Graft vs Leukemia Effect

移植物对白血病的影响
  • 文章类型: Journal Article
    背景:在急性髓系白血病(AML)的异基因造血干细胞移植中,供体T细胞通过移植物抗白血病(GVL)效应对抗白血病,同时,它们还存在通过与受体细胞相互作用引发危及生命的移植物抗宿主病(GVHD)的风险。GVHD的发作取决于供体T细胞和受体抗原呈递细胞(APC)之间的相互作用,激发T细胞激活。然而,缺乏平衡GVHD和GVL的有效方法。
    方法:在我们的研究中,我们通过将聚阳离子胺化明胶和聚阴离子藻酸盐层叠到T细胞表面来制作纳米胶囊,检查其基本生理功能的潜在变化。随后,我们建立了AML小鼠模型,并用骨髓细胞(BMC)联合包裹T细胞移植治疗,以研究包裹T细胞的GVL和抗GVHD作用.体外共培养被用来探索封装对免疫突触的影响,共刺激分子,和肿瘤杀伤途径。
    结果:将BMC与选择性包裹在AML小鼠体内的供体T细胞联合移植可显着减轻GVHD症状,同时保留基本的GVL效应。封装的T细胞通过阻碍与受体APC的免疫突触的形成来发挥免疫调节作用,从而下调共刺激信号,如CD28-CD80、ICOS-ICOSL、和CD40L-CD40。接受包囊T细胞移植的小鼠供体Ly-5.1-BMC细胞数量显着增加,伴随着穿孔素和颗粒酶B的体内表达水平不变。虽然在单细胞纳米封装后在体外观察到供体T细胞在肿瘤微环境中的细胞毒性的瞬时抑制,随后恢复正常的抗肿瘤活性,归因于包封的囊泡壳的选择性渗透性和材料降解。此外,在白血病肿瘤细胞中仍观察到凋亡蛋白和FAS-FAS配体通路在正常水平的表达。
    结论:封装的供体T细胞通过早期免疫分离使与APC的共刺激信号最小化,从而有效缓解GVHD,同时保留GVL效应。随后降解纳米胶囊恢复T细胞对AML细胞的细胞毒性功效,由细胞毒性途径介导。使用移植封装的T细胞提供了抑制GVHD同时保持GVL效应的有希望的策略。
    BACKGROUND: In allogeneic-hematopoietic stem cell transplantation for acute myeloid leukemia (AML), donor T cells combat leukemia through the graft-versus-leukemia (GVL) effect, while they also pose a risk of triggering life-threatening graft-versus-host disease (GVHD) by interacting with recipient cells. The onset of GVHD hinges on the interplay between donor T cells and recipient antigen-presenting cells (APCs), sparking T-cell activation. However, effective methods to balance GVHD and GVL are lacking.
    METHODS: In our study, we crafted nanocapsules by layering polycationic aminated gelatin and polyanionic alginate onto the surface of T cells, examining potential alterations in their fundamental physiological functions. Subsequently, we established an AML mouse model and treated it with transplantation of bone marrow cells (BMCs) combined with encapsulated T cells to investigate the GVL and anti-GVHD effects of encapsulated T cells. In vitro co-culture was employed to probe the effects of encapsulation on immune synapses, co-stimulatory molecules, and tumor-killing pathways.
    RESULTS: Transplantation of BMCs combined with donor T cells selectively encapsulated onto AML mice significantly alleviates GVHD symptoms while preserving essential GVL effects. Encapsulated T cells exerted their immunomodulatory effects by impeding the formation of immune synapses with recipient APCs, thereby downregulating co-stimulatory signals such as CD28-CD80, ICOS-ICOSL, and CD40L-CD40. Recipient mice receiving encapsulated T-cell transplantation exhibited a marked increase in donor Ly-5.1-BMC cell numbers, accompanied by unaltered in vivo expression levels of perforin and granzyme B. While transient inhibition of donor T-cell cytotoxicity in the tumor microenvironment was observed in vitro following single-cell nanoencapsulation, subsequent restoration to normal antitumor activity ensued, attributed to selective permeability of encapsulated vesicle shells and material degradation. Moreover, the expression of apoptotic proteins and FAS-FAS ligand pathway at normal levels was still observed in leukemia tumor cells.
    CONCLUSIONS: Encapsulated donor T cells effectively mitigate GVHD while preserving the GVL effect by minimizing co-stimulatory signaling with APCs through early immune isolation. Subsequent degradation of nanocapsules restores T-cell cytotoxic efficacy against AML cells, mediated by cytotoxic pathways. Using transplant-encapsulated T cells offers a promising strategy to suppress GVHD while preserving the GVL effect.
    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
    异基因造血细胞移植(alloHCT)为血液恶性肿瘤提供了潜在的治愈性治疗。治疗性移植物抗白血病(GvL)效应由供体T细胞攻击患者造血(恶性)细胞诱导。然而,如果健康的非造血组织成为目标,移植物抗疾病(GvHD)可能发展。HLA匹配的alloHCT后,GvL和GvHD由识别患者细胞上HLA呈递的多态肽的供体T细胞诱导,所谓的次要组织相容性抗原(MiHA)。GvL和GvHD之间的平衡取决于MiHA的组织分布和靶向这些MiHA的T细胞频率。针对广泛表达的MiHAs的T细胞诱导GvL和GvHD,而那些靶向具有造血限制表达的MiHA的诱导GvL而没有GvHD。最近,alloHCT后在天然免疫反应中鉴定的MiHA库扩展到159个HLA-I限制性的总MiHA,包括14个限制造血的MiHAs.这篇综述探讨了它们与预测的潜在相关性,监视器,并操纵GvL和GvHD以改善HLA匹配的alloHCT后的临床结果。
    Allogeneic hematopoietic cell transplantation (alloHCT) provides a potential curative treatment for haematological malignancies. The therapeutic Graft-versus-Leukaemia (GvL) effect is induced by donor T cells attacking patient hematopoietic (malignant) cells. However, if healthy non-hematopoietic tissues are targeted, Graft-versus-Disease (GvHD) may develop. After HLA-matched alloHCT, GvL and GvHD are induced by donor T cells recognizing polymorphic peptides presented by HLA on patient cells, so-called minor histocompatibility antigens (MiHAs). The balance between GvL and GvHD depends on the tissue distribution of MiHAs and T-cell frequencies targeting these MiHAs. T cells against broadly expressed MiHAs induce GvL and GvHD, whereas those targeting MiHAs with hematopoietic-restricted expression induce GvL without GvHD. Recently, the MiHA repertoire identified in natural immune responses after alloHCT was expanded to 159 total HLA-I-restricted MiHAs, including 14 hematopoietic-restricted MiHAs. This review explores their potential relevance to predict, monitor, and manipulate GvL and GvHD for improving clinical outcome after HLA-matched alloHCT.
    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:急性移植物抗宿主病(aGVHD),主要由同种异体T细胞介导,是异基因造血干细胞移植(allo-HCT)成功的决定性因素。临床患者对aGVHD的预防不能令人满意,对新方法仍有巨大的未满足需求。淫羊藿苷(ICA)显示出有效的抗炎活性并抑制T细胞介导的免疫反应。因此,ICA是预防aGVHD的潜在药物。然而,没有数据评估allo-HCT后ICA对aGVHD的影响。
    目的:本研究旨在探讨ICA对aGVHD的保护作用及其机制。此外,评估了ICA对移植物抗白血病(GVL)效应和供体造血和免疫细胞植入的影响.
    方法:开发了不同的小鼠allo-HCT模型来研究ICA对GVHD和GVL效应的影响。流式细胞术用于分析白血病细胞的生长,不同免疫细胞的改变,和凋亡。使用CCK-8测定法测定细胞增殖。进行RNA测序和定量蛋白质组学分析以阐明潜在的机制,通过聚合酶链反应或功能实验进一步验证。
    结果:不同浓度的ICA表现出相反的作用:低浓度的ICA促进,高浓度抑制T细胞的增殖和功能。在allo-HCT后3至5天的高剂量ICA给药可以缓解鼠aGVHD,但不影响慢性GVHD(cGVHD)的进程,GVL对急性髓系和淋巴细胞白血病的作用,或供体血液和免疫细胞的恢复。ICA广泛压制扩张,函数,和供体同种反应性T细胞的浸润,同时保留调节性T细胞(Tregs)和骨髓来源的抑制细胞(MDSC)。定量蛋白质组学分析表明,整合素连接激酶(ILK)和淋巴细胞胞浆蛋白2(LCP2)表达的下调可能与ICA介导的aGVHD保护作用有关。此外,ILK的抑制剂,这可以减轻小鼠aGVHD在allo-HCT后早期给药。
    结论:这些发现表明ICA的生物活性与其浓度相关,并且ICA可以有效缓解aGVHD,而不会失去GVL活性或供体造血和免疫细胞的植入。因此,ICA可能是临床上预防aGVHD的有前途的药物。
    BACKGROUND: Acute graft-versus-host disease (aGVHD), which is mainly mediated by allogeneic T cells, is a decisive factor in the success of allogeneic hematopoietic stem cell transplantation (allo-HCT). Prophylaxis for aGVHD in clinical patients is unsatisfactory, and there is still a huge unmet need for novel approaches. Icariin (ICA) shows potent anti-inflammatory activity and suppresses T cell-mediated immune responses. Thus, ICA is a potential drug for the prevention of aGVHD. However, there is no data assessing the impact of ICA on aGVHD after allo-HCT.
    OBJECTIVE: This study aimed to investigate the protective effect of ICA against aGVHD and its mechanisms. Moreover, the impact of ICA on the graft-versus-leukemia (GVL) effect and engraftment of donor hematopoietic and immune cells were assessed.
    METHODS: Different murine models of allo-HCT were developed to study the influence of the ICA on GVHD and GVL effect. Flow cytometry was used to analyze the growth of leukemia cells, alterations in different immune cells, and apoptosis. Cell proliferation was determined using a CCK-8 assay. RNA sequencing and quantitative proteomic analysis were performed to elucidate the underlying mechanisms, which were further verified by polymerase chain reaction or functional experiments.
    RESULTS: Different concentrations of ICA exhibited opposite effects: low-concentration ICA promoted, while high concentrations suppressed the proliferation and function of T cells. A high dose of ICA administration during days +3 to +5 post-allo-HCT can alleviate murine aGVHD but does not affect the course of chronic GVHD (cGVHD), the GVL effect against both acute myeloid and lymphoblastic leukemia, or the recovery of donor hematological and immune cells. ICA extensively represses the expansion, function, and infiltration of donor alloreactive T cells, while preserving regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSC). Quantitative proteomic analysis showed that downregulation of integrin-linked kinase (ILK) and lymphocyte cytosolic protein 2 (LCP2) expression was possibly associated with ICA-mediated aGVHD protective effects. Furthermore, an inhibitor of ILK, which can alleviate murine aGVHD administered early after allo-HCT.
    CONCLUSIONS: These findings suggest that the bioactivities of ICA are associated with its concentration and that ICA can effectively mitigate aGVHD without losing GVL activity or engraftment of donor hematopoietic and immune cells. Thus, ICA may be a promising drug for preventing aGVHD in clinical settings.
    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: Clinical Trial, Phase II
    背景:在患有血液系统恶性肿瘤的儿科患者移植后复发仍然是一个挑战。用于疾病控制的清髓治疗方案与急性和长期不良反应相关。我们使用CD45RA耗尽的单倍体移植物过继转移记忆T细胞并结合NK细胞回输,并假设最大化移植物抗白血病(GVL)效应可能会降低预处理方案的强度。
    方法:在本II期临床试验(NCT01807611)中,72例血液恶性肿瘤患者(完全缓解(CR)1:25,≥CR2:28,难治性疾病:19)接受了富含CD34的单倍体和CD45RA的造血祖细胞移植物,然后进行NK细胞输注。调理包括氟达拉滨,Thiotepa,melphalan,环磷酰胺,总淋巴照射,移植物抗宿主病(GVHD)的预防包括短程西罗莫司或霉酚酸酯,无需血清疗法。
    结果:CR1患者的3年总生存率(OS)和无事件生存率(EFS)分别为92%(95%CI:72-98)和88%(95%CI:67-96);≥CR2为81%(95%CI:61-92)和68%(95%CI:47-82),难治性疾病为32%(95%CI:54-6)。所有患者形态CR的3年EFS为77%(95%CI:64-87),在有或没有微小残留病的接受者之间没有差异(P=0.2992)。免疫重建很快,在第30天,平均CD3和CD4T细胞计数为410/μL和140/μL。急性GVHD和慢性GVHD的累积发生率分别为36%和26%,但大多数急性GVHD患者通过治疗迅速恢复。NK细胞同种反应性供体观察到III-IV级急性GVHD的发生率较低(P=0.004),母体供者的中度/重度慢性GVHD发生率更高(P=0.035)。
    结论:CD45RA耗尽的移植物和NK细胞回补的组合导致了强大的免疫重建,最大限度地提高了GVL效应,并允许使用清髓性下,与优秀的EFS相关的无TBI预处理方案在该高危人群中产生有希望的长期结果。该试验在ClinicalTrials.gov(NCT01807611)注册。
    BACKGROUND: Relapse remains a challenge after transplantation in pediatric patients with hematological malignancies. Myeloablative regimens used for disease control are associated with acute and long-term adverse effects. We used a CD45RA-depleted haploidentical graft for adoptive transfer of memory T cells combined with NK-cell addback and hypothesized that maximizing the graft-versus-leukemia (GVL) effect might allow for reduction in intensity of conditioning regimen.
    METHODS: In this phase II clinical trial (NCT01807611), 72 patients with hematological malignancies (complete remission (CR)1: 25, ≥ CR2: 28, refractory disease: 19) received haploidentical CD34 + enriched and CD45RA-depleted hematopoietic progenitor cell grafts followed by NK-cell infusion. Conditioning included fludarabine, thiotepa, melphalan, cyclophosphamide, total lymphoid irradiation, and graft-versus-host disease (GVHD) prophylaxis consisted of a short-course sirolimus or mycophenolate mofetil without serotherapy.
    RESULTS: The 3-year overall survival (OS) and event-free-survival (EFS) for patients in CR1 were 92% (95% CI:72-98) and 88% (95% CI: 67-96); ≥ CR2 were 81% (95% CI: 61-92) and 68% (95% CI: 47-82) and refractory disease were 32% (95% CI: 11-54) and 20% (95% CI: 6-40). The 3-year EFS for all patients in morphological CR was 77% (95% CI: 64-87) with no difference amongst recipients with or without minimal residual disease (P = 0.2992). Immune reconstitution was rapid, with mean CD3 and CD4 T-cell counts of 410/μL and 140/μL at day + 30. Cumulative incidence of acute GVHD and chronic GVHD was 36% and 26% but most patients with acute GVHD recovered rapidly with therapy. Lower rates of grade III-IV acute GVHD were observed with NK-cell alloreactive donors (P = 0.004), and higher rates of moderate/severe chronic GVHD occurred with maternal donors (P = 0.035).
    CONCLUSIONS: The combination of a CD45RA-depleted graft and NK-cell addback led to robust immune reconstitution maximizing the GVL effect and allowed for use of a submyeloablative, TBI-free conditioning regimen that was associated with excellent EFS resulting in promising long-term outcomes in this high-risk population. The trial is registered at ClinicalTrials.gov (NCT01807611).
    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
    白血病复发是异基因造血细胞移植(allo-HCT)后死亡的主要原因。我们测试了靶向TIM-3改善移植物抗白血病(GVL)作用的潜力。当造血干细胞过表达某些致癌驱动突变时,我们观察到TIM-3配体的差异表达。抗TIM-3Ab治疗改善了具有癌基因诱导的TIM-3配体表达的白血病小鼠的存活率。相反,具有低配体表达的白血病细胞具有抗TIM-3治疗抗性。体外,CD8+T细胞(Tc)中的TIM-3阻断或遗传缺失增强了Tc的激活,增殖和IFN-γ产生,同时增强GVL效应,预防Tc耗尽,改善Tc细胞毒性和体内糖酵解。相反,骨髓细胞中的TIM-3缺失在体外不影响同种异体Tc的增殖和活化,提示抗TIM-3治疗介导的GVL效应是Tc诱导的。与抗PD-1和抗CTLA-4治疗相反,抗TIM-3治疗未增强急性移植物抗宿主病(aGVHD).TIM-3及其配体经常在同种异体HCT复发患者的急性髓细胞性白血病(AML)细胞中表达。我们破译了在AML和TIM-3配体表达中发现的致癌突变之间的联系,并确定了抗TIM-3治疗作为通过代谢和转录Tc重编程增强GVL效应的策略。不会加剧aGVHD。我们的发现支持在allo-HCT后AML复发患者中进行抗TIM-3Abs的临床试验。
    Leukemia relapse is a major cause of death after allogeneic hematopoietic cell transplantation (allo-HCT). We tested the potential of targeting T cell (Tc) immunoglobulin and mucin-containing molecule 3 (TIM-3) for improving graft-versus-leukemia (GVL) effects. We observed differential expression of TIM-3 ligands when hematopoietic stem cells overexpressed certain oncogenic-driver mutations. Anti-TIM-3 Ab treatment improved survival of mice bearing leukemia with oncogene-induced TIM-3 ligand expression. Conversely, leukemia cells with low ligand expression were anti-TIM-3 treatment resistant. In vitro, TIM-3 blockade or genetic deletion in CD8+ Tc enhanced Tc activation, proliferation, and IFN-γ production while enhancing GVL effects, preventing Tc exhaustion, and improving Tc cytotoxicity and glycolysis in vivo. Conversely, TIM-3 deletion in myeloid cells did not affect allogeneic Tc proliferation and activation in vitro, suggesting that anti-TIM-3 treatment-mediated GVL effects are Tc induced. In contrast to anti-programmed cell death protein 1 (anti-PD-1) and anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA-4) treatment, anti-TIM-3-treatment did not enhance acute graft-versus-host disease (aGVHD). TIM-3 and its ligands were frequently expressed in acute myeloid leukemia (AML) cells of patients with post-allo-HCT relapse. We decipher the connections between oncogenic mutations found in AML and TIM-3 ligand expression and identify anti-TIM-3 treatment as a strategy for enhancing GVL effects via metabolic and transcriptional Tc reprogramming without exacerbation of aGVHD. Our findings support clinical testing of anti-TIM-3 Ab in patients with AML relapse after allo-HCT.
    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
    异基因造血细胞移植是治疗恶性血液病的有效方法,但是移植物抗宿主病(GVHD)等并发症会限制其获益.移植前的调理方案,包括化疗或放疗,可以引发内质网应激。IRE-1α是一种主要的内质网应激介质,可以进一步激活剪接的XBP-1(XBP-1s)和调节的IRE-1依赖性衰变(RIDD)。IRE-1α-XBP-1s信号传导控制树突状细胞(DC)分化和Ag呈递,在GVHD进展中至关重要。在这项研究中,我们使用DC特异性XBP-1缺陷小鼠作为供体或受体,观察到XBP-1s对GVHD诱导中的宿主DC至关重要,但对移植物抗白血病反应不重要.为了特异性靶向宿主中的IRE-1α,我们在骨髓移植前用IRE-1α抑制剂B-I09处理受体小鼠3天,显着抑制GVHD的发展,同时保持移植物抗白血病的作用。XBP-1缺陷或BI09治疗的接受者在照射和骨髓移植后显示DC存活率降低。抑制IRE-1α也导致DC同种反应性降低,随后降低同种异体T细胞的增殖和活化。随着使用RIDD缺陷型DCs的进一步研究,我们观察到RIDD也是最佳DC激活所必需的.一起来看,XBP-1s和RIDD均促进宿主DC存活和同种异体反应性,从而促进GVHD发展。
    Allogeneic hematopoietic cell transplantation is an effective treatment for hematologic malignancies, but the complications such as graft-versus-host disease (GVHD) can limit its benefit. The conditioning regimens before transplant, including chemotherapy or irradiation, can trigger endoplasmic reticulum stress. IRE-1α is a major endoplasmic reticulum stress mediator that can further activate both spliced XBP-1 (XBP-1s) and regulated IRE-1-dependent decay (RIDD). IRE-1α-XBP-1s signaling controls dendritic cell (DC) differentiation and Ag presentation, crucial in GVHD progression. In this study, we used DC-specific XBP-1-deficient mice as donors or recipients and observed that XBP-1s was crucial for host DCs in the induction of GVHD but dispensable for the graft-versus-leukemia response. To specifically target IRE-1α in the host, we treated recipient mice with the IRE-1α inhibitor B-I09 for 3 d prior to bone marrow transplantation, which significantly suppressed GVHD development while maintaining the graft-versus-leukemia effect. XBP-1-deficient or BI09-treated recipients showed reduced DC survival after irradiation and bone marrow transplantation. Inhibition of IRE-1α also led to a reduction in DC alloreactivity, subsequently decreasing the proliferation and activation of allogeneic T cells. With further study using RIDD-deficient DCs, we observed that RIDD was also required for optimal DC activation. Taken together, XBP-1s and RIDD both promote host DC survival and alloreactivity that contribute to GVHD development.
    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
    异基因造血细胞移植(HCT)在过去的几十年中通过增强的支持性治疗而发生了变化,降低强度调节(RIC),改善人类白细胞抗原(HLA)分型,和新型移植物抗宿主病(GVHD)的预防和治疗策略。最值得注意的是,移植后环磷酰胺(PTCy)的实施显著提高了这种救命疗法的安全性和可用性.鉴于这些进展降低了非复发死亡率(NRM),HCT社区更加重视开发减少复发的方法-复发是HCT后死亡的主要原因.使用RICHCT时,防止复发主要依赖于移植物抗白血病(GVL)反应.供者淋巴细胞输注(DLI),过继细胞疗法,检查点抑制,和HCT后维持策略代表了正在研究的旨在增强或协同HCT的GVL效应的方法。优化供体选择算法以利用GVL代表另一个活跃的研究领域。这些策略中的许多都试图利用T细胞的作用,几十年来,人们认为这是GVL的主要介质,也是减少复发的研究重点。然而,人们对利用自然杀伤(NK)细胞产生有效抗肿瘤作用的能力越来越感兴趣。基于NK细胞的方法优于T细胞介导的方法的潜在优势是除了复发之外还降低NRM的潜力。通过减少感染,在不增加GVHD风险的情况下,NK细胞可以减轻NRM,同时通过识别和清除癌细胞仍然可以减少复发。大多数以T细胞为重点的复发预防策略必须权衡减少复发的益处与GVHD引起的NRM风险增加。相比之下,NK细胞有可能减少两者,可能会使天平显着有利于生存。这里,我们将回顾NK细胞在GVL中的作用,优化NK细胞匹配或错配,以及NK细胞疗法的新兴研究领域,例如过继转移和嵌合抗原受体(CAR)NK细胞。
    Allogeneic hematopoietic cell transplantation (HCT) has transformed over the past several decades through enhanced supportive care, reduced intensity conditioning (RIC), improved human leukocyte antigen (HLA) typing, and novel graft-versus-host disease (GVHD)-prevention and treatment strategies. Most notably, the implementation of post-transplantation cyclophosphamide (PTCy) has dramatically increased the safety and availability of this life-saving therapy. Given reductions in nonrelapse mortality (NRM) with these advances, the HCT community has placed even greater emphasis on developing ways to reduce relapse - the leading cause of death after HCT. When using RIC HCT, protection from relapse relies predominantly on graft-versus-leukemia (GVL) reactions. Donor lymphocyte infusion (DLI), adoptive cellular therapy, checkpoint inhibition, and post-HCT maintenance strategies represent approaches under study that aim to augment or synergize with the GVL effects of HCT. Optimizing donor selection algorithms to leverage GVL represents another active area of research. Many of these strategies seek to harness the effects of T cells, which for decades were felt to be the primary mediators of GVL and the focus of investigation in relapse reduction. However, there is growing interest in capitalizing on the ability of natural killer (NK) cells to yield potent anti-tumor effects. A potential advantage of NK cell-based approaches over T cell-mediated is the potential to reduce NRM in addition to relapse. By decreasing infection, without increasing the risk of GVHD, NK cells may mitigate NRM, while still yielding relapse reduction through identification and clearance of cancer cells. Most T cell-focused relapse-prevention strategies must weigh the benefits of relapse reduction against the increased risk of NRM from GVHD. In contrast, NK cells have the potential to reduce both, potentially tipping the scales significantly in favor of survival. Here, we will review the role of NK cells in GVL, optimization of NK cell match or mismatch, and burgeoning areas of research in NK cell therapy such as adoptive transfer and chimeric antigen receptor (CAR) NK cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    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
    异基因造血干细胞移植(allo-HSCT)是许多血液系统恶性肿瘤的唯一治愈疗法,移植物抗白血病(GVL)效应在控制复发中起关键作用。然而,移植物抗宿主病(GVHD)阻碍了GVL的成功,供体T细胞攻击受体中的健康组织。天然调节性T细胞(Treg)抑制免疫应答的能力已被用作针对GVHD的治疗选择。尽管如此,评估Treg抑制GVHD的能力是否不损害GVL的益处是至关重要的。动物模型的初步研究表明,Treg可以减弱GVHD,同时保留GVL,但结果因肿瘤类型而异。使用Treg作为GVHD预防或治疗的人体试验显示了有希望的结果,强调输注时机和Treg/Tcon比率的重要性。在这次审查中,我们讨论了可用于增强Treg输注后GVL的策略,以及对过继性Treg转移维持GVL效应的拟议机制。为了优化allo-HSCT中Treg给药的治疗结果,未来的工作应集中在纯化用于输注的Treg来源,并评估其对介导GVHD的抗原的特异性,同时保留GVL应答.
    Allogeneic Hematopoietic Stem Cell Transplantation (allo-HSCT) is the only curative therapy for many hematologic malignancies, whereby the Graft-versus-Leukemia (GVL) effect plays a pivotal role in controlling relapse. However, the success of GVL is hindered by Graft-versus-Host Disease (GVHD), where donor T cells attack healthy tissues in the recipient. The ability of natural regulatory T cells (Treg) to suppress immune responses has been exploited as a therapeutical option against GVHD. Still, it is crucial to evaluate if the ability of Treg to suppress GVHD does not compromise the benefits of GVL. Initial studies in animal models suggest that Treg can attenuate GVHD while preserving GVL, but results vary according to tumor type. Human trials using Treg as GVHD prophylaxis or treatment show promising results, emphasizing the importance of infusion timing and Treg/Tcon ratios. In this review, we discuss strategies that can be used aiming to enhance GVL post-Treg infusion and the proposed mechanisms for the maintenance of the GVL effect upon the adoptive Treg transfer. In order to optimize the therapeutic outcomes of Treg administration in allo-HSCT, future efforts should focus on refining Treg sources for infusion and evaluating their specificity for antigens mediating GVHD while preserving GVL 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: English Abstract
    造血细胞移植(HCT)被认为是血液恶性肿瘤的治愈性治疗方法。然而,HCT接受者经常面临并发症,例如移植物抗宿主病(GVHD)和疾病复发。年龄和HLA差异等临床因素被认为是GVHD的风险。值得注意的是,性别不匹配的HCT,特别是女性捐赠者和男性接受者(F→M),据报道会增加慢性GVHD的风险。F→MHCT的这种不利影响被认为是由针对男性受体Y染色体上编码的次要组织相容性抗原(HY抗原)的同种异体免疫反应引起的。的确,在F→MHCT后三个月检测到针对HY抗原(HY-Ab)的抗体,HY-Abs的累积数量与慢性GVHD和非复发死亡率的风险增加显著相关.这篇综述的重点是F→MHCT,阐明其在几种临床环境中的影响,并提供其同种异体反应的临床证据,包括GVHD和移植物抗白血病(GVL)效应。此外,将讨论减轻F→MHCT不良反应的潜在临床选择。需要进一步的研究来改善临床结果并了解F→MHCT后的同种异体免疫重建。
    Hematopoietic cell transplantation (HCT) is considered a curative treatment for hematological malignancies. However, HCT recipients often face complications such as graft-versus-host disease (GVHD) and disease relapse. Clinical factors like age and HLA disparity are recognized as risks for GVHD. Notably, sex-mismatched HCT, particularly with female donors and male recipients (F→M), is reported to increase the risk of chronic GVHD. This adverse effect of F→M HCT is thought to result from allogeneic immune response against minor histocompatibility antigens encoded on the Y-chromosome of a male recipient (HY-antigens). Indeed, antibodies against HY-antigens (HY-Abs) were detected three months after F→M HCT, and the cumulative number of HY-Abs was significantly associated with increased risks of chronic GVHD and non-relapse mortality. This review focuses on F→M HCT, shedding light on its impact in several clinical settings and presenting clinical evidence of its allogeneic response, encompassing GVHD and graft-versus-leukemia (GVL) effects. Additionally, potential clinical options to mitigate adverse effects in F→M HCT will be discussed. Further investigation is required to improve clinical outcomes and understand allogenic immunological reconstitution after F→M HCT.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号