关键词: Mesenchymal stem cells Non-viral gene modification Peritoneal carcinomatosis

Mesh : Animals Mesenchymal Stem Cells / metabolism cytology Peritoneal Neoplasms / therapy secondary genetics pathology Humans Mesenchymal Stem Cell Transplantation Pentosyltransferases / genetics metabolism Transgenes Cell Line, Tumor Interferon-beta / metabolism genetics Xenograft Model Antitumor Assays Cytosine Deaminase / genetics metabolism Mice Female

来  源:   DOI:10.1186/s12967-024-05278-5   PDF(Pubmed)

Abstract:
BACKGROUND: Mesenchymal stem cells (MSCs) have garnered significant interest for their tumor-tropic property, making them potential therapeutic delivery vehicles for cancer treatment. We have previously shown the significant anti-tumour activity in mice preclinical models and companion animals with naturally occurring cancers using non-virally engineered MSCs with a therapeutic transgene encoding cytosine deaminase and uracil phosphoribosyl transferase (CDUPRT) and green fluorescent protein (GFP). Clinical studies have shown improved response rate with combinatorial treatment of 5-fluorouracil and Interferon-beta (IFNb) in peritoneal carcinomatosis (PC). However, high systemic toxicities have limited the clinical use of such a regime.
METHODS: In this study, we evaluated the feasibility of intraperitoneal administration of non-virally engineered MSCs to co-deliver CDUPRT/5-Flucytosine prodrug system and IFNb to potentially enhance the cGAS-STING signalling axis. Here, MSCs were engineered to express CDUPRT or CDUPRT-IFNb. Expression of CDUPRT and IFNb was confirmed by flow cytometry and ELISA, respectively. The anti-cancer efficacy of the engineered MSCs was evaluated in both in vitro and in vivo model. ES2, HT-29 and Colo-205 were cocultured with engineered MSCs at various ratio. The cell viability with or without 5-flucytosine was measured with MTS assay. To further compare the anti-cancer efficacy of the engineered MSCs, peritoneal carcinomatosis mouse model was established by intraperitoneal injection of luciferase expressing ES2 stable cells. The tumour burden was measured through bioluminescence tracking.
RESULTS: Firstly, there was no changes in phenotypes of MSCs despite high expression of the transgene encoding CDUPRT and IFNb (CDUPRT-IFNb). Transwell migration assays and in-vivo tracking suggested the co-expression of multiple transgenes did not impact migratory capability of the MSCs. The superiority of CDUPRT-IFNb over CDUPRT expressing MSCs was demonstrated in ES2, HT-29 and Colo-205 in-vitro. Similar observations were observed in an intraperitoneal ES2 ovarian cancer xenograft model. The growth of tumor mass was inhibited by ~ 90% and 46% in the mice treated with MSCs expressing CDUPRT-IFNb or CDUPRT, respectively.
CONCLUSIONS: Taken together, these results established the effectiveness of MSCs co-expressing CDUPRT and IFNb in controlling and targeting PC growth. This study lay the foundation for the development of clinical trial using multigene-armed MSCs for PC.
摘要:
背景:间充质干细胞(MSCs)因其亲肿瘤特性而引起了极大的兴趣,使它们成为癌症治疗的潜在治疗载体。我们先前已经使用具有编码胞嘧啶脱氨酶和尿嘧啶磷酸核糖转移酶(CDUPRT)和绿色荧光蛋白(GFP)的治疗性转基因的非病毒工程化MSC在患有天然存在的癌症的小鼠临床前模型和伴侣动物中显示出显著的抗肿瘤活性。临床研究表明,在腹膜癌(PC)中,5-氟尿嘧啶和干扰素-β(IFNb)的组合治疗可改善反应率。然而,高的全身毒性限制了这种药物的临床应用。
方法:在本研究中,我们评估了腹膜内给予非病毒工程化MSCs共同递送CDUPRT/5-氟胞嘧啶前药系统和IFNb以潜在增强cGAS-STING信号轴的可行性.这里,将MSC工程化以表达CDUPRT或CDUPRT-IFNb。流式细胞术和ELISA证实CDUPRT和IFNb的表达,分别。在体外和体内模型中评估工程化MSC的抗癌功效。将ES2、HT-29和Colo-205与工程化MSC以各种比例共培养。用MTS测定法测量有或没有5-氟胞嘧啶的细胞活力。为了进一步比较工程MSCs的抗癌功效,通过腹腔注射表达荧光素酶的ES2稳定细胞建立腹膜癌变小鼠模型。通过生物发光跟踪测量肿瘤负荷。
结果:首先,尽管编码CDUPRT和IFNb(CDUPRT-IFNb)的转基因高表达,但MSCs的表型没有变化。Transwell迁移测定和体内跟踪表明,多种转基因的共表达不会影响MSC的迁移能力。在体外ES2、HT-29和Colo-205中证明了CDUPRT-IFNb优于表达CDUPRT的MSC。在腹膜内ES2卵巢癌异种移植模型中观察到类似的观察结果。在用表达CDUPRT-IFNb或CDUPRT的MSCs治疗的小鼠中,肿瘤块的生长被抑制约90%和46%,分别。
结论:综合来看,这些结果确立了共表达CDUPRT和IFNb的MSC在控制和靶向PC生长中的有效性。本研究为开发使用多基因武装的MSCs进行PC的临床试验奠定了基础。
公众号