Intratumoral immunotherapy

肿瘤内免疫治疗
  • 文章类型: Journal Article
    背景:放疗(RT)与免疫检查点阻断(ICB)协同作用。肿瘤微环境中的CD1c(BDCA-1)/CD141(BDCA-3)髓样树突状细胞(myDC)在启动效应T细胞反应和对ICB的反应中是必不可少的。
    方法:在这项II期临床试验中,抗PD-1ICB预处理的寡转移患者(肿瘤无关者)接受了白细胞去除术,然后分离CD1c(BDCA-1)+/CD141(BDCA-3)+myDC.在低分割立体定向体RT(3×8Gy)之后,患者被随机分组(3:1).分别,在手臂A(立即治疗),肿瘤内(IT)ipilimumab(10mg)和avelumab(40mg)联合静脉(IV)pembrolizumab(200mg),然后IT注射myDC;随后,继续静脉注射派姆单抗和ITipilimumab/avelumab(q3W)。在B臂(当代控制臂)中,患者接受静脉注射pembrolizumab,有可能在进展中交叉。主要终点是1年无进展生存率(PFS)。次要终点是安全性,可行性,客观反应率,PFS,总生存率(OS)。
    结果:13例患者(A组10例,八种非小细胞肺癌,和五个黑色素瘤)被登记。两个病人交叉。A臂的一年PFS率为10%,B臂为0%。一名患者获得了稳定的疾病作为最佳反应。在B臂,一名患者获得了SD。中位PFS和OS分别为21.8周(A组)和24.9周(B组),和62.7对57.9周,分别。医源性气胸是唯一的3级治疗相关不良事件。
    结论:SBRT和派姆单抗联合或不联合ITavelumab/ipilimumab和ITmyDC在寡转移患者中是安全可行的,具有临床意义的肿瘤反应率。然而,该研究未能达到主要终点.
    背景:临床试验:NCT04571632(2020年8月9日)。
    2019-003668-32。注册日期:2019年12月17日,2021年3月1:6日,2022年2月2:4日。
    BACKGROUND: Radiotherapy (RT) synergizes with immune checkpoint blockade (ICB). CD1c(BDCA-1)+/CD141(BDCA-3)+ myeloid dendritic cells (myDC) in the tumor microenvironment are indispensable at initiating effector T-cell responses and response to ICB.
    METHODS: In this phase II clinical trial, anti-PD-1 ICB pretreated oligometastatic patients (tumor agnostic) underwent a leukapheresis followed by isolation of CD1c(BDCA-1)+/CD141(BDCA-3)+ myDC. Following hypofractionated stereotactic body RT (3 × 8 Gy), patients were randomized (3:1). Respectively, in arm A (immediate treatment), intratumoral (IT) ipilimumab (10 mg) and avelumab (40 mg) combined with intravenous (IV) pembrolizumab (200 mg) were administered followed by IT injection of myDC; subsequently, IV pembrolizumab and IT ipilimumab/avelumab were continued (q3W). In arm B (contemporary control arm), patients received IV pembrolizumab, with possibility to cross-over at progression. Primary endpoint was 1-year progression-free survival rate (PFS). Secondary endpoints were safety, feasibility, objective response rate, PFS, and overall survival (OS).
    RESULTS: Thirteen patients (10 in arm A, eight non-small cell lung cancer, and five melanoma) were enrolled. Two patients crossed over. One-year PFS rate was 10% in arm A and 0% in arm B. Two patients in arm A obtained a partial response, and one patient obtained a stable disease as best response. In arm B, one patient obtained a SD. Median PFS and OS were 21.8 weeks (arm A) versus 24.9 (arm B), and 62.7 versus 57.9 weeks, respectively. An iatrogenic pneumothorax was the only grade 3 treatment-related adverse event.
    CONCLUSIONS: SBRT and pembrolizumab with or without IT avelumab/ipilimumab and IT myDC in oligometastatic patients are safe and feasible with a clinically meaningful tumor response rate. However, the study failed to reach its primary endpoint.
    BACKGROUND: Clinicaltrials.gov: NCT04571632 (09 AUG 2020).
    UNASSIGNED: 2019-003668-32. Date of registration: 17 DEC 2019, amendment 1: 6 MAR 2021, amendment 2: 4 FEB 2022.
    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
    癌症的射频消融(RFA)诱导抗肿瘤免疫,这不足以防止复发。在老鼠身上,通过粒细胞-巨噬细胞集落刺激因子(GM-CSF)和卡介苗芽孢杆菌进行RFA-肿瘤内免疫治疗可导致转移完全消退。人类感染风险需要更换活疫苗。肿瘤内纯化蛋白衍生物(PPD)从未在消化系统癌症中进行过测试。RFA后肿瘤内免疫治疗的安全性尚未在人类模型中得到验证.我们研究了使用免疫粘膜粘附热凝胶(IMT)在转移性结直肠癌(CRC)小鼠模型中联合射频消融(RFA)和肿瘤内免疫疗法(ITI)的治疗效果,以及该方法在猪模型中的安全性。使用磁共振成像(MRI)和生物发光成像评估免疫凝胶的瘤内稳定性。用RFA单独或与肿瘤内IMT组合治疗74只携带CT26肿瘤的雌性BALB/c小鼠。监测远处转移和存活的消退60天。六只接受肝脏射频和病灶内IMT注射的猪随访15天。使用血管内方法治疗实验性凝胶栓塞。在肿瘤中证实了IMT的相关流变学,通过MRI中3天和生物发光成像中7天的信号稳定性。在老鼠身上,RFA-肿瘤内免疫治疗的切除效应导致在第16天完成的远处病变消退。RFA组在第25天的体积为350±99.3mm3,在60天的存活率为10倍。在猪中,在没有临床的情况下,容量调整后,在肝脏RFA区域注射免疫凝胶是安全的,血液学,和肝脏生物学紊乱。流式细胞术显示D7时CD3TCRγδ+T细胞早期增加(p<0.05),D15时CD29+-CD8T细胞晚期减少(p<0.05),反映了炎症状态的变化。未检测到系统性GM-CSF释放。通过经皮导管插入和冷血清输注治疗实验性腔和肺热凝胶栓塞。RFA-肿瘤内免疫治疗作为一种高效、安全的微创介入肿瘤学能够改善结直肠癌肝转移的消融治疗。
    Radiofrequency ablation (RFA) of cancer induces an anti-tumor immunity, which is insufficient to prevent recurrences. In mice, RFA-intratumoral immunotherapy by granulocyte-macrophage colony-stimulating factor (GM-CSF) and Bacillus Calmette-Guerin resulted in complete metastases regression. Infectious risk in human needs replacement of live vaccines. Intratumoral purified protein derivatives (PPD) have never been tested in digestive cancers, and the safety of intratumoral immunotherapy after RFA has not yet been validated in human models. We investigated the therapeutic efficacy of combined radiofrequency ablation (RFA) and intratumoral immunotherapy (ITI) using an immune-muco-adherent thermogel (IMT) in a mouse model of metastatic colorectal cancer (CRC) and the safety of this approach in a pig model. Intratumoral stability of the immunogel was assessed using magnetic resonance imaging (MRI) and bioluminescent imaging. Seventy-four CT26 tumor-bearing female BALB/c mice were treated with RFA either alone or in combination with intratumoral IMT. Regression of distant metastasis and survival were monitored for 60 days. Six pigs that received liver radiofrequency and intralesional IMT injections were followed for 15 days. Experimental gel embolisms were treated using an intravascular approach. Pertinent rheology of IMT was confirmed in tumors, by the signal stability during 3 days in MRI and 7 days in bioluminescence imaging. In mice, the abscopal effect of RFA-intratumoral immunotherapy resulted in regression of distant lesions completed at day 16 vs. a volume of 350 ± 99.3 mm3 in the RFA group at day 25 and a 10-fold survival rate at 60 days. In pigs, injection of immunogel in the liver RFA area was safe after volume adjustment without clinical, hematological, and liver biology disorder. Flow cytometry showed an early increase in CD3 TCRγδ+T cells at D7 (p < 0.05) and a late decrease in CD29+-CD8 T cells at D15 (p < 0.05), reflecting the inflammation status changes. Systemic GM-CSF release was not detectable. Experimental caval and pulmonary thermogel embolisms were treated by percutaneous catheterism and cold serum infusion. RFA-intratumoral immunotherapy as efficient and safe mini-invasive interventional oncology is able to improve ablative treatment of colorectal liver metastases.
    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
    癌症的免疫检查点疗法(ICT)可以产生巨大的临床反应;然而,这些可能只在少数患者中观察到。这些反应可进一步受到随后的疾病复发和抗性的限制。正在开发组合免疫疗法策略以克服这些限制。我们以前曾报道过瘤内cow豆花叶病毒免疫疗法(CPMVIIT)和ICT方法的联合功效增强。淋巴细胞活化基因-3(LAG-3)是下一代抑制性免疫检查点,其在多个免疫细胞亚群中具有广泛表达。其表达在活化的T细胞上增加并有助于T细胞耗尽。我们在黑素瘤的小鼠模型中观察到CPMVIIT和抗LAG-3联合治疗的功效增强。Further,发现瘤内CPMV施用后TME内的LAG-3表达增加。CPMVIIT与LAG-3抑制的整合具有通过同时诱导全面的抗肿瘤免疫反应来改善治疗结果的显着潜力。增强局部免疫激活,减轻T细胞衰竭。
    Immune checkpoint therapy (ICT) for cancer can yield dramatic clinical responses; however, these may only be observed in a minority of patients. These responses can be further limited by subsequent disease recurrence and resistance. Combination immunotherapy strategies are being developed to overcome these limitations. We have previously reported enhanced efficacy of combined intratumoral cowpea mosaic virus immunotherapy (CPMV IIT) and ICT approaches. Lymphocyte-activation gene-3 (LAG-3) is a next-generation inhibitory immune checkpoint with broad expression across multiple immune cell subsets. Its expression increases on activated T cells and contributes to T cell exhaustion. We observed heightened efficacy of a combined CPMV IIT and anti-LAG-3 treatment in a mouse model of melanoma. Further, LAG-3 expression was found to be increased within the TME following intratumoral CPMV administration. The integration of CPMV IIT with LAG-3 inhibition holds significant potential to improve treatment outcomes by concurrently inducing a comprehensive anti-tumor immune response, enhancing local immune activation, and mitigating T cell exhaustion.
    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)可以与免疫系统一起消除癌症。它可以引起免疫原性细胞死亡并促进肿瘤新抗原呈递,从而促进肿瘤特异性T淋巴细胞的交叉引发。将照射的肿瘤转化为原位疫苗。越来越多的临床前和临床证据表明,RT与ICB联合导致全身抗肿瘤免疫反应,从而激发了人们对使用ICB克服原发性和获得性癌症对放射疗法的耐药性的兴趣。然而,迄今为止获得的全身性效应(abscopal效应)远不能为临床翻译所接受.在这种情况下,多个临床前小鼠模型已经证明,多种免疫治疗剂可以局部递送,从而以局部和全身方式增强抗肿瘤免疫力.在同基因免疫活性宿主(C57BL/6)中使用两个略微异步且解剖学上远处的皮下B16OVA肿瘤,我们描述了结合外照射给予局部免疫治疗的可行性,在肿瘤内递送后在小鼠和人类中发挥免疫介导的抗肿瘤作用。随着微小的变化,同样的技术可以很容易地应用于各种小鼠可移植的肿瘤。
    Radiotherapy (RT) can work together with the immune system to eliminate cancer. It can cause immunogenic cell death and facilitate tumor neoantigen presentation and thereby the cross-priming of tumor-specific T-lymphocytes, turning irradiated tumors into in-situ vaccines. Accumulating preclinical and clinical evidence indicates that RT in conjunction with ICB leads to systemic anti-tumor immune responses, thus stimulating interest in using ICB to overcome primary and acquired cancer resistance to radiotherapy. However, the systemic effects (abscopal effects) obtained to date are far from being acceptable for clinical translation. In this context, multiple preclinical mouse models have demonstrated that a variety of immunotherapy agents can be delivered locally to enhance antitumor immunity both in a local and systemic fashion. Using two slightly asynchronous and anatomically distant subcutaneous B16OVA tumors in syngeneic immunocompetent hosts (C57BL/6), we describe the feasibility of a local immunotherapy treatment given in combination with external beam irradiation, which exerts immune-mediated antitumor effects in mice and humans upon intratumoral delivery. With minor variations, the same technique can be easily applied to a variety of mouse transplantable tumors.
    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
    可以想象,抑制性受体NKG2A与非经典MHC-I分子HLA-E或小鼠Qa-1b的相互作用在肿瘤微环境中抑制了NK细胞对癌症的反应性。我们发现,只有在与抗NKG2A和抗Qa-1b阻断性单克隆抗体共同注射针对实体小鼠肿瘤模型时,NK细胞的瘤内递送才能达到显着的治疗效果。这种治疗活性取决于内源性CD8T细胞和1型常规树突细胞(cDC1)。此外,与全身性抗PD-1mAb治疗联合治疗后,抗肿瘤作用得到增强,并对远处未注射的肿瘤取得部分外移疗效.在携带表达HLA-E的人类癌细胞的异种移植小鼠中,肿瘤内共注射活化的同种异体人NK细胞和临床级抗NKG2AmAb(monalizumab)协同实现了治疗效果。总之,这些研究为基于肿瘤内NK细胞的免疫治疗的临床潜力提供了证据,这些免疫治疗通过引发内源性T细胞反应而发挥抗肿瘤功效.
    NK-cell reactivity against cancer is conceivably suppressed in the tumor microenvironment by the interaction of the inhibitory receptor NKG2A with the non-classical MHC-I molecules HLA-E in humans or Qa-1b in mice. We found that intratumoral delivery of NK cells attains significant therapeutic effects only if co-injected with anti-NKG2A and anti-Qa-1b blocking monoclonal antibodies against solid mouse tumor models. Such therapeutic activity was contingent on endogenous CD8 T cells and type-1 conventional dendritic cells (cDC1). Moreover, the anti-tumor effects were enhanced upon combination with systemic anti-PD-1 mAb treatment and achieved partial abscopal efficacy against distant non-injected tumors. In xenografted mice bearing HLA-E-expressing human cancer cells, intratumoral co-injection of activated allogeneic human NK cells and clinical-grade anti-NKG2A mAb (monalizumab) synergistically achieved therapeutic effects. In conclusion, these studies provide evidence for the clinical potential of intratumoral NK cell-based immunotherapies that exert their anti-tumor efficacy as a result of eliciting endogenous T-cell responses.
    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
    缺乏评估新型药物的最佳模型正在延迟针对人类乳腺癌(BC)的有效免疫疗法的开发。在这项前瞻性开放标签研究中,我们应用新辅助瘤内免疫疗法与空cow豆花叶病毒样颗粒(eCPMV)对11只诊断为犬乳腺癌(CMC)的伴侣犬,类似于人类BC的自发性肿瘤。我们发现,两次新辅助肿瘤内注射eCPMV可导致所有患者的注射肿瘤以及位于注射犬同侧和对侧乳腺链中的非注射肿瘤的肿瘤减少。肿瘤缩小与临床分期无关,肿瘤大小,组织病理学分级,和肿瘤分子亚型。注射肿瘤的基于RNA-seq的分析表明DNA复制活性降低,肿瘤微环境中激活的树突状细胞浸润增加。免疫组织化学分析显示肿瘤内中性粒细胞显著增加,T和B淋巴细胞,和浆细胞。eCPMV肿瘤内免疫治疗显示抗肿瘤疗效,无任何不良反应。这种新型免疫疗法具有改善人类BC患者预后的潜力。
    The lack of optimal models to evaluate novel agents is delaying the development of effective immunotherapies against human breast cancer (BC). In this prospective open label study, we applied neoadjuvant intratumoral immunotherapy with empty cowpea mosaic virus-like particles (eCPMV) to 11 companion dogs diagnosed with canine mammary cancer (CMC), a spontaneous tumor resembling human BC. We found that two neoadjuvant intratumoral eCPMV injections resulted in tumor reduction in injected tumors in all patients and in noninjected tumors located in the ipsilateral and contralateral mammary chains of injected dogs. Tumor reduction was independent of clinical stage, tumor size, histopathologic grade, and tumor molecular subtype. RNA-seq-based analysis of injected tumors indicated a decrease in DNA replication activity and an increase in activated dendritic cell infiltration in the tumor microenvironment. Immunohistochemistry analysis demonstrated significant intratumoral increases in neutrophils, T and B lymphocytes, and plasma cells. eCPMV intratumoral immunotherapy demonstrated antitumor efficacy without any adverse effects. This novel immunotherapy has the potential for improving outcomes for human BC patients.
    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
    近年来,肿瘤免疫治疗在临床上取得了显著进展。尽管年龄是癌症发展的最大主要风险因素之一,老年人代表了大多数癌症患者,只有少数新的癌症免疫治疗干预措施在老年动物中进行了临床前测试.因此,缺乏关注癌症免疫治疗过程中年龄依赖性效应的临床前研究,可能会导致幼年和老年动物的治疗结果不同,并且未来会对人体临床试验进行修改.这里,我们比较了以前开发和测试的肿瘤内免疫疗法的疗效,基于多糖甘露聚糖的组合,toll样受体配体,和抗CD40抗体(MBTA免疫治疗),在患有实验性嗜铬细胞瘤(PHEO)的年轻(6周)和老年(71周)小鼠中。所提出的结果指出,尽管PHEO在老年小鼠中生长更快,但MBTA肿瘤内免疫疗法是没有年龄依赖性的有效方法,并且可能是增强老年和年轻宿主对嗜铬细胞瘤和其他肿瘤类型的免疫反应的可能治疗干预措施之一。
    Cancer immunotherapy has shown remarkable clinical progress in recent years. Although age is one of the biggest leading risk factors for cancer development and older adults represent a majority of cancer patients, only a few new cancer immunotherapeutic interventions have been preclinically tested in aged animals. Thus, the lack of preclinical studies focused on age-dependent effect during cancer immunotherapy could lead to different therapeutic outcomes in young and aged animals and future modifications of human clinical trials. Here, we compare the efficacy of previously developed and tested intratumoral immunotherapy, based on the combination of polysaccharide mannan, toll-like receptor ligands, and anti-CD40 antibody (MBTA immunotherapy), in young (6 weeks) and aged (71 weeks) mice bearing experimental pheochromocytoma (PHEO). The presented results point out that despite faster growth of PHEO in aged mice MBTA intratumoral immunotherapy is effective approach without age dependence and could be one of the possible therapeutic interventions to enhance immune response to pheochromocytoma and perhaps other tumor types in aged and young hosts.
    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
    植物病毒纳米颗粒可作为药物载体,成像试剂,疫苗携带者,瘤内原位癌疫苗制剂中的免疫佐剂。一个例子是cow豆花叶病毒(CPMV),一种无包膜病毒,具有二分正链RNA基因组,每种RNA分别包装到相同的蛋白质衣壳中。根据它们的密度差异,例如,携带RNA-1(6kb)的组分表示为底部(B)组分或携带RNA-2(3.5kb)表示为中间(M)组分可以彼此分离并与顶部(T)组分分离。没有任何RNA。以前的临床前小鼠研究和犬癌试验使用CPMV的混合群体(含有B,M,和T分量),因此,目前尚不清楚粒子类型是否在其功效上有所不同。已知CPMVRNA基因组通过激活TLR7而有助于免疫刺激。为了确定具有不同大小和不相关序列的两个RNA基因组是否引起不同的免疫刺激,我们比较了B和M组分和普通CPMV在体外和小鼠癌症模型中的治疗效果。我们发现分离的B和M颗粒的行为与混合的CPMV相似,激活先天性免疫细胞以诱导促炎细胞因子如IFNα的分泌,IFNγ,IL-6和IL-12,同时抑制免疫抑制细胞因子如TGF-β和IL-10。在黑色素瘤和结肠癌的小鼠模型中,混合和分离的CPMV颗粒均显着降低了肿瘤的生长,延长了生存期,但没有显着差异。这表明,即使B颗粒具有比M颗粒多40%的RNA,特定RNA基因组也类似地刺激免疫系统;每种CPMV颗粒类型可以用作针对癌症的有效佐剂,具有与天然混合CPMV相同的功效。从平移的角度来看,B或M组分相对于混合CPMV制剂的使用提供了这样的优点,即单独分离的B或M对植物是非传染性的,因此提供了农艺安全性。
    Plant virus nanoparticles can be used as drug carriers, imaging reagents, vaccine carriers, and immune adjuvants in the formulation of intratumoral in situ cancer vaccines. One example is the cowpea mosaic virus (CPMV), a nonenveloped virus with a bipartite positive-strand RNA genome with each RNA packaged separately into identical protein capsids. Based on differences in their densities, the components carrying RNA-1 (6 kb) denoted as the bottom (B) component or carrying RNA-2 (3.5 kb) denoted as the middle (M) component can be separated from each other and from a top (T) component, which is devoid of any RNA. Previous preclinical mouse studies and canine cancer trials used mixed populations of CPMV (containing B, M, and T components), so it is unclear whether the particle types differ in their efficacies. It is known that the CPMV RNA genome contributes to immunostimulation by activation of TLR7. To determine whether the two RNA genomes that have different sizes and unrelated sequences cause different immune stimulation, we compared the therapeutic efficacies of B and M components and unfractionated CPMV in vitro and in mouse cancer models. We found that separated B and M particles behaved similarly to the mixed CPMV, activating innate immune cells to induce the secretion of pro-inflammatory cytokines such as IFNα, IFNγ, IL-6, and IL-12, while inhibiting immunosuppressive cytokines such as TGF-β and IL-10. In murine models of melanoma and colon cancer, the mixed and separated CPMV particles all significantly reduced tumor growth and prolonged survival with no significant difference. This shows that the specific RNA genomes similarly stimulate the immune system even though B particles have 40% more RNA than M particles; each CPMV particle type can be used as an effective adjuvant against cancer with the same efficacy as native mixed CPMV. From a translational point of view, the use of either B or M component vs the mixed CPMV formulation offers the advantage that separated B or M alone is noninfectious toward plants and thus provides agronomic safety.
    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
    癌症免疫疗法是一个引起人们极大兴趣的领域,由免疫检查点抑制剂的临床成功推动。与传统的癌症疗法相反,免疫疗法通过增强先天和适应性免疫来控制癌症进展,从而利用宿主的免疫系统。尽管取得了这些令人激动的进展,只有一部分患者对这些药物有反应,和免疫疗法经常导致免疫相关的毒性。克服这些挑战的一种方法是肿瘤内施用治疗以使全身毒性最小化并使治疗效果最大化。肿瘤内癌症治疗在治疗和远处未治疗的肿瘤中显示出相似或优异的抗肿瘤功效。与常规治疗方法相比,获益风险比得到了广泛改善。在这里,我们回顾了肿瘤内肿瘤基因免疫治疗的现状。
    免疫疗法是旨在激活患者自身免疫系统以对抗癌症的药物。在美国FDA批准第一类此类药物后,该领域的研究激增。它们通过阻断癌细胞隐藏身体免疫系统的能力来发挥作用。不幸的是,当药物被输送到全身时,只有一些患者有反应,许多患者会出现副作用。克服这些问题的一种方法是将这些药物直接递送到患者的肿瘤中以限制副作用,同时保持积极作用。在这篇综述中,我们描述了在全身给药中直接将这些类型的药物给予肿瘤的益处。我们总结了当前的临床数据,并解释了每种药物背后的机制。
    Cancer immunotherapy is a field that garners significant interest, fueled by the clinical success of immune checkpoint inhibitors. In contrast to conventional cancer therapies, immunotherapies leverage the host\'s immune system by enhancing innate and adaptive immunity to control cancer progression. Despite these exciting advances, only a subset of patients respond to these drugs, and immunotherapies frequently result in immune-related toxicity. One approach to overcome these challenges is intratumoral administration of treatment to minimize systemic toxicities and maximize therapeutic effects. Intratumoral cancer therapies have shown similar or superior antitumor efficacy in both treated and distant untreated tumors, with a widely improved benefit-risk ratio over conventional therapeutic approaches. Herein, we review the current landscape of intratumoral cancer gene immunotherapy.
    Immunotherapies are drugs designed to activate a patient’s own immune system to fight cancer. Research in this field has soared following the US FDA’s approval of the first class of these drugs. They work by blocking cancer cells’ ability to hide from the body’s immune system. Unfortunately, only some patients respond and many experience side effects when the medicine is delivered to the whole body. One approach to overcome these problems is to deliver these drugs directly into a patient’s tumor to limit side effects while maintaining the positive effects. In this review we describe the benefits of giving these types of drugs directly into tumors over whole-body administration. We summarize the current clinical data and explain the mechanisms behind each drug.
    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
    系统管理的免疫疗法彻底改变了癌症患者的护理;然而,对于许多癌症类型,大多数患者没有表现出客观的反应。肿瘤内免疫治疗是一种新兴的策略,旨在提高整个恶性肿瘤范围内癌症免疫疗法的有效性。通过对肿瘤本身进行局部免疫激活疗法,肿瘤微环境中的免疫抑制屏障可以被打破。此外,对于全身给药过于有效的疗法可以安全地施用到目标位置,以最大化疗效和最小化毒性.为了使这些疗法有效,虽然,它们必须被有效地递送到靶肿瘤病灶中。在这次审查中,我们总结了肿瘤内免疫疗法的现状,并强调了影响肿瘤内递送的关键概念,引申开来,功效。我们还提供了已批准的微创递送装置的广度和深度的概述,这些装置可以被认为可以改善肿瘤内治疗的递送。
    Systemically administered immunotherapies have revolutionized the care of patients with cancer; however, for many cancer types, most patients do not exhibit objective responses. Intratumoral immunotherapy is a burgeoning strategy that is designed to boost the effectiveness of cancer immunotherapies across the spectrum of malignancies. By locally administering immune-activating therapies into the tumor itself, immunosuppressive barriers in the tumor microenvironment can be broken. Moreover, therapies too potent for systemic delivery can be safely administered to target location to maximize efficacy and minimize toxicity. In order for these therapies to be effective, though, they must be effectively delivered into the target tumor lesion. In this review, we summarize the current landscape of intratumoral immunotherapies and highlight key concepts that influence intratumoral delivery, and by extension, efficacy. We also provide an overview of the breadth and depth of approved minimally invasive delivery devices that can be considered to improve delivery of intratumoral therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号