hematological malignancies

血液恶性肿瘤
  • 文章类型: Journal Article
    Amdizalisib,也被称为HMPL-689,一种新型的选择性和有效的PI3Kδ抑制剂,目前在中国正在进行II期临床开发,用于治疗血液系统恶性肿瘤。在体外和体内广泛表征了amdizalisib的临床前药代动力学(PK),以支持amdizalisib的进一步发展。我们表征了血浆蛋白结合,血-血浆分配比,细胞通透性,肝微粒体代谢稳定性,和药物-药物相互作用潜力的amdizalisib使用体外实验。在小鼠中进行体内PK评估,老鼠,狗,和猴子在单次静脉内或口服给予amdizalisib后。在大鼠中评估了氨扎利布的组织分布和排泄。临床前物种(小鼠,老鼠,狗,和猴子)用于使用异速缩放(AS)方法进行人类PK投影。Amdizalisib吸收良好,在小鼠中显示出低至中等的清除率,老鼠,狗,还有猴子.它具有高细胞通透性,没有P-糖蛋白(P-gp)或乳腺癌耐药蛋白(BCRP)底物。amdizalisib的血浆蛋白结合高(约90%)。它分布广泛,但在大鼠中的脑与血浆暴露率低。Amdizalisib在体内广泛代谢,粪便中原型药物的回收率较低。Amdizalisib和/或其代谢物主要通过大鼠的胆汁和尿液排泄。Amdizalisib对P-gp显示抑制潜力,但对BCRP没有抑制,观察到抑制CYP2C8和CYP2C9,IC50值为30.4和10.7μM。分别。它对CYP1A2,CYP2B6,CYP3A4和CYP2C9表现出诱导电位。这些ADME研究的临床前数据证明了安迪扎利布的良好药代动力学特征,这有望支持amdizalisib作为一种有前途的抗癌药物的未来临床开发。
    Amdizalisib, also named HMPL-689, a novel selective and potent PI3Kδ inhibitor, is currently under Phase II clinical development in China for treating hematological malignancies. The preclinical pharmacokinetics (PK) of amdizalisib were extensively characterized in vitro and in vivo to support the further development of amdizalisib. We characterized the plasma protein binding, blood-to-plasma partition ratio, cell permeability, hepatic microsomal metabolic stability, and drug-drug interaction potential of amdizalisib using in vitro experiments. In vivo PK assessment was undertaken in mice, rats, dogs, and monkeys following a single intravenous or oral administration of amdizalisib. The tissue distribution and excretion of amdizalisib were evaluated in rats. The PK parameters (CL and Vss) of amdizalisib in preclinical species (mice, rats, dogs, and monkeys) were utilized for the human PK projection using the allometric scaling (AS) approach. Amdizalisib was well absorbed and showed low-to-moderate clearance in mice, rats, dogs, and monkeys. It had high cell permeability without P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP) substrate liability. Plasma protein binding of amdizalisib was high (approximately 90%). It was extensively distributed but with a low brain-to-plasma exposure ratio in rats. Amdizalisib was extensively metabolized in vivo, and the recovery rate of the prototype drug was low in the excreta. Amdizalisib and/or its metabolites were primarily excreted via the bile and urine in rats. Amdizalisib showed inhibition potential on P-gp but not on BCRP and was observed to inhibit CYP2C8 and CYP2C9 with IC50 values of 30.4 and 10.7 μM, respectively. It exhibited induction potential on CYP1A2, CYP2B6, CYP3A4, and CYP2C9. The preclinical data from these ADME studies demonstrate a favorable pharmacokinetic profile for amdizalisib, which is expected to support the future clinical development of amdizalisib as a promising anti-cancer agent.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    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
    血液系统恶性肿瘤(HM)患者患严重冠状病毒病2019(COVID-19)的风险很高,也在Omicron时期。
    回顾性单中心研究,包括2022年1月至2023年3月患有严重急性呼吸道综合症冠状病毒2(SARS-CoV2)感染的HM患者。研究结果为呼吸衰竭(RF),机械通气(MV),和COVID相关死亡率,根据SARS-CoV2血清学比较患者。
    请注意,包括112例患者:39%的SARS-CoV2血清学阴性。血清学年龄较大(71.5vs.65.0年,p=0.04),更常见的是淋巴样肿瘤(88.6%vs.69.1%,p=0.02),接受抗CD20治疗(50.0%vs.30.9%p=0.04),患有严重疾病的频率更高(23.0%vs.3.0%,p=0.02)比血清阳性。Kaplan-Meier显示血清阴性患者的RF风险较高(p=0.014),MV(p=0.044),和COVID相关死亡率(p=0.021)。SARS-CoV2血清状态阴性导致RF的危险因素(危险比[HR]2.19,95%置信区间[CI]1.03-4.67,p=0.04),MV(HR3.37,95%CI1.06-10.68,p=0.04),和COVID相关死亡率(HR4.26,95%CI1.09-16.71,p=0.04)。
    :SARS-CoV2血清学阴性的HM患者,尽管接种了疫苗和以前的感染,与Omicron时代的血清反应阳性的患者相比,临床结果更差。使用血清学诊断SARS-CoV2可能是识别容易发生并发症的患者的简单工具。
    UNASSIGNED: Patients with hematological malignancies (HM) have a high risk of severe coronavirus disease 2019 (COVID-19), also in the Omicron period.
    UNASSIGNED: Retrospective single-center study including HM patients with severe acute respiratory syndrome Coronavirus 2 (SARS-CoV2) infection from January 2022 to March 2023. Study outcomes were respiratory failure (RF), mechanical ventilation (MV), and COVID-related mortality, comparing patients according to SARS-CoV2 serology.
    UNASSIGNED: Note that, 112 patients were included: 39% had negative SARS-CoV2 serology. Seronegative were older (71.5 vs. 65.0 years, p = 0.04), had more often a lymphoid neoplasm (88.6% vs. 69.1%, p = 0.02), underwent anti-CD20 therapy (50.0% vs. 30.9% p = 0.04) and had more frequently a severe disease (23.0% vs. 3.0%, p = 0.02) than seropositive.Kaplan-Meier showed a higher risk for seronegative patients for RF (p = 0.014), MV (p = 0.044), and COVID-related mortality (p = 0.021). Negative SARS-CoV2 serostatus resulted in a risk factor for RF (hazards ratio [HR] 2.19, 95% confidence interval [CI] 1.03-4.67, p = 0.04), MV (HR 3.37, 95% CI 1.06-10.68, p = 0.04), and COVID-related mortality (HR 4.26, 95% CI 1.09-16.71, p = 0.04).
    UNASSIGNED: : HM patients with negative SARS-CoV2 serology, despite vaccinations and previous infections, have worse clinical outcomes compared to seropositive patients in the Omicron era. The use of serology for SARS-CoV2 diagnosis could be an easy tool to identify patients prone to developing complications.
    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
    恶性血液病的免疫治疗是近年来发展迅速的领域,主要包括嵌合抗原受体T细胞(CAR-T)疗法,免疫检查点抑制剂,和其他方式。然而,其临床疗效仍然有限,和耐药性构成了重大挑战。因此,需要确定新的免疫治疗靶点和药物。最近,N6-甲基腺苷(m6A),最普遍的RNA表位修饰,已经成为各种恶性肿瘤的关键因素。据报道,m6A突变影响血液系统恶性肿瘤的免疫微环境,导致免疫逃避和损害血液系统恶性肿瘤的抗肿瘤免疫反应。在这次审查中,我们全面总结了目前确定的m6A修饰在各种血液恶性肿瘤中的作用,特别关注它们对免疫微环境的影响。此外,我们概述了在开发用于血液肿瘤治疗的m6A靶向药物方面取得的研究进展,提供新的临床见解。
    Immunotherapy for hematological malignancies is a rapidly advancing field that has gained momentum in recent years, primarily encompassing chimeric antigen receptor T-cell (CAR-T) therapies, immune checkpoint inhibitors, and other modalities. However, its clinical efficacy remains limited, and drug resistance poses a significant challenge. Therefore, novel immunotherapeutic targets and agents need to be identified. Recently, N6-methyladenosine (m6A), the most prevalent RNA epitope modification, has emerged as a pivotal factor in various malignancies. Reportedly, m6A mutations influence the immunological microenvironment of hematological malignancies, leading to immune evasion and compromising the anti-tumor immune response in hematological malignancies. In this review, we comprehensively summarize the roles of the currently identified m6A modifications in various hematological malignancies, with a particular focus on their impact on the immune microenvironment. Additionally, we provide an overview of the research progress made in developing m6A-targeted drugs for hematological tumor therapy, to offer novel clinical insights.
    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
    嵌合抗原受体(CAR)疗法是血液系统恶性肿瘤治疗中最前所未有的进步之一。尤其是B细胞恶性肿瘤.这种疗法成功背后的基本概念是产生能够重定向T淋巴细胞以对抗癌细胞的合成蛋白(CAR)。对血液恶性肿瘤的遗传和分子基础的新见解最近引起了靶向治疗的发展。CART细胞疗法是这些免疫治疗技术之一,最近受到了很多关注,并为有效治愈复发性和难治性血液恶性肿瘤和一些实体恶性肿瘤奠定了希望。今天的研究人员可能不知道CAR-T细胞疗法的未来,但是从迄今为止所做的任何研究来看,尽管有其局限性,但这种疗法已被证明是成功的,可以假设其应用范围与日俱增。
    Chimeric antigen receptor (CAR) therapy is one of the most unprecedented advancements in the treatment of hematological malignancies, especially B-cell malignancies. The fundamental notion behind the success of this therapy is to generate a synthetic protein (CAR) capable of redirecting T lymphocytes to act against cancer cells. New insights into the genetic and molecular base of hematological malignancies have more recently given rise to the development of targeted treatments. CAR T-cell therapy is one of these immunological treatment techniques that has recently received a lot of attention and paved a light of hope for the effective cure of relapsed and refractory hematological malignancies and some solid malignancies. Researchers of today might not know what the future holds for CAR T-cell therapy, but from whatever research has been done so far, this therapy has proven to be a success despite its limitations, and it can be assumed that the spectrum of its application is expanding with each passing day.
    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
    由多重耐药生物体如肺炎克雷伯菌引起的血流感染是管理血液恶性肿瘤的重大挑战。本研究旨在描述肺炎克雷伯菌血流感染的流行病学特征,特别是在血液系统恶性肿瘤患者中,描绘初始抗生素治疗的模式,评估耐药菌株的患病率,确定这些耐药菌株的风险因素,并评估影响患者预后的因素。回顾性分析2017年1月至2020年12月在单个中心进行的,重点是182例发生肺炎克雷伯菌血流感染的血液恶性肿瘤患者。我们比较了接受适当和不适当抗生素治疗的患者30天死亡率,包括单药和联合治疗的有效性。采用Kaplan-Meier生存分析和多因素logistic和Cox回归分析确定影响死亡风险的因素。所有患者30天全因死亡率为30.2%。在接受不适当的初始治疗和适当的初始治疗的患者中,30天全因死亡率分别为77.2%和8.8%(p<0.001)。不适当的初始治疗显着影响死亡率,并且是30天死亡率的关键预测指标。以及感染性休克和以前的重症监护病房(ICU)。与CSKP组相比,耐碳青霉烯类肺炎克雷伯菌(CRKP)血流感染的患者表现出更严重的临床症状。该研究表明,碳青霉烯的经验性给药与CRKP和多药耐药肺炎克雷伯菌(MDR-KP)感染的患病率上升之间存在显着关联。此外,该研究发现初始抗生素治疗不适当,感染性休克,和入住ICU是30天死亡率的独立危险因素。
    Bloodstream infections caused by multidrug-resistant organisms such as Klebsiella pneumoniae are a significant challenge in managing hematological malignancies. This study aims to characterize the epidemiology of Klebsiella pneumoniae bloodstream infections specifically in patients with hematological malignancies, delineate the patterns of initial antibiotic therapy, assess the prevalence of resistant strains, identify risk factors for these resistant strains, and evaluate factors influencing patient outcomes. A retrospective analysis was conducted at a single center from January 2017 to December 2020, focusing on 182 patients with hematological malignancies who developed Klebsiella pneumoniae bloodstream infections. We compared the 30-day mortality rates between patients receiving appropriate and inappropriate antibiotic treatments, including the effectiveness of both single-drug and combination therapies. Kaplan-Meier survival analysis and multivariate logistic and Cox regression were used to identify factors influencing mortality risk. The 30-day all-cause mortality rate was 30.2% for all patients. The 30-day all-cause mortality rates were 77.2% and 8.8% in patients who received inappropriate initial treatment and appropriate initial treatment (p < 0.001). Inappropriate initial treatment significantly influenced mortality and was a key predictor of 30-day mortality, along with septic shock and previous intensive care unit (ICU) stays. Patients with carbapenem-resistant Klebsiella pneumoniae (CRKP) bloodstream infections exhibited more severe clinical symptoms compared to the CSKP group. The study demonstrates a significant association between empirical carbapenem administration and the escalating prevalence of CRKP and multidrug-resistant K. pneumoniae (MDR-KP) infections. Furthermore, the study identified inappropriate initial antibiotic therapy, septic shock, and ICU admission as independent risk factors for 30-day mortality.
    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
    与没有恶性血液病(HMs)的患者相比,患有恶性血液病(HMs)的患者感染COVID-19并经历严重后果的风险明显更高。包括潜在的恶性肿瘤,免疫抑制治疗,和患者相关因素。值得注意的是,通常用于HM治疗的免疫抑制方案可以导致B细胞和T细胞的消耗,这与这些患者的COVID-19相关并发症和死亡率增加有关。随着大流行转变为流行状态,承认和解决患有HMs的个人的持续风险仍然至关重要。在这篇综述中,我们的目标是总结目前的证据,以加强我们对HMs对COVID-19风险和结果的影响的理解,识别特别脆弱的个人,并强调需要专门的临床关注和管理。此外,在这些患者中观察到的对COVID-19疫苗接种的免疫反应受损,强调了实施其他缓解策略的重要性.如所示,这可以包括靶向预防和用抗病毒剂和单克隆抗体治疗。提供实际指导和考虑,我们提出了两个说明性的案例,以强调照顾HMs患者的医生所面临的现实生活中的挑战,强调需要根据疾病严重程度进行个性化管理,type,以及每个病人的独特情况。
    Patients with hematologic malignancies (HMs) are at a significantly higher risk of contracting COVID-19 and experiencing severe outcomes compared to individuals without HMs. This heightened risk is influenced by various factors, including the underlying malignancy, immunosuppressive treatments, and patient-related factors. Notably, immunosuppressive regimens commonly used for HM treatment can lead to the depletion of B cells and T cells, which is associated with increased COVID-19-related complications and mortality in these patients. As the pandemic transitions into an endemic state, it remains crucial to acknowledge and address the ongoing risk for individuals with HMs. In this review, we aim to summarize the current evidence to enhance our understanding of the impact of HMs on COVID-19 risks and outcomes, identify particularly vulnerable individuals, and emphasize the need for specialized clinical attention and management. Furthermore, the impaired immune response to COVID-19 vaccination observed in these patients underscores the importance of implementing additional mitigation strategies. This may include targeted prophylaxis and treatment with antivirals and monoclonal antibodies as indicated. To provide practical guidance and considerations, we present two illustrative cases to highlight the real-life challenges faced by physicians caring for patients with HMs, emphasizing the need for individualized management based on disease severity, type, and the unique circumstances of each patient.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Systematic Review
    简介:FLT3突变与血液系统和实体恶性肿瘤的发生密切相关,尤其是急性髓系白血病.目前,几种FLT3抑制剂正在临床试验中,有的已在临床应用。然而,安全,这些FLT3抑制剂的疗效和药效学之前尚未进行过系统分析.方法:我们检索并回顾了13种FLT3抑制剂单药治疗的临床试验报告,包括索拉非尼,列妥替尼,Midostaurin,gilteritinib,quizartinib,舒尼替尼,Crenolanib,坦度替尼,卡博替尼,帕西达替尼,帕克替尼,法米替尼,和TAK-659在2023年5月31日之前患有血液系统和实体恶性肿瘤的患者中。结果:我们的结果显示,最常见的不良事件(AE)是胃肠道不良反应,包括腹泻,手足综合征和恶心,而最常见的血液学AE是发热性中性粒细胞减少症,贫血,和血小板减少症.根据公布的数据,平均总生存期(OS)和平均无进展生存期(PFS)分别为9.639和5.905个月,分别。总反应率(ORR)的发生率,完全缓解(CR),部分响应(PR),所有这些FLT3抑制剂的稳定疾病(SD)为29.0%,8.7%,16.0%,和42.3%,分别。FLT3抑制剂在恶性血液病和实体瘤中的ORR分别为40.8%和18.8%,分别,表明FLT3抑制剂对血液系统恶性肿瘤比实体瘤更有效.此外,这些FLT3抑制剂的最大血浆浓度(Tmax)的时间范围为0.7-12.0小时,但消除半衰期(T1/2)范围是高度可变的,从6.8到151.8h。讨论:FLT3抑制剂单药治疗在临床上显示出明显的抗肿瘤作用,通过联合用药可以进一步提高疗效。
    Introduction: FLT3 mutations are closely associated with the occurrence of hematological and solid malignancies, especially with acute myeloid leukemia. Currently, several FLT3 inhibitors are in clinical trials, and some have been applied in clinic. However, the safety, efficacy and pharmacodynamics of these FLT3 inhibitors have not been systemically analyzed before. Methods: We searched and reviewed clinical trial reports on the monotherapy of 13 FLT3 inhibitors, including sorafenib, lestaurtinib, midostaurin, gilteritinib, quizartinib, sunitinib, crenolanib, tandutinib, cabozantinib, pexidartinib, pacritinib, famitinib, and TAK-659 in patients with hematological and solid malignancies before May 31, 2023. Results: Our results showed the most common adverse events (AEs) were gastrointestinal adverse reactions, including diarrhea, hand-foot syndrome and nausea, while the most common hematological AEs were febrile neutropenia, anemia, and thrombocytopenia. Based on the published data, the mean overall survival (OS) and the mean progression-free survival (PFS) were 9.639 and 5.905 months, respectively. The incidence of overall response rate (ORR), complete remission (CR), partial response (PR), and stable disease (SD) for all these FLT3 inhibitors was 29.0%, 8.7%, 16.0%, and 42.3%, respectively. The ORRs of FLT3 inhibitors in hematologic malignancies and solid tumors were 40.8% and 18.8%, respectively, indicating FLT3 inhibitors were more effective for hematologic malignancies than for solid tumors. In addition, time to maximum plasma concentration (Tmax) in these FLT3 inhibitors ranged from 0.7-12.0 hours, but the elimination half-life (T1/2) range was highly variable, from 6.8 to 151.8 h. Discussion: FLT3 inhibitors monotherapy has shown significant anti-tumor effect in clinic, and the effectiveness may be further improved through combination medication.
    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
    基于普鲁士蓝纳米粒子的光热疗法(PBNP-PTT)是能够引发抗肿瘤免疫应答的有效肿瘤治疗。受PBNP-PTT增强内源性免疫应答能力的激励,我们最近证明,PBNP-PTT可用于离体产生针对胶质母细胞瘤(GBM)细胞系的肿瘤特异性T细胞,作为过继性T细胞疗法(ATCT).在这项研究中,我们进一步开发了这个有前途的T细胞开发平台。首先,我们评估了使用PBNP-PTT产生的T细胞的表型和功能。我们观察到PBNP-PTT促进了CD8+T细胞从健康供体PBMC扩增,这些PBMC分泌IFNγ和TNFα,并上调了CD107a,以响应与靶U87细胞的参与。提示特异性抗肿瘤T细胞活化和脱颗粒。Further,与U87细胞共培养后,CD8+效应和效应记忆T细胞群体显着扩增,与肿瘤特异性效应反应一致。在体内原位植入的U87GBM肿瘤中,PBNP-PTT衍生的T细胞有效减少U87肿瘤生长,并在第100天时在>80%的荷瘤小鼠中产生长期存活,相比之下,用PBS处理的小鼠为0%,非特异性T细胞,或从裂解的U87细胞扩增的T细胞,证明了该ATCT平台的抗肿瘤功效增强。最后,我们通过产生靶向髓母细胞瘤的T细胞(D556)来测试我们方法的普遍性,乳腺癌(MDA-MB-231),神经母细胞瘤(SH-SY5Y),和急性单核细胞白血病(THP-1)细胞系。所产生的T细胞分泌IFNγ并发挥相对于对照增加的肿瘤特异性细胞溶解功能,证明PBNP-PTT在产生ATCT肿瘤特异性T细胞方面的多功能性。
    Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) is an effective tumor treatment capable of eliciting an antitumor immune response. Motivated by the ability of PBNP-PTT to potentiate endogenous immune responses, we recently demonstrated that PBNP-PTT could be used ex vivo to generate tumor-specific T cells against glioblastoma (GBM) cell lines as an adoptive T cell therapy (ATCT). In this study, we further developed this promising T cell development platform. First, we assessed the phenotype and function of T cells generated using PBNP-PTT. We observed that PBNP-PTT facilitated CD8+ T cell expansion from healthy donor PBMCs that secreted IFNγ and TNFα and upregulated CD107a in response to engagement with target U87 cells, suggesting specific antitumor T cell activation and degranulation. Further, CD8+ effector and effector memory T cell populations significantly expanded after co-culture with U87 cells, consistent with tumor-specific effector responses. In orthotopically implanted U87 GBM tumors in vivo, PBNP-PTT-derived T cells effectively reduced U87 tumor growth and generated long-term survival in >80% of tumor-bearing mice by Day 100, compared to 0% of mice treated with PBS, non-specific T cells, or T cells expanded from lysed U87 cells, demonstrating an enhanced antitumor efficacy of this ATCT platform. Finally, we tested the generalizability of our approach by generating T cells targeting medulloblastoma (D556), breast cancer (MDA-MB-231), neuroblastoma (SH-SY5Y), and acute monocytic leukemia (THP-1) cell lines. The resulting T cells secreted IFNγ and exerted increased tumor-specific cytolytic function relative to controls, demonstrating the versatility of PBNP-PTT in generating tumor-specific T cells for ATCT.
    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
    血液恶性肿瘤(HMs)包括具有显著发病率和死亡率的不同组的血液肿瘤。免疫疗法已成为HMs患者的一种有效和关键的治疗方式。尽管在过去十年中,在理解和实施HMs的免疫疗法方面取得了显著进步,几个挑战依然存在。这些挑战包括免疫相关的不良反应,治疗性抗原在体内的精确生物分布和消除,肿瘤的免疫耐受,和肿瘤细胞在肿瘤微环境(TME)内的免疫逃避。纳米技术,具有在纳米尺度上操纵材料特性的能力,有可能通过改善药物靶向和稳定性等各个方面来解决这些障碍并彻底改变治疗结果。纳米技术和免疫治疗的融合催生了纳米免疫治疗,抗肿瘤治疗的一个专门分支。纳米技术已经在嵌合抗原受体T细胞(CAR-T)治疗中找到了应用,癌症疫苗,免疫检查点抑制剂,以及其他针对HMs的免疫治疗策略。在这篇综述中,我们描述了最近的发展,并讨论了目前在纳米免疫治疗领域的挑战,为这些疾病的基于纳米技术的治疗方法的潜力提供了新的见解。
    Hematological malignancies (HMs) encompass a diverse group of blood neoplasms with significant morbidity and mortality. Immunotherapy has emerged as a validated and crucial treatment modality for patients with HMs. Despite notable advancements having been made in understanding and implementing immunotherapy for HMs over the past decade, several challenges persist. These challenges include immune-related adverse effects, the precise biodistribution and elimination of therapeutic antigens in vivo, immune tolerance of tumors, and immune evasion by tumor cells within the tumor microenvironment (TME). Nanotechnology, with its capacity to manipulate material properties at the nanometer scale, has the potential to tackle these obstacles and revolutionize treatment outcomes by improving various aspects such as drug targeting and stability. The convergence of nanotechnology and immunotherapy has given rise to nano-immunotherapy, a specialized branch of anti-tumor therapy. Nanotechnology has found applications in chimeric antigen receptor T cell (CAR-T) therapy, cancer vaccines, immune checkpoint inhibitors, and other immunotherapeutic strategies for HMs. In this review, we delineate recent developments and discuss current challenges in the field of nano-immunotherapy for HMs, offering novel insights into the potential of nanotechnology-based therapeutic approaches for these diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号