bystander effect

旁观者效应
  • 文章类型: Journal Article
    2019年冠状病毒病(COVID-19)可能会影响艾滋病毒携带者(PLWH)的疾病进展,包括那些有效的联合抗逆转录病毒治疗(cART)。这些个体经常经历以CD4+记忆T细胞和组织巨噬细胞中的前病毒潜伏期或低水平病毒复制为特征的慢性病症。促炎细胞因子,如TNF-α,IL-1β,IL-6和IFN-γ,可以在原代细胞和细胞系中重新激活原病毒表达。这些细胞因子通常在感染SARS-CoV-2的个体中升高,SARS-CoV-2是导致COVID-19的病毒。然而,尚不清楚SARS-CoV-2是否可以调节感染细胞中的HIV再激活。这里,我们报道了慢性HIV-1感染的髓样细胞系U1暴露于两种不同的SARS-CoV-2病毒分离株(祖先和BA.5)在24小时后逆转了其潜伏状态。我们还观察到SARS-CoV-2暴露于人类原代单核细胞衍生的巨噬细胞(MDM)最初导致其极化为M1表型,随着时间的推移,它向M2转移。这种效应与在M1极化初期释放的可溶性因子有关,这些因子在U1细胞中重新激活了HIV的产生。如用TLR激动剂瑞喹莫特刺激的MDM。我们的研究表明,SARS-CoV-2诱导的全身性炎症和与巨噬细胞的相互作用可能会影响PLWH中前病毒HIV-1在骨髓细胞中的潜伏期。
    Coronavirus disease 2019 (COVID-19) might impact disease progression in people living with HIV (PLWH), including those on effective combination antiretroviral therapy (cART). These individuals often experience chronic conditions characterized by proviral latency or low-level viral replication in CD4+ memory T cells and tissue macrophages. Pro-inflammatory cytokines, such as TNF-α, IL-1β, IL-6, and IFN-γ, can reactivate provirus expression in both primary cells and cell lines. These cytokines are often elevated in individuals infected with SARS-CoV-2, the virus causing COVID-19. However, it is still unknown whether SARS-CoV-2 can modulate HIV reactivation in infected cells. Here, we report that exposure of the chronically HIV-1-infected myeloid cell line U1 to two different SARS-CoV-2 viral isolates (ancestral and BA.5) reversed its latent state after 24 h. We also observed that SARS-CoV-2 exposure of human primary monocyte-derived macrophages (MDM) initially drove their polarization towards an M1 phenotype, which shifted towards M2 over time. This effect was associated with soluble factors released during the initial M1 polarization phase that reactivated HIV production in U1 cells, like MDM stimulated with the TLR agonist resiquimod. Our study suggests that SARS-CoV-2-induced systemic inflammation and interaction with macrophages could influence proviral HIV-1 latency in myeloid cells in PLWH.
    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
    产前接触毒素会对后代的长期健康结果产生不利影响。虽然化学疗法现在是在怀孕期间治疗癌症患者的标准护理,已知某些化合物会穿过胎盘并损害胎盘组织。对胎儿的影响在很大程度上是未知的。在这里,我们检查了组织培养中新生儿脐带血单核细胞对两种化疗药物的反应,环磷酰胺和表柔比星,当直接接触这些药物时,或间接穿过胎盘滋养层双层屏障后。与直接暴露于环磷酰胺相比,暴露于从环磷酰胺暴露的滋养层屏障获得的条件培养基的脐带血单核细胞显示出核ROS水平显着增加2.4倍。间接暴露于表阿霉素暴露的滋养层屏障不仅提高了核ROS水平,而且显着增加了具有双链断裂的脐带血细胞的分数。相对于直接暴露的细胞。直接或间接暴露于环磷酰胺或表柔比星后,脐带单核细胞的凋亡或增殖标志物均不受影响。我们的数据表明,暴露于环磷酰胺或表阿霉素的滋养层细胞可能会引起间接的“旁观者”效应,并可能加重胎儿隔室的遗传毒性。
    Prenatal exposure to toxins can adversely affect long-term health outcomes of the offspring. Though chemotherapeutics are now standard of care for treating cancer patients during pregnancy, certain compounds are known to cross the placenta and harm placental tissue. The consequences for the fetus are largely unexplored. Here we examined the responses of newborn cord blood mononuclear cells in tissue culture to two chemotherapeutic drugs, cyclophosphamide and epirubicin, when either directly exposed to these drugs, or indirectly after crossing a placenta trophoblast bilayer barrier. Cord blood mononuclear cells exposed to the conditioned media obtained from cyclophosphamide-exposed trophoblast barriers showed a significant 2.4-fold increase of nuclear ROS levels compared to direct exposure to cyclophosphamide. Indirect exposure to epirubicine-exposed trophoblast barriers not only enhanced nuclear ROS levels but also significantly increased the fraction of cord blood cells with double strand breaks, relative to directly exposed cells. Neither apoptosis nor proliferation markers were affected in cord mononuclear blood cells upon direct or indirect exposure to cyclophosphamide or epirubicin. Our data suggests that trophoblast cells exposed to cyclophosphamide or epirubicine may induce an indirect \'bystander\' effect and can aggravate genotoxicity in the fetal compartment.
    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
    在因流感样疾病住院的患者中,病毒共感染占很大一部分。随着我们对病毒共感染的认识增加,研究人员已经开始通过实验检查这些感染背后的一些病毒-病毒相互作用。病毒之间相互作用的一种机制是通过先天免疫应答。这似乎主要是通过干扰素反应发生的,在附近未感染的细胞中产生抗病毒状态,一种被称为旁观者效应的现象。这里,我们建立了两个病毒通过旁观者效应相互作用的数学模型。我们发现,当细胞去除到受保护状态的速率很高时,第一种病毒的生长受到抑制,虽然第二种病毒只能进入受保护的细胞,促进其成长。相反,如果第二种病毒感染受保护细胞的能力有限,则可以完全抑制其生长。
    Viral coinfections are responsible for a significant portion of cases of patients hospitalized with influenza-like illness. As our awareness of viral coinfections has increased, researchers have started to experimentally examine some of the virus-virus interactions underlying these infections. One mechanism of interaction between viruses is through the innate immune response. This seems to occur primarily through the interferon response, which generates an antiviral state in nearby uninfected cells, a phenomenon know as the bystander effect. Here, we develop a mathematical model of two viruses interacting through the bystander effect. We find that when the rate of removal of cells to the protected state is high, growth of the first virus is suppressed, while the second virus enjoys sole access to the protected cells, enhancing its growth. Conversely, growth of the second virus can be fully suppressed if its ability to infect the protected cells is limited.
    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
    关于辐射诱导的基质因子和治疗性辐射后胞外基质的可能降解知之甚少。这项研究的目的是确定辐射是否可以在特定部位切割胶原蛋白,诱导潜在的生物活性肽对抗软骨细胞。评价作为3D模型培养的软骨细胞的细胞外基质产生。在X照射的软骨细胞的条件培养基中体外分析旁观者分子。通过质谱法分析胶原蛋白多肽中的优先断裂位点,并针对软骨细胞测试所得肽。软骨细胞的3D模型显示出能够维持结构的轻质细胞外基质。辐射和旁观者软骨细胞在低剂量下表现出令人惊讶的辐射敏感性,存在旁观者因素的特征,特别是在0.1Gy之后。观察到甘氨酸-脯氨酸肽键是辐射后胶原蛋白多肽的优先切割位点和可能的弱点。从针对软骨细胞培养物分析的46种胶原肽,在0.1至1μg/ml的最高浓度下,20种肽诱导了生存力的降低,5种肽诱导了生存力的增加。我们得出的结论是,辐照促进了胶原蛋白的位点特异性降解。潜在产生的肽诱导软骨细胞生长的负或正调节。一起来看,这些结果表明,电离辐射会导致软骨蛋白的降解,暴露后导致软骨稳态的功能失衡,导致软骨功能障碍。
    Little is known regarding radiation-induced matrikines and the possible degradation of extracellular matrix following therapeutic irradiation. The goal of this study was to determine if irradiation can cut collagen proteins at specific sites, inducing potentially biologically active peptides against cartilage cells. Chondrocytes cultured as 3D models were evaluated for extracellular matrix production. Bystander molecules were analyzed in vitro in the conditioned medium of X-irradiated chondrocytes. Preferential breakage sites were analyzed in collagen polypeptide by mass spectrometry and resulting peptides were tested against chondrocytes. 3D models of chondrocytes displayed a light extracellular matrix able to maintain the structure. Irradiated and bystander chondrocytes showed a surprising radiation sensitivity at low doses, characteristic of the presence of bystander factors, particularly following 0.1 Gy. The glycine-proline peptidic bond was observed as a preferential cleavage site and a possible weakness of the collagen polypeptide after irradiation. From the 46 collagen peptides analyzed against chondrocytes culture, 20 peptides induced a reduction of viability and 5 peptides induced an increase of viability at the highest concentration between 0.1 and 1 µg/ml. We conclude that irradiation promoted a site-specific degradation of collagen. The potentially resulting peptides induce negative or positive regulations of chondrocyte growth. Taken together, these results suggest that ionizing radiation causes a degradation of cartilage proteins, leading to a functional unbalance of cartilage homeostasis after exposure, contributing to cartilage dysfunction.
    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:大体积肿瘤的放射治疗通常会导致疗效不佳的缓解。放化疗引起的免疫抑制使预后恶化,对生存产生负面影响。新型部分肿瘤照射(PTI)旨在保留肿瘤周围免疫微环境(PIM),并与免疫活性峰同步递送。从而通过免疫刺激增强局部和远处的肿瘤控制。
    方法:目前的原则证明试验纳入了26例巨大肿瘤患者,比较在免疫活性峰和低谷施用的治疗之间的结果。主要终点是局部旁观和远端横观反应率。次要终点包括整体,无进展-,癌症特异性生存率,新辅助和免疫调节潜能。
    结果:所有测量结果均受治疗时机的显著影响。旁观者和隔壁反应率分别为77%和41%,分别。PTI显著上调促炎和诱导细胞死亡的途径,提高高度复杂肿瘤的放疗疗效。
    结论:这项研究强调了PTI对以前认为超出治疗希望的高度姑息性患者队列的深远影响。这些患者中有41%在中位随访50个月后仍然存活,PTI为那些面临先进的人提供了潜在的生命线,治疗抗性癌症。这种方法还产生了远处的免疫原性抗肿瘤反应,为晚期癌症的治疗提供了一条有希望的新途径。
    Radiotherapy for bulky tumors often results in palliation with suboptimal outcomes. The prognosis is worsened by immunosuppression caused by radio-chemotherapy, negatively impacting on survival. Novel Partial Tumor Irradiation (PTI) was designed to spare the Peritumoral Immune Microenvironment (PIM) and to be delivered synchronously with immune activity peaks, thus enhancing both local and distant tumor control through immunostimulation.
    Present proof-of-principle trial enrolled 26 patients with bulky tumors, comparing outcomes between treatments administered at immune activity peaks versus troughs. The primary endpoint was local-bystander and distal-abscopal response-rate. Secondary endpoints included overall-, progression-free-, cancer-specific survival, neoadjuvant and immunomodulatory potential.
    All measured outcomes were significantly influenced by treatment-timing. The bystander and abscopal response rates were 77% and 41%, respectively. PTI significantly upregulated pro-inflammatory and cell-death-inducing pathways improving the efficacy of radiotherapy by highly complex tumors.
    This study highlights the profound impact PTI can have on a highly palliative patient cohort previously deemed beyond therapeutic hope. With 41 % of these patients still alive after a median follow-up of 50 months, PTI offers a potential lifeline for those facing advanced, treatment-resistant cancers. This approach generated also distant immunogenic anti-tumor responses, offering a promising new avenue for the treatment of advanced 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
    盐霉素(Sal)在肿瘤治疗领域引起了广泛的关注,尤其是对肿瘤干细胞(CSCs)和耐药肿瘤细胞的抑制作用。然而,其溶解性和靶向特异性对其药物开发提出了重大挑战。Sal-A6,一种新型的肽-药物偶联物(PDC),通过使用特异性接头将靶向CSC标记CD44的肽A6与Sal连接而形成。这种共轭显着增强了Sal的物理化学性质,与Sal相比,Sal-A6表现出显著增加的抗卵巢癌活性。此外,Sal-A6,使用二硫键作为接头,表现出旁观者的杀戮效果。此外,除了增强耐药卵巢癌细胞的化学敏感性外,它还对癌症干细胞和耐药细胞具有实质性的细胞毒性作用。总之,结果表明,Sal-A6是一种衍生自Sal的新型PDC,在卵巢癌和耐药患者的治疗中具有潜在的治疗应用。此外,这一发现为以Sal为基础开发PDC型药物提供了见解.
    Salinomycin (Sal) has attracted considerable attention in the field of tumor treatment, especially for its inhibitory effect on cancer stem cells (CSCs) and drug-resistant tumor cells. However, its solubility and targeting specificity pose significant challenges to its pharmaceutical development. Sal-A6, a novel peptide-drug conjugate (PDC), was formed by linking the peptide A6 targeting the CSC marker CD44 with Sal using a specific linker. This conjugation markedly enhances the physicochemical properties of Sal and compared to Sal, Sal-A6 demonstrated a significantly increased activity against ovarian cancer. Furthermore, Sal-A6, employing a disulfide bond as a linker, exhibited bystander killing effect. Moreover, it induces substantial cytotoxic effect on both cancer stem cells and drug-resistant cells in addition to enhance chemosensitivity of resistant ovarian cancer cells. In summary, the results indicated that Sal-A6, a novel PDC derived from Sal, has potential therapeutic applications in the treatment of ovarian cancer and drug-resistant patients. Additionally, this discovery offers insights for developing PDC-type drugs using Sal as a foundation.
    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:几种HER2靶向抗体-药物偶联物(ADC)已获得市场批准用于治疗HER2表达转移。已经报道了新一代ADC对其他HER2靶向疗法反应不佳的患者的有希望的反应。然而,这些ADC仍然面临与其特定有效负载毒素相关的抗性和/或严重副作用的挑战。Eribulin,一种治疗转移性乳腺癌和脂肪肉瘤的治疗剂,是ADC有效载荷的新选择,具有独特的作用机制和安全性。
    方法:我们已经生成了一种新型的HER2标记含有eribulin的ADC,BB-1701.在体外和体内测试BB-1701对抗其中HER2表达水平在大范围内变化的癌细胞的效力。还测试了BB-1701的旁观者杀伤作用和毒素诱导的免疫原性细胞死亡(ICD)。
    结果:与具有DM1和Dxd有效载荷的靶向HER2的ADC相比,含有艾瑞布林的ADC在HER2低癌细胞系中显示出更高的体外细胞毒性。BB-1701还有效抑制对含有DM1或Dxd的ADC具有抗性的模型中的肿瘤。作用模式研究表明,BB-1701对与HER2高细胞相邻的HER2无效细胞具有显著的旁观者效应。此外,BB-1701处理诱导ICD。在非人类灵长类动物中重复剂量的BB-1701在预期的临床剂量下显示出良好的药代动力学和安全性,给药途径,和时间表。
    结论:临床前数据支持BB-1701在各种HER2表达癌症患者中的测试,包括对其他HER2靶向ADC耐药的患者。BB-1701(NCT04257110)在患者中的I期临床试验目前正在进行中。
    BACKGROUND: Several HER2-targeting antibody-drug conjugates (ADC) have gained market approval for the treatment of HER2-expressing metastasis. Promising responses have been reported with the new generation of ADCs in patients who do not respond well to other HER2-targeting therapeutics. However, these ADCs still face challenges of resistance and/or severe adverse effects associated with their particular payload toxins. Eribulin, a therapeutic agent for the treatment of metastatic breast cancer and liposarcoma, is a new choice of ADC payload with a distinct mechanism of action and safety profile.
    METHODS: We\'ve generated a novel HER2-tageting eribulin-containing ADC, BB-1701. The potency of BB-1701 was tested in vitro and in vivo against cancer cells where HER2-expressing levels vary in a large range. Bystander killing effect and toxin-induced immunogenic cell death (ICD) of BB-1701 were also tested.
    RESULTS: In comparison with HER2-targeting ADCs with DM1 and Dxd payload, eribulin-containing ADC demonstrated higher in vitro cytotoxicity in HER2-low cancer cell lines. BB-1701 also effectively suppressed tumors in models resistant to DM1 or Dxd containing ADCs. Mode of action studies showed that BB-1701 had a significant bystander effect on HER2-null cells adjacent to HER2-high cells. In addition, BB-1701 treatment induced ICD. Repeated doses of BB-1701 in nonhuman primates showed favorable pharmacokinetics and safety profiles at the intended clinical dosage, route of administration, and schedule.
    CONCLUSIONS: The preclinical data support the test of BB-1701 in patients with various HER2-expressing cancers, including those resistant to other HER2-targeting ADCs. A phase I clinical trial of BB-1701 (NCT04257110) in patients is currently underway.
    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
    几十年来积累的体外和体内观察结果已经明确表明,电离辐射的生物学效应可以从受辐射的细胞/组织传播到非靶向细胞/组织。氧化还原调节的细胞间通讯机制,包括分泌因子和缝隙连接的作用,可以调解这些非靶向效应。显然,这种效应的表达及其对子代细胞的传递对辐射防护相关问题有影响。他们的阐明也与增强癌症放射治疗的功效和减少其对正常组织毒性发展的影响有关。此外,非靶向效应的研究与我们对氧化应激条件下细胞间通讯的基本理解有关.这篇综述将追溯辐射非靶向效应的历史,从早期的远视效应报道开始,该报道描述了远离辐射暴露部位的组织中辐射诱导的效应。相关作用涉及辐照后血浆中裂解因子的产生,这可以诱导未辐照细胞的染色体损伤。尽管这些早期报告表明辐射的非靶向效应,在原子核中需要直接沉积能量的经典范式仍然占主导地位。描述辐射引起的旁观者效应的论文对这种范式提出了挑战。这篇综述将涵盖辐射引起的旁观者效应的机制以及对辐射防护和放射治疗的潜在影响。
    In vitro and in vivo observations accumulated over several decades have firmly shown that the biological effects of ionizing radiation can spread from irradiated cells/tissues to non-targeted cells/tissues. Redox-modulated intercellular communication mechanisms that include a role for secreted factors and gap junctions, can mediate these non-targeted effects. Clearly, the expression of such effects and their transmission to progeny cells has implications for issues related to radiation protection. Their elucidation is also relevant towards enhancing the efficacy of cancer radiotherapy and reducing its impact on the development of normal tissue toxicities. In addition, the study of non-targeted effects is pertinent to our basic understanding of intercellular communications under conditions of oxidative stress. This review will trace the history of non-targeted effects of radiation starting with early reports of abscopal effects which described radiation induced effects in tissues distant from the site of radiation exposure. A related effect involved the production of clastogenic factors in plasma following irradiation which can induce chromosome damage in unirradiated cells. Despite these early reports suggesting non-targeted effects of radiation, the classical paradigm that a direct deposition of energy in the nucleus was required still dominated. This paradigm was challenged by papers describing radiation induced bystander effects. This review will cover mechanisms of radiation-induced bystander effects and the potential impacts on radiation protection and radiation therapy.
    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
    MPSIIIC是由乙酰肝素-α-氨基葡萄糖苷N-乙酰转移酶(HGSNAT)突变引起的溶酶体贮积病,没有可用的治疗方法。因为HGSNAT是一种反式溶酶体膜蛋白,MPSIIIC的基因治疗需要转导尽可能多的细胞以获得最大的益处.所有细胞连续释放细胞外囊泡(EV)并通过经由EV运输交换生物分子进行通信。为了解决未满足的需求,我们开发了一种rAAV-hHGSNATEV载体,在3UTR中具有EV-mRNA包装信号,以促进旁观者效应,并在体外MPSIIIC模型中进行了测试。在人类MPSIIIC细胞中,rAAV-hHGSNATEV增强HGSNATmRNA和蛋白表达,EV-hHGSNAT-mRNA包装,并清除了GAG存储。重要的是,用EV孵育导致受体MPSIIIC细胞中hHGSNAT蛋白表达和GAG内容物清除。Further,rAAV-hHGSNATEV转导导致MPSIIIC细胞中病理性EV降低至正常水平,表明更广泛的治疗益处。这些数据表明,将EV-mRNA包装信号掺入rAAV-hHGSNAT载体可增强hHGSNAT-mRNA的EV包装,可以转运到非转导细胞并翻译成功能性rHGSNAT蛋白,促进疾病病理的交叉矫正。这项研究支持rAAVEV对MPSIIIC的治疗潜力,和广泛的疾病,不必转导每个细胞。
    MPS IIIC is a lysosomal storage disease caused by mutations in heparan-α-glucosaminide N-acetyltransferase (HGSNAT), for which no treatment is available. Because HGSNAT is a trans-lysosomal-membrane protein, gene therapy for MPS IIIC needs to transduce as many cells as possible for maximal benefits. All cells continuously release extracellular vesicles (EVs) and communicate by exchanging biomolecules via EV trafficking. To address the unmet need, we developed a rAAV-hHGSNATEV vector with an EV-mRNA-packaging signal in the 3\'UTR to facilitate bystander effects, and tested it in an in vitro MPS IIIC model. In human MPS IIIC cells, rAAV-hHGSNATEV enhanced HGSNAT mRNA and protein expression, EV-hHGSNAT-mRNA packaging, and cleared GAG storage. Importantly, incubation with EVs led to hHGSNAT protein expression and GAG contents clearance in recipient MPS IIIC cells. Further, rAAV-hHGSNATEV transduction led to the reduction of pathological EVs in MPS IIIC cells to normal levels, suggesting broader therapeutic benefits. These data demonstrate that incorporating the EV-mRNA-packaging signal into a rAAV-hHGSNAT vector enhances EV packaging of hHGSNAT-mRNA, which can be transported to non-transduced cells and translated into functional rHGSNAT protein, facilitating cross-correction of disease pathology. This study supports the therapeutic potential of rAAVEV for MPS IIIC, and broad diseases, without having to transduce every cell.
    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
    放射治疗的护理标准很多,在整个肿瘤中均匀分布的低剂量部分。正在审查的另一种策略是在一个或几个部分中应用空间分割的放射疗法(整个肿瘤的高剂量和低剂量),单独或随后进行常规辐射分馏。空间分割允许显著保留正常组织,在两种情况下,接受亚致死剂量的肿瘤或正常组织区域都会产生有益的旁观者效应。旁观者效应被广泛定义为生物反应,其明显大于基于所接收的辐射剂量的预期。通常,这些作用是通过活性氧的扩散和各种细胞因子的分泌引发的。正如文献中所证明的,空间分级分离相关的旁观者效应可以在细胞与细胞之间局部发生,在所谓的“队列效应”中,“倾向于采取脉管系统重组的形式,增强的免疫浸润,和免疫记忆的发展。其他旁观者效应可能发生在遥远的地方,这就是所谓的“abscopal效应”。“虽然这些事件很少见,它们是由免疫系统介导的,可以导致根除继发性和转移性疾病。目前,由于这些旁观者效应的复杂性和可变性,他们没有被彻底理解,但随着知识水平的提高,它们可能为改善临床结局提供重要机会.
    The standard of care for radiation therapy is numerous, low-dose fractions that are distributed homogeneously throughout the tumor. An alternative strategy under scrutiny is to apply spatially fractionated radiotherapy (high and low doses throughout the tumor) in one or several fractions, either alone or followed by conventional radiation fractionation . Spatial fractionation allows for significant sparing of normal tissue, and the regions of tumor or normal tissue that received sublethal doses can give rise to beneficial bystander effects in both cases. Bystander effects are broadly defined as biological responses that are significantly greater than would be anticipated based on the radiation dose received. Typically these effects are initiated by diffusion of reactive oxygen species and secretion of various cytokines. As demonstrated in the literature, spatial fractionation related bystander effects can occur locally from cell to cell and in what are known as \"cohort effects,\" which tend to take the form of restructuring of the vasculature, enhanced immune infiltration, and development of immunological memory. Other bystander effects can take place at distant sites in what are known as \"abscopal effects.\" While these events are rare, they are mediated by the immune system and can result in the eradication of secondary and metastatic disease. Currently, due to the complexity and variability of these bystander effects, they are not thoroughly understood, but as knowledge improves they may present significant opportunities for improved clinical outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号