Abscopal effect

腹腔镜效应
  • 文章类型: Case Reports
    我们介绍了一个64岁的转移性肾细胞癌患者的病例,尽管接受了多种系统治疗,但仍经历了疾病进展,包括免疫检查点抑制剂(ICI)。在对他的脑部病变进行立体定向放射外科手术两个月后,当患者没有接受任何全身治疗时,重新扫描显示,原发性肾脏病变和转移部位几乎完全消退,这归因于横观效应,由暴露于ICI和放射疗法介导。虽然其机制尚未完全了解,它被认为源于辐射诱导的肿瘤免疫抑制和免疫原性。
    We present the case of a 64-year-old patient with metastatic renal cell carcinoma, who experienced disease progression despite undergoing multiple lines of systemic therapy, including immune checkpoint inhibitors (ICI). Two months after stereotactic radiosurgery to his brain lesions and while the patient was not on any systemic therapy, restaging scans demonstrated a dramatic near complete regression of the primary renal lesion and metastatic sites, which was attributed to the abscopal effect, mediated by the exposure to ICI and radiotherapy. While its mechanisms are not fully understood, it is believed to stem from the tumor immunosuppression and immunogenicity induced by radiation.
    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
    放射治疗(RT)控制局部病变,同时它有能力诱导全身反应以抑制远处,转移性,非放射性肿瘤,这被称为“离谱效应”。人们普遍认为放疗可以激发全身免疫反应。这为免疫治疗联合放疗(iRT)提供了令人信服的理论依据。的确,在临床治疗中也观察到这种现象,为患者带来显著的临床益处,一系列基础研究正在进行中,以扩大这种影响。然而,RT诱导免疫应答的分子机制,确定iRT的最佳治疗方案,以及如何放大横观效应。为了在临床管理中放大和利用这种效应,这些关键问题需要得到很好的解决;在这次审查中,我们全面总结了不断增长的共识,并强调了用放疗或免疫疗法增强腹腔镜疗效的新出现的局限性。最后,我们讨论了当前临床转化应用的前景和障碍。
    Radiotherapy (RT) controls local lesions, meantime it has the capability to induce systemic response to inhibit distant, metastatic, non-radiated tumors, which is referred to as the \"abscopal effect\". It is widely recognized that radiotherapy can stimulate systemic immune response. This provides a compelling theoretical basis for the combination of immune therapy combined with radiotherapy(iRT). Indeed, this phenomenon has also been observed in clinical treatment, bringing significant clinical benefits to patients, and a series of basic studies are underway to amplify this effect. However, the molecular mechanisms of immune response induced by RT, determination of the optimal treatment regimen for iRT, and how to amplify the abscopal effect. In order to amplify and utilize this effect in clinical management, these key issues require to be well addressed; In this review, we comprehensively summarize the growing consensus and emphasize the emerging limitations of enhancing the abscopal effect with radiotherapy or immunotherapy. Finally, we discuss the prospects and barriers to the current clinical translational 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
    放射生物学已从基于DNA损伤和响应因子的机械模型发展成为更复杂的模型,其中包括对免疫系统和肿瘤微环境(TME)的影响。照射具有免疫调节作用,其可以表现为增加的抗肿瘤免疫或免疫抑制。照射通过释放促炎细胞因子和内皮损伤促进炎症微环境,将免疫系统细胞招募到受照射的区域。辐射诱导的免疫原性细胞死亡(ICD),以损伤相关分子模式(DAMPs)和肿瘤抗原的释放为特征,引发先天免疫和适应性免疫的抗肿瘤免疫反应。抗肿瘤免疫可以在远离照射区域的地方表现出来,一种被称为横观效应(AE)的现象,涉及树突状细胞和CD8+T细胞。照射还产生由肿瘤相关巨噬细胞(TAMs)和调节性T淋巴细胞(Tregs)介导的免疫抑制作用,抵消免疫刺激作用。在这项工作中,我们回顾了辐射诱导的免疫反应的机制,支持RT和免疫治疗的联合治疗,聚焦,在可能的情况下,关于妇科癌症。
    Radiobiology has evolved from a mechanistic model based on DNA damage and response factors into a more complex model that includes effects on the immune system and the tumor microenvironment (TME). Irradiation has an immunomodulatory effect that can manifest as increased anti-tumor immunity or immunosuppression. Irradiation promotes an inflammatory microenvironment through the release of pro-inflammatory cytokines and endothelial damage, which recruit immune system cells to the irradiated area. Radiation-induced immunogenic cell death (ICD), characterized by the release of damage-associated molecular patterns (DAMPs) and tumor antigens, triggers an anti-tumor immune response of both innate and adaptive immunity. Anti-tumor immunity can manifest at a distance from the irradiated area, a phenomenon known as the abscopal effect (AE), which involves dendritic cells and CD8+ T cells. Irradiation also produces an immunosuppressive effect mediated by tumor-associated macrophages (TAMs) and regulatory T lymphocytes (Tregs), which counterbalances the immunostimulatory effect. In this work, we review the mechanisms involved in the radiation-induced immune response, which support the combined treatment of RT and immunotherapy, focusing, where possible, on gynecologic cancer.
    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
    贝伐单抗与阿替珠单抗的组合通过激活免疫应答增强治疗的抗肿瘤效果。该组合被批准用于治疗不可切除的肝细胞癌(HCC)。远视效应与由辐射诱导的免疫原性细胞死亡引发的免疫反应有关,基于实验模型。因此,联合放疗和免疫疗法有望诱导外科学效应。然而,由于临床病例很少,免疫治疗在腹腔镜作用中的临床意义仍然未知。在这里,我们报告一例晚期肝癌合并肺和肾上腺转移。立体定向放疗(SBRT)后,阿妥珠单抗和贝伐单抗(atezo/bev)的抗肿瘤疗效增强,尽管在SBRT之前,atezo/bev没有产生足够的治疗反应。此外,在单用阿特珠单抗期间未观察到SBRT后的脓肿效应,但在恢复贝伐单抗与阿特珠单抗组合后引起,最终患者达到完全反应状态。这些发现表明,放疗后的免疫激活可能与临床实践和实验环境中的远视效应的诱导有关。在HCC的病例中,放疗后联合免疫疗法与贝伐单抗可以引起异常效应,即使单独使用免疫检查点抑制剂可能不足。
    Combining bevacizumab with atezolizumab enhances the antitumor effects of the treatment by activating an immune response. This combination is approved for the treatment of unresectable hepatocellular carcinoma (HCC). An abscopal effect is associated with an immune response triggered by radiation-induced immunogenic cell death, based on experimental models. Thus, combining radiotherapy and immunotherapy is expected to induce an abscopal effect. However, the clinical significance of immunotherapy in the abscopal effect remains unknown due to the rarity of clinical cases. Herein, we report a case of advanced HCC with lung and adrenal metastases. The antitumor efficacy of atezolizumab and bevacizumab (atezo/bev) was enhanced following stereotactic body radiotherapy (SBRT), although atezo/bev did not yield a sufficient therapeutic response pre-SBRT. Furthermore, an abscopal effect following SBRT was not observed during atezolizumab alone but was evoked after resuming bevacizumab in combination with atezolizumab, culminating in the patient achieving a complete response status. These findings suggest that immune activation following radiotherapy may be related to the induction of an abscopal effect in clinical practice as well as in experimental settings, and combining immunotherapy with bevacizumab post-radiotherapy could evoke an abscopal effect in a case of HCC, even though immune checkpoint inhibitor use alone may be insufficient.
    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
    非小细胞肺癌(NSCLC)的治疗指南因病理分期等多种因素而异。患者候选人资格,和治疗目标。NSCLC临床医生工具包中有许多治疗方法和更多组合,与治疗优化相比,许多问题仍然没有答案。虽然一些研究已经开始探索NSCLC治疗-手术切除的许多支柱之间的相互作用,放射治疗,化疗,和免疫疗法-除了每种成分疗法的调节之外,不同疗法形式的大量组合和排列在原本快速发展的领域中留下了很多期望。鉴于NSCLC的高发病率和致死率,联合治疗可能赋予的协同益处的实验提供了一个成熟的发展目标和增加的理解,而没有新药开发的成本和负担。这篇综述介绍了,合成,并比较了突出的NSCLC疗法,强调治疗类型之间的相互作用以及一些组合疗法在过去几年中已经证明的协同益处。
    Treatment guidelines for non-small cell lung cancer (NSCLC) vary by several factors including pathological stage, patient candidacy, and goal of treatment. With many therapeutics and even more combinations available in the NSCLC clinician\'s toolkit, a multitude of questions remain unanswered vis-a-vis treatment optimization. While some studies have begun exploring the interplay among the many pillars of NSCLC treatment-surgical resection, radiotherapy, chemotherapy, and immunotherapy-the vast number of combinations and permutations of different therapy modalities in addition to the modulation of each constituent therapy leaves much to be desired in a field that is otherwise rapidly evolving. Given NSCLC\'s high incidence and lethality, the experimentation of synergistic benefits that combinatorial treatment may confer presents a ripe target for advancement and increased understanding without the cost and burden of novel drug development. This review introduces, synthesizes, and compares prominent NSCLC therapies, placing emphasis on the interplay among types of therapies and the synergistic benefits some combinatorial therapies have demonstrated over the past several years.
    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
    免疫检查点抑制剂(ICIs)对许多晚期恶性肿瘤有效。然而,许多患者对免疫疗法没有反应,克服这种对治疗的抵抗力很重要。硼中子俘获疗法(BNCT)是一种局部放化疗疗法,结合了在癌症中选择性积累的硼药物和癌症部位的中子辐照。这里,我们报道了第一个硼中子免疫疗法(B-NIT),结合BNCT和ICI免疫疗法,这是在放射性抗性和免疫疗法抗性晚期B16F10黑色素瘤小鼠模型上进行的。BNCT组显示局部肿瘤抑制,但抗PD-1抗体免疫治疗组未显示肿瘤抑制。只有B-NIT组在BNCT治疗和屏蔽的远处部位均显示出强的肿瘤生长抑制作用。B-NIT组瘤内CD8+T细胞浸润和血清高迁移率组1(HMGB1)水平较高。对肿瘤浸润淋巴细胞(TIL)中CD8+T细胞的分析显示,在B-NIT组中,CD62L-CD44+效应记忆T细胞和CD69+早期活化T细胞主要增加。向B-NIT组施用CD8消耗mAb完全抑制了增强的治疗效果。这表明B-NIT具有有效的免疫诱导的外视效应,用BNCT直接破坏肿瘤,诱导抗原扩散效应,保护正常组织.B-NIT,免疫疗法联合BNCT,是克服恶性黑色素瘤免疫疗法耐药性的第一种治疗方法。在未来,因为它的治疗功效不仅在黑色素瘤中得到证实,而且在其他免疫疗法抗性恶性肿瘤中也得到证实,B-NIT可成为治疗晚期癌症的新候选药物。
    Immune checkpoint inhibitors (ICIs) are effective against many advanced malignancies. However, many patients are nonresponders to immunotherapy, and overcoming this resistance to treatment is important. Boron neutron capture therapy (BNCT) is a local chemoradiation therapy with the combination of boron drugs that accumulate selectively in cancer and the neutron irradiation of the cancer site. Here, we report the first boron neutron immunotherapy (B-NIT), combining BNCT and ICI immunotherapy, which was performed on a radioresistant and immunotherapy-resistant advanced-stage B16F10 melanoma mouse model. The BNCT group showed localized tumor suppression, but the anti-PD-1 antibody immunotherapy group did not show tumor suppression. Only the B-NIT group showed strong tumor growth inhibition at both BNCT-treated and shielded distant sites. Intratumoral CD8+ T-cell infiltration and serum high mobility group box 1 (HMGB1) levels were higher in the B-NIT group. Analysis of CD8+ T cells in tumor-infiltrating lymphocytes (TILs) showed that CD62L- CD44+ effector memory T cells and CD69+ early-activated T cells were predominantly increased in the B-NIT group. Administration of CD8-depleting mAb to the B-NIT group completely suppressed the augmented therapeutic effects. This indicated that B-NIT has a potent immune-induced abscopal effect, directly destroying tumors with BNCT, inducing antigen-spreading effects, and protecting normal tissue. B-NIT, immunotherapy combined with BNCT, is the first treatment to overcome immunotherapy resistance in malignant melanoma. In the future, as its therapeutic efficacy is demonstrated not only in melanoma but also in other immunotherapy-resistant malignancies, B-NIT can become a new treatment candidate for advanced-stage cancers.
    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
    骨肉瘤(OS),儿童和青少年骨最常见的原发性恶性肿瘤,免疫检查点抑制剂由于其抗肿瘤免疫应答不良而难以治疗。化疗和病毒疗法诱导免疫原性细胞死亡(ICD)和抗肿瘤免疫反应,在未经治疗的肿瘤中导致外镜效应。我们先前证明了端粒酶特异性复制型溶瘤腺病毒OBP-301和p53武装的OBP-702在人类OS细胞中的抗肿瘤活性。这里,我们展示了化疗药物(阿霉素,顺铂)和端粒酶特异性溶瘤腺病毒(OBP-301,p53武装的OBP-702)在人OS细胞中诱导ICD(U2OS,MNNG/HOS,SaOS-2)和鼠OS细胞(NHOS)。与化疗和OBP-301相比,OBP-702通过分泌三磷酸腺苷(ATP)和高迁移率族蛋白盒B1(HMGB1)在人OS细胞中诱导更深入的ICD。鼠NHOS细胞对OBP-702也比OBP-301更敏感。皮下NHOS肿瘤模型表明,与OBP-301相比,瘤内注射OBP-702显着增加了细胞毒性CD8T细胞的肿瘤浸润,并诱导了对未治疗肿瘤的外视作用。我们的结果表明,OBP-702是一种有前途的抗肿瘤试剂,可诱导ICD分泌ATP和HMGB1,并具有抗OS的作用。
    Osteosarcoma (OS), the most frequent primary malignant tumor of bone in children and adolescents, is refractory to immune checkpoint inhibitors due to its poor antitumor immune response. Chemotherapy and virotherapy induce immunogenic cell death (ICD) and antitumor immune responses, leading to the abscopal effect in untreated tumors. We previously demonstrated the antitumor activity of the telomerase-specific replication-competent oncolytic adenoviruses OBP-301 and p53-armed OBP-702 in human OS cells. Here, we show the therapeutic potential of chemotherapeutic drugs (doxorubicin, cisplatin) and telomerase-specific oncolytic adenoviruses (OBP-301, p53-armed OBP-702) to induce ICD in human OS cells (U2OS, MNNG/HOS, SaOS-2) and murine OS cells (NHOS). OBP-702 induced more profound ICD via the secretion of adenosine triphosphate (ATP) and high-mobility group box protein B1 (HMGB1) compared with chemotherapy and OBP-301 in human OS cells. Murine NHOS cells were also more sensitive to OBP-702 than OBP-301. Subcutaneous NHOS tumor models demonstrated that intratumoral injection of OBP-702 significantly increased the tumor infiltration of cytotoxic CD8+ T cells and induced the abscopal effect against non-treated tumors compared with OBP-301. Our results suggest that OBP-702 is a promising antitumor reagent to induce ICD with secretion of ATP and HMGB1 and the abscopal effect against OS.
    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
    EGFR突变的非小细胞肺癌(NSCLC)患者对免疫检查点抑制剂(ICIs)的反应较差。据报道,EGFR突变的NSCLC中CD8+T细胞的数量减少。然而,不同CD8+T细胞群体的异质性和效应子功能的程度尚未得到深入研究.此外,目前缺乏研究放疗和ICIs联合治疗是否能提高EGFR突变肺癌ICIs疗效的研究.使用单细胞RNA测序(scRNA-seq)来研究EGFR突变的NSCLC中CD8+T细胞群体的异质性。在EGFR突变和野生型细胞的低分割辐射后探索STING途径。将携带LLC-19del和LLC-EGFR肿瘤的小鼠用放射疗法加抗PD-L1治疗。scRNA-seq数据显示,在EGFR突变的NSCLC中,祖细胞耗尽的CD8+T细胞的百分比较低。此外,EGFR突变的NSCLC中的CD8+T细胞富集氧化磷酸化。在EGFR突变和野生型细胞中,8Gy×3增加募集T细胞并激活cGAS-STING途径的趋化因子的表达。在LLC-19del和LLC-EGFR小鼠模型中,放疗和抗PD-L1联合使用可显著抑制腹腔镜肿瘤的生长。增强的远视效应与全身性CD8T细胞浸润有关。这项研究为EGFR突变的NSCLC中CD8+T细胞的异质性和效应子功能提供了深入的理解。我们表明,大分割辐射和抗PD-L1的组合通过激活小鼠中的CD8T细胞显着增强了EGFR突变和野生型肺癌的远视反应。
    Patients with EGFR-mutated non-small cell lung cancer (NSCLC) respond poorly to immune checkpoint inhibitors (ICIs). It has been reported that the number of CD8+T cells is reduced in EGFR-mutated NSCLC. However, the extent of heterogeneity and effector function of distinct populations of CD8+T cells has not been investigated intensively. In addition, studies investigating whether a combination of radiotherapy and ICIs can improve the efficacy of ICIs in EGFR-mutated lung cancer are lacking. Single-cell RNA sequencing (scRNA-seq) was used to investigate the heterogeneity of CD8+T cell populations in EGFR-mutated NSCLC. The STING pathway was explored after hypofractionated radiation of EGFR-mutated and wild-type cells. Mice bearing LLC-19del and LLC-EGFR tumors were treated with radiotherapy plus anti-PD-L1. The scRNA-seq data showed the percentage of progenitor exhausted CD8+T cells was lower in EGFR-mutated NSCLC. In addition, CD8+T cells in EGFR-mutated NSCLC were enriched in oxidative phosphorylation. In EGFR-mutated and wild-type cells, 8 Gy × 3 increased the expression of chemokines that recruit T cells and activate the cGAS-STING pathway. In the LLC-19del and LLC-EGFR mouse model, the combination of radiation and anti-PD-L1 significantly inhibited the growth of abscopal tumors. The enhanced abscopal effect was associated with systemic CD8+T cell infiltration. This study provided an intensive understanding of the heterogeneity and effector functions of CD8+T cells in EGFR-mutated NSCLC. We showed that the combination of hypofractionated radiation and anti-PD-L1 significantly enhanced the abscopal responses in both EGFR-mutated and wild-type lung cancer by activating CD8+T cells in mice.
    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
    目的:前瞻性比较不可逆电穿孔(IRE)和机器人辅助根治性前列腺切除术(RARP)在局部中危前列腺癌(PCa)患者中诱导的全身抗肿瘤免疫反应。
    方法:在2021年2月至2022年6月之间,前瞻性收集30例局限性PCa患者治疗前后(第5、14和30天)的外周血样本。患者纳入标准为:国际泌尿外科病理学家学会2-3级,临床癌症分期≤T2c,前列腺特异性抗原水平<20ng/mL)。患者接受IRE(n=20)或RARP(n=10)治疗。使用流式细胞术确定淋巴细胞和骨髓免疫细胞亚群的频率和活化状态。通过干扰素-γ酶联免疫斑点测定(ELISpot)确定PCa特异性T细胞对前列腺酸性磷酸酶(PSAP)和癌症睾丸抗原(纽约食管鳞状细胞癌1[NY-ESO-1])的反应。使用重复测量的方差分析和双侧Student'st检验来比较随时间和治疗组之间的免疫应答。
    结果:除了年龄(中位数为68岁[IRE]和62岁[RARP],P=0.01)。IRE诱导全身调节性T细胞的消耗(P=0.0001)和激活的细胞毒性T淋巴细胞抗原4(CTLA-4)+分化簇(CD)4+(P<0.001)和CD8+(P=0.032)T细胞同时增加,与减少全身性免疫抑制一致,允许效应T细胞激活,IRE后14天达到峰值。效果与肿瘤体积/消融大小呈正相关。因此,IRE在8名免疫有能力的患者中的4名中诱导PSAP和/或NY-ESO-1特异性T细胞应答的扩增。暂时增加的活化的骨髓来源的抑制细胞频率(P=0.047)与RARP后的短暂免疫抑制一致。
    结论:不可逆电穿孔可诱导局部PCa患者的PCa特异性全身免疫反应,帮助肿瘤微环境转化为更免疫允许的状态。通过与CTLA-4检查点抑制相结合,可以进一步提高治疗效果。可能为高风险的局部或(寡)转移性疾病开辟新的协同治疗模式。
    OBJECTIVE: To prospectively compare systemic anti-tumour immune responses induced by irreversible electroporation (IRE) and robot-assisted radical prostatectomy (RARP) in patients with localised intermediate-risk prostate cancer (PCa).
    METHODS: Between February 2021 and June 2022, before and after treatment (at 5, 14 and 30 days) peripheral blood samples of 30 patients with localised PCa were prospectively collected. Patient inclusion criteria were: International Society of Urological Pathologists Grade 2-3, clinical cancer stage ≤T2c, prostate-specific antigen level <20 ng/mL). Patients were treated with IRE (n = 20) or RARP (n = 10). Frequency and activation status of lymphocytic and myeloid immune cell subsets were determined using flow cytometry. PCa-specific T-cell responses to prostatic acid phosphatase (PSAP) and cancer testis antigen (New York oesophageal squamous cell carcinoma 1 [NY-ESO-1]) were determined by interferon-γ enzyme-linked immunospot assay (ELISpot). Repeated-measures analysis of variance and two-sided Student\'s t-tests were used to compare immune responses over time and between treatment cohorts.
    RESULTS: Patient and tumour characteristics were similar between the cohorts except for age (median 68 years [IRE] and 62 years [RARP], P = 0.01). IRE induced depletion of systemic regulatory T cells (P = 0.0001) and a simultaneous increase in activated cytotoxic T-lymphocyte antigen 4 (CTLA-4)+ cluster of differentiation (CD)4+ (P < 0.001) and CD8+ (P = 0.032) T cells, consistent with reduction of systemic immune suppression allowing for effector T-cell activation, peaking 14 days after IRE. Effects were positively correlated with tumour volume/ablation size. Accordingly, IRE induced expansion of PSAP and/or NY-ESO-1 specific T-cell responses in four of the eight immune competent patients. Temporarily increased activated myeloid derived suppressor cell frequencies (P = 0.047) were consistent with transient immunosuppression after RARP.
    CONCLUSIONS: Irreversible electroporation induces a PCa-specific systemic immune response in patients with localised PCa, aiding conversion of the tumour microenvironment into a more immune permissive state. Therapeutic efficacy might be further enhanced by combination with CTLA-4 checkpoint inhibition, potentially opening up a new synergistic treatment paradigm for high-risk localised or (oligo)metastatic disease.
    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
    在癌症治疗中,小分子药物的系统给药往往具有低疗效和全身毒性。此外,单一模式药物的应用通常导致不令人满意的治疗结果。目前,开发作用于不同途径而不增加副作用的多模式药物组合策略仍然是巨大的挑战.这里,我们开发了一种水凝胶系统,该系统共同递送糖酵解抑制剂芹菜素和化疗药物吉西他滨,以实现以最小的全身毒性对抗癌症的组合策略。我们证明了该系统不仅可以在原位消除肿瘤细胞,而且对各种肿瘤模型也有不同程度的影响。这些结果表明,我们的研究为临床癌症治疗提供了安全有效的策略。
    Systematic administration of small molecular drugs often suffered from the low efficacy and systemic toxicity in cancer therapy. In addition, application of single mode drug usually leads to unsatisfactory therapeutic outcomes. Currently, developing multimodal-drug combination strategy that acts on different pathways without increasing side effects remains great challenge. Here, we developed a hydrogel system that co-delivered glycolysis inhibitor apigenin and chemo-drug gemcitabine to realize combination strategy for combating cancer with minimal systemic toxicity. We demonstrated that this system can not only eliminate tumor cells in situ, but also induce abscopal effect on various tumor models. These results showed that our study provided a safe and effective strategy for clinical cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号