration of surviving patients is 24 months). The 2-year estimated overall survival for the cohort was 28.6% ± 12.1% while the treatment-related mortality (TRM) with this approach was 38.5% ± 13.5%. Haploidentical NK cell therapy as an adjunct to transplant is safe and needs further exploration in patients with AML. For refractory AML, post-transplant NK infusion and strategies to reduce TRM while using pre-transplant NK infusion merit exploration.
    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
    目前针对人类免疫缺陷病毒(HIV)的抗逆转录病毒疗法(ART)无法治愈,随着病毒在潜伏的水库中持续存在,需要终身坚持ART并增加合并症的风险。从潜伏的水库中重新激活HIV的“休克和杀死”方法,然后施用抗HIV药物代表了根除潜伏HIV的有希望的策略。为了实现有效的电击和杀戮,我们描述了一种根除HIV水库的策略,该策略结合了潜伏期逆转剂(LRA),广泛中和抗体(bnAbs),自然杀伤(NK)细胞该策略利用共包封LRA和bnAb的聚合物纳米储库(ND)以重新激活潜伏感染并从共施用的NK细胞引起增强的细胞毒性。
    使用纳米沉淀法合成聚(乳酸-共-乙醇酸)(PLGA)ND以将LRA(TNF-α)和bnAb(3BNC117)(TNF-α-3BNC117-NDs)共包封。ACH-2细胞用作潜伏HIV感染的细胞模型。NK92副线,经基因修饰以组成型表达Fc受体CD16,与TNF-α-3BNC117-ND组合施用于ACH-2细胞。通过流式细胞术和ELISA检测ACH-2细胞死亡和细胞外p24,分别。
    稳定的PLGAND以高效率共包封TNF-α和3BNC117,并在生理条件下释放这些药物。在存在TNF-α-3BNC117-NDs的情况下,NK92表型保持相似。从ND释放的TNF-α有效地重新激活ACH-2细胞中的HIV,细胞内p24阳性细胞的频率增加了3.0倍。释放3BNC117中和并结合了重新激活的病毒,靶向57.5%的ACH-2细胞。严重的,TNF-α-3BNC117-ND显着增强NK92细胞介导的对ACH-2细胞的杀伤(1.9倍),并将p24的细胞外水平降低至基线。
    这些研究结果表明,我们新的基于ND的三方策略可以从潜伏感染的细胞中重新激活HIV,为bnAb结合生成HIV特异性位点,并增强NK92细胞对重新激活的HIV感染靶细胞的杀伤作用。
    UNASSIGNED: Current antiretroviral therapies (ART) for human immunodeficiency virus (HIV) are not curative, as the virus persists in latent reservoirs, requiring lifelong adherence to ART and increasing the risk of co-morbidities. \"Shock and kill\" approaches to reactivate HIV from latent reservoirs followed by administration of anti-HIV drugs represent a promising strategy for eradicating latent HIV. To achieve effective shock and kill, we describe a strategy to eradicate the HIV reservoir that combines latency reversing agents (LRAs), broadly neutralizing antibodies (bnAbs), and natural killer (NK) cells. This strategy utilizes a polymer nanodepot (ND) that co-encapsulates the LRA and bnAb to reactivate latent infection and elicit enhanced cytotoxicity from co-administered NK cells.
    UNASSIGNED: Poly(lactic-co-glycolic acid) (PLGA) NDs were synthesized using the nanoprecipitation method to co-encapsulate an LRA (TNF-α) and a bnAb (3BNC117) (TNF-α-3BNC117-NDs). ACH-2 cells were used as a cellular model of latent HIV infection. An NK92 subline, genetically modified to constitutively express the Fc receptor CD16, was administered to ACH-2 cells in combination with TNF-α-3BNC117-NDs. ACH-2 cell death and extracellular p24 were measured via flow cytometry and ELISA, respectively.
    UNASSIGNED: Stable PLGA NDs co-encapsulated TNF-α and 3BNC117 with high efficiencies and released these agents in physiological conditions. NK92 phenotype remained similar in the presence of TNF-α-3BNC117-NDs. TNF-α released from NDs efficiently reactivated HIV in ACH-2 cells, as measured by a 3.0-fold increase in the frequency of intracellular p24 positive cells. Released 3BNC117 neutralized and bound reactivated virus, targeting 57.5% of total ACH-2 cells. Critically, TNF-α-3BNC117-NDs significantly enhanced NK92 cell-mediated killing of ACH-2 cells (1.9-fold) and reduced extracellular levels of p24 to baseline.
    UNASSIGNED: These findings suggest the therapeutic potential of our novel ND-based tripartite strategy to reactivate HIV from latently infected cells, generate an HIV-specific site for bnAb binding, and enhance the killing of reactivated HIV-infected target cells by NK92 cells.
    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
    NK细胞是免疫系统的第一个哨兵,可以识别和根除转化的细胞。在不需要额外信号传导的情况下,它们的激活引起了人们对NK细胞作为癌症免疫疗法中有希望的选择的极大关注。然而,大规模生产NK细胞用于成功的NK细胞治疗是一个需要解决的挑战。改造NK细胞以避免肿瘤逃逸并提高其抗肿瘤效力是近年来被广泛研究的另一个焦点问题。本文综述了干细胞来源的NK细胞技术的最新进展,并讨论了工程NK细胞在癌症免疫治疗中的临床应用潜力。
    NK细胞是我们身体防御系统中的重要细胞,可以发现并破坏肿瘤细胞。这些细胞是在骨髓(成人)或脐带(胚胎期)中由称为干细胞的细胞群产生的,然后释放到血液和淋巴中。干细胞是可以分化成多种细胞类型的早期祖先细胞。因为NK细胞在肿瘤环境中的数量和功能减少,因此,我们可以使用这些干细胞来制造大量的NK细胞用于治疗目的。科学家还可以通过将这些细胞更改为具有特殊传感器来使其更好地杀死肿瘤。最后,NK细胞就像对抗和杀死肿瘤细胞的超级英雄,使用干细胞来制造它们是一种非常有希望的方法来帮助治疗恶性疾病。
    NK cells are the first sentinels of the immune system that can recognize and eradicate transformed cells. Their activation without a need for additional signaling have attracted great attention on the use of NK cells as a promising option in cancer immunotherapy. However, the large-scale production of NK cells for successful NK cells therapy is a challenge that needs to be tackled. Engineering NK cells to avoid tumor escape and improve their antitumor potency are the other matters of focus that have widely been studied in the recent years. This paper reviews the most recent advances in the stem cell-derived NK cell technology and discusses the potential of the engineered NK cells for clinical applications in cancer immunotherapy.
    NK cells are important cells in our body\'s defense system that can find and destroy tumor cells. These cells are made in bone marrow (in adults) or umbilical cord (in the embryonic period) from a population of cells called stem cells, and then released into the blood and lymph. Stem cells are the early ancestral cells that can differentiate into multiple cell types. Because the number and function of NK cells in a tumor context are reduced, thus we can use these stem cells to make lots of NK cells for treatment purposes. Scientists can also make these cells even better at killing tumors by changing them to have special sensors. In the end, NK cells are like superheroes that fight and kill tumor cells, and using stem cells to make them is a really promising way to help treat malignant diseases.
    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
    尽管对恶性脑肿瘤患者进行了多模式治疗,他们的中位生存期<2年。最近,NK细胞通过其直接的天然细胞毒性和通过调节树突状细胞以增强肿瘤抗原的呈递并调节T细胞介导的抗肿瘤反应来提供癌症免疫监视。然而,这种治疗方式在脑肿瘤中的成功尚不清楚。主要原因是;脑肿瘤微环境,NK细胞的制备和给药,和捐赠者的选择。我们先前的研究表明,在动物模型中,颅内注射活化的单倍体NK细胞可根除胶质母细胞瘤肿瘤块,而没有任何肿瘤复发的证据。因此,在本研究中,我们评估了6例复发性胶质母细胞瘤(GBM)和恶性脑肿瘤对化疗/放疗耐药的患者手术腔内或脑脊液(CSF)内注射活体激活的单倍体相同NK细胞的安全性.我们的结果表明,活化的单倍体NK细胞表达激活剂和抑制剂标记,并可以杀死肿瘤细胞。然而,它们对患者来源的GBM(PD-GBM)的细胞毒性潜力大于其细胞系。此外,他们的输液使整体疾病控制率提高了约33.3%,平均存活400天.此外,我们表明,在恶性脑肿瘤中局部施用活化的单倍体NK细胞是安全的,可行,耐受较高剂量,和成本效益。
    Despite multi-modal therapies for patients with malignant brain tumors, their median survival is < 2 years. Recently, NK cells have provided cancer immune surveillance through their direct natural cytotoxicity and by modulating dendritic cells to enhance the presentation of tumor antigens and regulate T-cell-mediated antitumor responses. However, the success of this treatment modality in brain tumors is unclear. The main reasons are; the brain tumor microenvironment, the NK cell preparations and administration, and the donor selection. Our previous study showed that intracranial injection of activated haploidentical NK cells resulted in the eradication of glioblastoma tumor mass in the animal model without any evidence of tumor recurrence. Therefore, in the present study, we evaluated the safety of intra-surgical cavity or intra cerebrospinal fluid (CSF) Injectionofex vivoactivated haploidentical NK cells in six patients with recurrent glioblastoma multiform (GBM) and malignant brain tumors resistance to chemo/radiotherapy. Our results indicated that activated haploidentical NK cells express activator and inhibitor markers and can kill the tumor cells. However, their cytotoxic potential on patient-derived GBM (PD-GBM) was more than that of its cell line. Also, their infusion increased the overall disease control rate by about 33.3%, with a mean survival of 400 days. Moreover, we showed that local administration of the activated haploidentical NK cells in malignant brain tumors is safe, feasible, tolerated at higher doses, and cost-effective.
    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
    自然杀伤(NK)细胞有望在血液系统恶性肿瘤的治疗中产生希望。在过去的二十年中,过继转移的同种异体NK细胞的临床经验表明,CRS或ICANS的安全性和最小风险。与必须进行遗传改变以避免移植物抗宿主病(GVHD)的T细胞不同,可以输注HLA错配的NK细胞而没有GVHD风险。这使它们成为开发现成产品的理想选择。在这篇综述中,我们关注与癌症治疗相关的NK生物学,NK治疗白血病的轨迹,淋巴瘤和骨髓瘤;以及NK细胞平台的优势。我们还将讨论增强NK细胞靶向的新方法,持久性,并在肿瘤微环境中发挥作用。NK细胞治疗的未来取决于实现这些品质的新策略。
    Natural Killer (NK) cells yield promise in therapy of hematologic malignancies. The clinical experience with adoptively transferred allogeneic NK cells over past two decades has revealed safety and minimal risk of CRS or ICANS. Unlike T cells which have to be genetically altered to avoid graft vs host disease (GVHD), HLA mismatched NK cells can be infused without GVHD risk. This makes them ideal for the development of off-the-shelf products. In this review we focus on NK biology relevant to the cancer therapy, the trajectory of NK therapeutics for leukemia, lymphoma, and myeloma; and advantages of the NK cell platform. We will also discuss novel methods to enhance NK cell targeting, persistence, and function in the tumor microenvironment. The future of NK cell therapy depends on novel strategies to realize these qualities.
    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
    三阴性乳腺癌(TNBC)是乳腺癌干细胞(BCSC)含量最高且对常规治疗具有抵抗力的一种乳腺癌亚型。由于免疫抑制的肿瘤微环境和乳腺癌细胞的免疫原性,使用免疫细胞,特别是自然杀伤细胞(NK)在实体瘤的治疗中,包括乳腺癌,一直不令人满意。因此,确定新的治疗方法是乳腺癌治疗的必要条件。此外,癌症治疗的组合是提高治疗效果的有效策略。在这项研究中,我们用BIBR1532抑制端粒酶(hTERT),以刺激NK细胞对乳腺癌细胞的细胞毒性。用IC50水平的BIBR1532将MDA-MB-231细胞系治愈24h。用PBS洗涤细胞并与外周血NK细胞共培养5h。最后,我们评估了端粒酶抑制对NK细胞的细胞毒性和乳腺癌细胞凋亡的影响。此外,评估hTERT和凋亡相关基因的表达。数据显示,端粒酶的抑制增加了NK细胞对乳腺癌的细胞毒性。此外,端粒酶抑制和NK细胞通过抑制hTERT协同增强乳腺癌细胞的细胞死亡,bax的上调,糟糕的表情。总之,端粒酶抑制使乳腺癌细胞对NK细胞治疗更敏感。因此,端粒酶抑制和NK细胞的结合可用于乳腺癌细胞的治疗。
    Triple-negative breast cancer (TNBC) is a subtype of breast tumor with the highest breast cancer stem cells (BCSCs) content and resistance to conventional treatment. Due to the immunosuppressive tumor microenvironment and immunogenicity of breast cancer cells, the use of immune cells, especially natural killer cells (NK) in the treatment of solid tumors, including breast cancer, has been unsatisfactory. Therefore, identifying novel therapies is requisite for breast cancer treatment. Furthermore, the combination of cancer therapies is an effective strategy to improve therapeutic effectiveness. In this study, we inhibited telomerase (hTERT) with BIBR1532, in stimulating NK cell cytotoxicity against breast cancer cells. The MDA-MB-231 cell line was cured with IC50 level of BIBR1532 for 24 h. Afterward, cells were washed with PBS and were co-cultured with peripheral blood NK cell for 5h. Finally, we assessed the impact of telomerase inhibition on the cytotoxicity of NK cells and apoptosis of breast cancer. Also, the expression of hTERT and apoptotic-related genes were evaluated. The data revealed that inhibition of telomerase increases NK cell cytotoxicity against breast cancer. Furthermore, telomerase inhibition and NK cell synergistically enhanced cell death in breast cancer cells by suppressing hTERT, upregulation of bax, and bad expression. In conclusion, telomerase suppression makes breast cancer cells more sensitive to NK cell therapy. Consequently, the combination of telomerase inhibition and NK cells can be useful in the treatment of breast cancer 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
    过继性细胞疗法的最新进展极大地改变了癌症免疫疗法的范式。尽管目前的免疫疗法可以治愈许多多发性骨髓瘤(MM)患者,复发/难治性MM(RR/MM)在某些情况下仍然具有挑战性。自然杀伤(NK)细胞是先天免疫细胞,对恶性细胞如骨髓瘤细胞发挥有效的细胞毒活性。除了它们的抗肿瘤特性,NK细胞在移植后不诱导移植物抗宿主病。因此,它们为治疗RR/MM患者提供了一种有希望的方法。目前,已经尝试产生NK细胞的大规模和良好的制造规范(GMP)。源自自身患者或同种异体供体的离体扩增/活化的NK细胞是MM中NK细胞疗法的潜在选择。此外,新的基于细胞的产物如NK细胞系和嵌合抗原受体(CAR)-NK细胞可以为NK细胞治疗提供现成的来源。这里,我们总结了MM微环境中的NK细胞活性,并重点介绍了MM患者的不同NK细胞治疗方法。
    Recent advances in adoptive cell therapy have considerably changed the paradigm of cancer immunotherapy. Although current immunotherapies could cure many patients with multiple myeloma (MM), relapsed/refractory MM (RR/MM) is still challenging in some cases. Natural killer (NK) cells are innate immune cells that exert effective cytotoxic activity against malignant cells like myeloma cells. In addition to their antitumor properties, NK cells do not induce graft versus host disease following transplantation. Therefore, they provide a promising approach to treating RR/MM patients. Currently, attempts have been made to produce large-scale and good manufacturing practices (GMP) of NK cells. Ex vivo expanded/activated NK cells derived from the own patient or allogenic donors are potential options for NK cell therapy in MM. Besides, novel cell-based products such as NK cell lines and chimeric antigen receptor (CAR)-NK cells may provide an off-the-shelf source for NK cell therapy. Here, we summarized NK cell activity in the MM microenvironment and focused on different NK cell therapy methods for MM patients.
    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