Abstract The increasing incidence of central nervous system lymphoma (CNSL) is hindered by the blood-brain barrier and costly prolonged drug development. To overcome these obstacles, we developed a carboplatin lock-designed MOF (Pt-MOFs@Glu) targeting intestinal macrophages for brain-directed drug transport. Single-cell RNA sequencing analyses revealed that intestinal macrophages migrate to the brain in response to chemokines. Building on this insight, Pt-MOFs@Glu was designed to engage these cells for precise delivery. Comprising carboplatin, pyrazine-quinoxaline, and β-glucan, the system induces an “avalanche effect” in the CNSL microenvironment, promoting tumor apoptosis and inhibiting metastasis. Combining pyrazine-quinoxaline to lock carboplatin and β-glucan to boost targeting, immunity, and oral absorption, the system enables ROS-triggered drug release, efficiently crosses the gastrointestinal tract and BBB, and synergizes chemo-immunotherapy to enhance therapeutic efficacy. This approach redefines CNSL treatment by harnessing the gut-brain axis, offering a transformative pathway to overcoming therapeutic barriers and improving patient outcomes. __________________________________________________________________ Lock-designed MOF leverages gut-brain macrophage trafficking to trigger an avalanche effect against CNS lymphoma progression. INTRODUCTION Central nervous system lymphoma (CNSL) represents a formidable challenge in oncology, accounting for ~4% of intracranial malignant tumors and with an incidence of about seven cases per million ([40]1, [41]2). This aggressive cancer progresses rapidly if left untreated, with a mean survival time of just 1.5 months, underscoring the urgent need for effective therapeutic strategies ([42]3, [43]4). Current treatment modalities—which include high-dose chemotherapy, targeted therapy, radiotherapy, and immunotherapy—have not substantially improved outcomes, as evidenced by high recurrence and mortality rates in clinical trials. A major obstacle to effective treatment is the blood-brain barrier (BBB), a selective barrier that prevents many drugs from entering the central nervous system (CNS) ([44]5–[45]7). In addition, conventional therapies often result in severe side effects such as leukopenia, dermatological issues, alopecia, nausea, and fatigue, which can notably affect the quality of life of patients. Therefore, overcoming the BBB to facilitate efficient drug delivery to CNSL sites, while minimizing side effects and preventing recurrence, remains a critical clinical imperative ([46]8, [47]9). Traditionally, CNSL treatment has relied heavily on intravenous drug administration. While effective in some respects, intravenous therapy can be invasive, increase the risk of infection, and require clinical supervision ([48]10, [49]11). In contrast, oral anticancer medications offer numerous advantages, including painless administration, a lower risk of contamination, the ability for self-administration, and cost-effectiveness, making them a preferred option for many patients ([50]12, [51]13). However, oral drug delivery systems face notable physiological challenges posed by the gastrointestinal tract (GI) and the BBB, making the development of a safe and effective gut-brain axis drug delivery strategy a key objective ([52]14, [53]15). The concept of the gut-brain axis, introduced in 2011, describes the complex and bidirectional biochemical signaling between the GI and the CNS ([54]16, [55]17). This system involves multifaceted biochemical signaling pathways that enable the gut and the brain to influence each other’s functions, maintaining homeostasis and responding to various physiological and pathological stimuli. This axis includes connections from the gut to the brain through the CNS, immune system, enteric nervous system, vagus nerve, and gut microbiota ([56]18–[57]21). Recent studies have highlighted the notable role of the gut-brain axis in CNS pathologies. Intestinal immune cells, particularly those in the gut-associated lymphoid tissue, can become activated by gut microbes or dietary antigens. Once activated, these immune cells can migrate to the CNS, where they can cross the BBB and potentially influence the progression of neurological diseases through mechanisms such as neuroinflammation and immune modulation ([58]22–[59]25). This suggests that immune cells could potentially be harnessed as vehicles to transport orally administered drugs to the brain. By targeting immune cells in the gut with drug-loaded nanoparticles or other delivery systems, it may be possible to facilitate the efficient delivery of therapeutics across the BBB, thereby enhancing treatment efficacy for CNS diseases. Building on these insights, the study initially used bioinformatics and single-cell RNA sequencing (scRNA-seq) techniques to analyze CNSL. Results revealed that the CNSL microenvironment contains a significant presence of gut-derived macrophages, which can migrate from the intestine to the brain in response to chemokines, such as Cx3cr1 ([60]26–[61]29). Consequently, this study aims to develop a drug delivery platform that harnesses the gut-brain axis to bypass physiological barriers noninvasively, enabling macrophage-mediated transport from the intestine to directly target CNSL treatment. This approach seeks not only to eradicate tumor cells but also to activate the immune system synergistically, providing a comprehensive treatment strategy for CNSL. The proposed platform features a unique oral carboplatin padlock-designed metal-organic framework (MOFs), termed Pt-MOFs@Glu, which is designed to target and treat CNSL ([62]Fig. 1). This system comprises carboplatin as the metal center, pyrazine-quinoxaline as a molecular lock for carboplatin, and β-glucan as both the CNS-targeting and immune-activating component. Fig. 1. A noninvasive oral drug delivery system for gut-to-brain transport and its mechanism of action. [63]Fig. 1. [64]Open in a new tab (A) Bioinformatics and scRNA-seq analysis of human clinical CNSL samples and mouse CNSL samples, and the dashed marked portions are intestinal-derived macrophages in the tumors. (B) System relates to the construction and synthesis process of padlock-designed metal-organic frameworks (Pt-MOFs@Glu). (C) Upon oral administration, Pt-MOFs@Glu are taken up by intestinal M cells and subsequently endocytosed by resident macrophages (Mφ) in situ. These macrophages, carrying the Pt-MOFs@Glu, travel via the lymphatic system to the circulatory system and ultimately cross the blood-brain barrier (BBB) to reach the brain tumor site. Within the brain tumor, Pt-MOFs@Glu release carboplatin in response to high levels of ROS, triggering an avalanche effect to exert a therapeutic impact on the tumor. In addition, this system synergistically enhances chemotherapy and immune response, promotes tumor cell apoptosis, and effectively prevents tumor metastasis. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [65]https://BioRender.com/ef5jegu). The Pt-MOFs@Glu system uses a lock-and-key mechanism, using pyrazine-quinoxaline as a molecular lock to immobilize carboplatin, while reactive oxygen species (ROS) within the tumor microenvironment (TME) act as the “key” to trigger drug release. This dynamic interaction initiates an “avalanche effect,” significantly enhancing the therapeutic efficacy of Pt-MOFs@Glu. This mechanism enables precise drug delivery to the tumor site, thereby reducing systemic toxicity and improving treatment outcomes. Furthermore, the immune-stimulating damage-associated molecular pattern (DAMP) proteins released upon tumor cell death interact synergistically with β-glucan, a potent immune adjuvant, to activate the immune system ([66]30, [67]31). This combined chemotherapy-immunotherapy approach is finely tuned to the TME, offering a robust and comprehensive strategy for treating CNSL. The Pt-MOFs@Glu represents a notable advancement in CNSL treatment, leveraging the gut-brain axis to induce an avalanche effect within the CNSL microenvironment. By synergistically integrating the advantages of chemotherapy and immunotherapy and harnessing the distinctive dynamics of the gut-brain axis, this system heralds a groundbreaking approach to CNSL treatment, promising to significantly enhance patient outcomes. RESULTS Acquisition and analysis of scRNA-seq Single-cell transcriptome data for human CNSL and normal human brain samples were obtained as raw datasets from the ArrayExpress platform. The CellRanger software (version 2.0.0; [68]http://software.10xgenomics.com/single-cell/overview/welcome) ([69]32) was used to process each sample’s raw data, generating relevant gene expression profiles ([70]Fig. 2, A to C). Cell-type identification, based on distinct cell-specific markers, revealed the presence of B cell, dendritic cell, endothelial cell, macrophage, meningeal cell, cancer cells, and T and NK cell ([71]Fig. 2, B and C). Notably, macrophages represented the largest proportion of cell types when comparing normal brain samples to those of CNSL ([72]Fig. 2, D and I). Given their prominence as a central component of the TME, macrophages were isolated for detailed analysis. Fig. 2. Bioinformatics analysis of scRNA-seq in CNSL tissue and normal human brain, along with immunofluorescence from patients with CNSL. [73]Fig. 2. [74]Open in a new tab (A) Schematic representation of the experimental workflow. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [75]https://BioRender.com/ef5jegu). (B) UMAP plot for the analysis of cell subpopulations in the CNSL, which were divided into seven cell subsets. (C) UMAP plot for the analysis of cell subpopulations in the normal human brain, which were divided into seven cell subsets. (D) Comparison of intracellular macrophage content between CNSL tissues and normal brain tissues. Expression of intestinal macrophage markers in CNSL-associated macrophages, including CD11c (E) and CD14 (F). (G) Volcano plot highlighting differentially expressed genes between CNSL and normal brain samples, with significantly up-regulated and down-regulated genes marked in red and blue, respectively [P < 0.05, log[2](fold change) > 1]. (H) Gene Ontology (GO) annotation and KEGG pathway analysis of ESRP1 in CNSL using LinkedOmics. Confocal laser scanning microscopy (CLSM) images reveal that macrophages present in CNSL tissues are derived from intestinal macrophages. (l) UMAP visualization highlighting macrophage populations across human intestinal tissue and PBMCs. Although all three clusters were identified in both tissues, cluster 5 was predominantly found in PBMC-derived macrophages, whereas cluster 11 was more dominant in intestine-derived macrophages. (J) Annotated UMAP visualization of CD45^+ immune cell subsets derived from both human intestinal tissue and PBMCs. (K) UMAP visualization of CD45^+ immune cell subsets derived from either human intestinal tissue or PBMCs. (L) UMAP visualization highlighting macrophage populations across human intestinal tissue and PBMCs. (M) Heatmap showing the expression profiles of macrophage-related genes in human intestinal tissue and PBMCs. (N) Expression levels of intestinal macrophage markers in CNSL-associated macrophages, including LSTQ, LYZ, THBS1, HLA-DQA1, and HLA-DQB1. Our findings indicate a notably higher level of macrophage infiltration in CNSL tissue compared to normal brain tissue ([76]Fig. 2, B to D), suggesting a correlation between macrophage presence and CNSL development. Further analysis of 4346 qualified macrophage-related genes highlighted key markers indicative of the macrophages’ origin and behavior in CNSLs ([77]Fig. 2E). Genes associated with intestinal macrophages, including CD11c and CD14 (a key marker for human intestinal macrophages), exhibited increased expression in CNSL-associated macrophages. These results support a model in which intestinal macrophages are a primary source of macrophages within CNSLs. To explore the mechanisms underlying macrophage migration from the gut to CNSL, we performed gene set enrichment analysis to identify the top 20 significant Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. The analysis revealed that macrophage-related pathways in CNSLs are predominantly associated with interleukin signaling and immunomodulatory interactions within the innate immune system ([78]Fig. 2, F and G). These findings indicate a potential link between macrophage infiltration in CNSLs and the activation of the chemokine pathway. Encouraged by the favorable bioinformatics analysis of clinical samples, we also collected tissue samples from patients with clinical neurological lymphoma for immunofluorescence analysis ([79]Fig. 2I). The results revealed a significant presence of intestinal macrophage-associated proteins, such as CD11c and CD14, in the CNSL tissue samples ([80]Fig. 2I). These proteins were highly expressed in CNSLs. Although CD14 is commonly used as a marker for intestinal macrophages, its expression is not exclusive and may also be detected in other immune cell types. To refine the identification of intestinal macrophages, we further interrogated publicly available datasets and used dataset [81]GSE125527, which incorporated single-cell RNA sequencing data (scRNA-seq) data of CD45^+ immune cells from both human peripheral blood and intestinal tissue ([82]Fig. 2, J to N). Three distinct macrophage subsets—clusters 5, 10, and 11—were identified from the intestine and peripheral blood immune cells ([83]Fig. 2, J and K). Although all three clusters were identified in both tissues, cluster 5 was predominantly found in Peripheral blood mononuclear cell (PBMC)–derived macrophages, whereas cluster 11 was more dominant in intestine-derived macrophages ([84]Fig. 2l). Transcriptomic profiling of these clusters revealed that certain genes, such as LYZ and LST1, were highly expressed in both cluster 5 and cluster 11. In contrast, certain marker genes were specifically highly expressed in one cluster: THBS1 showed low expression in cluster 11 but was markedly elevated in cluster 5, whereas HLA-DQA1 and HLA-DQB1 were low in cluster 5 but highly expressed in cluster 11([85]Fig. 2M). We then validated those marker gene expression in CNSL dataset. LYZ, LST1, HLA-DQA1, and HLA-DQB1 were all highly expressed in CNSL-associated macrophages, while THBS1 expression remained low ([86]Fig. 2N). This finding suggests a shared similarity between intestine-derived macrophages and CNSL-associated macrophages, supporting the hypothesis that intestinal macrophage cells may infiltrate or be recruited into the CNSL microenvironment. These findings suggest that intestinal macrophages—particularly those characterized by unique expression of THBS1, HLA-DQA1, and HLA-DQB1—have the capacity to migrate and function within the CNSL. This lays a theoretical foundation for exploring the potential of intestinal macrophages as carriers for gut-brain axis–targeted drug delivery in future therapeutic strategies. To establish an orthotopic CNSL model, we injected A20 lymphoma cells into mouse brains. Comparative analysis between the brains of these CNSL model mice and healthy controls revealed the presence of enteric-derived macrophages, marked by CD11c and CD64 (a key marker of mouse intestinal macrophages), within CNSL tissue ([87]Fig. 3, A to F). Furthermore, we identified elevated expression of chemokine receptor–associated genes in these intestinal macrophages, including Cx3cr1, Ccr1, Ccr2, and Ccr5 ([88]Fig. 3, G to J). These findings suggest that macrophages in CNSLs primarily originate from the gut and are recruited to the brain via chemokine receptor signaling (Cx3cr1, Ccr1, Ccr2, and Ccr5). In addition, CNSL tissue samples from mice were collected for immunofluorescence analysis ([89]Fig. 2, K and L). The results revealed a significant presence of intestinal macrophage-associated proteins, including CD11c and CD64, within the CNSL tissues ([90]Fig. 2K). Moreover, these intestinal macrophages exhibited elevated expression of chemokine receptor–related genes, such as Cx3cr1 ([91]Fig. 2L). This evidence lays a promising foundation for using intestinal macrophages to deliver nanomedicine, such as padlock-designed MOFs, to the brain as a potential therapeutic strategy for CNSLs. Fig. 3. scRNA-seq analysis of mouse CNSL tissue and normal brain, along with immunofluorescence findings in patients with CNSL. [92]Fig. 3. [93]Open in a new tab (A) Schematic representation of the experimental workflow. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [94]https://BioRender.com/ef5jegu). (B) UMAP plot for the analysis of cell subpopulations in the CNSL, which were divided into six cell subsets. (C) UMAP plot for the analysis of cell subpopulations in the normal mouse brain, which were divided into six cell subsets. (D) Comparative analysis of macrophage content between CNSL and normal brain tissues. (E and F) Expression of mesenteric macrophage markers in CNSL-associated macrophages, including CD11c (E) and CD64 (F). (G to J) Expression of chemokine receptor-related genes in CNSL-associated macrophages, including Cx3cr1 (G), Ccr1 (H), Ccr2 (I), and Ccr5 (J). (K and L) CLSM images showing high expression of enteric-derived macrophage markers (CD11c and CD64) (K) and the chemokine receptor Cx3cr1 (L) in CNSL tissue macrophages. (M) UMAP plots showing the distribution of cell subsets in CNSL tissue, blood, and intestine. (N) UMAP plots highlighting macrophage populations from CNSL tissue, blood, and intestine. (O) Principal components analysis (PCA) plots of macrophages identified by scRNA-seq and color-coded by tissue of origin (blood, CNSL, or intestine). (P) Heatmap depicting the intergroup distances between macrophages from blood, CNSL, and intestine. (Q) Violin plots showing the number of unique transcripts and genes detected per cell across the scRNA-seq dataset; each dot represents an individual cell. (R) Schematic illustrating the injection of flow-sorted intestinal macrophages into the intestine and their subsequent migration and homing to brain tumor sites in CNSL-bearing mice. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [95]https://BioRender.com/ef5jegu). (S) Schematic representation of the flow sorting and labeling of intestinal macrophages prior to injection. (T) Confocal microscopy images of CNSL tissue following immunohistochemical staining to detect transplanted intestinal macrophages. DAPI, 4′,6-diamidino-2-phenylindole; SSC, side scatter; FSC, forward scatter. To strengthen the evidence for the intestinal origin of macrophages in CNSL, we carried out scRNA-seq on additional CNSL tissue, mouse intestinal tissue, and peripheral blood samples ([96]Fig. 3M). Diverse immune cell populations, particularly macrophages, were identified across all samples. To delineate the relationship among macrophages from different tissues, we extracted macrophage subsets and conducted uniform manifold approximation and projection (UMAP) analysis ([97]Fig. 3N), which revealed partial overlap between intestinal and CNSL-derived macrophages. Principal components analysis (PCA) ([98]Fig. 3O) further clarified this relationship, showing substantial overlap between brain- and intestine-derived macrophages, while blood-derived macrophages remained largely distinct. Quantitative assessment of cellular distances ([99]Fig. 3P) demonstrated that CNSL macrophages were more closely related to intestinal macrophages than to those from blood, suggesting a common or highly similar origin. Comparative analysis of surface markers ([100]Fig. 3Q) revealed that CD74 was ubiquitously expressed across macrophages from all tissues, Cd4 was predominantly restricted to blood macrophages, and Itgax (also known as CD11c) was highly expressed in both intestinal and CNSL macrophages. These findings indicate that intestinal macrophages expressing high levels of Itgax likely contribute to the macrophage population within CNSL lesions. To functionally validate the migratory potential of intestinal macrophages, we isolated CD45^+ CD11c^+ macrophages from the intestine via flow cytometry and surgically injected them into the intestinal lumen of recipient mice ([101]Fig. 3, R and S). Notably, within 4 hours postinjection, labeled macrophages were detected in the brain ([102]Fig. 3T), providing direct evidence that intestinal macrophages can traffic to the CNS. Together, these results support the concept that intestinal macrophages contribute to CNSL macrophage populations and establish a mechanistic basis for exploiting gut-derived macrophages in gut-brain axis–targeted therapies Characteristics of Pt-MOFs@glu To attain the desired attributes for effective cancer therapy, it is crucial to regulate the dimensions of nanoparticles. In this study, we synthesized padlock-designed Pt-MOFs through a hydrothermal process that involves carboplatin and pyrazinequinoxaline, with the specific synthesis procedure detailed in [103]Fig. 4A. The particle size was meticulously controlled by varying the reaction time. For enhanced tumor tissue penetration, the padlock-designed Pt-MOFs were synthesized at 180°C for 2 hours, resulting in an appropriate particle size of 130 to 150 nm ([104]Fig. 4B). It is common knowledge that diminutive nanoparticles are more readily internalized by cancer cells owing to their size and surface characteristics. Furthermore, tumor targeting can be accomplished by utilizing nanoparticles within the range of 50 to 200 nm. ([105]33) Fig. 4. Characteristics of as-synthesized Pt-MOFs@Glu. [106]Fig. 4. [107]Open in a new tab (A) Composition and structure: Details of the composition and structure of the proposed Pt-MOFs@Glu. Pt-MOFs were formed from carboplatin and pyrazine-quinoxaline by solvothermal and became ROS-responsive drug-releasing nanoparticles by linking glucan. (B) Scanning electron microscopy (SEM) image of Pt-MOFs: SEM image depicting the morphology of Pt-MOFs. (C) Ultraviolet-visible (UV-Vis) spectra: UV-Vis spectra comparison of carboplatin, Pt-MOFs, and Pt-MOFs@Glu. (D and E) Spectral analysis: UV-Vis spectra (D) and mass spectrometry (MS) data (E) of Pt-MOFs@Glu after simulating the TME. (F) XPS analysis: X-ray photoelectron spectroscopy (XPS) data of Pt-MOFs@Glu, both pre- and postsimulation of the TME. (G) Fourier-transform infrared (FTIR) spectra: FTIR spectra of pyrazino[2,3-f] quinoxaline, carboplatin, Pt-MOFs, glucan, and Pt-MOFs@Glu. (H) SEM image of Pt-MOFs@Glu: Detailed SEM image of Pt-MOFs@Glu showcasing its surface characteristics. (I) TEM image of Pt-MOFs@Glu: TEM image of Pt-MOFs@Glu for a closer view of its internal structure. (J) Elemental mapping: Elemental mapping images of Pt-MOFs@Glu nanoparticles, captured using aberration-corrected high angle annular dark field scanning transmission electron microscope (AC-HAADF-STEM), providing a comprehensive elemental distribution analysis. (K) X-ray diffraction (XRD) patterns: XRD patterns of Pt-MOFs@Glu. (L) TEM images of Pt-MOFs@Glu at various time points (0, 10, 20, and 30 min) under ROS conditions within the TME. a.u., absorbance unit; m/z, mass/charge ratio. To achieve uniform nanoparticles with consistent size and morphology, we selected a feed ratio of 1:1 for the synthesis process using dynamic light scattering (DLS) detection. The optimized nanoparticles exhibited a uniform spherical shape ([108]Fig. 4B) with a subtly uneven surface, boasting a particle size of 180.06 ± 10.52 nm and a zeta potential of −29.48 ± 1.16 mV (n = 3; fig. S2). The morphology of the padlock-designed Pt-MOFs remained virtually unaltered even after 1 month of storage in deionized water at 4°C, underscoring its remarkable stability during storage (fig. S3). To validate the efficacy of padlock-designed Pt-MOFs in harnessing the TME to induce the release of carboplatin, a comprehensive suite of analytical techniques was deployed, including DLS, ultraviolet (UV)–visible spectroscopy, mass spectrometry, and x-ray photoelectron spectroscopy (XPS). DLS analysis, as presented in fig. S4, revealed a notable increase in the particle size of padlock-designed Pt-MOFs within the TME. UV-visible spectroscopy results, shown in [109]Fig. 4C, demonstrated the absence of a characteristic absorption peak for carboplatin at 229 nm in a normal physiological conditions [phosphate-buffered saline (PBS) = 7.4] solution. This is attributable to the conjugated double bonds in pyrazine-quinoxaline, which produce a strong absorption peak at this wavelength, thereby giving pyrazine-quinoxaline their ultraviolet absorption properties. Notably, under normal conditions, there was minimal carboplatin release (fig. S5). In contrast, exposure to the TME, characterized by elevated hydrogen peroxide (H[2]O[2]) levels, led to the emergence of a prominent absorption peak for carboplatin at 229 nm, as illustrated in [110]Fig. 4D. In addition, evaluating a range of ROS concentrations in vitro provides a comprehensive assessment of the stability and responsiveness of Pt-MOFs@Glu under varying oxidative stress conditions (fig. S6). Our results demonstrate that higher ROS levels notably enhance the avalanche effect of Pt-MOFs@Glu, resulting in a more efficient and controlled release of carboplatin. Conversely, at lower ROS concentrations, Pt-MOFs@Glu shows limited capability to release carboplatin. This demonstrates a substantial release of carboplatin under tumor-specific conditions. The findings are further supported by mass spectrometry analysis and quantitative carboplatin release data, as illustrated in [111]Fig. 4E and fig. S7, providing robust confirmatory evidence. Concurrently, XPS was used to probe for any potential changes in the valence state of carboplatin within the TME ([112]Fig. 4F). The analysis indicated that the valence state change of divalent platinum (Pt) in the MOFs, designed with a platinum padlock, remained negligible before and after exposure to simulated TME conditions (fig. S8). This confirms the stability of carboplatin in these circumstances. The aforementioned findings comprehensively illustrate the efficacy and stability of the carboplatin/pyrazine-quinoxaline nanocomposite in facilitating the controlled release of carboplatin within the TME. The desired nanocomplex has been successfully obtained, yet a challenge remains in directing it to the brain via oral administration. In our prior research, we found that oral administration of β-glucan–conjugated drugs effectively targets intestinal macrophages via the intestinal epithelial barrier (IEB), capitalizing on the tumor homing effect to traverse the BBB and reach the site of brain tumors. ([113]14) To facilitate oral brain targeting of padlock-designed Pt-MOFs, we covalently linked them with thioethylated β-glucan and characterized their structure using Fourier transform infrared (FTIR) spectroscopy. The structural characteristics of the synthesized Pt-MOFs@Glu were analyzed using FTIR spectroscopy, which revealed a distinct absorption peak at 810 cm^−1, corresponding to the H-N-H stretching mode associated with cisplatin ([114]Fig. 4G). Notably, the spectrum of the synthesized padlock-designed MOFs exhibited a characteristic absorption peak of the drug at a similar wavenumber, along with a sulfhydryl absorption peak absent in the drug and ligand alone, signifying the successful synthesis of glucan padlock-designed Pt-MOFs (i.e., Pt-MOFs@Glu). Simultaneously, SEM revealed that padlock-designed Pt-MOFs exhibited a smoother surface upon glucan coating ([115]Fig. 4H). Moreover, the shell-core structure was observed via transmission electron microscopy (TEM; [116]Fig. 4I). The elemental profiling of STEM samples of presynthesized glucan-modified nanoparticles was conducted using energy-dispersive X-ray spectroscopy (EDS), indicating the presence of Pt, C, O, N, and S elements ([117]Fig. 4J). The nearly exclusive derivation of S elements from glucan further suggests the effective grafting of glucan onto padlock-designed Pt-MOFs. To confirm the crystal structure of padlock-designed Pt-MOFs and Pt-MOFs@Glu, PXRD analysis was conducted. [118]Figure 4K and fig. S9 demonstrate that the formation of Pt-MOFs@Glu, derived from glucan-modified padlock-designed Pt-MOFs, does not alter the crystal structure of padlock-designed Pt-MOFs. To further confirm its therapeutic potential, Pt-MOFs@Glu was tested for carboplatin release under ROS conditions within the TME, leveraging an avalanche effect. TEM image observations, as shown in [119]Fig. 4l, revealed that Pt-MOFs@Glu rapidly disintegrates under these ROS conditions, validating its ability to swiftly release carboplatin. These findings support the application of Pt-MOFs@Glu in treating CNSL by enabling accelerated carboplatin release through the avalanche effect triggered by TME ROS. Cellular uptake in Pt-MOFs@Glu and cytotoxicity To achieve phagocytosis by intestinal macrophages, facilitating subsequent entry into brain tumor sites, efficient absorption of padlock-designed MOFs by these macrophages is imperative. The mouse macrophage cell line (RAW264.7) displays a notable ability to recognize trehalose via its Dectin-1 receptor ([120]Fig. 5A and fig. S10). To assess the uptake of Pt-MOFs@Glu by RAW264.7, these padlock-designed MOFs were prelabeled with Cy5.5. Laser confocal microscopy revealed a rapid uptake of Pt-MOFs@Glu by RAW264.7, peaking at 3 hours ([121]Fig. 5B). Fig. 5. In vitro characteristics of Pt-MOFs@Glu. [122]Fig. 5. [123]Open in a new tab (A) Transport mechanism: Pt-MOFs@Glu are transported by macrophages across the BBB to the brain tumor. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [124]https://BioRender.com/ef5jegu). (B) Cellular uptake: CLSM images showing Pt-MOFs@Glu uptake by RAW264.7 macrophages. (C) Cytotoxicity to macrophages: Evaluation of the cytotoxicity of free carboplatin and Pt-MOFs@Glu on RAW264.7 macrophages. (D) Cytotoxicity to A20 cells: Assessment of the cytotoxicity of free carboplatin and Pt-MOFs@Glu on A20 lymphoma cells. (E) Tumor spheroid penetration: CLSM images demonstrating the penetration of free Pt-MOFs@Glu and macrophage-hitchhiked Pt-MOFs@Glu into lymphoma spheroids. * denotes statistically significant (P < 0.05). Contrastingly, the pretreatment of RAW264.7 with a Dectin-1 receptor inhibitor significantly diminished the uptake of Pt-MOFs@Glu (fig. S10). These findings strongly imply that the efficient absorption of padlock-designed MOFs by macrophages is facilitated by the Dectin-1 receptor. Furthermore, as proficient phagocytes, intestinal macrophages exhibit a remarkable ability to engulf a substantial number of padlock-designed MOFs. This underscores their potential as carriers for targeted CNSL drug delivery. Evaluating the cytotoxicity of Pt-MOFs@Glu on intestinal macrophages is paramount to ensure their biosafety during phagocytosis and subsequent transport to CNSL sites. Various concentrations of Pt-MOFs@Glu were incubated with intestinal macrophages, and cell viability was assessed using the methylthiazolyldiphenyl-tetrazolium bromide (MTT) method ([125]Fig. 5C). A comparable concentration of free drug (carboplatin) served as the control. The macrophage viability decreased significantly in a dose-dependent manner in the presence of free drugs. Conversely, macrophages treated with Pt-MOFs@Glu exhibited heightened activity, indicating superior biosafety during transportation. To further elucidate the in vitro cytotoxicity of Pt-MOFs@Glu against CNSL cells, diverse concentrations of these MOFs were incubated with the cells ([126]Fig. 5D). As a control, an equivalent concentration of free drug (platinum) was used. Following the cell viability experiment, MTT testing was conducted. Strikingly, Pt-MOFs@Glu demonstrated enhanced cytotoxicity against CNSL compared to free drugs. These findings suggest that Pt-MOFs@Glu exhibits excellent biosafety during transit. Once internalized by tumor cells, it can activate carboplatin through ROS triggers, initiating an avalanche effect that significantly amplifies tumor cell death ([127]Fig. 4L). Macrophages demonstrate the remarkable ability to infiltrate tumor tissues, facilitated by a cascade of chemical mediators emanating from hypoxic tumor environments. To assess the tumor tissue penetration of macrophage-conveyed Pt-MOFs@Glu, we established a tumor cell sphere model. In this model, free Pt-MOFs@Glu served as the control, predominantly adhering to the outer layer of the cell sphere upon co-incubation. Conversely, macrophage-facilitated Pt-MOFs@Glu exhibited superior penetration into the tumor cell sphere. Notably, over time, the Pt-MOFs@Glu gradually permeated the inner core of the cell sphere, as depicted in [128]Fig. 5E. These findings underscore the capacity of intestinal macrophages to enhance the diffusion of Pt-MOFs@Glu into the hypoxic regions of tumors, thereby addressing the limitations associated with conventional nanoparticles. Pt-MOFs@Glu stability assessment As a pivotal component of oral drug delivery, Pt-MOFs@Glu must endure the rigors of the GI with unwavering stability. To scrutinize this critical aspect, in vitro experiments were conducted, wherein the Pt-MOFs@Glu underwent immersion in simulated gastric fluid (SGF; pH 2.0) and simulated intestinal fluid (SIF; pH 7.0) at 37°C. As depicted in the fig. S11, the morphology and particle size of the Pt-MOFs@Glu following exposure to SGF and SIF closely mirrored those of the untreated counterparts. This compelling observation signifies that even amid the harsh milieu of the GI, the Pt-MOFs@Glu maintained their structural integrity, remaining steadfast and unscathed. Furthermore, an in vitro simulation of the TME was meticulously conducted using hydrogen peroxide, with glutathione and physiological conditions (PBS 7.0) serving as the control ([129]Fig. 4E). The results indicated that Pt-MOFs@Glu, in the presence of hydrogen peroxide, triggered an avalanche effect that rapidly released carboplatin. In contrast, it remained inert when exposed to glutathione (fig. S12). This behavior underscores the Pt-MOFs@Glu remarkable ability to selectively respond to hydrogen peroxide within the TME, thereby facilitating the targeted release of carboplatin—a pivotal advancement in tumor treatment strategies. Pathways of absorption in Pt-MOFs@glu In this study, macrophage recruitment plays a crucial role in the therapeutic approach using padlock-designed MOFs for CNSL. As shown in [130]Fig. 2, both bioinformatics analyses and examination of two additional clinical CNSL samples revealed significantly higher macrophage infiltration in patients with CNSL compared to normal human tissues. This elevated macrophage presence was also observed in a CNSL mouse model ([131]Fig. 3). These findings underscore the potential of using orally administered padlock-designed MOFs, which can be selectively taken up by macrophages and transported to the brain, as a promising treatment strategy for CNSL. To elucidate the intricate pathway of f-Pt-MOFs@Glu from the intestine to tumors, we meticulously examined their biodistribution following oral administration. Using euthanized mice, we extracted and processed both villi and Peyer’s patches from the GI, alongside CNSL specimens. These tissues were meticulously examined ex vivo using confocal laser scanning microscopy (CLSM) to delineate the distribution of f-Pt-MOFs@Glu, thereby unraveling the intricate nanoparticle gut-brain axis delivery pathways. Our findings unveiled a targeted affinity of Pt-MOFs@Glu toward M cells within Peyer’s patches ([132]Fig. 6A). Subsequently, these nanoparticles traversed into the villi, where they were efficiently engulfed by local macrophages ([133]Fig. 6B). These macrophages, acting as efficient carriers, facilitated the transportation of nanoparticles through the lymphatic ducts ([134]Fig. 6C), thus providing a conduit into the systemic circulation. Ultimately, these nanoparticles breached the BBB, accumulating at the site of CNSL ([135]Fig. 6, D and E). Fig. 6. The transport route of Pt-MOFs@Glu. [136]Fig. 6. [137]Open in a new tab (A to C) Colocalization in the intestinal tract: Schematic depictions and CLSM images showing the colocalization of Pt-MOFs@Glu with M cells (A), lymphatic vessels (B), and resident macrophages (C) in the intestinal tract. (D) Transport to brain tumors: Pt-MOFs@Glu are carried by macrophages into brain tumors. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [138]https://BioRender.com/ef5jegu). (E and F) Brain tumor imaging: CLSM images of brain tumors without (E) and with macrophage blockers (F). (G) Ex vivo IVIS imaging: Ex vivo images from the IVIS showing the accumulation of Pt-MOFs@Glu. (H) Real-time IVIS imaging: Real-time IVIS images depicting the accumulation of Pt-MOFs@Glu in brain tumors at specified times following oral administration in mice. (I) Radiant efficiency analysis: Corresponding total radiant efficiencies from ex vivo IVIS images showing the accumulation of Pt-MOFs@Glu. (J) Carboplatin content in brain tissue: Measurement of carboplatin levels in brain tissue across different groups: Control, carboplatin, Pt-MOFs + glucan, Pt-MOFs, and Pt-MOFs@Glu. Moreover, our study used intravenous injection of chlorophosphate liposomes to induce macrophage depletion, thereby inhibiting the uptake of Pt-MOFs@Glu by macrophages. This intervention served to corroborate the absorption pathway of Pt-MOFs@Glu (fig. S13). The absence of fluorescence observed in the brain ([139]Fig. 6F and fig. S14) confirmed that Pt-MOFs@Glu, upon engulfment by macrophages, indeed traversed through the lymphatic system into the bloodstream. Consequently, they successfully crossed the BBB, thereby penetrating the brain to combat CNSL. To further elucidate the mechanism governing macrophage migration, we investigated the role of the chemokine receptor Cx3Cr1, a key regulator of macrophage trafficking. To selectively inhibit Cx3Cr1 in intestinal macrophages, we administered JMS-17, a specific Cx3Cr1 inhibitor, to CNSL model mice. Following this intervention, we evaluated the targeted delivery of Pt-MOFs@Glu to brain tumor sites (fig. S14). Our findings revealed that Cx3Cr1 inhibition completely abrogated the accumulation of Pt-MOFs@Glu at the tumor site, confirming the critical role of Cx3Cr1 in mediating macrophage homing and recruitment from the intestine to CNSL. These results provide compelling evidence that CNSLs harbor a substantial population of gut-derived macrophages, which infiltrate the TME through Cx3Cr1-dependent pathways. This study underscores the significance of intestinal-immune interactions in CNSL pathogenesis and identifies Cx3Cr1 as a promising therapeutic target for modulating macrophage infiltration. In vivo distribution analysis of Pt-MOFs@Glu We explored the potential of Pt-MOFs@Glu as a gut-brain axis nanodelivery system using an orthotopic CNSL mouse model. The experimental group received orally administered Pt-MOFs@Glu, while the control group received oral deionized water. Using an in vivo imaging system (IVIS), we tracked the distribution of Pt-MOFs@Glu postadministration ([140]Fig. 6, G to I). Notably, no fluorescent signals were detected in brain tissues of the control group. In contrast, in the study, CNSL mice that were orally administered the platinum padlock nanodrug exhibited detectable fluorescence signals in the brain as early as 4 hours postadministration. Ex vivo imaging of tissues was performed 24 hours postadministration, with quantification of fluorescence intensity in each tissue ([141]Fig. 6G). These signals progressively intensified, culminating in peak fluorescence levels at 12 hours. Consequently, 12 hours postadministration was selected as the optimal time point for subsequent in vitro brain experiments. Ex vivo fluorescence analysis revealed the predominant accumulation of Pt-MOFs@Glu in brain tumors, liver, and kidneys following oral administration. These findings suggest that Pt-MOFs@Glu can surmount the formidable barriers to oral drug delivery, facilitated by intestinal macrophages traversing the IEB and the BBB, ultimately reaching the site of CNSL. To precisely assess the delivery efficiency of carboplatin in Pt-MOFs@Glu, we used inductively coupled plasma mass spectrometry (ICP-MS) to analyze carboplatin concentrations in CNSL. This analysis aimed to demonstrate the efficacy of carboplatin drugs in reaching CNSL following oral administration ([142]Fig. 6J). The experimental group received Pt-MOFs@Glu orally, while the control group received an equivalent dosage of free cisplatin. Results revealed that the concentration of Pt-MOFs@Glu at the brain tumor site in the experimental group exceeded that of the control group by 15-fold. This unequivocally confirms the capacity of Pt-MOFs@Glu to effectively leverage intestinal macrophages for targeted delivery to the CNSL site. Antitumor effectiveness Motivated by the exceptional performance demonstrated by Pt-MOFs@Glu, we proceeded to assess their anticancer efficacy in vivo. Primary CNSL was meticulously established through intracranial injection of A20 lymphoma cells into Balb/c mice. The resultant CNSLs were subsequently stratified into five distinct experimental groups for comprehensive investigation: an untreated control group receiving deionized water, a group receiving free carboplatin, a group with carboplatin equivalent to Pt-MOFs, a group administered free glucan, and a final group treated with Pt-MOFs@Glu ([143]Fig. 7A). Fig. 7. Antitumor efficacy of Pt-MOFs@Glu in mice bearing CNSL. [144]Fig. 7. [145]Open in a new tab (A) Study design: Schematic representation of the time course for the establishment of an orthotopic brain tumor model in mice and the treatment regimen. Treatment was administered on day 7 after modeling and the trial cycle ended on day 35. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [146]https://BioRender.com/ef5jegu). (B) Health metrics: Body weights and survival rates of mice following various treatments. (C) Brain pathology: Pathological sections of brain tissue from mice treated for CNSL. (D and E) Tumor analysis: TUNEL and Ki67 staining of tumor tissues showing pathological changes across different treatment groups. * denotes statistically significant (P < 0.05). Throughout the treatment period, we meticulously documented changes in body weight ([147]Fig. 7B) and survival rates ([148]Fig. 7B) across all groups. Upon completion of the experiment, the tumors were excised and subjected to hematoxylin and eosin (H&E) staining to evaluate the changes in tumor size posttreatment, as depicted in [149]Fig. 7C. While treatment with free carboplatin and free glucan exhibited some degree of antitumor activity, it was accompanied by significant reductions in body weight and poor survival rates, indicative of insufficient tumor growth inhibition. Notably, Pt-MOFs@Glu, when orally administered, traverses the body via M cells, undergoes phagocytosis, and is then transported by macrophages into the brain, where they accumulate at the CNSL site. Subsequently, the substantial quantity of ROS present in CNSL instigates an avalanche effect within the Pt-MOFs@Glu, resulting in the release of a significant amount of carboplatin, which effectively eradicates the tumor cells. Pt-MOFs@Glu therapy demonstrated significantly heightened antitumor efficacy compared to the control group, as evidenced by notable improvements in body weight, survival rates, and reductions in both tumor volume (P < 0.05). Furthermore, the oral administration of Pt-MOFs@Glu exhibited superior tumor cell eradication (H&E staining; [150]Fig. 7C), an increased presence of apoptotic cells [terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate nick end labeling (TUNEL) measurement; [151]Fig. 7D], and a lower proportion of proliferating cells (Ki67 staining; [152]Fig. 7E) compared to other control groups. These findings underscore the potency of Pt-MOFs@Glu, featuring multiple platinum padlocks, in effectively combating CNSL, offering promising prospects in cancer therapeutics. Inspired by the exceptional efficacy of Pt-MOFs@Glu in the treatment of CNSL in vivo, we sought to elucidate the underlying mechanisms contributing to its remarkable performance. We hypothesized that the effectiveness of Pt-MOFs@Glu stems from its dual action: The release of a substantial amount of carboplatin through an ROS-induced avalanche effect and the release of glucan, a potent immune adjuvant, which has been previously validated in cellular experiments (as shown in the fig. S15). Glucan is known to activate the body’s immune response, thereby aiding in the eradication of tumors and preventing recurrence postchemotherapy ([153]31). To test this hypothesis, we assessed the immune activation potential of Pt-MOFs@Glu in a CNSL mouse model ([154]Fig. 8A). Compared to the untreated CNSL control group, mice treated with Pt-MOFs@Glu for 2 weeks exhibited a marked increase in brain-infiltrating immune cells, including CD4^+ T cells ([155]Fig. 8B), CD8^+ T cells ([156]Fig. 8C), natural killer (NK) T cells (fig. S16), and effector T cells (fig. S17). Fig. 8. Pt-MOFs@Glu prevents relapse postchemotherapy through immune activation. [157]Fig. 8. [158]Open in a new tab (A) Immune activation mechanism: Pt-MOFs@Glu promotes the maturation of dendritic cells (DCs) and subsequently acts on lymph nodes to activate T cells. This activation leads to tumor cell apoptosis and suppression of tumor growth. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [159]https://BioRender.com/ef5jegu). (B and C) T cell analysis: Evaluation of intratumoral CD4^+ and CD8^+ T cell populations in CNSL mice following treatment with different therapeutic strategies. (D to G) Cytokine and immune marker analysis: Quantification of immune-related markers and cytokines in tumor tissues, including HMGB1 (high mobility group box 1) (D), CRT (calreticulin) (E), IL-12 (interleukin-12) (F), and TNF-α (tumor necrosis factor–α) (G) using ELISA kits. * denotes statistically significant (P < 0.05). To further substantiate our hypothesis regarding the immune activation mechanism induced by Pt-MOFs@Glu, we used immunofluorescence analysis. We conducted a comprehensive examination of high mobility group box 1 (HMGB1) ([160]Fig. 8D) and calreticulin (CRT) ([161]Fig. 8E) of DAMP proteins in the brains of CNSL mice subjected to various treatments: Pt-MOFs@Glu, Pt-MOFs with equivalent carboplatin content, free glucan, carboplatin, and untreated control groups. Our findings demonstrate that Pt-MOFs@Glu significantly up-regulates CRT, HMGB1, and other DAMP proteins in CNSL cells. Additional analysis explored whether the immunoadjuvant glucan, released upon tumor cell apoptosis via the avalanche effect, enhances the release of immunoinflammatory mediators, such as interferon-α (IFN-α) and interleukin-12 (IL-12), which collectively promote immune activation ([162]Fig. 8, F and G). These results indicate that Pt-MOFs@Glu treatment effectively stimulates the release of key immunoinflammatory mediators following tumor cell death. The preceding investigation used a primary CNSL mouse model; however, clinically, the majority of CNSL cases are secondary. Thus, we established a secondary CNSL model for further study ([163]Fig. 9A). This model involves subcutaneous lymphoma establishment followed by inoculation of secondary CNSL into the mice’s brains on the 14th day postestablishment. The results presented in [164]Fig. 9 (B to D) demonstrate that Pt-MOFs@Glu therapy exhibited superior antitumor efficacy in secondary CNSL compared to the control group. Notably, improvements in body weight, survival rate, and tumor volume were statistically significant (P < 0.05). Fig. 9. Efficacy of Pt-MOFs@Glu in the treatment of secondary CNSL. [165]Fig. 9. [166]Open in a new tab (A) Study design: Schematic representation of the time course for establishing a brain tumor model in mice and the treatment regimen. Treatment was performed on the fourth day after initial tumor inoculation, and secondary tumour modelling was performed on the 14th day, ending the experiment on the 21st day. Some elements of this figure were created in BioRender (Yang-Bao Miao, 2025; [167]https://BioRender.com/ef5jegu). (B) Measurement of mouse weight and survival rate: Changes in mouse weight and survival rate over time were observed following various treatments. (C) Photographs of resected tumors with H&E staining, along with (D) a quantitative chart illustrating the corresponding tumor volumes. (E and F) Histopathological analysis: TUNEL and Ki67 staining revealed pathological changes in tumor tissues across different treatment groups. (G) T cell analysis: Assessment of intratumoral CD4^+ and CD8^+ T cell populations in CNSL mice following various therapeutic interventions. * denotes statistically significant (P < 0.05). Moreover, the oral administration of Pt-MOFs@Glu significantly inhibits the growth of CNSL tumor cells ([168]Fig. 9, C and D), a reduced proportion of proliferating cells (as indicated by Ki67 staining, [169]Fig. 9E), and an increase in apoptotic cells (as observed in TUNEL measurements, [170]Fig. 9F), in contrast to other control groups. These findings underscore the potent efficacy of Pt-MOFs@Glu in effectively combatting CNSL. This promising efficacy holds notable potential for advancing cancer treatment strategies. Simultaneously, we validated the immune activation potential of Pt-MOFs@Glu in a secondary CNSL model. Notably, mice treated with Pt-MOFs@Glu for 2 weeks exhibited a significant increase in brain-infiltrating CD4^+ ([171]Fig. 9G) and CD8^+ T cells ([172]Fig. 9G) compared to the untreated CNSL control group. This result demonstrates that Pt-MOFs@Glu can effectively release carboplatin to eradicate tumors while simultaneously activating the immune system through the action of glucan and DAMP proteins ([173]Fig. 8, D and E). This dual mechanism of action substantially inhibits secondary CNSL. Unveiling the mechanisms behind the antitumor potential of Pt-MOFs@Glu To investigate the mechanism by which oral carboplatin lock-designed MOFs (Pt-MOFs@Glu) triggers an avalanche effect to promote CNSL cell death in the CNSL microenvironment via the gut-brain axis, we collected tissue samples from both treated (post-oral Pt-MOFs@Glu) and untreated CNSL cases. RNA was extracted from these samples for analysis. Venn analysis of gene expression (transcripts per million ≥ 1) revealed 57,180 coexpressed genes across the three groups. The data indicated that, following Pt-MOFs@Glu intervention, 1283 death-related genes were up-regulated and 1451 were down-regulated ([174]Fig. 10, A and B). A total of 2734 differential genes (both up- and down-regulated) were selected to identify the signaling pathways through which oral Pt-MOFs@Glu induces CNSL cell death. Fig. 10. Mechanism behind the antitumor potential of Pt-MOFs@Glu. [175]Fig. 10. [176]Open in a new tab (A) Thermogram of differentially expressed gene expression before and after treatment with Pt-MOFs@Glu. (B) Volcano plots highlighting significantly up-regulated (red) and down-regulated (blue) genes. (C) KEGG pathway enrichment analysis. (D) GO enrichment analysis, where the x axis represents the enrichment fraction, and the y axis corresponds to the pathway information. Larger bubbles indicate greater differences, while the color gradient from blue to red signifies a decreasing P value, indicating higher statistical significance. (E) Differential gene GO enrichment analysis between control and treatment groups. (F) Network mapping of functionally linked, differentially expressed antitumor potential associated proteins. (G) Diagram illustrating the antitumor mechanism based on gene sequencing results. JAK, Janus kinase; STAT, signal transducers and activators of transcription; PI3K, phosphatidylinositol 3-kinase. The Pt-MOFs@Glu promoted apoptosis in CNSL cells, with most of the related genes associated with immune function ([177]Fig. 10C). The results indirectly indicate that Pt-MOFs@Glu, traveling from the gut to the brain, initiates an avalanche effect in the CNSL microenvironment via ROS, thereby facilitating the targeted release of carboplatin to CNSL cells. The release of carboplatin directly kills CNSL cells, triggering the secretion of tumor necrosis factor. This binds with glucan, an immune adjuvant released during the avalanche effect, to activate a tumor-targeted immune response. Together, these actions robustly enhance the immune response against CNSL. KEGG pathway analysis revealed that many cross-signaling genes are involved in pathways related to tumor apoptosis and T cell activation ([178]Fig. 10, D and E). These genes are enriched in apoptosis pathways, the p53 signaling pathway, T helper 1 (T[H]1) and T[H]2 cell differentiation, and the IL-17 signaling pathway, underscoring the role of Pt-MOFs@Glu in inducing tumor cell apoptosis, inflammation, and immune activation following oral therapy for CNSL. Further analysis of protein-protein interactions identified 11 key hub proteins encoded by 9 up-regulated and 2 down-regulated genes, all implicated in critical antitumor processes ([179]Fig. 10E). Tyrosine protein kinases, in particular, were associated with 37 proteins encoded by differential genes and play an essential role in phosphorylating tyrosine residues in key signaling molecules. These kinases facilitate tumor cell apoptosis and stimulate antigen release, thereby activating the immune response against tumor cells. In addition, the network analysis of differentially expressed, functionally associated proteins revealed that Pt-MOFs@Glu primarily exerted its antitumor activity through mitochondrial dysfunction-induced oxidative stress ([180]Fig. 10F). Central nodes in the interaction network—including mt-Co1, mt-Co2, mt-Nd1, mt-Nd2, Ndufa1, Ndufb3, and Uqcrc1—were predominantly enriched in mitochondrial electron transport chain components. This suggests that Pt-MOFs@Glu substantially impaired mitochondrial function, leading to excessive generation of ROS, which in turn initiated intrinsic apoptosis via cytochrome c release and activation of proapoptotic proteins such as Bax and Bak. Concomitantly, Pt-MOFs@Glu triggered extensive immunomodulatory effects within the TME. The up-regulation of immune signaling regulators—such as Foxo3, Il17d, Jak3, Tyk2, Eif5a2, and Gbp6—indicated activation of IFN-related pathways, T[H]17 differentiation, and the Janus kinase/signal transducers and activators of transcription axis—hallmarks of enhanced antitumor immune responses. The engagement of immune-enhancing chemokines and cytokine regulators—including Ccl5, Cxcl, Wnt6, and Ptpn11—further supports the recruitment and activation of cytotoxic T lymphocytes and NK cells, thereby augmenting tumor immunosurveillance. Moreover, Pt-MOFs@Glu modulated key immunological and structural proteins associated with cell adhesion and immune evasion. The down-regulation of Spp1 and disruption of Lam5-integrin signaling contributed to the loss of cell-matrix interactions, promoting apoptosis through anoikis. Simultaneously, the presence of apoptosis-regulatory factors such as Trp^73 and Tp^53 within the network, alongside changes in metabolic regulators (e.g., Hmgcs1, Acat2, and Cyp51), suggested that Pt-MOFs@Glu reprogrammed tumor cell metabolism and reinforced proapoptotic signaling. These findings underscore the dual therapeutic capacity of Pt-MOFs@Glu in mediating both direct cytotoxicity through mitochondrial damage and ROS generation and indirect antitumor effects via immune activation and TME remodeling. This comprehensive mechanism highlights the effectiveness of Pt-MOFs@Glu in inducing an avalanche effect, where carboplatin release synergizes with the immunoadjuvant action of glucan to effectively eradicate CNSL. Biosafety evaluation of Pt-MOFs@Glu To assess the potential toxicity of Pt-MOFs@Glu, a comprehensive series of in vivo experiments were conducted, including examination of tissue sections from major organs, blood chemistry analysis, and hematological analysis. Methotrexate, a first-line clinical drug, was used as a positive control. As shown in figs. S18 to S20, the results revealed that methotrexate treatment induced significant pathological changes in the liver and kidneys of the mice, including cellular necrosis and inflammation, consistent with the well-established toxic effects of the drug. Biochemical analysis revealed a notable increase in blood urea nitrogen (BUN) levels, indicative of kidney dysfunction, while hematological analysis demonstrated a significant decrease in white blood cell (WBC) count, neutrophils (Neu), and platelet count (PLT), reflecting the hematopoietic toxicity typically associated with methotrexate therapy. In contrast, the group treated with Pt-MOFs@Glu exhibited no such abnormalities in organ histology or hematological parameters, highlighting the biocompatibility and lower toxicity of Pt-MOFs@Glu. The absence of these adverse effects suggests that Pt-MOFs@Glu not only provides effective therapeutic potential but also offers a superior safety profile compared to conventional treatments such as methotrexate. These findings emphasize the promise of Pt-MOFs@Glu as a safer alternative, potentially reducing the systemic toxicity commonly seen with current first-line CNSL treatments. This evidence strengthens the case for further clinical investigation of Pt-MOFs@Glu, with the goal of improving patient outcomes by minimizing off-target effects and enhancing the overall therapeutic index. DISCUSSION This study introduces a pioneering therapeutic strategy for the treatment of CNSL by using a platinum-based MOF (Pt-MOFs@Glu) with a padlock design to exploit the gut-brain axis for targeted drug delivery. CNSL treatment is inherently challenging due to the BBB, which notably hinders the effective delivery of therapeutic agents to brain tumors ([181]34). In addressing this limitation, our approach leverages scRNA-seq data to identify intestinal macrophages as potential drug carriers that can migrate from the gut to the brain via chemokine-mediated signaling. By designing Pt-MOFs@Glu, which incorporates carboplatin, pyrazine-quinoxaline, and β-glucan, we harness this migratory pathway, ensuring precise delivery of the drug directly to CNSL sites through macrophage-mediated transport. Key findings of this study include the identification of Cx3cr1 signaling as a critical pathway in macrophage migration, with experimental results showing that intestinal macrophages efficiently internalize Pt-MOFs@Glu through Dectin-1 receptors. This provides a robust transport mechanism for delivering the drug to CNSL sites. Cytotoxicity assays confirmed the favorable biosafety profile of Pt-MOFs@Glu, with enhanced cell viability compared to free carboplatin and effective, controlled drug release upon ROS-triggering. The induced avalanche effect resulted in significant tumor cell apoptosis, substantially enhancing therapeutic efficacy. In vivo studies further validated the oral administration route of Pt-MOFs@Glu, demonstrating its ability to traverse the GI, cross the BBB, and accumulate at CNSL sites. Imaging studies confirmed sustained brain accumulation, and immune profiling revealed that Pt-MOFs@Glu activated CD^4+, CD^8+ T cells, killer T cells, and effector T cells and enhanced proinflammatory cytokine expression, indicating an additional immune-mediated antitumor effect. Despite these promising results, several challenges must be addressed before clinical translation can be realized. First, while our scRNA-seq data and animal models have provided valuable insights into macrophage migration and Cx3cr1 signaling, the precise role of these pathways in human CNSL requires further investigation. Species-specific differences in the gut-brain axis dynamics present a critical challenge, as the mechanisms observed in animal models may not entirely reflect those in humans. Humanized models or early-phase clinical trials will be essential to assess the pharmacokinetics, biodistribution, and efficacy of Pt-MOFs@Glu in humans. Understanding interspecies variability in drug transport and macrophage recruitment will be critical for ensuring the clinical relevance of these findings. Second, while the ROS-triggered avalanche effect has shown efficacy in preclinical models, the consistency of ROS production within the human CNSL microenvironment remains uncertain. ROS generation may vary across different TMEs and between individuals. Therefore, strategies to modulate ROS levels—such as combining Pt-MOFs@Glu with pro-oxidant drugs or photothermal therapies—could improve the reliability and effectiveness of this activation mechanism. However, these strategies will require extensive optimization and validation in human models to ensure their applicability and clinical success ([182]35–[183]37). Last, although the Pt-MOFs@Glu system demonstrated stability under gastrointestinal conditions, its long-term biocompatibility and potential systemic toxicity must be rigorously evaluated in preclinical and clinical studies. It is crucial to assess both the acute and chronic safety profiles of Pt-MOFs@Glu, including its long-term effects on organ function and overall health. Special attention must be given to immune activation and inflammation, as excessive immune responses could undermine therapeutic efficacy and induce adverse side effects. While the Pt-MOFs@Glu system leverages macrophages for targeted drug delivery, macrophages also play a crucial role in systemic immune surveillance, and their activation could potentially lead to off-target effects. These unintended interactions may result in immune responses in nontumor tissues, affecting the overall treatment efficacy and safety. Potential off-target effects should therefore be carefully considered, as the Pt-MOFs@Glu system may interact with unintended tissues or cells, leading to unforeseen consequences. These off-target interactions could affect nontumor tissues, potentially causing unintended toxicity or immunological responses that could limit the therapeutic efficacy and safety of this system. To address these concerns, potential solutions include incorporating macrophage-targeting strategies with enhanced selectivity for tumor-associated macrophages (TAMs) rather than those involved in systemic immune surveillance. This could be achieved by designing Pt-MOFs@Glu to preferentially bind to specific surface markers expressed by TAMs, reducing the risk of off-target immune activation. In addition, incorporating “shielding” strategies, such as coating nanoparticles with immunologically inert or anti-inflammatory agents, could help mitigate undesired immune responses. Advanced in vivo imaging and profiling techniques will be critical for monitoring macrophage activity in real time and guiding the refinement of these delivery systems. Comprehensive studies focused on identifying and mitigating these off-target effects will be essential to ensure the clinical feasibility of this therapeutic platform. In summary, this study proposes a promising gut-to-brain delivery system using Pt-MOFs@Glu, offering notable potential for overcoming the treatment barriers of CNSL. Although the preclinical data are compelling, several critical challenges remain to be addressed before advancing to clinical application. Future research should focus on validating these findings in humanized models, optimizing the system’s pharmacokinetics, and evaluating long-term safety and efficacy in clinical trials. The development of this novel therapeutic strategy holds great promise for improving CNSL treatment and potentially other brain-related diseases. In conclusion, the padlock-designed Pt-MOFs@Glu system offers a highly promising approach to overcoming the challenges of CNSL treatment. By harnessing the gut-brain axis and a lock-and-key mechanism, this platform enables targeted, noninvasive drug delivery across the BBB. The combination of carboplatin, pyrazine-quinoxaline, and β-glucan ensures precise tumor targeting, ROS-triggered drug release, and immune activation, resulting in an avalanche effect that not only enhances therapeutic efficacy but also minimizes systemic toxicity. While the findings are promising, it is important to acknowledge that this study is preclinical, and further research is required to address crucial factors such as long-term safety, pharmacokinetics in humans, and the potential immunogenicity of the MOF formulation. Nevertheless, this work lays the foundation for the future of precision therapies, and with continued advancements, the promise of transforming the treatment landscape for CNSL and other CNS diseases becomes increasingly tangible. MATERIALS AND METHODS Materials The following compounds were sourced from Sigma-Aldrich in St. Louis, Missouri, USA: pyrazine-quinoxaline, carboplatin, hydrogen peroxide (30%, w/w), yeast, triphenylphosphine (PPh[3]), and diisopropyl azodicarboxylate. The Alexa Fluor 633 N-hydroxysuccinimide ester was obtained from Thermo Fisher Scientific in Waltham, Massachusetts, USA. The A20 and RAW264.7 cells used in this study were provided by the American Type Culture Collection in Manassas, VA. Cell culture reagents were supplied by Gibco, located in Grand Island, NY, USA. All compounds and reagents used in this research were of analytical grade. scRNA-seq datasets processing Quantitative parameters and cell screening criteria were applied following previously published literature. Briefly, the gene barcode counting matrix was analyzed using the Seurat R package (version 4.0.2). Cells with more than 200 genes and less than 10% mitochondrial gene content were selected for downstream analysis. After converting samples into Seurat objects, normalization and scaling were performed by regressing unique molecular identifier counts and mitochondrial gene percentages. For dimensionality reduction, the FindVariableGenes function identified the most variable genes, followed by PCA for dimensional reduction. The RunTSNE function in Seurat (version 3.1.3) was then used to generate the TSNE plot. Preparation and characterization of Pt-MOFs@Glu The padlock-designed Pt-MOFs were synthesized through a solvothermal reaction to achieve a highly efficient and structurally robust nanocarrier. In summary, a mixture of carboplatin (15 mg), pyrazino[2,3-f]quinoxaline (15 mg), and acetic acid (100 μl) was dissolved in N,N′-dimethylformamide (15 ml). The solution was then transferred into a 50-ml round-bottom flask, which was subsequently heated to 180°C for 4 hours to facilitate the solvothermal reaction. After the reaction was completed, the resulting mixture was centrifuged at 10,000 rpm for 10 min to separate the solid products from the solution. The solid nanoparticles were then washed sequentially with 30 ml of deionized (DI) water and ethanol to remove any unreacted precursors or by-products. Following the purification, the Pt-MOFs were dispersed in DI water (0.5 mg/ml) to obtain a stable nanoparticle suspension. To further functionalize the nanoparticles, mercaptoethylated β-glucan (0.5 mg/ml) was added to the dispersion, and the mixture was stirred overnight at room temperature. Last, to ensure complete the removal of any residual reagents and unreacted β-glucan, the nanoparticles underwent an additional washing step. The purified Pt-MOFs were then ready for further characterization and application in drug delivery and therapeutic studies. The morphology and structure of the nanoparticles were analyzed using scanning electron microscopy, transmission electron microscopy, FTIR, and ICP-MS. In addition, the particle size and surface charge of the Pt-MOFs@Glu were determined using DLS. To assess their long-term stability and gastrointestinal resilience, the particles were subjected to 4°C DI water and simulated gastric and intestinal fluids, with samples withdrawn at specific intervals for particle size analysis. Furthermore, to evaluate the responsiveness of the Pt-MOFs@Glu to ROS within the TME, a solution of 100 μM H[2]O[2] was prepared in PBS for simulation purposes. Drug release studies were conducted at 37°C, with supernatant samples collected at designated time intervals. The concentration of carboplatin in the supernatant was quantified by measuring absorbance values at 229 nm using a UV-visible spectrophotometer. Cell viability assay The cytotoxicity or cytocompatibility of the prepared nanoparticles was assessed by incubating them at different concentrations (0 to 400 μg/ml) with A20 cells or RAW264.7 macrophages (5 × 10^3 cells per well) in 96-well plates. Carboplatin at an equivalent concentration was used as a positive control. Following 48 hours of incubation, cell viability was determined using an MTT cell viability assay kit. Uptake by macrophage To visualize the uptake of Pt-MOFs@Glu by macrophage cells in vitro, RAW264.7 cells (1 × 10^6 cells/ml) were cultured with fluorescence-labeled Pt-MOFs@Glu (100 μg/ml), synthesized using Cy5.5 according to the manufacturer’s protocol. After incubation for predetermined time intervals (0, 1, 2, 4, or 6 hours), the cells were collected, washed with PBS, stained with 4′,6-diamidino-2-phenylindole, and subsequently examined using CLSM. Penetration of CNSL by Pt-MOFs@Glu Three-dimensional CNSL spheroids composed of A20 cells were generated. These spheroids were then incubated with Pt-MOFs@Glu or macrophages following predetermined co-incubation periods (3, 6, or 12 hours). Subsequently, they were rinsed with PBS, fixed in 4% paraformaldehyde (PFA), immunofluorescently stained with F4/80 antibody, transferred onto a confocal dish, and examined using CLSM. Animal study BALB/c mice (8 weeks old, weighing 18 to 20 g) were procured from SiPeiFu. All animal experiments were conducted in accordance with the guidelines outlined in the “Guide for the Care and Use of Laboratory Animals,” formulated by the Institute of Laboratory Animal Resources, National Research Council and published by the National Academy Press in 2011. Animal study protocols were approved by the Institutional Animal Care and Use Committee of Sichuan Provincial People’s Hospital (protocol no. 2024416). To establish the brain tumor model, test mice were intracranially inoculated with 1 × 10^7 A20 cells suspended in 5 μl of PBS. Biodistribution of Pt-MOFs@Glu In the biodistribution study, mice bearing tumors and fasted overnight were orally administered Pt-MOFs@Glu. At predetermined intervals (0, 2, 4, 6, or 8 hours) posttreatment, the accumulation of Pt-MOFs@Glu in the brain tumor and major organs—including the heart, lungs, liver, spleen, and kidneys—was quantified using IVIS and ICP-MS, respectively. Untreated mice and those receiving an equivalent dose of free cisplatin served as controls. Transport route of Pt-MOFs@Glu To investigate the transport route of Pt-MOFs@Glu in vivo, mice fasted overnight were orally administered Pt-MOFs@Glu (n = 3). In the inhibitor group, mice were intraperitoneally injected with cycloheximide (3 mg/kg, n = 3). After six hours, small intestines (villi, Peyer’s patches, and lymphatic vessels) and brain tumors were harvested from euthanized mice. The tissues were cryosectioned and immunofluorescently stained with anti-M cell antibody, F4/80 antibody, and anti-LYVE-1 antibody to identify M cells, intestinal macrophages, and lymphatic vessels, respectively. CLSM was used to detect the colocalization of Pt-MOFs@Glu with M cells, intestinal macrophages, and lymphatic vessels. In vivo antitumor efficacy To evaluate the antitumor efficacy of each formulation, tumor-bearing mice fasted overnight were randomly allocated to five test groups: untreated control, free drug (carboplatin), and Pt-MOFs@Glu. Each mouse in the treatment groups received an equivalent dose of carboplatin (20 mg/kg). The body weights of tumor-bearing mice were monitored every other day throughout the study. At the study’s conclusion, mice were euthanized, and tumor tissues were retrieved, fixed with 4% PFA, embedded in paraffin wax, and then sectioned and stained with H&E, Ki67 monoclonal antibody, or TUNEL. In vivo toxicity evaluation To assess in vivo toxicity, vital organs from each study group were retrieved at the study’s conclusion, fixed in 4% PFA, embedded in paraffin blocks, and stained with H&E for histopathological examination. Organs obtained from healthy mice served as controls. Serum samples collected from mice were used to measure the levels of aspartate aminotransferase and BUN to evaluate potential liver and kidney toxicities. Statistical analysis All data are presented as the means ± SD, unless otherwise specified. Each experiment was conducted with a minimum of three repetitions. Statistical comparisons were performed using one-way or two-way analysis of variance (ANOVA), followed by Bonferroni multiple comparisons posttest. The survival curves were compared using the log-rank test (Mantel-Cox). Statistical significance was set at *P < 0.05 for all analyses. All statistical calculations were conducted using Prism 8 (GraphPad Software). Acknowledgments