Adoptive T cell therapy

过继性 T 细胞疗法
  • 文章类型: Journal Article
    多瘤病毒是一组小的,无包裹,可以感染各种宿主的双链DNA病毒,包括人类。已知BKPyV会导致人类多瘤病毒相关肾病(HPyVAN)等疾病,人多瘤病毒相关出血性膀胱炎(HPyVHC),和人类多瘤病毒相关的尿路上皮癌(HPyVUC)。JCPyV,另一方面,负责进行性多灶性白质脑病(PML),严重的中枢神经系统脱髓鞘疾病。PML主要影响免疫受损个体,包括那些感染艾滋病毒的人,某些免疫抑制疗法的接受者,移植患者。HPyV感染的治疗选择有限,但是病毒特异性T细胞(VST)治疗的最新进展显示出了希望。虽然VST疗法在治疗BKPyV和JCPyV感染方面显示出希望,仍然存在一些挑战。其中包括耗时且昂贵的VST准备工作,需要先进的生产设施,以及最佳细胞类型和输注频率的不确定性。据我们所知,85例出血性膀胱炎,27例BKPyV病毒血症,2例BKPyV肾炎,14例出血性膀胱炎和BKPyV病毒血症,文献中32例PML患者接受VST治疗。总体反应是82、33、35和10,局部,无反应,和无结果报告(NA),分别。总之,这篇综述强调了VST治疗作为多瘤病毒感染的一种有希望的治疗方法的重要性,强调需要持续的研究和临床试验,以完善和扩大这种创新的免疫治疗策略。
    Polyomaviruses are a group of small, non-enveloped, double-stranded DNA viruses that can infect various hosts, including humans. BKPyV is known to cause conditions such as human polyomavirus-associated nephropathy (HPyVAN), human polyomavirus-associated hemorrhagic cystitis (HPyVHC), and human polyomavirus-associated urothelial cancer (HPyVUC). JCPyV, on the other hand, is responsible for progressive multifocal leukoencephalopathy (PML), a severe demyelinating disease of the central nervous system. PML primarily affects immunocompromised individuals, including those with HIV, recipients of certain immunosuppressive therapies, and transplant patients. The treatment options for HPyV infections have been limited, but recent developments in virus-specific T cell (VST) therapy have shown promise. While VST therapy has shown promise in treating both BKPyV and JCPyV infections, several challenges remain. These include the time-consuming and costly preparation of VSTs, the need for sophisticated production facilities, and uncertainties regarding the optimal cell type and infusion frequency. To the best of our knowledge, 85 patients with hemorrhagic cystitis, 27 patients with BKPyV viremia, 2 patients with BKPyV nephritis, 14 patients with hemorrhagic cystitis and BKPyV viremia, 32 patients with PML were treated with VST in the literature. The overall response was 82, 33, 35, and 10 complete, partial, non-response, and no-outcome-reported (NA), respectively. In conclusion, this review underscores the importance of VST therapy as a promising treatment approach for polyomavirus infections, emphasizing the need for continued research and clinical trials to refine and expand this innovative immunotherapeutic strategy.
    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:爱泼斯坦-巴尔病毒特异性细胞毒性T淋巴细胞(EBV-CTL)是一种自体过继性T细胞免疫疗法,由个体血液产生,未经遗传修饰而制造。在先前的局部复发或转移性鼻咽癌(R/MNPC)患者的2期试验中,一线吉西他滨、卡铂(GC)和EBV-CTL联合用药显示出客观的抗肿瘤EBV-CTL活性和良好的安全性.本研究探讨了这种联合的一线化学免疫治疗策略与常规化疗治疗相比是否会产生更好的临床疗效和更好的生活质量。
    方法:这个多中心,随机化,3期试验评估了GC和EBV-CTL的疗效和安全性单独GC作为R/MNPC患者的一线治疗。新加坡30个临床地点,马来西亚,台湾,泰国,包括美国(US)。受试者以1:1的比例随机接受一线GC(4个循环)和EBV-CTL(6个循环)或GC(6个循环)。主要结局是总生存期(OS),次要结局包括无进展生存期,客观反应率,临床获益率,生活质量,和安全。
    结果:gov标识符:NCT02578641。
    结果:纳入330例鼻咽癌患者。两个治疗组中的大多数受试者接受≥4个周期的化疗,GC+EBV-CTL组中的大多数受试者接受≥2次EBV-CTL输注。中央良好生产规范(GMP)设施为94%的GC+EBV-CTL受试者生产足够的EBV-CTL。GC+EBV-CTL组的中位OS为25.0个月,GC组为24.9个月(风险比=1.19;95%CI:0.91,1.56;P=0.194)。只有1名受试者经历了与EBV-CTL相关的2级严重不良事件。
    结论:在患有R/MNPC的受试者中,GC+EBV-CTL表现出良好的安全性,但OS与OS相比没有整体改善化疗。这是迄今为止在实体瘤中报道的最大的过继性T细胞治疗试验。
    BACKGROUND: Epstein-Barr virus-specific cytotoxic T lymphocyte (EBV-CTL) is an autologous adoptive T cell immunotherapy generated from the blood of individuals and manufactured without genetic modification. In a previous Phase 2 trial of locally recurrent or metastatic nasopharyngeal cancer (R/M NPC) patients, first-line gemcitabine and carboplatin (GC) and EBV-CTL combination demonstrated objective anti-tumor EBV-CTL activity and a favorable safety profile. The present study explored whether this combined first-line chemo-immunotherapy strategy would produce superior clinical efficacy and better quality of life compared to conventional chemotherapy treatment.
    METHODS: This multicenter, randomized, Phase 3 trial evaluated the efficacy and safety of GC followed by EBV-CTL vs. GC alone as first-line treatment for R/M NPC patients. Thirty clinical sites in Singapore, Malaysia, Taiwan, Thailand, and the United States (US) were included. Subjects were randomized to first-line GC (4 cycles) and EBV-CTL (6 cycles) or GC (6 cycles) in a 1:1 ratio. The primary outcome was overall survival (OS) and secondary outcomes included progression-free survival, objective response rate, clinical benefit rate, quality of life, and safety.
    RESULTS: gov identifier: NCT02578641.
    RESULTS: 330 subjects with NPC were enrolled. Most subjects in both treatment arms received ≥4 cycles of chemotherapy and most subjects in the GC+EBV-CTL group received ≥2 infusions of EBV-CTL. The central Good Manufacturing Practices (GMP) facility produced sufficient EBV-CTL for 94% of GC+EBV-CTL subjects. The median OS was 25.0 months in the GC+EBV-CTL group and 24.9 months in the GC group (hazard ratio = 1.19; 95% CI: 0.91, 1.56; P = 0.194). Only 1 subject experienced a Grade 2 serious adverse event related to EBV-CTL.
    CONCLUSIONS: GC+EBV-CTL in subjects with R/M NPC demonstrated a favorable safety profile but no overall improvement in OS vs. chemotherapy. This is the largest adoptive T cell therapy trial reported in solid tumors to date.
    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
    人工抗原呈递细胞(aAPC)的发展导致过继性T细胞疗法(ACT)的改善,免疫疗法,癌症治疗。APC有助于简化T细胞的一致生产和扩增,从而减少与ACT相关的时间和成本。然而,ACT仍然存在几个问题,例如T细胞效力不足,这降低了ACT的翻译潜力。虽然aAPC主要用于提高ACT的T细胞的生产效率,基于生物材料的aAPC的固有特性可能会影响T细胞的表型和功能.在CD8+T细胞中,活性氧(ROS)和氧化应激积累可以激活叉头盒蛋白O1(FOXO1)转录抗氧化剂,从而减少ROS并改善记忆形成。海藻酸盐,生物相容性和抗氧化剂丰富的生物材料,有望掺入aAPC制剂中以调节T细胞表型。为了调查它的效用,开发了一种新型的基于藻酸盐的aAPC平台,该平台优先扩增具有记忆相关特征的CD8+T细胞.基于藻酸盐的aAPC允许更好地控制CD8+T细胞质量,包括,显着改善体内持久性和增强体内抗肿瘤T细胞反应。
    The development of artificial Antigen Presenting Cells (aAPCs) has led to improvements in adoptive T cell therapy (ACT), an immunotherapy, for cancer treatment. aAPCs help to streamline the consistent production and expansion of T cells, thus reducing the time and costs associated with ACT. However, several issues still exist with ACT, such as insufficient T cell potency, which diminishes the translational potential for ACT. While aAPCs have been used primarily to increase production efficiency of T cells for ACT, the intrinsic properties of a biomaterial-based aAPC may affect T cell phenotype and function. In CD8+ T cells, reactive oxygen species (ROS) and oxidative stress accumulation can activate Forkhead box protein O1 (FOXO1) to transcribe antioxidants which reduce ROS and improve memory formation. Alginate, a biocompatible and antioxidant rich biomaterial, is promising for incorporation into an aAPC formulation to modulate T cell phenotype. To investigate its utility, a novel alginate-based aAPC platform was developed that preferentially expanded CD8+ T cells with memory related features. Alginate-based aAPCs allowed for greater control of CD8+ T cell qualities, including, significantly improved in vivo persistence and augmented in vivo anti-tumor T cell responses.
    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
    生物医学研究见证了制造嵌合抗原受体T细胞(CAR-T)疗法的重大进展,标志着细胞免疫疗法的变革时代。然而,现有的自体细胞疗法的制造方法仍然存在与成本相关的几个挑战,免疫细胞来源,安全风险,和可扩展性。这些挑战促使最近努力使用自动化封闭系统生物反应器和使用人工智能创建的模型来优化细胞疗法的工艺开发和制造。同时,非病毒基因转移方法,如mRNA,CRISPR基因组编辑,转座子正被用于改造T细胞和其他免疫细胞,如巨噬细胞和自然杀伤细胞。正在开发原始免疫细胞和干细胞的替代来源,以产生普遍的,同种异体疗法,信号从当前的自体范式转变。这些多方面的制造业创新强调了集体努力推动这种治疗方法朝着更广泛的临床采用和改善癌症治疗领域不断发展的患者结果。这里,我们回顾了当前的CAR免疫细胞制造策略,并强调了细胞疗法扩大规模的最新进展,自动化,过程开发,和工程。
    Biomedical research has witnessed significant strides in manufacturing chimeric antigen receptor T cell (CAR-T) therapies, marking a transformative era in cellular immunotherapy. Nevertheless, existing manufacturing methods for autologous cell therapies still pose several challenges related to cost, immune cell source, safety risks, and scalability. These challenges have motivated recent efforts to optimize process development and manufacturing for cell therapies using automated closed-system bioreactors and models created using artificial intelligence. Simultaneously, non-viral gene transfer methods like mRNA, CRISPR genome editing, and transposons are being applied to engineer T cells and other immune cells like macrophages and natural killer cells. Alternative sources of primary immune cells and stem cells are being developed to generate universal, allogeneic therapies, signaling a shift away from the current autologous paradigm. These multifaceted innovations in manufacturing underscore a collective effort to propel this therapeutic approach toward broader clinical adoption and improved patient outcomes in the evolving landscape of cancer treatment. Here, we review current CAR immune cell manufacturing strategies and highlight recent advancements in cell therapy scale-up, automation, process development, and engineering.
    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
    与初始T细胞相比,作为供体淋巴细胞输注的记忆T选择细胞(CD45RA-/RO)产生同种异体反应性和移植物抗宿主病(GvHD)的能力较低。本研究的目的是评估异基因造血细胞移植(HCT)后大剂量记忆(CD45RA-/RO)供体淋巴细胞输注(mDLI)的安全性和有效性。mDLI的适应症为“根据需要”和“作为预防方案”。“61名被诊断为恶性(82%)和非恶性疾病(18%)的儿童接受了241mDLIs。患者接受mDLI的中位输注3次(范围1-13),中位输注剂量为1.35×107/kgCD45RO,含8.96×106/kgCD3CD45RO和3.81×103/kgCD3CD45RA。在4%的mDLI输注后,7名患者发生了从头GvHD。在mDLI之前患有GvHD的患者中,在3例II-IV级急性GvHD患者中进行6次输注(11%)后,这种情况恶化。65%的mDLI输注后,巨细胞病毒载量降低。总队列的两年总生存率(OS)为64%(95%CI57%-72%)。对于接受预防性mDLI的患者,两年内非复发死亡率为10%(95%CI9%~11%).总之,大剂量mDLI是可行和安全的,即使在活动性GvHD患者中,严重GvHD的风险也相对较低。重要的是,mDLI与积极效应相关,包括增强对CMV病毒血症的控制。
    Memory T selected cells (CD45RA-/RO+) as donor lymphocyte infusion are less capable of producing alloreactivity and graft versus host disease (GvHD) compared with naïve T cells. The objective of this study was to evaluate the safety and efficacy of high-dose memory (CD45RA-/RO+) donor lymphocyte infusion (mDLI) after allogeneic hematopoietic cell transplantation (HCT). Indications for mDLI were \"as needed\" and \"as prophylactic regimen.\" Sixty-one children diagnosed with malignant (82%) and non-malignant diseases (18%) received 241 mDLIs. Patients received a median of three infusions (range 1‒13) of mDLI with a median infused dose of 1.35 × 107/kg CD45RO+ containing 8.96 × 106/kg CD3+CD45RO+ and 3.81 × 103/kg CD3+CD45RA+. De novo GvHD developed in 7 patients following 4% of the mDLI infusions. Among patients with GvHD before mDLI, this condition worsened following 6 infusions (11%) in the 3 patients with grade II-IV acute GvHD. A decrease in cytomegalovirus viral load followed 65% of mDLI infusions. Two-year overall survival (OS) for the total cohort was 64% (95% CI 57%‒72%). For patients receiving prophylactic mDLI, the two-year non-relapse mortality was 10% (95% CI 9%‒11%). In summary, high-dose mDLI is feasible and safe, with a relatively low risk of severe GvHD even in patients with active GvHD. Importantly, mDLI was associated with positive effects, including enhanced control of CMV viremia.
    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
    多发性骨髓瘤(MM)是一种浆细胞疾病,具有优先的骨髓(BM)嗜性。组织特异性趋化因子受体的强制表达已被证明可以成功地将过继转移的CARNK细胞引导到实体癌的恶性环境中。也包括BM居民AML和MM。为了重定向到BM相关的趋化因子CXCL12,我们用野生型CXCR4或功能获得突变体CXCR4R334X的异位表达对BCMACAR-NK-92以及原代NK细胞进行了装甲。我们的数据显示,配备有CXCR4的BCMCAR-NK-92和原代NK细胞在体外获得了向CXCL12迁移的改善的能力。除了协调趋化性的经典作用之外,CXCR4已被证明参与T细胞共刺激,这促使我们检查CXCR4共转导的BCMA-CARNK细胞的功能。异位CXCR4表达增强BCMACAR-NK细胞的细胞毒性能力,如通过体外消除表达BCMA的靶细胞系和原代MM细胞的能力以及通过加速的溶细胞颗粒释放所证明的。我们表明CXCR4共修饰延长了BCMACAR表面沉积,在汽车参与之后,增强了ZAP-70的招募,和加速远端信号转导动力学。BCMACAR对抗原的敏感性通过增强的ZAP-70募集到免疫突触而增强,揭示了在CXCR4过表达时CAR被触发的倾向增加。出乎意料的是,在不存在CXCL12配体刺激的情况下发生通过CXCR4的共刺激。总的来说,我们的研究结果暗示,CAR-NK细胞与组织相关趋化因子受体的共修饰对过继性NK细胞治疗的影响超出了在肿瘤部位内的转运和滞留改善.
    Multiple myeloma (MM) is a plasma cell disease with a preferential bone marrow (BM) tropism. Enforced expression of tissue-specific chemokine receptors has been shown to successfully guide adoptively-transferred CAR NK cells towards the malignant milieu in solid cancers, but also to BM-resident AML and MM. For redirection towards BM-associated chemokine CXCL12, we armored BCMA CAR-NK-92 as well as primary NK cells with ectopic expression of either wildtype CXCR4 or a gain-of-function mutant CXCR4R334X. Our data showed that BCMA CAR-NK-92 and -primary NK cells equipped with CXCR4 gained an improved ability to migrate towards CXCL12 in vitro. Beyond its classical role coordinating chemotaxis, CXCR4 has been shown to participate in T cell co-stimulation, which prompted us to examine the functionality of CXCR4-cotransduced BCMA-CAR NK cells. Ectopic CXCR4 expression enhanced the cytotoxic capacity of BCMA CAR-NK cells, as evidenced by the ability to eliminate BCMA-expressing target cell lines and primary MM cells in vitro and through accelerated cytolytic granule release. We show that CXCR4 co-modification prolonged BCMA CAR surface deposition, augmented ZAP-70 recruitment following CAR-engagement, and accelerated distal signal transduction kinetics. BCMA CAR sensitivity towards antigen was enhanced by virtue of an enhanced ZAP-70 recruitment to the immunological synapse, revealing an increased propensity of CARs to become triggered upon CXCR4 overexpression. Unexpectedly, co-stimulation via CXCR4 occurred in the absence of CXCL12 ligand-stimulation. Collectively, our findings imply that co-modification of CAR-NK cells with tissue-relevant chemokine receptors affect adoptive NK cell therapy beyond improved trafficking and retention within tumor sites.
    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
    过继性T细胞疗法(ACT)用于治疗实体瘤的功效仍然具有挑战性。除了受实体瘤周围物理屏障限制的效应T(Teff)细胞浸润不良外,另一个主要障碍是调节性T(Treg)细胞的广泛浸润,一个主要的免疫抑制免疫细胞亚群,在肿瘤微环境中。这里,我们开发了一种用于增强ACT的凹槽微针贴片,旨在同时克服身体和免疫抑制障碍。微针通过冰模板方法进行工程改造,以产生用于足够的T细胞负载的沟槽结构。此外,随着趋化因子CCL22的表面修饰,MNs不仅可以通过物理渗透直接将肿瘤特异性T细胞递送到实体瘤中,而且还通过细胞因子浓度梯度将Treg细胞从肿瘤微环境转移到微针表面,导致小鼠黑素瘤模型中Teff细胞/Treg细胞比例增加。因此,这种T细胞受体T细胞或嵌合抗原受体T细胞通过CCL22修饰的沟槽微针作为局部小生境的局部递送策略可以显著增强ACT的抗肿瘤功效并降低其在靶肿瘤外毒性.本文受版权保护。保留所有权利。
    The efficacy of adoptive T cell therapy (ACT) for the treatment of solid tumors remains challenging. In addition to the poor infiltration of effector T (Teff) cells limited by the physical barrier surrounding the solid tumor, another major obstacle is the extensive infiltration of regulatory T (Treg) cells, a major immunosuppressive immune cell subset, in the tumor microenvironment. Here, this work develops a grooved microneedle patch for augmenting ACT, aiming to simultaneously overcome physical and immunosuppressive barriers. The microneedles are engineered through an ice-templated method to generate the grooved structure for sufficient T-cell loading. In addition, with the surface modification of chemokine CCL22, the MNs could not only directly deliver tumor-specific T cells into solid tumors through physical penetration, but also specifically divert Treg cells from the tumor microenvironment to the surface of the microneedles via a cytokine concentration gradient, leading to an increase in the ratio of Teff cells/Treg cells in a mouse melanoma model. Consequently, this local delivery strategy of both T cell receptor T cells and chimeric antigen receptor T cells via the CCL22-modified grooved microneedles as a local niche could significantly enhance the antitumor efficacy and reduce the on-target off-tumor toxicity of ACT.
    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
    基于普鲁士蓝纳米粒子的光热疗法(PBNP-PTT)是能够引发抗肿瘤免疫应答的有效肿瘤治疗。受PBNP-PTT增强内源性免疫应答能力的激励,我们最近证明,PBNP-PTT可用于离体产生针对胶质母细胞瘤(GBM)细胞系的肿瘤特异性T细胞,作为过继性T细胞疗法(ATCT).在这项研究中,我们进一步开发了这个有前途的T细胞开发平台。首先,我们评估了使用PBNP-PTT产生的T细胞的表型和功能。我们观察到PBNP-PTT促进了CD8+T细胞从健康供体PBMC扩增,这些PBMC分泌IFNγ和TNFα,并上调了CD107a,以响应与靶U87细胞的参与。提示特异性抗肿瘤T细胞活化和脱颗粒。Further,与U87细胞共培养后,CD8+效应和效应记忆T细胞群体显着扩增,与肿瘤特异性效应反应一致。在体内原位植入的U87GBM肿瘤中,PBNP-PTT衍生的T细胞有效减少U87肿瘤生长,并在第100天时在>80%的荷瘤小鼠中产生长期存活,相比之下,用PBS处理的小鼠为0%,非特异性T细胞,或从裂解的U87细胞扩增的T细胞,证明了该ATCT平台的抗肿瘤功效增强。最后,我们通过产生靶向髓母细胞瘤的T细胞(D556)来测试我们方法的普遍性,乳腺癌(MDA-MB-231),神经母细胞瘤(SH-SY5Y),和急性单核细胞白血病(THP-1)细胞系。所产生的T细胞分泌IFNγ并发挥相对于对照增加的肿瘤特异性细胞溶解功能,证明PBNP-PTT在产生ATCT肿瘤特异性T细胞方面的多功能性。
    Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) is an effective tumor treatment capable of eliciting an antitumor immune response. Motivated by the ability of PBNP-PTT to potentiate endogenous immune responses, we recently demonstrated that PBNP-PTT could be used ex vivo to generate tumor-specific T cells against glioblastoma (GBM) cell lines as an adoptive T cell therapy (ATCT). In this study, we further developed this promising T cell development platform. First, we assessed the phenotype and function of T cells generated using PBNP-PTT. We observed that PBNP-PTT facilitated CD8+ T cell expansion from healthy donor PBMCs that secreted IFNγ and TNFα and upregulated CD107a in response to engagement with target U87 cells, suggesting specific antitumor T cell activation and degranulation. Further, CD8+ effector and effector memory T cell populations significantly expanded after co-culture with U87 cells, consistent with tumor-specific effector responses. In orthotopically implanted U87 GBM tumors in vivo, PBNP-PTT-derived T cells effectively reduced U87 tumor growth and generated long-term survival in >80% of tumor-bearing mice by Day 100, compared to 0% of mice treated with PBS, non-specific T cells, or T cells expanded from lysed U87 cells, demonstrating an enhanced antitumor efficacy of this ATCT platform. Finally, we tested the generalizability of our approach by generating T cells targeting medulloblastoma (D556), breast cancer (MDA-MB-231), neuroblastoma (SH-SY5Y), and acute monocytic leukemia (THP-1) cell lines. The resulting T cells secreted IFNγ and exerted increased tumor-specific cytolytic function relative to controls, demonstrating the versatility of PBNP-PTT in generating tumor-specific T cells for ATCT.
    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
    微流控技术在器官芯片技术和合成细胞研究中起着举足轻重的作用,特别是在人工细胞模型的开发和分析中。然而,使用合成细胞作为微流控系统的整体功能组件来塑造芯片上培养的天然活细胞的微环境的方法尚未被探索。这里,我们将基于胶体体的合成细胞集成到3D微流体设备中,开创了芯片上器官的基于细胞的合成微环境的概念。我们设计了一些方法来创建致密而稳定的二氧化硅胶体网络,被支持的脂质双层包裹,在微流体通道内。这些网络促进与芯片上培养的细胞的受体-配体相互作用。此外,我们引入了一种将生长因子从合成细胞控制释放到通道中的技术,使用基于海藻酸钙的水凝胶在胶体体内形成。为了证明这项技术的潜力,我们提出了一个模块化即插即用的淋巴结芯片原型,该原型通过刺激T细胞上的受体配体和调节其细胞因子环境来指导原代人T细胞的扩增.将合成细胞整合到微流体系统中,为芯片上器官技术提供了新的方向,并为潜在的治疗应用提供了进一步的探索途径。本文受版权保护。保留所有权利。
    Microfluidics plays a pivotal role in organ-on-chip technologies and in the study of synthetic cells, especially in the development and analysis of artificial cell models. However, approaches that use synthetic cells as integral functional components for microfluidic systems to shape the microenvironment of natural living cells cultured on-chip are not explored. Here, colloidosome-based synthetic cells are integrated into 3D microfluidic devices, pioneering the concept of synthetic cell-based microenvironments for organs-on-chip. Methods are devised to create dense and stable networks of silica colloidosomes, enveloped by supported lipid bilayers, within microfluidic channels. These networks promote receptor-ligand interactions with on-chip cultured cells. Furthermore, a technique is introduced for the controlled release of growth factors from the synthetic cells into the channels, using a calcium alginate-based hydrogel formation within the colloidosomes. To demonstrate the potential of the technology, a modular plug-and-play lymph-node-on-a-chip prototype that guides the expansion of primary human T cells by stimulating receptor ligands on the T cells and modulating their cytokine environment is presented. This integration of synthetic cells into microfluidic systems offers a new direction for organ-on-chip technologies and suggests further avenues for exploration in potential therapeutic applications.
    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
    多发性硬化是一种自身炎性病症,其导致受影响的患者中的有髓神经元的损伤。虽然疾病改善治疗已成功减缓复发缓解疾病的进展,大多数患者仍进展为继发性进行性疾病,对疾病改善治疗基本无反应.同样,目前对于原发性进行性MS患者尚无有效的治疗方法。CNS中的固有和适应性免疫细胞在启动自身免疫攻击和维持驱动疾病进展的慢性炎症中起关键作用。在这次审查中,我们将重点关注具有调节功能的T细胞在抑制MS进展中的作用,and,更重要的是,促进中枢神经系统MS病变的髓鞘再生和修复。我们将讨论基因重新编程调节性T细胞以实现免疫抑制并增强组织损伤部位局部修复的令人兴奋的潜力,同时保留中枢神经系统外完全胜任的免疫系统。在未来,具有确定的特异性和功能的重新编程的调节性T细胞可能提供生命药物,这些药物可以在患者体内持续存在,并在一个治疗周期后实现持久的疾病抑制。
    Multiple sclerosis is an autoinflammatory condition that results in damage to myelinated neurons in affected patients. While disease-modifying treatments have been successful in slowing the progression of relapsing-remitting disease, most patients still progress to secondary progressive disease that is largely unresponsive to disease-modifying treatments. Similarly, there is currently no effective treatment for patients with primary progressive MS. Innate and adaptive immune cells in the CNS play a critical role in initiating an autoimmune attack and in maintaining the chronic inflammation that drives disease progression. In this review, we will focus on recent insights into the role of T cells with regulatory function in suppressing the progression of MS, and, more importantly, in promoting the remyelination and repair of MS lesions in the CNS. We will discuss the exciting potential to genetically reprogram regulatory T cells to achieve immune suppression and enhance repair locally at sites of tissue damage, while retaining a fully competent immune system outside the CNS. In the future, reprogramed regulatory T cells with defined specificity and function may provide life medicines that can persist in patients and achieve lasting disease suppression after one cycle of treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号