首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 771 毫秒
1.
自然杀伤(natural killer, NK)细胞是固有免疫的重要组成部分,可通过表面抑制性受体与激活性受体的协同作用活化后,直接识别杀伤肿瘤细胞;可分泌细胞因子募集树突状细胞(dendritic cells, DCs),促进DCs成熟,增强适应性免疫应答;可杀伤肿瘤干细胞(cancer stem cells, CSCs)与循环肿瘤细胞(circulating tumor cells, CTCs),维持肿瘤细胞休眠,抑制肿瘤转移。NK细胞具备独特的炎症趋向性,可响应肿瘤部位释放的细胞因子与趋化因子迁徙至肿瘤部位,使其在抗肿瘤靶向治疗中占据重要优势。因此, NK细胞载体、NK细胞膜包被仿生化载体和NK细胞外囊泡(NK cell extracellular vesicles, NKEVs)的肿瘤靶向治疗研究,受到越来越多的关注。本文将重点介绍NK细胞抗肿瘤作用机制及其在肿瘤靶向治疗中的应用研究进展。  相似文献   

2.
T/B细胞相互作用是引起机体适应性免疫应答的关键,其中CD40和CD40L的协同作用不仅对体液免疫和细胞免疫应答具有重要的调节作用,而且在抗感染、抗病毒和抗肿瘤免疫应答及炎症反应中也起重要作用.CD40主要表达于B细胞、抗原提呈细胞和一些肿瘤细胞表面.因此,CD40分子介导的信号通路与大多数恶性肿瘤、自身免疫疾病、免疫缺陷病有关.以CD40为靶标进行肿瘤免疫治疗已成为免疫研究热点之一.此文主要阐述了CD40的生物学特征和功能、CD40相关恶性肿瘤以及治疗性抗CD40单克隆抗体在肿瘤治疗中的应用.  相似文献   

3.
抗肿瘤免疫以细胞免疫为主,共刺激分子发挥了相当重要的作用。B7-CD28是激发机体抗肿瘤免疫应答的共刺激信号,由于肿瘤细胞缺乏B7分子的表达,机体抗肿瘤作用显著降低。转基因治疗后,可以使机体重新获得对肿瘤的主动免疫应答能力。  相似文献   

4.
肿瘤是严重威胁人类生命的一种疾病,大多肿瘤均可表达肿瘤相关抗原(tumor-associated antigens,TAA).肿瘤免疫以细胞免疫为主,抗原呈递细胞(APC)可以捕获TAA,经加工处理后将抗原信号呈递给细胞毒性T细胞,从而引发一系列免疫应答,这一过程是诱导细胞免疫的关键所在.树突状细胞(dendritic cells,DC)是目前已知的功能最强的抗原呈递细胞,体内DC广泛分布,但数量很少,仅占外周血单核细胞(PBMC)的1%以下,是唯一能够呈递蛋白质抗原,并致敏初始记忆型T细胞的APC.肿瘤患者大多表现为局部DC功能低下,数目减少,容易产生免疫耐受,是肿瘤细胞逃脱免疫防御的众多原因之一.机体识别并捕获单一的肿瘤抗原无法诱导出有效的抗肿瘤反应,只有通过免疫组化方法将DC与肿瘤细胞抗原融合后,才能有效抑制免疫耐受,诱导机体对肿瘤细胞的免疫应答,因此DC的深入研究对肿瘤免疫治疗有着重要意义.  相似文献   

5.
主要组织相容性复合体Ⅰ相关分子A(MICA)在多种肿瘤细胞中如乳腺癌、肺癌、胃肠道肿瘤、前列腺癌等中均有表达,被认为与恶性转化相关。但在这些肿瘤中NKG2D配体信号并不能有效诱导抗肿瘤的免疫应答。肿瘤细胞表面表达MICA分子可能有助于机体自然杀伤(NK)细胞与其结合并产生杀瘤活性。  相似文献   

6.
<正>现阶段,肿瘤治疗越来越趋于综合化,方法包括手术、化疗、放疗。在临床中,肿瘤患者死亡的原因主要为复发、远处转移。在肿瘤治疗中,单纯化疗具有一定的疗效,但并不理想。而免疫治疗作为一种新兴治疗方式,能够使机体免疫应答被激活,从而有效识别肿瘤细胞,达到灭杀肿瘤细胞的目的。在肿瘤治疗中联合应用放疗与免疫治疗,可取得良好的效果,成为了临床研究的重要课题。1免疫系统作用免疫系统可促进机体抗肿瘤,同时诱导肿瘤细  相似文献   

7.
张华竹  刘晓波 《药学研究》2021,40(1):40-43,59
细胞程序性死亡因子1(programmed death-1,PD-1)及其配体1(programmed death-ligand 1,PD-L1)是一对共刺激分子,激活细胞程序性死亡因子1及其配体1信号通路可抑制肿瘤特异性T细胞活性,有助于肿瘤细胞逃避免疫监视。而阻断该通路可以激活机体抗肿瘤免疫应答,抑制肿瘤细胞的生长。近年来,针对细胞程序性死亡因子1及其配体1通路的免疫疗法在黑色素瘤的治疗中已取得显著效果。本文对细胞程序性死亡因子1及其配体1通路在黑色素瘤中的表达调控、细胞程序性死亡因子1及其配体1抑制剂在黑色素瘤治疗中的临床研究进行综述,讨论细胞程序性死亡因子1作为一种新的免疫疗法在临床的应用前景和不良反应。  相似文献   

8.
<正>目前,在恶性肿瘤的治疗中,生物治疗已与手术、放疗、化疗并列为恶性肿瘤综合治疗的四大方式。DC-CIK细胞肿瘤生物治疗技术是一种通过体外分离单个核细胞并分别培养,从而诱导成熟的DC细胞和CIK细胞并将两者联合起来用于治疗肿瘤的技术[1]。DC-CIK细胞不仅具有杀死肿瘤细胞、激发增强肿瘤患者的特异性肿瘤免疫应答,诱发免疫记忆获得长期抗肿瘤预防肿瘤复发的作  相似文献   

9.
树突状细胞(DC)是最强的抗原递呈细胞,可以激活幼稚T细胞,打破外周免疫耐受,进而诱导肿瘤免疫应答。对于复发或者进展性前列腺癌患者,以DC为基础的免疫治疗可延长患者生存期。本文综述DC疫苗临床治疗研究进展。  相似文献   

10.
程樱  张立 《中国药业》2013,22(17):103-104
目的 介绍中药抗肿瘤机制及药物研究进展.方法 参阅近年来文献资料,进行归纳总结.结果 中药抗肿瘤机制主要包括直接抑制肿瘤细胞生长与增殖、诱导肿瘤细胞凋亡、诱导肿瘤细胞分化、抑制肿瘤血管生长、增强机体免疫力等.结论 开发中药抗肿瘤药物对于抢救肿瘤患者的生命具有重要意义.  相似文献   

11.
Introduction: Immunogenic cell death inducers (ICD inducers) are a diverse group of therapeutic molecules capable of eliciting an adaptive immune response against the antigens present on the surface of dying cancer cells. Most of these molecules suffer from low bioavailability, high toxicity and poor pharmacokinetics which limit their application. It is believed that nanotechnology, in particular nano-sized nanocarriers, can address most of the issues that limit the use of ICD inducers.

Area covered: The mechanism of action of ICD inducers and their limitations is discussed. In addition, we cover the novel possibilities arising from the use of nanotechnology to improve delivery of ICD inducers to the target tissue as well as the restrictions of modern nanotechnology.

Expert opinion: At present, nanocarrier formulations suffer from low bioavailability, poor pharmacokinetics and stability issues. Nonetheless, there is a tremendous future for combinatorial immune-pharmacological treatments of human tumors based on nanocarrier delivery of ICD inducers.  相似文献   


12.
《药学学报(英文版)》2022,12(6):2740-2750
Immunogenic cell death (ICD) plays a major role in cancer immunotherapy by stimulating specific T cell responses and restoring the antitumor immune system. However, effective type II ICD inducers without biotoxicity are still very limited. Herein, a tentative drug- or photosensitizer-free strategy was developed by employing enzymatic self-assembly of the peptide F-pY-T to induce mitochondrial oxidative stress in cancer cells. Upon dephosphorylation catalyzed by alkaline phosphatase overexpressed on cancer cells, the peptide F-pY-T self-assembled to form nanoparticles, which were subsequently internalized. These affected the morphology of mitochondria and induced serious reactive oxygen species production, causing the ICD characterized by the release of danger-associated molecular patterns (DAMPs). DAMPs enhanced specific immune responses by promoting the maturation of DCs and the intratumoral infiltration of tumor-specific T cells to eradicate tumor cells. The dramatic immunotherapeutic capacity could be enhanced further by combination therapy of F-pY-T and anti-PD-L1 agents without visible biotoxicity in the main organs. Thus, our results revealed an alternative strategy to induce efficient ICD by physically promoting mitochondrial oxidative stress.  相似文献   

13.
《药学学报(英文版)》2022,12(9):3694-3709
Amplifying “eat me signal” during tumor immunogenic cell death (ICD) cascade is crucial for tumor immunotherapy. Inspired by the indispensable role of adenosine triphosphate (ATP, a necessary “eat me signal” for ICD), a versatile ICD amplifier was developed for chemotherapy-sensitized immunotherapy. Doxorubicin (DOX), ATP and ferrous ions (Fe2+) were co-assembled into nanosized amplifier (ADO-Fe) through π?π stacking and coordination effect. Meanwhile, phenylboric acid-polyethylene glycol-phenylboric acid (PBA-PEG-PBA) was modified on the surface of ADO-Fe (denoted as PADO-Fe) by the virtue of d-ribose unit of ATP. PADO-Fe could display active targetability against tumor cells via sialic acid/PBA interaction. In acidic microenvironment, PBA-PEG-PBA would dissociate from amplifier. Moreover, high H2O2 concentration would induce hydroxyl radical (·OH) and oxygen (O2) generation through Fenton reaction by Fe2+. DOX and ATP would be released from the amplifier, which could induce ICD effect and “ICD adjuvant” to amplify this process. Together with programmed death ligands 1 (PD-L1) checkpoint blockade immunotherapy, PADO-Fe could not only activate immune response against primary tumor, but also strong abscopal effect against distant tumor. Our simple and multifunctional ICD amplifier opens a new window for enhancing ICD effect and immune checkpoint blockade therapy.  相似文献   

14.
Photothermal therapy (PTT) has brought hope for cancer treatments, with hyperthermia-induced immunogenic cell death (ICD), which is a critical part of therapeutically induced antitumor immune responses. Limited immune stimulation response in PTT is the primary reason for incomplete tumor ablation, therefore demonstrating urgent requirements for ICD amplifier. Herein, a sub-10 nm supramolecular nanoassembly was formed by co-assembly of clinically approved aluminum adjuvant and commonly used indocyanine green (ICG) under the assistance of lignosulfonate (LS, a green and sustainable multifunctional lignin derivative) for localized photothermal-immunotherapy of breast cancer. The overall results revealed that LS-Al-ICG is capable of inducing amplified ICD, efficiently eliciting solid immune responses through dendritic cells (DCs) activation and cytotoxic T-cell responses initiation for tumor killing. Moreover, anti-PD-1 therapy blocked the PD-1 pathway and led to remarkable anti-tumor efficacy against laser-irradiated primary tumors and distant tumors by potentiating systemic tumor specific T cell immunity. The results of this study demonstrate a handy and extensible approach for engineering green natural lignin nanoparticles for cancer immunotherapy, which shows promise for delivering other therapeutics in biomedical applications.  相似文献   

15.
Immune checkpoint blockade therapy has become a first-line treatment in various cancers. But there are only a small percent of colorectal patients responding to PD-1/PD-L1 blockage immunotherapy. How to increase their treatment efficacy is an urgent and clinically unmet need. It is acknowledged that immunogenic cell death (ICD) induced by some specific chemotherapy can enhance antitumor immunity. Chemo-based combination therapy can yield improved outcomes by activating the immune system to eliminate the tumor, compared with monotherapy. Here, we develop a PD-L1-targeting immune liposome (P-Lipo) for co-delivering irinotecan (IRI) and JQ1, and this system can successfully elicit antitumor immunity in colorectal cancer through inducing ICD by IRI and interfering in the immunosuppressive PD-1/PD-L1 pathway by JQ1. P-Lipo increases intratumoral drug accumulation and promotes DC maturation, and thereby facilitates adaptive immune responses against tumor growth. The remodeling tumor immune microenvironment was reflected by the increased amount of CD8+ T cells and the release of IFN-γ, and the reduced CD4+Foxp3+ regulatory T cells (Tregs). Collectively, the P-Lipo codelivery system provides a chemo-immunotherapy strategy that can effectively remodel the tumor immune microenvironment and activate the host immune system and arrest tumor growth.  相似文献   

16.
《药学学报(英文版)》2022,12(1):107-134
The immune system is involved in the initiation and progression of cancer. Research on cancer and immunity has contributed to the development of several clinically successful immunotherapies. These immunotherapies often act on a single step of the cancer–immunity cycle. In recent years, the discovery of new nanomaterials has dramatically expanded the functions and potential applications of nanomaterials. In addition to acting as drug-delivery platforms, some nanomaterials can induce the immunogenic cell death (ICD) of cancer cells or regulate the profile and strength of the immune response as immunomodulators. Based on their versatility, nanomaterials may serve as an integrated platform for multiple drugs or therapeutic strategies, simultaneously targeting several steps of the cancer–immunity cycle to enhance the outcome of anticancer immune response. To illustrate the critical roles of nanomaterials in cancer immunotherapies based on cancer–immunity cycle, this review will comprehensively describe the crosstalk between the immune system and cancer, and the current applications of nanomaterials, including drug carriers, ICD inducers, and immunomodulators. Moreover, this review will provide a detailed discussion of the knowledge regarding developing combinational cancer immunotherapies based on the cancer–immunity cycle, hoping to maximize the efficacy of these treatments assisted by nanomaterials.  相似文献   

17.
Cancer immunotherapy, which suppresses cancer progression by activating the anti-cancer immunity of patients, shows utility in treating multiple types of cancers. Immunogenic cell death (ICD) induced by most clinical treatment modalities plays a critical role in promoting cancer immunotherapy by releasing tumor-associated antigens and neoantigens and exposing “danger signals” to stimulate immune cells. This comment article presents the different roles of nanoparticles in various treatment modalities of cancers, including chemotherapy, radiotherapy, photodynamic and photothermal therapies, and therapy with radiated tumor cell-released nanoparticles, which often activate anti-cancer immunological effects by inducing ICD of cancer cells, and highlights the challenges and opportunities of ICD-related cancer immunotherapy in the clinic.  相似文献   

18.
Immunogenic cell death (ICD) plays an important role in sensitizing tumor cells to antigen-presenting cells followed by antitumor immunity. However, a successful antitumor response by ICD requires both apoptotic tumor microenvironments and activated immune systems. Ivermectin (IVM) has been shown to induce cell apoptosis through autophagy which can be a great candidate for ICD therapy. However, a single treatment of IVM-free drug is not an ideal anticancer therapy due to its anti-inflammatory effects and cytotoxicity. In the present study, IVM was shown to enhance the ICD process in addition to the treatment of resiquimod (R848), a TLR7/8 agonist, when co-loaded in a squalene-based nanoemulsion (NE). R848-IVM co-loaded NE was developed and characterized in vitro. Antitumor activity of R848-IVM NE was also evaluated in vitro and in vivo. R848-IVM NE exhibited long-term stability and reduced cytotoxicity by IVM. In vivo studies demonstrated that IVM significantly augments the ICD by upregulating Cd8a and releasing HMGB1 in tumor tissue, which could enhance R848-driven antitumor immunity. R848-IVM NE treatment showed strong antitumor activity with over 80% tumor growth inhibition, suggesting a potential combination therapy of systemic co-delivering IVM with TLR agonists against solid cancer.  相似文献   

19.
Cancer immunotherapy is a strategy that is moving to the frontier of cancer treatment in the current decade. In this study, we show evidence that 3-(2-nitrophenyl) propionic acid-paclitaxel nanoparticles (NPPA-PTX NPs), act as immunogenic cell death (ICD) inducers, stimulating an antitumor response which results in synergistic antitumor activity by combining anti-PD-L1 antibody (aPD-L1) in vivo. To investigate the antitumor immunity induced by NPPA-PTX NPs, the expression of both ICD marker calreticulin (CRT) and high mobility group box 1 (HMGB1) were analyzed. In addition, the antitumor activity of NPPA-PTX NPs combined with aPD-L1 in vivo was also investigated. The immune response was also measured through quantitation of the infiltration of T cells and the secretion of pro-inflammatory cytokines. The results demonstrate that NPPA-PTX NPs induce ICD of MDA-MB-231 and 4T1 cells through upregulation of CRT and HMGB1, reactivating the antitumor immunity via recruitment of infiltrating CD3+, CD4+, CD8+ T cells, secreting IFN-γ, TNF-α, and the enhanced antitumor activity by combining with aPD-L1. These data suggest that the combined therapy has a synergistic antitumor activity and has the potential to be developed into a novel therapeutic regimen for cancer patients.  相似文献   

20.
In addition to maintaining cell homeostasis under physiological and stress conditions, some heat shock proteins (HSPs) are potent inducers of immunity and have been harnessed as vaccine adjuvants targeted to cancers and infections. HSPs are a group of ubiquitous intracellular molecules that function as molecular chaperones in numerous processes, such as protein folding and transport, and are induced under stress conditions, such as fever and radiation. Certain HSPs are potent inducers of innate and antigen-specific immunity. They activate dendritic cells partly through toll-like receptors, activate natural killer cells, increase presentation of antigens to effector cells and augment T-cell and humoral immune responses against their associated antigens. Their roles in priming multiple host defense pathways are being exploited in vaccine development for cancer and infectious diseases.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号