低剂量白细胞介素-2扩大调节性T细胞并调节轻中度阿尔茨海默病的生物标志物:2a期随机试验显示安全性和有希望的信号

低剂量白细胞介素-2扩大调节性T细胞并调节轻中度阿尔茨海默病的生物标志物:2a期随机试验显示安全性和有希望的信号

亮点

– 每4周一次皮下注射低剂量IL-2(1 x 10^6 IU/天,连续5天)在轻中度阿尔茨海默病(AD)患者中是安全且耐受良好的。

– 每4周一次IL-2显著增加了循环中的调节性T细胞(Tregs)数量,并提高了Foxp3平均荧光强度(MFI),与安慰剂相比具有增强的抑制功能。

– 每4周一次IL-2在多个血浆炎症介质(减少CCL2、CCL11、IL-15;增加IL-4、CCL13)和脑脊液Aβ42(第148天,p = 0.045 vs 安慰剂)方面产生了有利变化;脑脊液NfL保持稳定,而安慰剂组有所上升(p = 0.060)。

– 该试验在第22周通过ADAS-Cog评估发现了认知衰退减缓的信号(p = 0.061),支持进行更大规模、更长时间的研究。

背景:疾病负担和研究理由

阿尔茨海默病(AD)是一种以认知衰退、淀粉样蛋白-β(Aβ)和tau病理积累、突触丢失以及神经炎症为特征的进行性神经退行性疾病。炎症和免疫调节功能障碍已成为疾病进展的重要贡献者,并逐渐被视为可干预的治疗靶点。调节性T细胞(Tregs)是关键的免疫调节因子,能够抑制过度炎症并维持稳态。先前的研究已记录了AD患者Treg数量和/或功能受损,外周和中枢免疫谱型向促炎转变,以及免疫失调与临床进展之间的关联。

低剂量白细胞介素-2(IL-2)在体内优先扩增Tregs,已在多种自身免疫和炎症性疾病中进行了研究,作为恢复免疫调节的策略。将这种方法应用于AD旨在减少适应不良的炎症,保护神经元免受免疫介导的损伤,并可能影响AD的核心病理生物学。

研究设计

本报告描述了一项2a期、随机、双盲、安慰剂对照试验(ClinicalTrials.gov NCT06096090),共招募了38名轻至中度AD患者。参与者被随机分配到三个组之一:每4周一次IL-2(n = 9)、每2周一次IL-2(n = 10)或安慰剂(n = 19)。IL-2给药方案包括每天皮下注射1 × 10^6 IU,连续5天,每4周或每2周重复一次,持续21周的治疗期,随后进行9周的观察期。

主要终点是安全性和耐受性——不良事件(AEs)的发生率和严重程度。次要终点评估了免疫药理学动力学:Treg频率和抑制功能的变化,以及Foxp3 MFI。探索性终点包括45种血浆炎症介质的纵向测量、脑脊液(CSF)AD生物标志物(Aβ42、神经丝蛋白轻链[NfL])和临床量表(如ADAS-Cog)。

主要发现

安全性和耐受性

所有38名参与者均完成了试验。未报告严重的不良事件或死亡。两种IL-2给药方案总体上均耐受良好。在这一老年AD人群中,低剂量IL-2短期疗程的安全性信号缺失支持了其可行性,但需要更大的样本量来定义罕见的不良事件和长期安全性。

免疫学效应:Treg扩增和功能

每4周一次和每2周一次IL-2均增加了循环中的Treg数量并增强了抑制活性,相对于基线和安慰剂。值得注意的是,每4周一次IL-2在Treg百分比和Foxp3 MFI方面的增加优于每2周一次方案。这些读数不仅表明数量上的扩展,还表明Treg区室的表型强化(更高的Foxp3表达),这在机制研究中与抑制能力相关。

外周炎症介质

对45种血浆炎症标志物的纵向分析显示,每4周一次IL-2对几种与神经炎症和AD相关的介质产生了最显著的影响。具体而言,每4周一次IL-2减少了CCL2(MCP-1)、CCL11(嗜酸性粒细胞趋化因子-1)和IL-15,同时增加了IL-4和CCL13。这些变化表明单核细胞/趋化因子驱动的炎症募集(CCL2、CCL11)减弱,以及向抗炎或调节性细胞因子环境倾斜(IL-4)。IL-15的减少可能反映了细胞毒性或促炎淋巴细胞激活的减少。这种模式在生物学上是合理的,即通过Treg介导的外周免疫音调调节间接影响中枢免疫。

脑脊液生物标志物

在第148天,每4周一次IL-2组的脑脊液Aβ42水平显著高于安慰剂组(p = 0.045)。解释需要细致入微:在症状性AD中,低脑脊液Aβ42通常被解释为在大脑淀粉样蛋白聚集物中隔离。免疫调节疗法后脑脊液Aβ42的升高可能合理地反映了从大脑到脑脊液的可溶性Aβ物种清除的增加、沉积减少或隔室间平衡的改变。需要额外的淀粉样PET验证才能确定实质淀粉样蛋白的清除。

脑脊液NfL(神经轴索损伤的标志物)在安慰剂组增加了217 pg/ml,而在每4周一次IL-2组保持稳定(p = 0.060 IL-2 vs 安慰剂),这是一个接近显著的结果,表明Treg扩增可能通过减少免疫介导的轴突损伤而具有潜在的神经保护作用。

临床结果

在第22周,与安慰剂相比,每4周一次IL-2组的ADAS-Cog从基线调整后的平均变化显示出减缓进展的趋势(p = 0.061)。尽管未达到常规统计学意义,但在生物标志物、免疫学终点和认知方面的方向性一致提供了支持进一步确认的连贯生物学信号。

专家评论:解释、机制和局限性

解释。这项2a期试验表明,在研究的AD队列中,间歇性低剂量IL-2是安全的,可以扩大并功能性增强Treg,调节外周炎症介质,并产生与疾病修饰假设一致的生物标志物和临床信号。免疫、生化和认知措施之间的一致性加强了生物学合理性。

假定机制。低剂量IL-2优先刺激Treg(CD25hi)上表达的高亲和力IL-2受体,促进Treg的存活、增殖和抑制功能。恢复的外周免疫调节可能减少促炎单核细胞和淋巴细胞进入中枢神经系统,减少小胶质细胞活化,并减轻下游神经元损伤。趋化因子(如CCL2和CCL11)的变化与减少外周炎症细胞的募集一致;IL-4的增加可能反映向2型/调节性反应的转变。

为什么每4周一次优于每2周一次?每4周一次方案的优越性能可能反映了Treg诱导和收缩的稳态动态:脉冲给药与恢复间隔相结合可能允许Treg区室更持久的重新编程,而较短的重复间隔可能仅促进短暂的扩增而没有表型成熟。未来研究中的药理学建模和机制免疫表型分析可以阐明最佳给药方案。

局限性。关键局限性包括样本量较小(n = 38),尤其是在活性组内,这降低了功效并增加了I型/II型错误的风险。多项探索性生物标志物比较增加了假阳性的可能性;脑脊液Aβ42的发现(p = 0.045)和接近显著的NfL和ADAS-Cog结果需要复制。研究持续时间相对较短,不足以提出疾病修饰的主张;AD病理演变需要数年时间,因此需要更长时间的随访来评估对临床进展和成像生物标志物的持久影响。试验人群和纳入/排除标准可能限制了普遍性,特别是对于合并症或晚期疾病的患者。缺乏淀粉样或tau PET限制了脑脊液变化与实质病理之间的解释。

意义和下一步行动

这些结果证明了进行更大规模、充分功效的随机试验的必要性,设计考虑因素包括:更长的治疗和随访时间(≥12-18个月),根据基线淀粉样/tau负荷分层(理想情况下使用PET或CSF谱型),结合成像生物标志物(淀粉样和tau PET,体积MRI),更详细的免疫表型(Treg亚群,迁移标记),以及对合并症老年人的仔细安全性监测。剂量寻找和给药优化研究可以确定最大化持久Treg功能并最小化给药负担的方案。如果IL-2在有效调节下游神经炎症的同时,特定疾病药物解决蛋白质病,则可以考虑联合方法(例如,IL-2与抗淀粉样或抗tau疗法)。

结论

在这项2a期随机试验中,每4周一次的间歇性低剂量IL-2是安全的,可以扩大并功能性增强调节性T细胞,改变血浆炎症介质的方向与减少促炎音调一致,增加脑脊液Aβ42,相对于安慰剂稳定脑脊液NfL,并在认知衰退方面产生有利趋势。尽管初步,这些一致的信号支持通过更大规模、更长时间的多模态生物标志物终点试验进一步研究针对Treg的免疫疗法在AD中的应用。

资金和试验注册

试验注册:ClinicalTrials.gov 标识符:NCT06096090,注册日期:2023年10月17日。资金来源见原始出版物(Faridar等,Alzheimers Res Ther. 2025)。

参考文献

Faridar A, Gamez N, Li D, Wang Y, Boradia R, Thome AD, Zhao W, Beers DR, Thonhoff JR, Nakawah MO, Román GC, Volpi JJ, Toledo JB, George M, Davis CS, Pascual B, Grundman M, Masdeu JC, Appel SH. 低剂量白细胞介素-2在轻中度阿尔茨海默病患者中的应用:一项随机临床试验。Alzheimers Res Ther. 2025 Jul 4;17(1):146. doi: 10.1186/s13195-025-01791-x. PMID: 40615880; PMCID: PMC12231701。

关于低剂量IL-2和Treg疗法在人类疾病中的基础文献可以在临床免疫学和转化医学文献中找到;读者可参阅上述主要试验报告以获取完整的方法、安全性表格和补充分析。

Low‑Dose Interleukin‑2 Expands Regulatory T Cells and Modulates Biomarkers in Mild–Moderate Alzheimer’s Disease: Phase 2a Randomized Trial Shows Safety and Promising Signals

Low‑Dose Interleukin‑2 Expands Regulatory T Cells and Modulates Biomarkers in Mild–Moderate Alzheimer’s Disease: Phase 2a Randomized Trial Shows Safety and Promising Signals

Highlights

– Low‑dose subcutaneous IL‑2 (1 x 10^6 IU/day for 5 days) administered every 4 weeks (IL‑2 q4wks) was safe and well tolerated in people with mild–moderate Alzheimer’s disease (AD).

– IL‑2 q4wks significantly expanded circulating regulatory T cells (Tregs) and increased Foxp3 mean fluorescent intensity (MFI), with enhanced suppressive function versus placebo.

– IL‑2 q4wks produced favorable shifts in multiple plasma inflammatory mediators (reduced CCL2, CCL11, IL‑15; increased IL‑4, CCL13) and increased CSF Aβ42 (p = 0.045 vs placebo) on Day 148; CSF NfL remained stable versus a rise in placebo (p = 0.060).

– The trial identified a signal toward slower cognitive decline by ADAS‑Cog at week 22 (p = 0.061) supporting larger, longer studies.

Background: disease burden and rationale

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, accumulation of amyloid‑β (Aβ) and tau pathology, synaptic loss, and neuroinflammation. Inflammation and dysfunctional immune regulation have emerged as central contributors to disease progression and are increasingly viewed as actionable therapeutic targets. Regulatory T cells (Tregs) are key immune modulators that suppress excessive inflammation and maintain homeostasis. Prior work has documented compromised Treg numbers and/or function in AD, a shift toward pro‑inflammatory peripheral and central immune profiles, and associations between immune dysregulation and clinical progression.

Low‑dose interleukin‑2 (IL‑2) preferentially expands Tregs in vivo and has been investigated in several autoimmune and inflammatory disorders as a strategy to restore immune regulation. Translating this approach to AD seeks to reduce maladaptive inflammation, protect neurons from immune‑mediated injury, and potentially influence core AD pathobiology.

Study design

This report describes a phase 2a, randomized, double‑blind, placebo‑controlled trial (ClinicalTrials.gov NCT06096090) enrolling 38 participants with mild to moderate AD. Participants were randomized to one of three arms: IL‑2 q4wks (n = 9), IL‑2 q2wks (n = 10), or placebo (n = 19). The IL‑2 regimen consisted of subcutaneous injections at a dose of 1 × 10^6 IU/day for five consecutive days, repeated either every 4 weeks or every 2 weeks, over a 21‑week treatment period followed by 9 weeks of observation.

Primary endpoints were safety and tolerability — incidence and severity of adverse events (AEs). Secondary endpoints assessed immunologic pharmacodynamics: changes in Treg frequency and suppressive function, and Foxp3 MFI. Exploratory endpoints included longitudinal measurement of 45 plasma inflammatory mediators, cerebrospinal fluid (CSF) AD biomarkers (Aβ42, neurofilament light chain [NfL]), and clinical scales including the ADAS‑Cog.

Key findings

Safety and tolerability

All 38 participants completed the trial. There were no serious adverse events or deaths reported. Both IL‑2 dosing schedules were generally well tolerated. The absence of serious safety signals in this older, AD cohort supports feasibility of short courses of low‑dose IL‑2 in this population, though larger samples are required to define uncommon adverse events and long‑term safety.

Immunologic effects: Treg expansion and function

Both IL‑2 q4wks and q2wks increased circulating Treg numbers and enhanced suppressive activity relative to baseline and placebo. Notably, IL‑2 q4wks produced superior increases in Treg percentage and Foxp3 MFI compared with the q2wks regimen. These readouts indicate not only quantitative expansion but also phenotypic strengthening of the Treg compartment (higher Foxp3 expression), which correlates with suppressive capacity in mechanistic studies.

Peripheral inflammatory mediators

Longitudinal profiling of 45 plasma inflammatory markers revealed that IL‑2 q4wks had the most pronounced effects on several mediators implicated in neuroinflammation and AD. Specifically, IL‑2 q4wks decreased CCL2 (MCP‑1), CCL11 (eotaxin‑1), and IL‑15 while increasing IL‑4 and CCL13. These shifts suggest a dampening of monocyte/chemokine‑driven inflammatory recruitment (CCL2, CCL11) and a tilt toward anti‑inflammatory or regulatory cytokine milieu (IL‑4). IL‑15 reduction may reflect decreased cytotoxic or pro‑inflammatory lymphocyte activation. The pattern is biologically plausible for a Treg‑mediated modulation of peripheral immune tone that could impact central immunity indirectly.

CSF biomarkers

On Day 148, IL‑2 q4wks recipients showed a significant increase in CSF Aβ42 compared with placebo (p = 0.045). Interpretation requires nuance: in symptomatic AD, low CSF Aβ42 is classically interpreted as sequestration into brain amyloid aggregates. A rise in CSF Aβ42 after an immunomodulatory therapy could plausibly reflect enhanced clearance of soluble Aβ species from brain to CSF, reduced deposition, or altered equilibrium between compartments. Additional corroboration with amyloid PET would be needed to firmly conclude clearance of parenchymal amyloid.

CSF NfL, a marker of neuroaxonal injury, increased by 217 pg/ml in the placebo arm but remained stable in the IL‑2 q4wks group (p = 0.060 IL‑2 q4wks vs placebo), a near‑significant finding that signals potential neuroprotective effects of Treg expansion through reduction of immune‑mediated axonal injury.

Clinical outcomes

The adjusted mean change from baseline in ADAS‑Cog at week 22 favored IL‑2 q4wks with a trend toward slower progression versus placebo (p = 0.061). Although not reaching conventional statistical significance, the directionality across biomarkers, immunologic endpoints, and cognition provides a coherent biological signal warranting confirmation.

Expert commentary: interpretation, mechanisms, and limitations

Interpretation. This phase 2a trial demonstrates that intermittent low‑dose IL‑2 is safe in the studied AD cohort, expands and functionally augments Tregs, modulates peripheral inflammatory mediators, and produces biomarker and clinical signals consistent with disease modification hypotheses. The coherence across immune, biochemical, and cognitive measures strengthens biological plausibility.

Putative mechanism. Low‑dose IL‑2 preferentially stimulates the high‑affinity IL‑2 receptor expressed on Tregs (CD25hi), promoting Treg survival, proliferation, and suppressive function. Restored peripheral immune regulation may reduce trafficking of pro‑inflammatory monocytes and lymphocytes into the CNS, decrease microglial activation, and mitigate downstream neuronal injury. Changes in chemokines such as CCL2 and CCL11 are consistent with reduced recruitment of peripheral inflammatory cells; increased IL‑4 may reflect a shift toward type‑2/regulatory responses.

Why q4wks > q2wks? The superior performance of the q4wks schedule could reflect homeostatic dynamics of Treg induction and contraction: pulse dosing with recovery intervals might allow more durable reprogramming of the Treg compartment, whereas shorter repeat intervals could favor transient expansion without phenotypic maturation. Pharmacodynamic modeling and mechanistic immunophenotyping in future studies could clarify optimal scheduling.

Limitations. Key limitations include small sample size (n = 38), especially within active arms, which reduces power and increases risk for type I/II errors. Multiple exploratory biomarker comparisons raise the possibility of false positives; the CSF Aβ42 finding (p = 0.045) and near‑significant NfL and ADAS‑Cog results require replication. Study duration was relatively short for disease‑modifying claims; AD pathology evolves over years, so longer follow‑up is necessary to assess durable effects on clinical progression and imaging biomarkers. The trial population and inclusion/exclusion criteria may limit generalizability, particularly to patients with comorbidities or later‑stage disease. The absence of amyloid or tau PET limits interpretation of CSF changes in relation to parenchymal pathology.

Implications and next steps

These results justify a larger, adequately powered randomized trial with several design considerations: longer treatment and follow‑up (≥12–18 months), stratification by baseline amyloid/tau burden (ideally PET or CSF profiles), incorporation of imaging biomarkers (amyloid and tau PET, volumetric MRI), more granular immunophenotyping (Treg subsets, trafficking markers), and careful safety monitoring in older adults with comorbidities. Dose‑finding and schedule optimization studies could identify regimens that maximize durable Treg function with minimal dosing burden. Combination approaches (e.g., IL‑2 with anti‑amyloid or anti‑tau therapies) could be considered if IL‑2 proves to modulate downstream neuroinflammation effectively while disease‑specific agents address proteinopathy.

Conclusion

In this phase 2a randomized trial, intermittent low‑dose IL‑2 administered every 4 weeks was safe, expanded and functionally strengthened regulatory T cells, altered plasma inflammatory mediators in a direction consistent with reduced pro‑inflammatory tone, increased CSF Aβ42, stabilized CSF NfL relative to placebo, and produced a favorable trend in cognitive decline. While preliminary, these convergent signals support further investigation of Treg‑targeted immunotherapy in AD through larger and longer trials with multimodal biomarker endpoints.

Funding and trial registration

Trial registration: ClinicalTrials.gov Identifier: NCT06096090, Registration Date: 10‑17‑2023. Funding sources are reported in the original publication (Faridar et al., Alzheimers Res Ther. 2025).

References

Faridar A, Gamez N, Li D, Wang Y, Boradia R, Thome AD, Zhao W, Beers DR, Thonhoff JR, Nakawah MO, Román GC, Volpi JJ, Toledo JB, George M, Davis CS, Pascual B, Grundman M, Masdeu JC, Appel SH. Low‑dose interleukin‑2 in patients with mild to moderate Alzheimer’s disease: a randomized clinical trial. Alzheimers Res Ther. 2025 Jul 4;17(1):146. doi: 10.1186/s13195-025-01791-x. PMID: 40615880; PMCID: PMC12231701.

Additional foundational literature on low‑dose IL‑2 and Treg therapy in human disease can be found in the clinical immunology and translational medicine literature; readers are referred to the primary trial report above for full methods, safety tables, and supplementary analyses.

Comments

No comments yet. Why don’t you start the discussion?

发表回复