嵌合抗原受体-巨噬细胞疗法进入临床:HER2+实体瘤的首次人体试验

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
MedComm Pub Date : 2025-08-27 DOI:10.1002/mco2.70374
Wenxue Ma, Catriona Jamieson
{"title":"嵌合抗原受体-巨噬细胞疗法进入临床:HER2+实体瘤的首次人体试验","authors":"Wenxue Ma,&nbsp;Catriona Jamieson","doi":"10.1002/mco2.70374","DOIUrl":null,"url":null,"abstract":"<p>In a recent study published in Nature Medicine (2025), Reiss et al. [<span>1</span>] reported the first-in-human Phase 1 trial of CT-0508, a chimeric antigen receptor macrophage (CAR-M) therapy targeting HER2-overexpressing solid tumors. This trial demonstrated safety, feasibility, and early immune activity in heavily pretreated patients, marking the first clinical validation of macrophage-based immunotherapy. This milestone highlights the potential of engineered macrophages to overcome tumor microenvironment (TME)-mediated resistance, signaling a paradigm shift for treating solid tumors refractory to current therapies.</p><p>Fourteen patients with HER2-overexpressing tumors, including breast, gastroesophageal, and salivary duct carcinomas, were enrolled and received CT-0508 without prior lympho-depleting chemotherapy. Two dosing regimens were explored: fractionated (Group 1) and bolus (Group 2). CAR-Ms were generated from autologous monocytes using a replication-incompetent adenoviral vector (Ad5.F35), achieving high viability, purity, and CAR expression across all manufactured products. Preclinical assays demonstrated HER2-specific cytotoxicity, phagocytosis, and secretion of proinflammatory cytokines upon antigen engagement.</p><p>Clinically, CT-0508 exhibited a favorable safety profile. No dose-limiting toxicities, Grade ≥3 cytokine release syndrome (CRS), or immune effector cell-associated neurotoxicity syndrome (ICANS) were observed. Grade 1–2 CRS occurred in nine patients and was managed without corticosteroids or intensive supportive care. Importantly, the trial avoided the use of lympho-depleting regimens such as cyclophosphamide or fludarabine, which are standard in CAR-T therapies, thereby supporting a more tolerable, outpatient-based delivery model. This distinction represents a major advantage in the solid tumor space, where toxicity concerns and logistical burdens have limited broader access to cell therapy.</p><p>Although no objective responses (per RECIST v1.1) were observed, four of nine patients (44.5%) with HER2 immunohistochemistry (IHC) 3<sup>+</sup> tumors achieved stable disease (SD), while all five patients with HER2 IHC 2<sup>+</sup> tumors exhibited progressive disease (PD). Tumor volume reductions were noted in 41% of measurable lesions, and circulating tumor DNA (ctDNA) levels decreased in 62% of patients, including all those with SD. These molecular and radiographic evidence indicate biological activity, although transient. Interestingly, some patients exhibited ctDNA rebound at later timepoints, raising questions about the durability of response and the potential need for maintenance dosing or sequential therapeutic strategies. These observations reinforce the value of integrating longitudinal molecular monitoring in early-phase trials to better understand the kinetics of response and identify early markers of relapse. A graphical overview summarizing the clinical workflow, immunologic changes, and future directions of CAR-M therapy is provided in Figure 1.</p><p>A striking feature of the study is the clear dependence of clinical benefit on HER2 expression intensity. Responses were exclusively observed in patients with HER2 IHC 3<sup>+</sup> tumors, characterized by strong membranous staining. Non-clinical benefit was observed among HER2 IHC 2<sup>+</sup> patients. Furthermore, more than half of screening failures were attributed to HER2 downregulation at the time of enrollment, highlighting the plasticity of HER2 expression and its susceptibility to prior treatment pressure or tumor evolution [<span>1</span>]. These findings highlight the critical need for rigorous and real-time biomarker evaluation to ensure optimal patient selection. As target expression heterogeneity continues to challenge the field, developing more dynamic patient stratification models, including serial biopsies or advanced imaging modalities, will be essential for identifying those most likely to benefit from CAR-M therapy.</p><p>Mechanistically, CT-0508 induced significant remodeling of the TME. Single-cell RNA sequencing (scRNA-seq) analysis and paired biopsies revealed increased antigen presentation gene signatures and enhanced infiltration of CD8<sup>+</sup> T cells, many of which displayed clonotypic expansion and cytotoxic markers (granzyme B, perforin 1, IFN-γ). These features suggest antigen spreading and reactivation of endogenous T-cell responses. Patients achieving SD also showed higher baseline CD4:CD8 ratios and lower systemic inflammation, pointing to possible predictive markers for immunologic responsiveness. Importantly, these immune signatures resemble those seen in checkpoint blockade and suggest that CAR-M may prime the TME for combinatorial strategies.</p><p>Despite these promising mechanistic insights, the therapeutic effects were transient. Most patients experienced disease progression within a few months. Limited CAR-M persistence in circulation and low infiltration levels within tumor tissues, as assessed by RNAScope in situ RNA technology, likely contributed to this outcome. This raises critical questions about how to enhance CAR-M retention, tumor homing, and intratumoral survival. Future approaches may include genetic modifications to enhance lifespan, co-expression of chemokines or homing receptors, and integration with depot-based delivery systems or repeated dosing strategies.</p><p>Importantly, this trial demonstrates that macrophages, historically difficult to transduce, can be successfully engineered using adenoviral platforms without compromising their phenotype or function. This technical advance overcomes a major bottleneck and paves the way for future iterations of CAR-M with enhanced payloads, such as cytokine release constructs, antigen-presenting modules, or immune checkpoint regulators [<span>2</span>]. Additionally, the manufacturing process, performed without ex vivo expansion, enhances the feasibility and scalability of autologous CAR-M therapy for broader clinical application.</p><p>Beyond HER2 targeting, the broader implications of this trial lie in its validation of macrophages as a viable immune effector cell type for adoptive immunotherapy. Unlike T cells, macrophages are innate immune cells capable of infiltrating tumor stroma, performing phagocytosis, and activating adaptive immunity through antigen presentation. Their myeloid lineage reduces susceptibility to exhaustion and supports potential for off-the-shelf, allogeneic applications. As engineering platforms evolve, CAR-Ms may be tailored to address multiple tumor antigens, modulate polarization states, or carry synthetic payloads to reprogram the TME.</p><p>From a translational perspective, this trial highlights several critical considerations for advancing CAR-M platforms. These include (i) optimizing patient selection based on both tumor antigen density and immune contexture; (ii) integrating real-time, multiomic monitoring to guide therapeutic decisions; and (iii) rationally designing combination regimens, particularly with ICIs, toll-like receptor (TLR) agonists, or metabolic modulators to enhance efficacy and overcome resistance. Collaborative efforts between academic centers and industry partners will be essential to accelerate innovation and clinical translation.</p><p>Moving forward, enhancing the persistence and expansion of CAR-Ms, possibly through cytokine support, synthetic constructs, or combination with immune checkpoint blockade, may improve clinical outcomes [<span>3</span>]. Biomarker-guided patient stratification, especially based on HER2 IHC scores and immune fitness signatures, will be critical in optimizing therapeutic outcomes. Furthermore, strategies to convert early immune activation into sustained tumor regression, such as in vivo reprogramming, combination immunotherapies, or CAR-M modifications for improved antigen retention, are essential for long-term efficacy.</p><p>In summary, this first-in-human trial of CT-0508 establishes a clinical proof-of-concept for CAR-M therapy in HER2-positive solid tumors. While limitations remain, the study demonstrates that macrophage-based therapies can be safely administered, remodel the TME, and engage adaptive immunity. CAR-Ms represent a promising new frontier in cancer immunotherapy, offering a unique mechanism of action that may complement existing T-cell-based approaches [<span>4</span>].</p><p>Wenxue Ma: conceptualization, investigation, writing—original draft, validation, visualization, writing—review and editing, software, formal analysis, project administration, data curation, supervision, resources. Catriona Jamieson: conceptualization, funding acquisition, writing—review and editing, project administration, supervision, resources, validation. Both authors have read and approved the final manuscript.</p><p>The authors have nothing to report.</p><p>The authors declare no conflicts of interest.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"6 9","pages":""},"PeriodicalIF":10.7000,"publicationDate":"2025-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70374","citationCount":"0","resultStr":"{\"title\":\"Chimeric Antigen Receptor-Macrophage Therapy Enters the Clinic: The First-in-Human Trial for HER2+ Solid Tumors\",\"authors\":\"Wenxue Ma,&nbsp;Catriona Jamieson\",\"doi\":\"10.1002/mco2.70374\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>In a recent study published in Nature Medicine (2025), Reiss et al. [<span>1</span>] reported the first-in-human Phase 1 trial of CT-0508, a chimeric antigen receptor macrophage (CAR-M) therapy targeting HER2-overexpressing solid tumors. This trial demonstrated safety, feasibility, and early immune activity in heavily pretreated patients, marking the first clinical validation of macrophage-based immunotherapy. This milestone highlights the potential of engineered macrophages to overcome tumor microenvironment (TME)-mediated resistance, signaling a paradigm shift for treating solid tumors refractory to current therapies.</p><p>Fourteen patients with HER2-overexpressing tumors, including breast, gastroesophageal, and salivary duct carcinomas, were enrolled and received CT-0508 without prior lympho-depleting chemotherapy. Two dosing regimens were explored: fractionated (Group 1) and bolus (Group 2). CAR-Ms were generated from autologous monocytes using a replication-incompetent adenoviral vector (Ad5.F35), achieving high viability, purity, and CAR expression across all manufactured products. Preclinical assays demonstrated HER2-specific cytotoxicity, phagocytosis, and secretion of proinflammatory cytokines upon antigen engagement.</p><p>Clinically, CT-0508 exhibited a favorable safety profile. No dose-limiting toxicities, Grade ≥3 cytokine release syndrome (CRS), or immune effector cell-associated neurotoxicity syndrome (ICANS) were observed. Grade 1–2 CRS occurred in nine patients and was managed without corticosteroids or intensive supportive care. Importantly, the trial avoided the use of lympho-depleting regimens such as cyclophosphamide or fludarabine, which are standard in CAR-T therapies, thereby supporting a more tolerable, outpatient-based delivery model. This distinction represents a major advantage in the solid tumor space, where toxicity concerns and logistical burdens have limited broader access to cell therapy.</p><p>Although no objective responses (per RECIST v1.1) were observed, four of nine patients (44.5%) with HER2 immunohistochemistry (IHC) 3<sup>+</sup> tumors achieved stable disease (SD), while all five patients with HER2 IHC 2<sup>+</sup> tumors exhibited progressive disease (PD). Tumor volume reductions were noted in 41% of measurable lesions, and circulating tumor DNA (ctDNA) levels decreased in 62% of patients, including all those with SD. These molecular and radiographic evidence indicate biological activity, although transient. Interestingly, some patients exhibited ctDNA rebound at later timepoints, raising questions about the durability of response and the potential need for maintenance dosing or sequential therapeutic strategies. These observations reinforce the value of integrating longitudinal molecular monitoring in early-phase trials to better understand the kinetics of response and identify early markers of relapse. A graphical overview summarizing the clinical workflow, immunologic changes, and future directions of CAR-M therapy is provided in Figure 1.</p><p>A striking feature of the study is the clear dependence of clinical benefit on HER2 expression intensity. Responses were exclusively observed in patients with HER2 IHC 3<sup>+</sup> tumors, characterized by strong membranous staining. Non-clinical benefit was observed among HER2 IHC 2<sup>+</sup> patients. Furthermore, more than half of screening failures were attributed to HER2 downregulation at the time of enrollment, highlighting the plasticity of HER2 expression and its susceptibility to prior treatment pressure or tumor evolution [<span>1</span>]. These findings highlight the critical need for rigorous and real-time biomarker evaluation to ensure optimal patient selection. As target expression heterogeneity continues to challenge the field, developing more dynamic patient stratification models, including serial biopsies or advanced imaging modalities, will be essential for identifying those most likely to benefit from CAR-M therapy.</p><p>Mechanistically, CT-0508 induced significant remodeling of the TME. Single-cell RNA sequencing (scRNA-seq) analysis and paired biopsies revealed increased antigen presentation gene signatures and enhanced infiltration of CD8<sup>+</sup> T cells, many of which displayed clonotypic expansion and cytotoxic markers (granzyme B, perforin 1, IFN-γ). These features suggest antigen spreading and reactivation of endogenous T-cell responses. Patients achieving SD also showed higher baseline CD4:CD8 ratios and lower systemic inflammation, pointing to possible predictive markers for immunologic responsiveness. Importantly, these immune signatures resemble those seen in checkpoint blockade and suggest that CAR-M may prime the TME for combinatorial strategies.</p><p>Despite these promising mechanistic insights, the therapeutic effects were transient. Most patients experienced disease progression within a few months. Limited CAR-M persistence in circulation and low infiltration levels within tumor tissues, as assessed by RNAScope in situ RNA technology, likely contributed to this outcome. This raises critical questions about how to enhance CAR-M retention, tumor homing, and intratumoral survival. Future approaches may include genetic modifications to enhance lifespan, co-expression of chemokines or homing receptors, and integration with depot-based delivery systems or repeated dosing strategies.</p><p>Importantly, this trial demonstrates that macrophages, historically difficult to transduce, can be successfully engineered using adenoviral platforms without compromising their phenotype or function. This technical advance overcomes a major bottleneck and paves the way for future iterations of CAR-M with enhanced payloads, such as cytokine release constructs, antigen-presenting modules, or immune checkpoint regulators [<span>2</span>]. Additionally, the manufacturing process, performed without ex vivo expansion, enhances the feasibility and scalability of autologous CAR-M therapy for broader clinical application.</p><p>Beyond HER2 targeting, the broader implications of this trial lie in its validation of macrophages as a viable immune effector cell type for adoptive immunotherapy. Unlike T cells, macrophages are innate immune cells capable of infiltrating tumor stroma, performing phagocytosis, and activating adaptive immunity through antigen presentation. Their myeloid lineage reduces susceptibility to exhaustion and supports potential for off-the-shelf, allogeneic applications. As engineering platforms evolve, CAR-Ms may be tailored to address multiple tumor antigens, modulate polarization states, or carry synthetic payloads to reprogram the TME.</p><p>From a translational perspective, this trial highlights several critical considerations for advancing CAR-M platforms. These include (i) optimizing patient selection based on both tumor antigen density and immune contexture; (ii) integrating real-time, multiomic monitoring to guide therapeutic decisions; and (iii) rationally designing combination regimens, particularly with ICIs, toll-like receptor (TLR) agonists, or metabolic modulators to enhance efficacy and overcome resistance. Collaborative efforts between academic centers and industry partners will be essential to accelerate innovation and clinical translation.</p><p>Moving forward, enhancing the persistence and expansion of CAR-Ms, possibly through cytokine support, synthetic constructs, or combination with immune checkpoint blockade, may improve clinical outcomes [<span>3</span>]. Biomarker-guided patient stratification, especially based on HER2 IHC scores and immune fitness signatures, will be critical in optimizing therapeutic outcomes. Furthermore, strategies to convert early immune activation into sustained tumor regression, such as in vivo reprogramming, combination immunotherapies, or CAR-M modifications for improved antigen retention, are essential for long-term efficacy.</p><p>In summary, this first-in-human trial of CT-0508 establishes a clinical proof-of-concept for CAR-M therapy in HER2-positive solid tumors. While limitations remain, the study demonstrates that macrophage-based therapies can be safely administered, remodel the TME, and engage adaptive immunity. CAR-Ms represent a promising new frontier in cancer immunotherapy, offering a unique mechanism of action that may complement existing T-cell-based approaches [<span>4</span>].</p><p>Wenxue Ma: conceptualization, investigation, writing—original draft, validation, visualization, writing—review and editing, software, formal analysis, project administration, data curation, supervision, resources. Catriona Jamieson: conceptualization, funding acquisition, writing—review and editing, project administration, supervision, resources, validation. Both authors have read and approved the final manuscript.</p><p>The authors have nothing to report.</p><p>The authors declare no conflicts of interest.</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":\"6 9\",\"pages\":\"\"},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2025-08-27\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70374\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.70374\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, RESEARCH & EXPERIMENTAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.70374","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

摘要

在Nature Medicine(2025)最近发表的一项研究中,Reiss等人报道了针对her2过表达实体瘤的嵌合抗原受体巨噬细胞(CAR-M)疗法CT-0508的首次人体一期试验。该试验证明了大量预处理患者的安全性、可行性和早期免疫活性,标志着巨噬细胞免疫疗法的首次临床验证。这一里程碑强调了工程巨噬细胞克服肿瘤微环境(TME)介导的耐药的潜力,标志着目前治疗难治性实体瘤的范式转变。纳入14例her2过表达肿瘤患者,包括乳腺癌、胃食管癌和唾液管癌,并接受CT-0508治疗,之前未接受淋巴消耗化疗。研究了两种给药方案:分剂(组1)和丸剂(组2)。CAR-Ms由自体单核细胞生成,使用无复制能力的腺病毒载体(Ad5)。F35),在所有制成品中实现高活力、纯度和CAR表达。临床前试验显示her2特异性细胞毒性、吞噬作用和促炎细胞因子在抗原接触时的分泌。临床,CT-0508表现出良好的安全性。未观察到剂量限制性毒性、≥3级细胞因子释放综合征(CRS)或免疫效应细胞相关神经毒性综合征(ICANS)。9例患者发生1-2级CRS,未使用皮质类固醇或强化支持治疗。重要的是,该试验避免使用CAR-T疗法标准的淋巴消耗方案,如环磷酰胺或氟达拉滨,从而支持更耐受的门诊递送模式。这种区别代表了实体肿瘤领域的一个主要优势,在实体肿瘤领域,毒性问题和后勤负担限制了细胞治疗的广泛应用。虽然没有观察到客观反应(根据RECIST v1.1),但9例HER2免疫组化(IHC) 3+肿瘤患者中有4例(44.5%)实现了疾病稳定(SD),而5例HER2免疫组化(IHC) 2+肿瘤患者均表现出疾病进展(PD)。在可测量的病变中,有41%的肿瘤体积缩小,62%的患者(包括所有SD患者)循环肿瘤DNA (ctDNA)水平下降。这些分子和放射学证据表明生物活性,尽管是短暂的。有趣的是,一些患者在较晚的时间点表现出ctDNA反弹,这提出了关于反应持久性和维持剂量或顺序治疗策略的潜在需求的问题。这些观察结果加强了在早期试验中整合纵向分子监测的价值,以更好地了解反应动力学并识别早期复发标志物。图1提供了CAR-M治疗的临床工作流程、免疫学变化和未来方向的图形概述。该研究的一个显著特征是临床获益明显依赖于HER2表达强度。仅在HER2 IHC 3+肿瘤患者中观察到应答,其特征是强膜染色。在HER2 IHC 2+患者中观察到非临床获益。此外,超过一半的筛选失败归因于入组时HER2下调,突出了HER2表达的可塑性及其对先前治疗压力或肿瘤演变的易感性[1]。这些发现强调了严格和实时生物标志物评估的迫切需要,以确保最佳的患者选择。随着靶表达异质性不断挑战该领域,开发更动态的患者分层模型,包括连续活检或先进的成像模式,对于识别最有可能从CAR-M治疗中获益的患者至关重要。机制上,CT-0508诱导TME显著重构。单细胞RNA测序(scRNA-seq)分析和配对活检显示抗原呈递基因特征增加,CD8+ T细胞浸润增强,其中许多细胞表现出克隆型扩增和细胞毒性标记(颗粒酶B、穿孔素1、IFN-γ)。这些特征提示抗原扩散和内源性t细胞反应的再激活。达到SD的患者也显示出更高的基线CD4:CD8比率和更低的全身性炎症,这可能是免疫反应性的预测指标。重要的是,这些免疫特征与检查点阻断中看到的相似,表明CAR-M可能为组合策略启动TME。尽管这些有希望的机制见解,治疗效果是短暂的。大多数患者在几个月内出现疾病进展。通过RNAScope原位RNA技术评估,有限的CAR-M在循环中的持久性和肿瘤组织内的低浸润水平可能是导致这一结果的原因。 这就提出了关于如何增强CAR-M保留、肿瘤归巢和肿瘤内生存的关键问题。未来的方法可能包括基因修饰以延长寿命,趋化因子或归巢受体的共同表达,以及与基于储存库的给药系统或重复给药策略的整合。重要的是,这项试验表明,巨噬细胞,历史上难以转导,可以成功地工程利用腺病毒平台,而不损害其表型或功能。这一技术进步克服了一个主要的瓶颈,并为CAR-M的未来迭代铺平了道路,增强了有效载荷,如细胞因子释放构建体、抗原呈递模块或免疫检查点调节剂[2]。此外,该制造过程无需体外扩增,提高了自体CAR-M治疗的可行性和可扩展性,可用于更广泛的临床应用。除了HER2靶向,该试验更广泛的意义在于它验证了巨噬细胞作为过继免疫治疗的可行免疫效应细胞类型。与T细胞不同,巨噬细胞是一种先天免疫细胞,能够浸润肿瘤基质,进行吞噬,并通过抗原呈递激活适应性免疫。它们的髓系谱系减少了对衰竭的易感性,并支持了现成的异体应用的潜力。随着工程平台的发展,CAR-Ms可能会针对多种肿瘤抗原、调节极化状态或携带合成有效载荷来重编程TME。从翻译的角度来看,该试验突出了推进CAR-M平台的几个关键考虑因素。这些包括(i)根据肿瘤抗原密度和免疫环境优化患者选择;(ii)整合实时、多组监测以指导治疗决策;(iii)合理设计联合方案,特别是与ICIs、toll样受体(TLR)激动剂或代谢调节剂联合使用,以提高疗效并克服耐药性。学术中心和行业合作伙伴之间的合作对于加速创新和临床翻译至关重要。展望未来,增强CAR-Ms的持久性和扩张性,可能通过细胞因子支持、合成结构物或与免疫检查点阻断联合,可能改善临床结果。生物标志物引导的患者分层,特别是基于HER2免疫组化评分和免疫适应度特征,将是优化治疗结果的关键。此外,将早期免疫激活转化为持续肿瘤消退的策略,如体内重编程、联合免疫疗法或CAR-M修饰以改善抗原保留,对于长期疗效至关重要。总之,CT-0508的首次人体试验为CAR-M治疗her2阳性实体瘤建立了临床概念验证。尽管局限性仍然存在,但该研究表明,基于巨噬细胞的疗法可以安全地给予,重塑TME,并参与适应性免疫。CAR-Ms代表了癌症免疫治疗的一个有前景的新领域,它提供了一种独特的作用机制,可以补充现有的基于t细胞的方法。马文学:概念化、调查、写作—初稿、验证、可视化、写作—评审与编辑、软件、形式分析、项目管理、数据管理、监督、资源。Catriona Jamieson:概念化,资金获取,写作审查和编辑,项目管理,监督,资源,验证。两位作者已经阅读并批准了最终的手稿。作者没有什么可报告的。作者声明无利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Chimeric Antigen Receptor-Macrophage Therapy Enters the Clinic: The First-in-Human Trial for HER2+ Solid Tumors

Chimeric Antigen Receptor-Macrophage Therapy Enters the Clinic: The First-in-Human Trial for HER2+ Solid Tumors

In a recent study published in Nature Medicine (2025), Reiss et al. [1] reported the first-in-human Phase 1 trial of CT-0508, a chimeric antigen receptor macrophage (CAR-M) therapy targeting HER2-overexpressing solid tumors. This trial demonstrated safety, feasibility, and early immune activity in heavily pretreated patients, marking the first clinical validation of macrophage-based immunotherapy. This milestone highlights the potential of engineered macrophages to overcome tumor microenvironment (TME)-mediated resistance, signaling a paradigm shift for treating solid tumors refractory to current therapies.

Fourteen patients with HER2-overexpressing tumors, including breast, gastroesophageal, and salivary duct carcinomas, were enrolled and received CT-0508 without prior lympho-depleting chemotherapy. Two dosing regimens were explored: fractionated (Group 1) and bolus (Group 2). CAR-Ms were generated from autologous monocytes using a replication-incompetent adenoviral vector (Ad5.F35), achieving high viability, purity, and CAR expression across all manufactured products. Preclinical assays demonstrated HER2-specific cytotoxicity, phagocytosis, and secretion of proinflammatory cytokines upon antigen engagement.

Clinically, CT-0508 exhibited a favorable safety profile. No dose-limiting toxicities, Grade ≥3 cytokine release syndrome (CRS), or immune effector cell-associated neurotoxicity syndrome (ICANS) were observed. Grade 1–2 CRS occurred in nine patients and was managed without corticosteroids or intensive supportive care. Importantly, the trial avoided the use of lympho-depleting regimens such as cyclophosphamide or fludarabine, which are standard in CAR-T therapies, thereby supporting a more tolerable, outpatient-based delivery model. This distinction represents a major advantage in the solid tumor space, where toxicity concerns and logistical burdens have limited broader access to cell therapy.

Although no objective responses (per RECIST v1.1) were observed, four of nine patients (44.5%) with HER2 immunohistochemistry (IHC) 3+ tumors achieved stable disease (SD), while all five patients with HER2 IHC 2+ tumors exhibited progressive disease (PD). Tumor volume reductions were noted in 41% of measurable lesions, and circulating tumor DNA (ctDNA) levels decreased in 62% of patients, including all those with SD. These molecular and radiographic evidence indicate biological activity, although transient. Interestingly, some patients exhibited ctDNA rebound at later timepoints, raising questions about the durability of response and the potential need for maintenance dosing or sequential therapeutic strategies. These observations reinforce the value of integrating longitudinal molecular monitoring in early-phase trials to better understand the kinetics of response and identify early markers of relapse. A graphical overview summarizing the clinical workflow, immunologic changes, and future directions of CAR-M therapy is provided in Figure 1.

A striking feature of the study is the clear dependence of clinical benefit on HER2 expression intensity. Responses were exclusively observed in patients with HER2 IHC 3+ tumors, characterized by strong membranous staining. Non-clinical benefit was observed among HER2 IHC 2+ patients. Furthermore, more than half of screening failures were attributed to HER2 downregulation at the time of enrollment, highlighting the plasticity of HER2 expression and its susceptibility to prior treatment pressure or tumor evolution [1]. These findings highlight the critical need for rigorous and real-time biomarker evaluation to ensure optimal patient selection. As target expression heterogeneity continues to challenge the field, developing more dynamic patient stratification models, including serial biopsies or advanced imaging modalities, will be essential for identifying those most likely to benefit from CAR-M therapy.

Mechanistically, CT-0508 induced significant remodeling of the TME. Single-cell RNA sequencing (scRNA-seq) analysis and paired biopsies revealed increased antigen presentation gene signatures and enhanced infiltration of CD8+ T cells, many of which displayed clonotypic expansion and cytotoxic markers (granzyme B, perforin 1, IFN-γ). These features suggest antigen spreading and reactivation of endogenous T-cell responses. Patients achieving SD also showed higher baseline CD4:CD8 ratios and lower systemic inflammation, pointing to possible predictive markers for immunologic responsiveness. Importantly, these immune signatures resemble those seen in checkpoint blockade and suggest that CAR-M may prime the TME for combinatorial strategies.

Despite these promising mechanistic insights, the therapeutic effects were transient. Most patients experienced disease progression within a few months. Limited CAR-M persistence in circulation and low infiltration levels within tumor tissues, as assessed by RNAScope in situ RNA technology, likely contributed to this outcome. This raises critical questions about how to enhance CAR-M retention, tumor homing, and intratumoral survival. Future approaches may include genetic modifications to enhance lifespan, co-expression of chemokines or homing receptors, and integration with depot-based delivery systems or repeated dosing strategies.

Importantly, this trial demonstrates that macrophages, historically difficult to transduce, can be successfully engineered using adenoviral platforms without compromising their phenotype or function. This technical advance overcomes a major bottleneck and paves the way for future iterations of CAR-M with enhanced payloads, such as cytokine release constructs, antigen-presenting modules, or immune checkpoint regulators [2]. Additionally, the manufacturing process, performed without ex vivo expansion, enhances the feasibility and scalability of autologous CAR-M therapy for broader clinical application.

Beyond HER2 targeting, the broader implications of this trial lie in its validation of macrophages as a viable immune effector cell type for adoptive immunotherapy. Unlike T cells, macrophages are innate immune cells capable of infiltrating tumor stroma, performing phagocytosis, and activating adaptive immunity through antigen presentation. Their myeloid lineage reduces susceptibility to exhaustion and supports potential for off-the-shelf, allogeneic applications. As engineering platforms evolve, CAR-Ms may be tailored to address multiple tumor antigens, modulate polarization states, or carry synthetic payloads to reprogram the TME.

From a translational perspective, this trial highlights several critical considerations for advancing CAR-M platforms. These include (i) optimizing patient selection based on both tumor antigen density and immune contexture; (ii) integrating real-time, multiomic monitoring to guide therapeutic decisions; and (iii) rationally designing combination regimens, particularly with ICIs, toll-like receptor (TLR) agonists, or metabolic modulators to enhance efficacy and overcome resistance. Collaborative efforts between academic centers and industry partners will be essential to accelerate innovation and clinical translation.

Moving forward, enhancing the persistence and expansion of CAR-Ms, possibly through cytokine support, synthetic constructs, or combination with immune checkpoint blockade, may improve clinical outcomes [3]. Biomarker-guided patient stratification, especially based on HER2 IHC scores and immune fitness signatures, will be critical in optimizing therapeutic outcomes. Furthermore, strategies to convert early immune activation into sustained tumor regression, such as in vivo reprogramming, combination immunotherapies, or CAR-M modifications for improved antigen retention, are essential for long-term efficacy.

In summary, this first-in-human trial of CT-0508 establishes a clinical proof-of-concept for CAR-M therapy in HER2-positive solid tumors. While limitations remain, the study demonstrates that macrophage-based therapies can be safely administered, remodel the TME, and engage adaptive immunity. CAR-Ms represent a promising new frontier in cancer immunotherapy, offering a unique mechanism of action that may complement existing T-cell-based approaches [4].

Wenxue Ma: conceptualization, investigation, writing—original draft, validation, visualization, writing—review and editing, software, formal analysis, project administration, data curation, supervision, resources. Catriona Jamieson: conceptualization, funding acquisition, writing—review and editing, project administration, supervision, resources, validation. Both authors have read and approved the final manuscript.

The authors have nothing to report.

The authors declare no conflicts of interest.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信