Next Article in Journal
Preparation and Therapeutic Evaluation of Engineered Semaglutide and Statin–Lipid Conjugate-Based Nanoparticle
Previous Article in Journal
Nanostructured Lipid Carriers (NLC)-Based Topical Formulation of Hesperidin for Effective Treatment of Psoriasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases

Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(4), 479; https://doi.org/10.3390/pharmaceutics17040479
Submission received: 20 February 2025 / Revised: 25 March 2025 / Accepted: 3 April 2025 / Published: 7 April 2025
(This article belongs to the Section Nanomedicine and Nanotechnology)

Abstract

:
Matrix metalloproteinase-9 (MMP-9) is implicated in tumor progression and vascular diseases, contributing to angiogenesis, metastasis, and extracellular matrix degradation. This review comprehensively examines the relationship between MMP-9 and these pathologies, exploring the underlying molecular mechanisms and signaling pathways involved. Specifically, we discuss the contribution of MMP-9 to tumor epithelial–mesenchymal transition, angiogenesis, and metastasis, as well as its involvement in a spectrum of vascular diseases, including macrovascular, cerebrovascular, and ocular vascular diseases. This review focuses on recent advances in MMP-9-targeted nanomedicine strategies, highlighting the design and application of responsive nanoparticles for enhanced drug delivery. These nanotherapeutic strategies leverage MMP-9 overexpression to achieve targeted drug release, improved tumor penetration, and reduced systemic toxicity. We explore various nanoparticle platforms, such as liposomes and polymer nanoparticles, and discuss their mechanisms of action, including degradation, drug release, and targeting specificity. Finally, we address the challenges posed by the heterogeneity of MMP-9 expression and their implications for personalized therapies. Ultimately, this review underscores the diagnostic and therapeutic potential of MMP-9-targeted nanomedicines against tumors and vascular diseases.

Graphical Abstract

1. Introduction

Matrix metalloproteinases (MMPs) constitute a family of calcium-dependent, zinc-containing endopeptidases. These enzymes are initially synthesized as inactive zymogens by connective tissues, such as the endothelium and fibroblasts, as well as by inflammatory cells, including macrophages and neutrophils. These precursors are secreted into the extracellular matrix and subsequently activated through proteolytic cleavage by enzymes such as fibrinolytic enzymes [1]. Currently, the human MMP family comprises 23 identified proteins [2]. Table 1 presents a comprehensive overview of their classification and functional roles, adapted from Wang et al. [3]. Notably, MMP-9, secreted by cells such as neutrophils, macrophages, and endothelial cells, plays a crucial role in the degradation of the extracellular matrix, thereby facilitating tumor invasion and angiogenesis. These unique functions have attracted significant research interest. The MMP-9 gene, located on chromosome 20q11.1-13.1, encodes a protein composed of four domains: (1) an N-terminal propeptide domain that binds to the catalytic zinc ion and maintains the stability of the pro-MMP-9 zymogen; (2) a catalytic domain containing zinc and calcium ions, which forms the enzymatic active site and includes a hinge region involved in collagen or gelatin binding; (3) a hinge region; and (4) a C-terminal hemopexin-like domain essential for collagenase activity [4,5,6,7]. The MMP-9 promoter binds multiple transcription factors, including NF-κB, AP-1, and SP-1, and its expression is regulated by various signaling pathways. MMP-9 significantly contributes to tumor progression and metastasis through its involvement in tumor angiogenesis, invasion, metastasis, and extracellular matrix degradation [8,9,10]. Furthermore, MMP-9 significantly influences the pathogenesis of vascular diseases such as atherosclerosis and abdominal aortic aneurysm by destabilizing atherosclerotic plaques and degrading the aortic wall [11,12]. As a secreted MMP, MMP-9 is readily accessible to extracellularly targeted nanomedicines, offering advantages over membrane-bound or intracellular MMPs. Its widespread expression in various cancers and vascular diseases has led to its investigation as a biomarker for patient stratification and personalized diagnostics and therapeutics [13,14,15,16].
Tumors are leading causes of global mortality, with approximately 10 million deaths in 2020, according to the World Health Organization (WHO). Lung, colorectal, liver, gastric, and breast cancers account for the majority of cancer-related deaths [17]. Chemotherapy remains a primary treatment for tumors, aiming to control tumor growth, prevent metastasis, and enhance the success rates of solid tumor surgeries while reducing the risk of recurrence. However, conventional chemotherapy drugs for tumors often face limitations such as poor bioavailability, low tissue specificity, high systemic toxicity, and reduced efficacy against highly heterogeneous tumors.
Vascular diseases encompass a diverse range of pathologies affecting the arterial, venous, and lymphatic systems, ultimately compromising tissue function due to impaired perfusion and drainage. The global burden of vascular disease is substantial. Cardiovascular diseases, predominantly including coronary artery disease and stroke, account for an estimated 17.9 million deaths annually [18], with stroke being a major cause of long-term disability [19]. Furthermore, microvascular complications, such as diabetic retinopathy, affect approximately 22% of individuals with diabetes, significantly impacting their quality of life [20]. Current therapeutic strategies for vascular diseases encounter several key challenges. These include the difficulty in achieving adequate drug concentrations at the target lesion, limited drug penetration across the vessel wall, non-specific drug accumulation leading to systemic side effects, and the complex interplay of underlying pathophysiological mechanisms, often necessitating multi-targeted therapeutic interventions.
Nanomedicine involves the application of nanotechnology to medicine. It leverages the unique physicochemical properties of materials at the nanoscale to address the current limitations in disease diagnosis and treatment [21]. Nanoparticles, defined as particles with diameters ranging from 1 to 100 nm, possess a high surface area-to-volume ratio, which confers unique physicochemical properties [22]. Nanoparticles have emerged as powerful tools in biomedicine, with applications in cancer therapy, drug delivery, vaccine delivery, gene therapy, the treatment of inflammatory and autoimmune diseases, imaging, diagnostics, tissue engineering, and antibacterial materials. Figure 1, originally published by Zhang et al., illustrates the mechanism by which various nanoparticles recognize and bind to cancer-associated molecules, thereby enhancing tumor diagnosis [23]. Compared with conventional chemotherapeutics, nanodrugs offer the potential for targeted accumulation and controlled release within the tumor microenvironment, enhancing local drug uptake and minimizing systemic side effects. Nanoparticles can be classified based on material and size, as shown in Table 2 and Table 3. The gelatinase activity of MMP-9 has spurred the development of numerous nanocarriers that utilize MMP-9 as a regulatory or responsive target. This review aims to elucidate the regulatory mechanisms of MMP-9 through diverse signaling pathways, consolidate the understanding of MMP-9’s involvement in tumors and vascular diseases, and highlight the recent advancements in MMP-9-targeted nanomedicine strategies, ultimately providing valuable insights into the diagnostic and therapeutic potential of nanomedicines for tumors and vascular diseases.

2. Relationship Between MMP-9 and Tumors and Vascular Diseases

2.1. Relationship Between MMP-9 and Tumors

2.1.1. Relationship Between MMP-9 and Tumor Epithelial–Mesenchymal Transition

MMP-9, a gelatinase capable of degrading major ECM components including type IV collagen, plays a critical role in the epithelial–mesenchymal transition (EMT) of cancer cells. EMT is the process by which epithelial cells acquire mesenchymal stem cell-like characteristics. This complex process involves the loss of epithelial traits, such as intercellular adhesion and cell polarity, and the gain of mesenchymal properties, including motility and invasiveness [77]. EMT is essential for tumor invasion and progression. It facilitates the detachment of cancer cells from the primary tumor, enabling the infiltration of surrounding tissues (direct infiltration). Furthermore, mesenchymal cancer cells can invade the vasculature, disseminate through the circulatory system, and establish distant metastases through the mesenchymal–epithelial transition (MET), the reverse process of EMT, at secondary sites, where they regain epithelial characteristics [78]. Cells undergoing EMT also acquire stem cell-like properties, contributing to their resistance to chemotherapy and targeted therapies [79]. Additionally, these cells interact with the tumor microenvironment, promoting immunosuppression and metabolic adaptation [80].
MMP-9, through ECM degradation, reduces cell–cell adhesion, promoting cancer cell detachment and invasion. This degradation also facilitates cancer cell entry into the vasculature [9]. Furthermore, ECM degradation triggers the release of factors such as TGF-β and VEGF, which can lead to the downregulation of E-cadherin and the upregulation of N-cadherin, the hallmarks of EMT [81].

2.1.2. Relationship Between MMP-9 and Tumor Angiogenesis and Metastasis

MMP-9 also promotes angiogenesis, facilitating tumor growth and intravasation, ultimately leading to distant metastasis [8]. MMP-9 overexpression is closely linked to tumor angiogenesis and invasive metastatic behavior [10].
MMP-9 plays a significant role in vasculogenic mimicry and tumor invasion, impacting glioma patient survival [82]. Du et al. demonstrated that MMP-9 secreted by glioblastoma cells enhances VEGF utilization by bone marrow-derived CD45+ cells, promoting angiogenesis and regulating tumor invasion [83]. Long et al. found that melanoma induces MMP-9 and IL-10 secretion. MMP-9 increases vascular permeability, while IL-10 promotes immunosuppression. These effects contribute to the recruitment of granulocytic myeloid-derived suppressor cells (MDSCs) to the lungs, creating a pre-metastatic niche and facilitating melanoma lung metastasis [84]. Furthermore, non-VEGF-driven intussusceptive angiogenesis, observed in human melanoma metastases with high MMP-9 expression and immune cell infiltration near intravascular pillars, is rare in mouse patient-derived xenografts (PDXs). MMP-9 inhibition suppresses intravascular pillar formation [85]. Bruno et al. observed a persistent elevation of MMP-9, its inhibitors TIMP-1/2, and angiopoietin in colorectal cancer, unlike in intestinal inflammatory diseases. The TIMP-1/2/MMP-9 axis correlates with angiogenesis and invasion, and MMP-9 inhibits natural killer (NK) cell function, thereby regulating tumor metastasis [86]. Therefore, targeting MMP-9 is crucial for research on anti-tumor angiogenesis, invasion, and metastasis.

2.2. Relationship Between MMP-9 and Vascular Diseases

Many factors, including infection, hyperlipidemia, hyperglycemia, surgery, and autoimmune diseases [87], have the potential to impair the vascular wall and even induce neovascularization. MMP-9 levels and related signaling pathways are closely related to vascular pathologies. MMP-9’s gelatinase activity degrades key extracellular matrix components such as collagen and elastin, disrupting vascular wall integrity, including the blood–brain and blood–retinal barriers [88]. MMP-9 also plays a role in the formation and progression of atherosclerotic plaques [12] and promotes thrombosis [89], contributing to cardiovascular events. In the process of MMP-9-mediated vascular damage, various factors such as inflammation and hypoxia upregulate the levels of pro-angiogenic factors including VEGF and FGF. Consequently, endothelial cells are activated, leading to neovascularization [90].

2.2.1. Relationship Between MMP-9 and Macrovascular Diseases

The chronic overexpression of MMP-2/9 in vascular smooth muscle cells and macrophages within the infrarenal aorta degrades the elastic matrix, weakening vascular wall elasticity and contributing to abdominal aortic aneurysm development [91,92]. Wang et al. demonstrated that VEGF significantly enhances MMP-9 expression in vascular smooth muscle cells via its receptor flt-1, promoting vascular basement membrane degradation, smooth muscle cell migration, and angiogenesis, ultimately contributing to atherosclerosis [93].

2.2.2. Relationship Between MMP-9 and Cerebrovascular Diseases

MMP-9 plays a significant role in the pathogenesis of various cerebrovascular diseases, with elevated MMP-9 levels consistently correlating with poor clinical prognosis [94,95]. The blood–brain barrier (BBB), comprising the cerebral microvascular endothelium, pericytes, and astrocytes, is vulnerable to disruption under pathological conditions [96]. For instance, Polavarapu et al. demonstrated that the intracerebral injection of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) activates MMP-9 and the upstream NF-κB pathway, thereby increasing BBB permeability [97]. Similarly, interleukin-1β (IL-1β) also induces MMP-9 secretion and the degradation of tight junction proteins in pericytes. Conversely, melatonin has been shown to protect the BBB integrity by regulating Notch-3 expression, the NF-κB pathway, and NF-κB nuclear translocation, resulting in the downregulation of MMP-9 and the preservation of tight junction proteins [98]. Furthermore, Machida et al. reported that thrombin, acting through its receptor PAR-1 and the PKCθ-Akt and PKCδ-ERK1/2 pathways, triggers substantial MMP-9 release from pericytes, leading to BBB dysfunction [99]. Chen et al. demonstrated that the glutamate receptor NMDAR upregulates MMP-9 expression in mouse cerebrovascular endothelium via ERK1/2 phosphorylation, disrupting occludin in the BBB [100]. In ischemic stroke, reactive nitrogen species, such as nitric oxide and peroxynitrite, activate MMPs by downregulating caveolin-1 and activating nitric oxide synthase, thereby exacerbating BBB damage and ischemia/reperfusion injury. This highlights MMPs as potential therapeutic targets [101]. Metformin has been shown to protect the BBB in a mouse model of middle cerebral artery occlusion by downregulating the JNK pathway, reducing MMP-9 expression, and preserving the tight junction protein ZO-1, ultimately reducing infarct size [102]. Subarachnoid hemorrhage also induces MMP-9 expression and activation. Increased cyclophilin A in pericytes activates the NF-κB pathway and MMP-9 via the CD147 receptor, disrupting tight junctions [103]. Conversely, glucocorticoids prevent intracranial hemorrhage in preterm infants by inhibiting MMP-9 and caspase-3 activity in the lateral ventricular germinal matrix of fetal animals [104].

2.2.3. Relationship Between MMP-9 and Ocular Vascular Diseases

MMP-9 is a critical protein in ocular research, particularly concerning fundus diseases, due to its activity against type IV collagen, a major component of retinal endothelial cell basement membranes and Bruch’s membrane [105]. In a rat model of diabetes, elevated MMP-9 levels, decreased vascular endothelial-cadherin, and increased retinal vascular permeability were observed within two weeks [106]. Kowluru et al. demonstrated that high glucose increases MMP-9 expression in cultured retinal endothelial cells by inhibiting sirtuin 1 activity via oxidative stress and enhancing p65 (NF-κB subunit) binding to the MMP-9 promoter, contributing to diabetic retinopathy [107]. Furthermore, Mishra et al. found that MMP-9 enters the mitochondria in human diabetic retinopathy endothelial cells via HSP70-mediated mechanisms, damaging mitochondrial DNA, disrupting mitochondrial membranes, releasing cytochrome c, and ultimately inducing apoptosis [108]. In retinal endothelial cells, high glucose activates H-Ras, leading to increased NF-κB and MMP-9 expression and accelerated capillary cell apoptosis. Simvastatin, an HMG-CoA reductase inhibitor, inhibits H-Ras membrane anchoring, downregulating its activity and modulating MMP-9 expression [109]. Lambert et al. observed an increased MMP-9 expression five days after laser-induced choroidal neovascularization (CNV) in mice, coinciding with macrophage infiltration and Bruch’s membrane rupture [105]. In a mouse model of laser-induced CNV, MMP-2/9 double knockout mice exhibited minimal CNV compared to single knockouts, suggesting a synergistic effect of MMP-2 and MMP-9 [110]. Additionally, Kinoshita et al. found that oral genistein administration reduced MMP-9 and ICAM-1 levels in the RPE–choroidal complex in a mouse model of laser-induced CNV, inhibiting neovascularization [111].
Given MMP-9’s crucial role in tumor progression—driving EMT, angiogenesis, growth, and metastasis—and its involvement in the pathogenesis of vascular diseases, the development of MMP-9-related nanomedicines represents a promising therapeutic avenue.

3. Signal Pathways Regulating MMP-9 Expression

MMP-9 expression is regulated by a variety of signaling pathways, summarized in Table 4 and Figure 2. Figure 2 illustrates the regulatory network of these pathways on tumor progression and vascular diseases.
The nuclear transcription factor NF-κB directly promotes MMP-9 gene transcription by binding to its promoter. Consequently, various signaling pathways can regulate MMP-9 expression through NF-κB. For example, platelet-activating factor (PAF) activates NF-κB via the Ca2+/PI3K and ERK signaling pathways, upregulating MMP-9 expression [112].
The transcriptional activator AP-1, composed of c-Fos and c-Jun heterodimers, also binds to the MMP-9 promoter. Hyperglycemia-induced inhibition of the deacetylase sirtuin 1 leads to AP-1 hyperacetylation and increased MMP-9 promoter binding, promoting MMP-9 expression. Conversely, sirtuin 1 activation downregulates MMP-9 [113].
Poly(ADP-ribose) polymerase-1 (PARP-1) forms complexes with transcription factors such as NF-κB and AP-1 within the MMP-9 promoter region to enhance transcription [108]. Mishra et al. demonstrated that PARP-1 inhibition in high-glucose-cultured retinal microvascular endothelial cells reduces NF-κB and AP-1 promoter binding, decreasing MMP-9 expression. Notably, PARP-1 is also regulated by sirtuin 1-mediated acetylation [108].
Mitogen-activated protein kinases (MAPKs), including JNK, ERK1/2, and p38 MAPK, play crucial roles in regulating MMP-9 expression [115]. Interleukin-20 (IL-20) treatment of human endothelial cells induces JNK, ERK1/2, and p38 MAPK phosphorylation, promoting the transcription of various factors, including MMP-9, and stimulating angiogenesis [114]. In human umbilical vein endothelial cells (HUVECs), TNF-α upregulates ERK1/2 phosphorylation and downstream AP-1 activation mediated by endoplasmic reticulum protein disulfide isomerase (Endo-PDI) overexpression, increasing MMP-9 transcription and angiogenesis [115]. The antifungal drug miconazole prevents hemorrhagic stroke in zebrafish and mesenteric hemorrhage in mammals by downregulating ERK phosphorylation and MMP-9 expression [117]. Conversely, the knockout of tumor vascular transient receptor potential vanilloid 4 (TRPV4) upregulates ERK phosphorylation and MMP-9 expression, promoting tumor angiogenesis and metastasis [10]. Similarly, under TNF-α, high glucose, or lipopolysaccharide (LPS) stimulation, the overexpression of the orphan C-family G protein-coupled receptor 5B (GPRC5B) phosphorylates ERK1/2, activates NF-κB, and enhances MMP-9 expression in the vascular wall, contributing to inflammation and atherosclerosis [118]. However, Miyoshi et al. found that JNK, but not ERK or p38 MAPK, inhibition almost completely suppressed TNF-α-induced MMP-9 expression in human endothelial cells [116]. The soluble CD40 ligand promotes MMP-9 secretion and angiogenesis in endothelial progenitor cells via the p38 MAPK pathway [119].
The PI3K/Akt pathway also regulates MMP-9 expression and is associated with vasculogenic mimicry in invasive tumors. Tenascin-C (TNC) promotes Akt phosphorylation, upregulating MMP-9 expression and facilitating vasculogenic mimicry and glioma invasion [82]. Jin et al. discovered that fibronectin induces MMP-9 secretion through the JNK, ERK, and PI3K/Akt pathways, mediated by AP-1, leading to collagen degradation [120]. In contrast, Bhowmik et al. demonstrated that estrogen receptor pathway activation in MCF7 breast cancer cells inhibits ERK, p38 MAPK, and PI3K/Akt signaling, decreasing NF-κB and MMP-9 expression, thereby exhibiting an anti-metastatic effect [121].
Thrombin and its receptor protease-activated receptor 1 (PAR-1) activate pericytes, stimulating PKC signaling and promoting MMP-9 secretion via the PKCθ/Akt and PKCδ/ERK pathways, affecting the blood–brain barrier integrity [99]. In mammalian endothelial cells, PKC-α inhibitors reduce ERK1/2 activation and MMP-9 secretion [122].
Rho signaling acts upstream of MMP-9. Renault et al. showed that the morphogen Sonic Hedgehog (Shh) regulates angiogenesis in endothelial cells by upregulating proteins such as MMP-9 through Rho and Rho-associated protein kinase (ROCK) [123]. Chatterjee et al. demonstrated that ROCKII facilitates Smad nuclear localization, upregulates MMP-9, and promotes tumor angiogenesis and lung metastasis in mouse melanoma [124].
The histone methyltransferase Ezh2 regulates MMP-9. Under high glucose conditions, increased Ezh2 activity in retinal endothelial cells leads to MMP-9 promoter methylation, upregulating MMP-9 transcription and triggering mitochondrial damage and apoptosis [125]. However, during normal mouse embryogenesis, Ezh2 inhibits MMP-9 activation, protecting vascular integrity [126].
Melanoma-derived exosomal miR-155 downregulates suppressor of cytokine signaling 1 (SOCS1), an inhibitor of the JAK2/STAT3 pathway, activating JAK2/STAT3 signaling, promoting MMP-9 expression, and enhancing tumor angiogenesis [127]. In a mouse stroke model, tissue plasminogen activator (tPA) binds to low-density lipoprotein receptor-related protein (LRP), upregulating MMP-9 expression in brain microvascular endothelial cells and causing cerebral hemorrhage [128].
Table 5 summarizes the pathways relevant to the relationship between MMP-9 and tumor progression and vascular diseases.

4. Nanomedicine and MMP-9-Targeted Nanoparticles

Nanomedicine leverages nanotechnology—the manipulation of materials at the nanoscale (1 to 100 nm)—to advance medical diagnosis and treatment. Nanoparticles, characterized by their high surface area-to-volume ratio, possess unique physicochemical properties that make them powerful tools in biomedicine.

4.1. Comparison of MMP-9-Targeted and Non-Targeted Nanomedicines

Non-targeted nanomedicines can enhance drug solubility, prolong circulation time, and improve chemotherapeutic efficacy while mitigating systemic toxicity through the enhanced permeability and retention (EPR) effect. However, tumor heterogeneity often limits their efficacy, hindering optimal accumulation within deep tumor tissues. In contrast, MMP-9-targeted nanomedicines employ active targeting, binding to overexpressed MMP-9 in the tumor microenvironment. This approach enhances specificity and local penetration, improves therapeutic outcomes, and potentially overcomes chemotherapy resistance [137]. Nonetheless, heterogeneous MMP-9 expression necessitates personalized diagnostic and treatment strategies based on individual MMP-9 biomarker profiles [14,15,16]. Furthermore, the design and synthesis of MMP-9-targeted nanoparticles are more complex than their non-targeted counterparts.

4.2. MMP-9 Overexpression and MMP-9-Responsive Nanoparticles

MMP-9, frequently overexpressed in certain cancer types, plays a crucial role in tumor microenvironment remodeling. Many nanomedicines exploit MMP-9 overexpression to modify their physical properties, achieving targeted controlled release. For example, MMP-9 cleavage can facilitate deeper tumor penetration and enhance permeability [138]. Smaller nanoparticles, generated through this cleavage, can be internalized via endocytosis rather than phagocytosis [139]. The nanoparticle surface charge is also critical; a positive charge facilitates cellular penetration and uptake through interaction with the negatively charged cell membrane [140]. Furthermore, the nanomedicine composition significantly impacts drug delivery. Hydrophobic drugs are often encapsulated within a hydrophobic core. Upon exposure to MMP-9, this core can be exposed, triggering drug release [141].

4.3. Mechanisms of Action of MMP-9-Responsive Nanoparticles

In general, MMP-9-responsive nanoparticles operate through several mechanisms: degradation, drug release, and targeting specificity.

4.3.1. Degradation

(a)
Direct cleavage: Nanoparticles composed of MMP-9-cleavable materials, such as gelatin, are degraded in MMP-9-rich environments such as tumor microenvironments and vasculopathies, facilitating local drug release [139,141].
(b)
Cross-linking disruption: MMP-9-sensitive linkers maintain nanoparticle stability. Overexpressed MMP-9 disrupts these linkages, destabilizing the structure and releasing the encapsulated drug [137,141].
(c)
Matrix degradation: Nanoparticles embedded within the ECM are released upon MMP-9-mediated matrix degradation [142].

4.3.2. Drug Release

(a)
Degradation-triggered release: MMP-9 activity degrades the nanoparticles, altering their composition, size, and cross-linking density, thereby modulating drug release rates [139].
(b)
Enhanced permeability: MMP-9 increases tumor blood vessel and tissue permeability, enhancing nanoparticle penetration into the tumor tissues [142,143].

4.3.3. Targeting Specificity

(a)
Active targeting: Nanoparticles decorated with MMP-9-targeting ligands (e.g., antibodies, peptides, small molecules) bind to specific regions such as the active site of MMP-9 [144,145,146], enhancing the accumulation at MMP-9 overexpression sites and minimizing the off-target effects.
(b)
Passive targeting: Nanoparticles passively accumulate in tumors due to the EPR effect, exploiting leaky tumor vasculature and impaired lymphatic drainage [147].

4.4. Heterogeneity of MMP-9 Expression and Targeted Therapy Efficacy

MMP-9 expression varies considerably across tumors and vascular diseases, impacting targeted nanomedicine efficacy. Tumors with a high MMP-9 expression (e.g., breast, glioblastoma, colorectal, melanoma, and lung cancers) are particularly amenable to targeted therapies [143,144,146,148]. In tumors with a moderate MMP-9 expression (e.g., prostate, pancreatic, and head and neck cancers), nanoprobes and other diagnostic tools can assess MMP-9 levels to guide personalized strategies [149], and MMP-9-targeted nanomedicines may still impede the disease progression. However, in lesions with a low MMP-9 expression, the potential for significant off-target effects limits the utility of MMP-9-targeted therapies.

5. MMP-9-Related Nanomedicines in Tumor Angiogenesis and Metastasis

5.1. MMP-9-Targeted Nanomedicine in Anti-Tumor Angiogenesis and Metastasis Therapy

Liposomes represent one of the most clinically advanced nanodrug delivery systems. Gao et al. modified cationic liposomes with low-molecular-weight gelatin (an MMP-9 substrate) to reduce the interstitial fluid pressure, thereby improving drug delivery and enabling the MMP-9-dependent release of doxorubicin, quercetin, and imatinib. This approach significantly promoted apoptosis and inhibited metastasis in breast cancer cells [143]. Similarly, Han et al. designed cationic liposomes modified with an MMP-9-cleavable peptide (OMPE), a glutamate-rich fragment, and hydrophobic oleic acid, forming anionic nanohybrids. MMP-9 cleavage reversed the surface charge to cationic, enhancing drug endocytosis [140]. Zhang et al. constructed hybrid nanovesicles with surfaces composed of cancer cell membranes and MMP-9-responsive liposome membranes. Within the tumor microenvironment, MMP-9 activation of the nanovesicle surface peptides increased their uptake, and they subsequently released siRNAs and chemotherapeutic drugs to effectively inhibit non-small cell lung cancer growth [150]. Deng et al. designed liposomal spherical nucleic acids (SNAs) composed of DOPE, adriamycin, CpG, and MMP-9-responsive peptides. In the MMP-9- and glutathione-rich tumor microenvironment, the MMP-9-mediated degradation of the peptides released chemotherapeutic drugs, enhancing dendritic cell activation, T cell expansion, and inhibiting tumor growth [151].
Polymer nanoparticles offer flexible composition and structure, enabling the encapsulation of hydrophobic drugs for solubilization and drug loading via chemical conjugation. This multifunctional delivery approach enhances controlled release and reduces chemotherapeutic toxicity. Porta et al. constructed paclitaxel-loaded PDMS–PMOXA polymerosomes with surfaces modified with MMP-9-cleavable SRL peptides. These polymerosomes accumulated in the liver and tail vein of zebrafish embryos, significantly reducing tumor cells in MCF7-transplanted embryos [147]. Battistella et al. prepared an amphiphilic diblock copolymer comprising a hydrophilic MMP-9-responsive peptide and a hydrophobic Toll-like receptor agonist. In circulation, the polymer self-assembled into spheres. In the tumor environment, the MMP-9-mediated hydrolysis of the hydrophilic peptide exposed and retained the hydrophobic fragment in the 4T1 tumor. This accumulation formed micron-sized scaffolds, releasing the immunostimulatory drug, increasing pro-inflammatory cytokines (IP-10, MCP-1, IL-6), and inhibiting tumor growth and metastasis [152]. Li et al. developed hyaluronic acid/poly(lactic-co-glycolic acid)-poly(ethyleneimine) nanoparticles (PP-HA/NPs) co-delivering doxorubicin/quercetin, which downregulated Akt phosphorylation and MMP-9 expression, significantly inhibiting breast cancer cell invasion [153]. Jiang et al. designed a sequential delivery strategy for synergistic breast cancer treatment: combretastatin A4 nanoparticles (a vascular disrupting agent) disrupted neovascularization and exacerbated tumor hypoxia, upregulating MMP-9 and subsequently triggering MMP-9-activated doxorubicin-loaded nanoparticles to enhance chemotherapy targeting [145]. Poly(N-vinylpyrrolidone)-block-poly(ε-caprolactone) (PVP-b-PCL) nanoparticles loaded with tetrandrine downregulated MMP-2/9 expression and upregulated TIMP-3 via a “Trojan strategy,” effectively inhibiting non-small cell lung cancer cell invasion [154]. Low-molecular-weight heparin-tocopheryl succinate (TOS) nanoparticles inhibited G-MDSC-produced MMP-9 and prevented early lung recruitment of tumor-induced G-MDSCs, impeding tumor invasion and colonization [84]. Yu et al. designed MMP-2/9-sensitive, folate receptor-targeting nanoparticles with a sandwich structure. The upregulation of MMP-2/9 in the tumor environment removed the PEG shell, exposing folate, enhancing nanoparticle endocytosis by B16 melanoma cells, improving drug targeting, and promoting intracellular chemotherapy uptake [146]. α-TOS-loaded self-assembled polymer nanoparticles induced intracellular ROS accumulation and endothelial cell apoptosis, reducing MMP-9 expression and inhibiting angiogenesis and tumor invasion in head and neck squamous carcinoma [155]. Ehrsam et al. constructed self-assembled nanoparticles composed of paclitaxel, hemisuccinic acid, and MMP-9-sensitive peptides, demonstrating MMP-9-dependent cytotoxicity in glioblastoma cells [144]. S-triazine-based dendrimers targeted and inhibited MMP-2/9 activity and could be conjugated with cancer cell-specific ligands and anticancer drugs to inhibit hepatocellular carcinoma growth [156]. Nanovesicles containing MMP-9-sensitive lipopeptide and PEG groups released gemcitabine upon MMP-9- and glutathione-mediated hydrolysis, inhibiting pancreatic ductal carcinoma growth [141]. PLGA–PEG polymer particles with outer long-chain PEG- and MMP-9-cleavable linkers created smaller PLGA–b-PEG nanoparticles for enhanced uptake by pancreatic cancer cells [139]. Doxorubicin-conjugated RGD peptide nanoparticles, responsive to MMP-9 and pH, transformed from spheres to rods in the tumor environment, enhancing penetration and doxorubicin release and inducing apoptosis [138]. MMP-2/9-sensitive TGK enzyme-sensitive polymer nanomicelles exhibited specific degradation, improved stability, and higher drug uptake than mPEG2K–α-TOS nanomicelles, achieving ideal local accumulation and targeted release [157].
Inorganic nanoparticles, offering good dispersibility, stability, and unique electromagnetic and optical properties, are promising for thermal therapy and imaging. Magnetic nanoparticles with a surface-modified glucose oxidase layer amplified oxidative stress, promoting MMP-9 expression and facilitating tumor extracellular matrix degradation, which enhanced nanoparticle penetration and inhibited breast tumor growth [142]. Magnetic heating with methotrexate-coupled magnetic nanoparticles reduced angiogenic signals (MMP-9, VEGF-R1) in bladder tumors, decreasing tumor size, promoting tumor destruction, and preventing recurrence [158]. Gold nanoparticles conjugated with cytotoxic protein NKCT1 inhibited MMP-9 expression and metastasis by suppressing p38 MAPK, ERK1/2, PI3K/Akt phosphorylation and NF-κB nuclear translocation in MCF-7 cells [121]. CuS@mSiO2-PEG photothermal nanoparticles reduced HeLa cell metastasis and enhanced survival in tumor-bearing mice by downregulating MMP-2/9 and inhibiting the non-receptor tyrosine kinase/focal adhesion kinase pathway [159]. Gallic acid-loaded gold nanoparticles inhibited EGF-induced Akt and c-Jun phosphorylation in triple-negative breast cancer, suppressing MMP-9 transcription and hindering invasion and metastasis [160]. Macrophage membrane-camouflaged hollow bismuth selenide nanoparticles loaded with quercetin downregulated Akt phosphorylation and MMP-9 expression, inhibiting breast cancer metastasis [161]. MMP-9-sensitive SNAs modified with avidin on mesoporous silica nanoparticles selectively released cisplatin and bortezomib in Kras-mutant lung tumors, synergistically inducing cell death [148]. Gold nanoparticles inhibited MMP-9 activity in prostate cancer cells, promoting anti-cancer cytokine secretion and exerting cytotoxic effects [162]. Zwitterionic tetrapeptides on nanoparticle surfaces, activated by MMP-9 cleavage, triggered nanoparticle auto-aggregation, enhancing drug uptake [137]. Metallofullerenol Gd@C82(OH)22 nanoparticles selectively inhibited MMP-9 activity, treating angiogenesis and invasion in pancreatic cancer [163,164]. Han et al. constructed a dual-enzyme-sensitive nanocarrier with a PEG modification for enhanced circulatory stability. In the pancreatic cancer environment, MMP-9 hydrolyzed the PEG, exposing the RGD-targeting ligand. Lysosomal cathepsin B then cleaved the GFLG peptide conjugated with gemcitabine and CdSe/ZnS quantum dots, facilitating drug release and increasing active gemcitabine intracellularly [165]. Carbon quantum dots/Cu2O complexes (CQDs/Cu2O) specifically reduced MMP-2/9, VEGF, and cytoskeletal component levels, inducing cell death and exhibiting anti-angiogenic effects in SKOV3 ovarian cancer cells [166].

5.2. MMP-9-Targeted Assays for Tumor Cells

Numerous studies utilize MMP-9-targeted vectors as nanoprobes for tumor cell detection, diagnosis, and assessment of invasiveness. Liu et al. developed Au@4-mercaptobenzonitrile@Ag@peptide nanoprobes that anchored to tumor cell membranes, weakening the surface-enhanced Raman scattering (SERS) signal on the cell membrane. This enabled SERS sensors to detect MMP-9 secretion during cellular communication and to assess breast and liver cancer cell invasiveness [13]. Furthermore, fluorescent nanoprobes have been constructed by conjugating pH-sensitive fluorescent dyes to nanoparticles, such as Fe3O4, via MMP-9-responsive peptide chains. MMP-9 cleavage induces fluorescence, enabling the visualization of abnormal pH and MMP-9 overexpression in tumor environments, facilitating tumor invasiveness assessment [14,15,16,167]. Nossier et al. used gelatin to modify colorimetric gold nanoparticles, stabilizing them against aggregation-inducing agents. However, exposure to the bladder cancer marker MMP-2/9 destabilized the nanoparticles, causing a color change. A subsequent interaction with 6-mercaptohexanol (6-MCH) further shifted the color from red to blue, enabling a sensitive, rapid, and non-invasive bladder cancer diagnosis [149]. Black et al. labeled gold nanoparticles with radionuclides I125 and In111 and modified them with MMP-9 substrate peptides to create a contrast agent for single-photon emission computed tomography (SPECT) imaging [168]. The pharmacokinetics of these nanocontrast agents allows for the observation of MMP-9 expression variations among tumors and the assessment of biological behaviors, including tumor invasiveness [169].

5.3. Effect of Tumor Heterogeneity on MMP-9 Targeting Efficacy

Tumor heterogeneity, characterized by diverse cell populations with varying phenotypes and functional traits, contributes to heterogeneous MMP-9 expression [170]. This heterogeneity challenges the effective delivery and distribution of MMP-9-targeted drugs, leading to inconsistent drug efficacy and promoting drug resistance [171]. Moreover, MMP-9’s pivotal role in the tumor microenvironment, including angiogenesis and metastasis, complicates the complete inhibition of MMP-9-associated tumor progression by targeted therapies [10].

6. MMP-9-Related Nanomedicines in Vascular Diseases

6.1. MMP-9-Related Nanomedicines in Macrovascular Diseases

6.1.1. Nanomedicines Regulating MMP-9 Expression in Macrovascular Diseases

Intimal hyperplasia, characterized by smooth muscle cell aggregation in the vascular wall, can lead to post-angioplasty restenosis [172,173]. Given MMP-9’s role in promoting vascular smooth muscle cell proliferation and migration, various nanomedicines aim to maintain endothelial stability by downregulating MMP-9 expression and related pathways. For instance, a docetaxel-loaded LDL-mimetic lipid nanoparticle significantly reduced the expression of MMP-9, NF-κB, and other proteins in a rabbit model of aortic atherosclerosis, achieving an 80% reduction in the atherosclerotic area compared to the controls [174]. Another study utilized a polyethylene glycol/polyethyleneimine nanoparticle (PEG-Et 1:1/shSmad3) loaded with Smad3 shRNA. This efficiently delivered shRNA to vascular smooth muscle cells, downregulating Smad3 and MMP-9 expression while upregulating the MMP-9 inhibitor TIMP-1, ultimately reducing intimal hyperplasia 14 days post-vascular injury [175]. Chen et al. demonstrated that Foxp1 overexpression in vascular endothelial cells inhibited MMP-9 and cyclin-dependent kinase inhibitor 1B (CDKN1B) expression, which regulates VSM proliferation and neointima formation. They restored intimal homeostasis in a Foxp1 knockout mouse model of femoral artery injury-induced neointima by delivering CDKN1B siRNA-loaded RGD peptide-conjugated magnetic nanoparticles to endothelial cells [176].

6.1.2. MMP-9-Responsive Nanocarriers in Macrovascular Diseases

A gadolinium-rich paramagnetic fluorescent nanomicelle conjugated with the NAP9 peptide, which specifically binds the MMP-9 inducer EMMPRIN, was developed. Combined with magnetic resonance (MR) imaging, this nanomicelle enabled the visualization of EMMPRIN content in the carotid artery wall, allowing the non-invasive analysis of MMP-9 secretion in macrophages and vascular smooth muscle cells post-endarterectomy [177]. Nguyen et al. designed an MMP-9-responsive nanocarrier comprising a brush peptide polymer amphiphile with a specific peptide chain. Intravenously injected, this nanocarrier sensed MMP-9, transforming from discrete spherical particles (15–20 nm) to a grid-like structure, remaining at the infarction site for up to four weeks, and facilitating long-term local accumulation, particularly during vascular leakage and MMP-9 overexpression post-acute myocardial infarction [178].

6.2. MMP-9-Related Nanomedicines in Cerebrovascular and Ocular Vascular Diseases

6.2.1. MMP-9-Related Nanomedicines in Cerebrovascular Diseases

Several studies have investigated MMP-9-related nanomedicines in cerebrovascular diseases. Foroshani et al., using a rat model of middle cerebral artery occlusion, found that fullerenol nanoparticles significantly reduced MMP-9 and IL-6 transcription, protected the BBB integrity, and attenuated cerebral edema following ischemia-reperfusion injury [179]. Cai et al. demonstrated that intravenously injected Momordica charantia-derived exosome-like nanoparticles (ELNs) delivered miR-5266, downregulating MMP-9, improving the BBB function, reducing infarction area, and mitigating neurological damage in a rat middle cerebral artery occlusion model [180].

6.2.2. MMP-9-Related Nanomedicines in Ocular Vascular Diseases

Nanoparticles targeting MMP-9 in ophthalmology are relatively scarce, with topical administration (eye drops) and intravitreal injections being the primary delivery routes. Miyagawa et al. designed GEH-RGD NP eye drops comprising epigallocatechin-3-gallate (EGCG), RGD peptide, gelatin, and a hyaluronic acid coating. GEH-RGD NPs suppressed HUVEC tube formation and MMP-9 activity in vitro. Daily topical application in a chemical burn-induced corneal neovascularization mouse model reduced MMP-9 and VEGF expression, inhibiting neovascularization [181]. Zeng et al. developed IL-12-PNP, a PLGA nanoparticle encapsulating IL-12, and administered it intravitreally in a diabetic retinopathy mouse model. IL-12-PNP showed superior MMP-9 and VEGF inhibition compared to IL-12 alone in rat endothelial cells in vitro and in vivo, reducing retinal neovascularization and improving retinal thickness [182]. Recently, C18PGM, a graphene oxide quantum dot (GOQD) nanoparticle modified with MMP-9-responsive peptide chains and loaded with minocycline, was developed. C18PGM was selectively cleaved by MMP-9, releasing minocycline and inhibiting MMP-9 expression in a laser-induced choroidal neovascularization (CNV) mouse model, suppressing choroidal inflammation and neovascularization [183].

6.2.3. Challenges of Blood–Brain Barrier and Blood–Retinal Barrier to Nanodrug Delivery

The BBB and blood–retinal barrier (BRB) stringently regulate molecular passage into brain and retinal tissues, posing significant drug delivery challenges. Tight junctions between endothelial cells restrict paracellular transport and macromolecule diffusion, hindering nanoparticle penetration. Efflux transporters actively restrict drug influx [184]. Additionally, limited and selective receptor-mediated transcytosis further impedes efficient cytoplasmic drug delivery [185].
Strategies to overcome these challenges include focused ultrasound for temporary, localized BBB/BRB disruption [186], intranasal administration for BBB bypass [187], and conjugation with penetrating peptides to enhance cellular uptake and endocytosis [188]. However, careful consideration is needed for potential off-target effects and the optimization of drug dispersion within the target tissue.

7. Effects of the Immune System on MMP-9-Targeted Nanoparticles

Systemically administered nanomedicines, typically via intravenous injection, must contend with immune system interactions. Preventing complement system activation is crucial to avoid nanoparticle clearance by the mononuclear phagocyte system [189]. Furthermore, allergic individuals may experience IgE antibody-mediated hypersensitivity reactions [190]. Certain nanoparticles can also induce excessive pro-inflammatory cytokine release, potentially leading to cytokine release syndrome and life-threatening consequences [191].
Combining MMP-9-targeted nanoparticles with immunotherapy holds considerable promise. These combined approaches can stimulate the immune system by modulating the tumor microenvironment [151], delivering relevant molecules [152], and enhancing anti-tumor immune cell infiltration deep within the tumor [192], facilitating tumor cell elimination or immunogenic cell death. MMP-9-targeted therapies can synergistically enhance anti-tumor efficacy as well [192]. Moreover, in vascular diseases where immune-mediated inflammation is central, MMP-9-targeted therapies can deliver anti-inflammatory agents to localized lesions, minimizing systemic side effects [178].

8. Conclusions

MMP-9 plays a crucial role in tumor and vascular disease development and progression, offering significant diagnostic and therapeutic potential. Nanomedicine provides innovative strategies to modulate MMP-9 activity and exploit its distribution and enzymatic properties for targeted drug delivery. Despite substantial progress in MMP-9-targeted nanomedicines, further research is needed to optimize their efficacy, safety, and clinical translatability across various organs.
Several challenges remain for nanoparticle-based drugs. First, some nanomaterials, particularly metal nanoparticles, exhibit inherent toxicity [193,194]. Furthermore, poorly biocompatible materials can trigger immune responses and inflammation, including complement activation and hypersensitivity reactions, compromising nanomedicine efficacy [195]. Meticulous material selection is therefore essential, prioritizing biocompatible materials and incorporating biomimetic substances or polyethylene glycol to reduce immunogenicity. Second, nanoparticles face challenges with non-specific uptake, even with targeting, due to factors such as tumor heterogeneity, leading to off-target effects. Refining drug delivery strategies, such as developing stimulus-responsive nanomedicines (as discussed herein), is crucial for enhancing controlled release specificity. Finally, the complex design and synthesis of nanoparticles result in high manufacturing costs. Ensuring drug quality and consistency during large-scale production remains challenging, necessitating comprehensive performance characterization. Clinical trials have demonstrated the feasibility of MMP-9-targeted nanomedicines [196].
Further refinements to MMP-9-targeted nanomedicines can enhance their specificity. Regarding the MMP-9 molecule, the computational design of inhibitors tailored to the specific MMP-9 crystal structure can minimize cross-reactivity with other MMP family members by targeting sites beyond the active region [197]. Developing MMP-9-activated prodrugs can also enhance selectivity and reduce off-target effects [198]. For drug delivery systems, besides stimulus-responsive nanomedicines, synergies with physical modalities (e.g., ultrasound for enhanced drug permeability [186]) and imaging technologies (e.g., MRI and PET for visualizing drug distribution [199]) can be explored. Utilizing exosomes or cell membrane-coated biomimetic nanoparticles can improve lesion permeability and targeting precision while reducing immune-mediated clearance [200]. These combined approaches hold promise for enhancing the selectivity of MMP-9-targeted nanomedicine release, minimizing off-target toxicity, and improving drug utilization.
MMP-9-targeted nanoparticles, as a highly effective drug delivery system, show significant promise when combined with conventional chemotherapy or immunotherapy. They efficiently deliver anti-tumor drugs deep into tumor tissue, enhancing their efficacy and reducing systemic toxicity. Nanotherapy can also mitigate drug resistance mechanisms such as drug efflux and enable the sequential release of multiple chemotherapeutic agents, further reducing the resistance potential.
Ultimately, advancing our understanding of MMP-9 biology and refining nanomedicine delivery systems will be critical for the clinical translation of these promising therapies.

Author Contributions

Investigation and writing, X.L.; conceptualization, validation, supervision, review & editing, Z.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Clinical Research Incubation Project, West China Hospital, Sichuan University, grant number 2021HXFH026; the Aier Ophthalmology–Sichuan University Scientific Research Fund Project, grant number 23JZH039; and the Sichuan Provincial Science and Technology Department Key R&D Projects, grant number 2023YFQ0103.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
4T1mouse mammary tumor cell line
6-MCH6-hydroxy-1-hexanethiol
Aktprotein kinase B
AP-1activator protein 1
bFGFbasic fibroblast growth factor
CDKN1Bcycling-dependent kinase inhibitor 1B
CNVchoroidal neovascularization
CpG5′-C-phosphate-G-3′
CQDscarbon quantum dots
DOPE, 11,2-dioleoyl-sn-glycero-3-phosphoethanolamine
EGCGepigallocatechin-3-gallate
ELNsexosome-like nanoparticles
EMMPRINextracellular matrix metalloproteinase inducer
EMTepithelial–mesenchymal transition
ERKextracellular regulated protein kinases
GFLGGly-Phe-Leu-Gly
G-MDSCgranulocyte-like myeloid derived suppressor cells
GOQDgraphene oxide quantum dots
GPRC5BG protein-coupled receptor 5B
GSTglutathione-S-transferase
HIVhuman immunodeficiency virus
HMG-CoA3-hydroxy-3-methylglutaryl coenzyme A
HSP70heat shock 70 kda protein
ICAM-1intercellular adhesion molecule-1
IL-10interleukin 10
IP-10interferon–γ–inducible protein 10
JAK2/STAT3janus kinase 2/signal transduction and activator of transcription 3
JNKC-jun N-terminal kinase
LDLlow-density lipoprotein
LRPlow density lipoprotein receptor-related protein
MAPKmitogen-activated protein kinase
MCF7a breast cancer cell line
MMPmatrix metalloproteinase
mPEGmethoxypolyethylene glycol
NF-κBnuclear factor kappa-light-chain-enhancer of activated B cells
NKCT1purified naja kaouthia protein toxin
NMDARN-methyl-D-aspartate receptor
PAFplatelet-activating factor
PAR-1protease-activated receptor 1
PARP-1poly [adp-ribose] polymerase 1
PDIprotein disulfide isomerase
PDMS–PMOXApoly(dimethylsiloxane)-poly-b-(methyloxazoline)
PEGpolyethylene glycol
PI3Kphosphoinositide 3-kinase
PKCprotein kinase C
PLGApoly(lactic-co-glycolic acid)
PP-HA/NPshyaluronic acid/poly(lactic acid)-glycolic acid-poly(ethyleneimine) nanoparticles
PTKpoly (5,5-dimethyl-4,6-dithio-propylene glycol azelate)
PVP-b-PCLpoly(N-vinylpyrrolidone)-block-poly(ε-caprolactone)
RGDL-Arginyl-Glycyl-L-Aspartic acid
ROCKrho-associated protein kinase
ROSreactive oxygen species
RPEretinal pigment epithelium
SERSsurface-enhanced Raman scattering
Shhsonic hedgehog protein
SKOV3
SMCs
an ovarian cancer cell line
smooth muscle cells
SNAsspherical nucleic acid
SPECTsingle-photon emission computed tomography
TAK1TGF-β-activated kinase 1
TGF-βtransforming growth factor β
TGKprotein-glutamine gamma-glutamyltransferase K
TNF-αtumor necrosis factor α
TOStocopheryl succinate
tPAtissue plasminogen activator
TRPV4transient receptor potential cation channel subfamily V member 4
VEGFvascular endothelial growth factor
VEGF-R1
VSM
vascular endothelial growth factor receptor 1
vascular smooth muscle
ZOzonula occludens
α-TPGSα-tocopherol polyethylene glycol succinate

References

  1. Verma, R.P.; Hansch, C. Matrix Metalloproteinases (MMPs): Chemical-Biological Functions and (Q)SARs. Bioorg. Med. Chem. 2007, 15, 2223–2268. [Google Scholar] [CrossRef] [PubMed]
  2. de Almeida, L.G.N.; Thode, H.; Eslambolchi, Y.; Chopra, S.; Young, D.; Gill, S.; Devel, L.; Dufour, A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol. Rev. 2022, 74, 712–768. [Google Scholar]
  3. Wang, X.; Khalil, R.A. Matrix Metalloproteinases, Vascular Remodeling, and Vascular Disease. Adv. Pharmacol. 2018, 81, 241–330. [Google Scholar] [PubMed]
  4. Nagase, H.; Woessner, J.F. Matrix Metalloproteinases. J. Biol. Chem. 1999, 274, 21491–21494. [Google Scholar] [PubMed]
  5. Van Wart, H.E.; Birkedal-Hansen, H. The Cysteine Switch: A Principle of Regulation of Metalloproteinase Activity with Potential Applicability to the Entire Matrix Metalloproteinase Gene Family. Proc. Natl. Acad. Sci. USA 1990, 87, 5578–5582. [Google Scholar]
  6. Bode, W. A Helping Hand for Collagenases: The Haemopexin-like Domain. Structure 1995, 3, 527–530. [Google Scholar] [PubMed]
  7. Vandooren, J.; Van den Steen, P.E.; Opdenakker, G. Biochemistry and Molecular Biology of Gelatinase B or Matrix Metalloproteinase-9 (MMP-9): The next Decade. Crit. Rev. Biochem. Mol. Biol. 2013, 48, 222–272. [Google Scholar] [PubMed]
  8. Buttacavoli, M.; Di Cara, G.; Roz, E.; Pucci-Minafra, I.; Feo, S.; Cancemi, P. Integrated Multi-Omics Investigations of Metalloproteinases in Colon Cancer: Focus on MMP2 and MMP9. Int. J. Mol. Sci. 2021, 22, 12389. [Google Scholar] [CrossRef]
  9. Barillari, G. The Impact of Matrix Metalloproteinase-9 on the Sequential Steps of the Metastatic Process. Int. J. Mol. Sci. 2020, 21, 4526. [Google Scholar] [CrossRef]
  10. Kanugula, A.K.; Adapala, R.K.; Jamaiyar, A.; Lenkey, N.; Guarino, B.D.; Liedtke, W.; Yin, L.; Paruchuri, S.; Thodeti, C.K. Endothelial TRPV4 Channels Prevent Tumor Growth and Metastasis via Modulation of Tumor Angiogenesis and Vascular Integrity. Angiogenesis 2021, 24, 647–656. [Google Scholar]
  11. Lindsey, M.L. Assigning Matrix Metalloproteinase Roles in Ischaemic Cardiac Remodelling. Nat. Rev. Cardiol. 2018, 15, 471–479. [Google Scholar] [PubMed]
  12. Sharifi, M.A.; Wierer, M.; Dang, T.A.; Milic, J.; Moggio, A.; Sachs, N.; von Scheidt, M.; Hinterdobler, J.; Müller, P.; Werner, J.; et al. ADAMTS-7 Modulates Atherosclerotic Plaque Formation by Degradation of TIMP-1. Circ. Res. 2023, 133, 674–686. [Google Scholar] [CrossRef]
  13. Liu, X.; Gu, J.; Wang, J.; Zhang, W.; Wang, Y.; Xu, Z. Cell Membrane-Anchored SERS Biosensor for the Monitoring of Cell-Secreted MMP-9 during Cell-Cell Communication. ACS Sens. 2023, 8, 4307–4314. [Google Scholar] [CrossRef] [PubMed]
  14. Ma, T.; Hou, Y.; Zeng, J.; Liu, C.; Zhang, P.; Jing, L.; Shangguan, D.; Gao, M. Dual-Ratiometric Target-Triggered Fluorescent Probe for Simultaneous Quantitative Visualization of Tumor Microenvironment Protease Activity and pH in Vivo. J. Am. Chem. Soc. 2018, 140, 211–218. [Google Scholar] [CrossRef]
  15. Rainu, S.; Parameswaran, S.; Krishnakumar, S.; Singh, N. Dual-Sensitive Fluorescent Nanoprobes for Detection of Matrix Metalloproteinases and Low pH in a 3D Tumor Microenvironment. J. Mater. Chem. B 2022, 10, 5388–5401. [Google Scholar] [CrossRef] [PubMed]
  16. Hou, Y.; Zhou, J.; Gao, Z.; Sun, X.; Liu, C.; Shangguan, D.; Yang, W.; Gao, M. Protease-Activated Ratiometric Fluorescent Probe for pH Mapping of Malignant Tumors. ACS Nano 2015, 9, 3199–3205. [Google Scholar] [CrossRef] [PubMed]
  17. Cancers. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer/ (accessed on 4 April 2025).
  18. Cardiovascular Diseases. Available online: https://www.who.int/health-topics/cardiovascular-diseases (accessed on 23 March 2025).
  19. World Stroke Day 2022. Available online: https://www.who.int/srilanka/news/detail/29-10-2022-world-stroke-day-2022 (accessed on 23 March 2025).
  20. Teo, Z.L.; Tham, Y.-C.; Yu, M.; Chee, M.L.; Rim, T.H.; Cheung, N.; Bikbov, M.M.; Wang, Y.X.; Tang, Y.; Lu, Y.; et al. Global Prevalence of Diabetic Retinopathy and Projection of Burden through 2045: Systematic Review and Meta-Analysis. Ophthalmology 2021, 128, 1580–1591. [Google Scholar] [CrossRef] [PubMed]
  21. Zuo, Y.; Sun, R.; Del Piccolo, N.; Stevens, M.M. Microneedle-Mediated Nanomedicine to Enhance Therapeutic and Diagnostic Efficacy. Nano Converg. 2024, 11, 15. [Google Scholar] [CrossRef]
  22. Ren, S.; Xu, Y.; Dong, X.; Mu, Q.; Chen, X.; Yu, Y.; Su, G. Nanotechnology-Empowered Combination Therapy for Rheumatoid Arthritis: Principles, Strategies, and Challenges. J. Nanobiotechnol. 2024, 22, 431. [Google Scholar] [CrossRef]
  23. Zhang, Y.; Li, M.; Gao, X.; Chen, Y.; Liu, T. Nanotechnology in Cancer Diagnosis: Progress, Challenges and Opportunities. J. Hematol. Oncol. 2019, 12, 137. [Google Scholar] [CrossRef]
  24. Gonnelli, A.; Gerbé de Thoré, M.; Ermini, M.L.; Frusca, V.; Zamborlin, A.; Signolle, N.; Bawa, O.; Clémenson, C.; Meziani, L.; Bergeron, P.; et al. Nonpersistent Nanoarchitectures Enhance Concurrent Chemoradiotherapy in an Immunocompetent Orthotopic Model of HPV+ Head/Neck Carcinoma. Adv. Mater. 2024, 36, e2400949. [Google Scholar] [CrossRef]
  25. Ye, Z.; Liu, J.; Liu, Y.; Zhao, Y.; Li, Z.; Xu, B.; Chen, D.; Wang, B.; Wang, Q.; Shen, Y. Hybrid Nanopotentiators with Dual Cascade Amplification for Glioma Combined Interventional Therapy. J. Control Release 2024, 372, 95–112. [Google Scholar] [CrossRef] [PubMed]
  26. Zhou, K.; Cai, W.; Tan, Y.; Zhao, Z.; Liu, J. Highly Controllable Nanoassemblies of Luminescent Gold Nanoparticles with Abnormal Disassembly-Induced Emission Enhancement for In Vivo Imaging Applications. Angew. Chem. Int. Ed. Engl. 2022, 61, e202212214. [Google Scholar] [CrossRef] [PubMed]
  27. Martorana, A.; Pitarresi, G.; Palumbo, F.S.; Catania, V.; Schillaci, D.; Mauro, N.; Fiorica, C.; Giammona, G. Fabrication of Silver Nanoparticles by a Diethylene Triamine-Hyaluronic Acid Derivative and Use as Antibacterial Coating. Carbohydr. Polym. 2022, 295, 119861. [Google Scholar] [PubMed]
  28. Chen, X.; Li, H.; Qiao, X.; Jiang, T.; Fu, X.; He, Y.; Zhao, X. Agarose Oligosaccharide- Silver Nanoparticle- Antimicrobial Peptide- Composite for Wound Dressing. Carbohydr. Polym. 2021, 269, 118258. [Google Scholar] [PubMed]
  29. Yu, B.; Wang, Y.; Bing, T.; Tang, Y.; Huang, J.; Xiao, H.; Liu, C.; Yu, Y. Platinum Prodrug Nanoparticles with COX-2 Inhibition Amplify Pyroptosis for Enhanced Chemotherapy and Immune Activation of Pancreatic Cancer. Adv. Mater. 2024, 36, e2310456. [Google Scholar] [CrossRef]
  30. Lv, K.; Hou, M.; Kou, Y.; Yu, H.; Liu, M.; Zhao, T.; Shen, J.; Huang, X.; Zhang, J.; Mady, M.F.; et al. Black Titania Janus Mesoporous Nanomotor for Enhanced Tumor Penetration and Near-Infrared Light-Triggered Photodynamic Therapy. ACS Nano 2024, 18, 13910–13923. [Google Scholar] [CrossRef]
  31. Guo, S.; Shu, G.; Luo, H.; Kuang, X.; Zheng, L.; Wang, C.; Zhou, C.-A.; Song, L.; Ma, K.; Yue, H. Low-Cytotoxic Core-Sheath ZnO NWs@TiO2-xNy Triggered Piezo-Photocatalytic Antibacterial Activity. ACS Appl. Mater. Interfaces 2024, 16, 24410–24420. [Google Scholar] [CrossRef] [PubMed]
  32. Jacqmarcq, C.; Picot, A.; Flon, J.; Lebrun, F.; Martinez de Lizarrondo, S.; Naveau, M.; Bernay, B.; Goux, D.; Rubio, M.; Malzert-Fréon, A.; et al. MRI-Based Microthrombi Detection in Stroke with Polydopamine Iron Oxide. Nat. Commun. 2024, 15, 5070. [Google Scholar] [CrossRef] [PubMed]
  33. Zhu, L.; Lian, W.; Yu, N.; Meng, J.; Zeng, H.; Wang, Y.; Wang, X.; Wen, M.; Chen, Z. Erythrocyte-Membrane-Camouflaged Magnetic Nanocapsules with Photothermal/Magnetothermal Effects for Thrombolysis. Adv. Healthc. Mater. 2024, 13, e2400127. [Google Scholar] [CrossRef]
  34. Xiao, H.; Meng, X.; Li, S.; Li, Z.; Fang, S.; Wang, Y.; Li, J.; Tang, J.; Ma, L. Combined Drug Anti-Deep Vein Thrombosis Therapy Based on Platelet Membrane Biomimetic Targeting Nanotechnology. Biomaterials 2024, 311, 122670. [Google Scholar]
  35. Pan, P.; Yue, Q.; Yang, X.; Ren, Y.; Alharthi, F.A.; Alghamdi, A.; Su, J.; Deng, Y. Structure Engineering of Yolk-Shell Magnetic Mesoporous Silica Microspheres with Broccoli-Like Morphology for Efficient Catalysis and Enhanced Cellular Uptake. Small 2021, 17, e2006925. [Google Scholar] [PubMed]
  36. Tanabe, M.; Ando, K.; Komatsu, R.; Morigaki, K. Nanofluidic Biosensor Created by Bonding Patterned Model Cell Membrane and Silicone Elastomer with Silica Nanoparticles. Small 2018, 14, e1802804. [Google Scholar] [CrossRef] [PubMed]
  37. Dassanayake, T.M.; Dassanayake, A.C.; Abeydeera, N.; Pant, B.D.; Jaroniec, M.; Kim, M.-H.; Huang, S.D. An Aluminum Lining to the Dark Cloud of Silver Resistance: Harnessing the Power of Potent Antimicrobial Activity of γ-Alumina Nanoparticles. Biomater. Sci. 2021, 9, 7996–8006. [Google Scholar] [PubMed]
  38. Yan, Y.; Liu, Z.; Pang, W.; Huang, S.; Deng, M.; Yao, J.; Huang, Q.; Jin, M.; Shui, L. Integrated Biosensor Array for Multiplex Biomarkers Cancer Diagnosis via In-Situ Self-Assembly Carbon Nanotubes with an Ordered Inverse-Opal Structure. Biosens. Bioelectron. 2024, 262, 116528. [Google Scholar]
  39. Sui, N.; Ji, Y.; Li, M.; Zheng, F.; Shao, S.; Li, J.; Liu, Z.; Wu, J.; Zhao, J.; Li, L.-J. Photoprogrammed Multifunctional Optoelectronic Synaptic Transistor Arrays Based on Photosensitive Polymer-Sorted Semiconducting Single-Walled Carbon Nanotubes for Image Recognition. Adv. Sci. 2024, 11, e2401794. [Google Scholar] [CrossRef] [PubMed]
  40. Behrouznejad, B.; Sadat, S.B.; Masaeli, E. The Orchestration of Sustained Drug Delivery by Bacterial Cellulose/Gelatin Nanocomposites Reinforced with Carboxylic Carbon Nanotubes. Carbohydr. Polym. 2024, 333, 121917. [Google Scholar] [CrossRef]
  41. Sun, J.; Jia, W.; Qi, H.; Huo, J.; Liao, X.; Xu, Y.; Wang, J.; Sun, Z.; Liu, Y.; Liu, J.; et al. An Antioxidative and Active Shrinkage Hydrogel Integratedly Promotes Re-Epithelization and Skin Constriction for Enhancing Wound Closure. Adv. Mater. 2024, 36, e2312440. [Google Scholar] [CrossRef]
  42. Zaręba, N.; Więcławik, K.; Kizek, R.; Hosnedlova, B.; Kepinska, M. The Impact of Fullerenes as Doxorubicin Nano-Transporters on Metallothionein and Superoxide Dismutase Status in MCF-10A Cells. Pharmaceutics 2022, 14, 102. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, L.; Chang, Z.; Guo, B.; Lu, Y.; Lu, X.; Ren, Q.; Lv, A.; Nie, J.; Ji, D.; Rotenberg, M.Y.; et al. Robust, Stretchable Bioelectronic Interfaces for Cardiac Pacing Enabled by Interfacial Transfer of Laser-Induced Graphene via Water-Response, Nonswellable PVA Gels. Biosens. Bioelectron. 2024, 261, 116453. [Google Scholar]
  44. Lu, J.; Chen, X.; Ding, X.; Jia, Z.; Li, M.; Zhang, M.; Liu, F.; Tang, K.; Yu, X.; Li, G. Droplet Micro-Sensor and Detection of Respiratory Droplet Transmission. Adv. Sci. 2024, 11, e2401940. [Google Scholar]
  45. Toro-González, M.; Akingbesote, N.; Bible, A.; Pal, D.; Sanders, B.; Ivanov, A.S.; Jansone-Popova, S.; Popovs, I.; Benny, P.; Perry, R.; et al. Development of 225Ac-Doped Biocompatible Nanoparticles for Targeted Alpha Therapy. J. Nanobiotechnol. 2024, 22, 306. [Google Scholar] [CrossRef]
  46. Baruah, N.; Ahamad, N.; Halder, P.; Koley, H.; Katti, D.S. Facile Synthesis of Multi-Faceted, Biomimetic and Cross-Protective Nanoparticle-Based Vaccines for Drug-Resistant Shigella: A Flexible Platform Technology. J. Nanobiotechnol. 2023, 21, 34. [Google Scholar]
  47. Blaya-Cánovas, J.L.; Griñán-Lisón, C.; Blancas, I.; Marchal, J.A.; Ramírez-Tortosa, C.; López-Tejada, A.; Benabdellah, K.; Cortijo-Gutiérrez, M.; Cano-Cortés, M.V.; Graván, P.; et al. Autologous Patient-Derived Exhausted Nano T-Cells Exploit Tumor Immune Evasion to Engage an Effective Cancer Therapy. Mol. Cancer 2024, 23, 83. [Google Scholar] [CrossRef]
  48. Pathan, S.; Jayakannan, M. Zwitterionic Strategy to Stabilize Self-Immolative Polymer Nanoarchitecture under Physiological pH for Drug Delivery In Vitro and In Vivo. Adv. Healthc. Mater. 2024, 13, e2304599. [Google Scholar] [CrossRef] [PubMed]
  49. Silva, J.C.; Udangawa, R.N.; Chen, J.; Mancinelli, C.D.; Garrudo, F.F.F.; Mikael, P.E.; Cabral, J.M.S.; Ferreira, F.C.; Linhardt, R.J. Kartogenin-Loaded Coaxial PGS/PCL Aligned Nanofibers for Cartilage Tissue Engineering. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 107, 110291. [Google Scholar] [CrossRef]
  50. Wang, K.; Gao, Y.; Wu, S.; Zhang, J.; Zhu, M.; Chen, X.; Fu, X.; Duan, X.; Men, K. Dual-mRNA Delivery Using Tumor Cell Lysate-Based Multifunctional Nanoparticles as an Efficient Colon Cancer Immunogene Therapy. Int. J. Nanomed. 2024, 19, 4779–4801. [Google Scholar] [CrossRef] [PubMed]
  51. Luo, X.; Yan, S.; Chen, G.; Wang, Y.; Zhang, X.; Lan, J.; Chen, J.; Yao, X. A Cavity Induced Mode Hybridization Plasmonic Sensor for Portable Detection of Exosomes. Biosens. Bioelectron. 2024, 261, 116492. [Google Scholar] [CrossRef] [PubMed]
  52. Goldstein, Y.; Cohen, O.T.; Wald, O.; Bavli, D.; Kaplan, T.; Benny, O. Particle Uptake in Cancer Cells Can Predict Malignancy and Drug Resistance Using Machine Learning. Sci. Adv. 2024, 10, eadj4370. [Google Scholar] [CrossRef] [PubMed]
  53. Hou, Y.; Zhu, L.; Ye, X.; Ke, Q.; Zhang, Q.; Xie, X.; Piao, J.-G.; Wei, Y. Integrated Oral Microgel System Ameliorates Renal Fibrosis by Hitchhiking Co-Delivery and Targeted Gut Flora Modulation. J. Nanobiotechnol. 2024, 22, 305. [Google Scholar] [CrossRef]
  54. Li, B.; Niu, H.; Zhao, X.; Huang, X.; Ding, Y.; Dang, K.; Yang, T.; Chen, Y.; Ma, J.; Liu, X.; et al. Targeted Anti-Cancer Therapy: Co-Delivery of VEGF siRNA and Phenethyl Isothiocyanate (PEITC) via cRGD-Modified Lipid Nanoparticles for Enhanced Anti-Angiogenic Efficacy. Asian J. Pharm. Sci. 2024, 19, 100891. [Google Scholar] [CrossRef] [PubMed]
  55. Jiang, W.; Peng, J.; Jiang, N.; Zhang, W.; Liu, S.; Li, J.; Duan, D.; Li, Y.; Peng, C.; Yan, Y.; et al. Chitosan Phytate Nanoparticles: A Synergistic Strategy for Effective Dental Caries Prevention. ACS Nano 2024, 18, 13528–13537. [Google Scholar] [CrossRef] [PubMed]
  56. Song, C.; Wu, X.; Wang, Y.; Wang, J.; Zhao, Y. Cuttlefish-Inspired Photo-Responsive Antibacterial Microparticles with Natural Melanin Nanoparticles Spray. Small 2024, 20, e2310444. [Google Scholar] [CrossRef]
  57. Liu, X.; Chen, W.; Zhao, D.; Liu, X.; Wang, Y.; Chen, Y.; Ma, X. Enzyme-Powered Hollow Nanorobots for Active Microsampling Enabled by Thermoresponsive Polymer Gating. ACS Nano 2022, 16, 10354–10363. [Google Scholar] [CrossRef]
  58. Zaritski, A.; Castillo-Ecija, H.; Kumarasamy, M.; Peled, E.; Sverdlov Arzi, R.; Carcaboso, Á.M.; Sosnik, A. Selective Accumulation of Galactomannan Amphiphilic Nanomaterials in Pediatric Solid Tumor Xenografts Correlates with GLUT1 Gene Expression. ACS Appl. Mater. Interfaces 2019, 11, 38483–38496. [Google Scholar] [CrossRef] [PubMed]
  59. Yu, K.; Liang, B.; Zheng, Y.; Exner, A.; Kolios, M.; Xu, T.; Guo, D.; Cai, X.; Wang, Z.; Ran, H.; et al. PMMA-Fe3O4 for Internal Mechanical Support and Magnetic Thermal Ablation of Bone Tumors. Theranostics 2019, 9, 4192–4207. [Google Scholar] [CrossRef]
  60. Dosta, P.; Dion, M.Z.; Prado, M.; Hurtado, P.; Riojas-Javelly, C.J.; Cryer, A.M.; Soria, Y.; Andrews Interiano, N.; Muñoz-Taboada, G.; Artzi, N. Matrix Metalloproteinase- and pH-Sensitive Nanoparticle System Enhances Drug Retention and Penetration in Glioblastoma. ACS Nano 2024, 18, 14145–14160. [Google Scholar] [CrossRef]
  61. Zhang, P.; Li, Z.; Cao, W.; Tang, J.; Xia, Y.; Peng, L.; Ma, J. A PD-L1 Antibody-Conjugated PAMAM Dendrimer Nanosystem for Simultaneously Inhibiting Glycolysis and Promoting Immune Response in Fighting Breast Cancer. Adv. Mater. 2023, 35, e2305215. [Google Scholar] [CrossRef] [PubMed]
  62. Ding, L.; Lyu, Z.; Perles-Barbacaru, T.-A.; Huang, A.Y.-T.; Lian, B.; Jiang, Y.; Roussel, T.; Galanakou, C.; Giorgio, S.; Kao, C.-L.; et al. Modular Self-Assembling Dendrimer Nanosystems for Magnetic Resonance and Multimodality Imaging of Tumors. Adv. Mater. 2024, 36, e2308262. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, X.; Meng, X.; Mao, K.; Chen, H.; Cong, X.; Liu, F.; Wang, J.; Liu, S.; Xin, Y.; Zhu, G.; et al. Maleimide as the PEG End-Group Promotes Macrophage-Targeted Drug Delivery of PEGylated Nanoparticles in Vivo by Enhancing Interaction with Circulating Erythrocytes. Biomaterials 2023, 300, 122187. [Google Scholar] [CrossRef] [PubMed]
  64. Schnorenberg, M.R.; Hawley, K.M.; Thomas-Toth, A.T.; Watkins, E.A.; Tian, Y.; Ting, J.M.; Leak, L.B.; Kucera, I.M.; Raczy, M.M.; Kung, A.L.; et al. Targeted Polymersome Delivery of a Stapled Peptide for Drugging the Tumor Protein P53:BCL-2-Family Axis in Diffuse Large B-Cell Lymphoma. ACS Nano 2023, 17, 23374–23390. [Google Scholar] [PubMed]
  65. Valsalakumari, R.; Pandya, A.D.; Prasmickaite, L.; Kvalvaag, A.; Myrann, A.G.; Åslund, A.K.O.; Kjos, M.S.; Fontecha-Cuenca, C.; Haroon, H.B.; Ribeiro, A.R.S.; et al. Preclinical Efficacy of Cabazitaxel Loaded Poly(2-Alkyl Cyanoacrylate) Nanoparticle Variants. Int. J. Nanomed. 2024, 19, 3009–3029. [Google Scholar]
  66. Park, J.S.; Seo, J.H.; Jeong, M.Y.; Yang, I.G.; Kim, J.S.; Kim, J.H.; Ho, M.J.; Jin, S.G.; Choi, M.K.; Choi, Y.S.; et al. Carboxymethyl Cellulose-Based Rotigotine Nanocrystals-Loaded Hydrogel for Increased Transdermal Delivery with Alleviated Skin Irritation. Carbohydr. Polym. 2024, 338, 122197. [Google Scholar]
  67. Rong, M.; Liu, D.; Xu, X.; Li, A.; Bai, Y.; Yang, G.; Liu, K.; Zhang, Z.; Wang, L.; Wang, K.; et al. A Superparamagnetic Composite Hydrogel Scaffold as In Vivo Dynamic Monitorable Theranostic Platform for Osteoarthritis Regeneration. Adv. Mater. 2024, 36, e2405641. [Google Scholar] [CrossRef] [PubMed]
  68. Xi, X.; Lei, F.; Gao, K.; Li, J.; Liu, R.; Karpf, A.R.; Bronich, T.K. Ligand-Installed Polymeric Nanocarriers for Combination Chemotherapy of EGFR-Positive Ovarian Cancer. J. Control Release 2023, 360, 872–887. [Google Scholar] [PubMed]
  69. Gao, Z.; He, T.; Zhang, P.; Li, X.; Zhang, Y.; Lin, J.; Hao, J.; Huang, P.; Cui, J. Polypeptide-Based Theranostics with Tumor-Microenvironment-Activatable Cascade Reaction for Chemo-Ferroptosis Combination Therapy. ACS Appl. Mater. Interfaces 2020, 12, 20271–20280. [Google Scholar]
  70. Han, J.; Choi, S.; Hong, J.; Gang, D.; Lee, S.; Shin, K.; Ko, J.; Kim, J.-U.; Hwang, N.S.; An, Y.-H.; et al. Superoxide Dismutase-Mimetic Polyphenol-Based Carbon Dots for Multimodal Bioimaging and Treatment of Atopic Dermatitis. ACS Appl. Mater. Interfaces 2024, 16, 24308–24320. [Google Scholar]
  71. Baek, G.W.; Kim, Y.J.; Kim, J.; Chang, J.H.; Kim, U.; An, S.; Park, J.; Yu, S.; Bae, W.K.; Lim, J.; et al. Memristive Switching Mechanism in Colloidal InP/ZnSe/ZnS Quantum Dot-Based Synaptic Devices for Neuromorphic Computing. Nano Lett. 2024, 24, 5855–5861. [Google Scholar]
  72. Balamur, R.; Eren, G.O.; Kaleli, H.N.; Karatum, O.; Kaya, L.; Hasanreisoglu, M.; Nizamoglu, S. A Retina-Inspired Optoelectronic Synapse Using Quantum Dots for Neuromorphic Photostimulation of Neurons. Adv. Sci. 2024, 11, e2401753. [Google Scholar] [CrossRef] [PubMed]
  73. Gupta, P.K.; Son, S.E.; Seong, G.H. Functionalized Ultra-Fine Bimetallic PtRu Alloy Nanoparticle with High Peroxidase-Mimicking Activity for Rapid and Sensitive Colorimetric Quantification of C-Reactive Protein. Mikrochim. Acta 2021, 188, 119. [Google Scholar] [CrossRef] [PubMed]
  74. Mu, Q.; Lin, G.; Stephen, Z.R.; Chung, S.; Wang, H.; Patton, V.K.; Gebhart, R.N.; Zhang, M. In Vivo Serum Enabled Production of Ultrafine Nanotherapeutics for Cancer Treatment. Mater Today 2020, 38, 10–23. [Google Scholar] [CrossRef]
  75. Thangudu, S.; Yu, C.-C.; Lee, C.-L.; Liao, M.-C.; Su, C.-H. Magnetic, Biocompatible FeCO3 Nanoparticles for T2-Weighted Magnetic Resonance Imaging of In Vivo Lung Tumors. J. Nanobiotechnol. 2022, 20, 157. [Google Scholar] [CrossRef]
  76. Nafee, N.; Gaber, D.M.; Abouelfetouh, A.; Alseqely, M.; Empting, M.; Schneider, M. Enzyme-Linked Lipid Nanocarriers for Coping Pseudomonal Pulmonary Infection. Would Nanocarriers Complement Biofilm Disruption or Pave Its Road? Int. J. Nanomed. 2024, 19, 3861–3890. [Google Scholar] [CrossRef]
  77. Dongre, A.; Weinberg, R.A. New Insights into the Mechanisms of Epithelial-Mesenchymal Transition and Implications for Cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84. [Google Scholar] [CrossRef]
  78. Mittal, V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu. Rev. Pathol. 2018, 13, 395–412. [Google Scholar] [CrossRef] [PubMed]
  79. Pan, G.; Liu, Y.; Shang, L.; Zhou, F.; Yang, S. EMT-Associated microRNAs and Their Roles in Cancer Stemness and Drug Resistance. Cancer Commun 2021, 41, 199–217. [Google Scholar] [CrossRef] [PubMed]
  80. Erin, N.; Grahovac, J.; Brozovic, A.; Efferth, T. Tumor Microenvironment and Epithelial Mesenchymal Transition as Targets to Overcome Tumor Multidrug Resistance. Drug Resist. Updat. 2020, 53, 100715. [Google Scholar] [CrossRef] [PubMed]
  81. Chen, J.; Ding, Z.-Y.; Li, S.; Liu, S.; Xiao, C.; Li, Z.; Zhang, B.-X.; Chen, X.-P.; Yang, X. Targeting Transforming Growth Factor-β Signaling for Enhanced Cancer Chemotherapy. Theranostics 2021, 11, 1345–1363. [Google Scholar] [CrossRef]
  82. Cai, H.-P.; Wang, J.; Xi, S.-Y.; Ni, X.-R.; Chen, Y.-S.; Yu, Y.-J.; Cen, Z.-W.; Yu, Z.-H.; Chen, F.-R.; Guo, C.-C.; et al. Tenascin-Cmediated Vasculogenic Mimicry Formation via Regulation of MMP2/MMP9 in Glioma. Cell Death Dis. 2019, 10, 879. [Google Scholar] [CrossRef] [PubMed]
  83. Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegué, E.; Song, H.; Vandenberg, S.; Johnson, R.S.; Werb, Z.; et al. HIF1alpha Induces the Recruitment of Bone Marrow-Derived Vascular Modulatory Cells to Regulate Tumor Angiogenesis and Invasion. Cancer Cell 2008, 13, 206–220. [Google Scholar] [CrossRef] [PubMed]
  84. Long, Y.; Lu, Z.; Xu, S.; Li, M.; Wang, X.; Zhang, Z.; He, Q. Self-Delivery Micellar Nanoparticles Prevent Premetastatic Niche Formation by Interfering with the Early Recruitment and Vascular Destruction of Granulocytic Myeloid-Derived Suppressor Cells. Nano Lett. 2020, 20, 2219–2229. [Google Scholar] [CrossRef] [PubMed]
  85. Pandita, A.; Ekstrand, M.; Bjursten, S.; Zhao, Z.; Fogelstrand, P.; Le Gal, K.; Ny, L.; Bergo, M.O.; Karlsson, J.; Nilsson, J.A.; et al. Intussusceptive Angiogenesis in Human Metastatic Malignant Melanoma. Am. J. Pathol. 2021, 191, 2023–2038. [Google Scholar] [CrossRef] [PubMed]
  86. Bruno, A.; Bassani, B.; D’Urso, D.G.; Pitaku, I.; Cassinotti, E.; Pelosi, G.; Boni, L.; Dominioni, L.; Noonan, D.M.; Mortara, L.; et al. Angiogenin and the MMP9-TIMP2 Axis Are Up-Regulated in Proangiogenic, Decidual NK-like Cells from Patients with Colorectal Cancer. FASEB J. 2018, 32, 5365–5377. [Google Scholar] [CrossRef]
  87. Ross, R. Cell Biology of Atherosclerosis. Annu. Rev. Physiol. 1995, 57, 791–804. [Google Scholar] [CrossRef] [PubMed]
  88. Fan, H.; Tian, H.; Jin, F.; Zhang, X.; Su, S.; Liu, Y.; Wen, Z.; He, X.; Li, X.; Duan, C. CypD Induced ROS Output Promotes Intracranial Aneurysm Formation and Rupture by 8-OHdG/NLRP3/MMP9 Pathway. Redox Biol. 2023, 67, 102887. [Google Scholar] [CrossRef] [PubMed]
  89. Meteva, D.; Vinci, R.; Seppelt, C.; Abdelwahed, Y.S.; Pedicino, D.; Nelles, G.; Skurk, C.; Haghikia, A.; Rauch-Kröhnert, U.; Gerhardt, T.; et al. Toll-like Receptor 2, Hyaluronan, and Neutrophils Play a Key Role in Plaque Erosion: The OPTICO-ACS Study. Eur. Heart J. 2023, 44, 3892–3907. [Google Scholar] [CrossRef]
  90. Tajali, R.; Eidi, A.; Ahmadi Tafti, H.; Pazouki, A.; Sharifi, A.M. Restoring the Angiogenic Capacity of the Human Diabetic Adipose-Derived Mesenchymal Stem Cells Primed with Deferoxamine as a Hypoxia Mimetic Agent: Role of HIF-1α. Adv. Pharm. Bull. 2023, 13, 350–360. [Google Scholar] [CrossRef]
  91. Venkataraman, L.; Sivaraman, B.; Vaidya, P.; Ramamurthi, A. Nanoparticulate Delivery of Agents for Induced Elastogenesis in Three-Dimensional Collagenous Matrices. J. Tissue Eng. Regen. Med. 2016, 10, 1041–1056. [Google Scholar] [CrossRef]
  92. Arnold, F.; Muzzio, N.; Patnaik, S.S.; Finol, E.A.; Romero, G. Pentagalloyl Glucose-Laden Poly(Lactide-Co-Glycolide) Nanoparticles for the Biomechanical Extracellular Matrix Stabilization of an In Vitro Abdominal Aortic Aneurysm Model. ACS Appl. Mater. Interfaces 2021, 13, 25771–25782. [Google Scholar] [CrossRef]
  93. Wang, H.; Keiser, J.A. Vascular Endothelial Growth Factor Upregulates the Expression of Matrix Metalloproteinases in Vascular Smooth Muscle Cells: Role of Flt-1. Circ. Res. 1998, 83, 832–840. [Google Scholar] [CrossRef]
  94. Gong, T.; Hong, Z.-Y.; Chen, C.-H.; Tsai, C.-Y.; Liao, L.-D.; Kong, K.V. Optical Interference-Free Surface-Enhanced Raman Scattering CO-Nanotags for Logical Multiplex Detection of Vascular Disease-Related Biomarkers. ACS Nano 2017, 11, 3365–3375. [Google Scholar] [CrossRef] [PubMed]
  95. Islam, Y.; Khalid, A.; Pluchino, S.; Sivakumaran, M.; Teixidò, M.; Leach, A.; Fatokun, A.A.; Downing, J.; Coxon, C.; Ehtezazi, T. Development of Brain Targeting Peptide Based MMP-9 Inhibiting Nanoparticles for the Treatment of Brain Diseases with Elevated MMP-9 Activity. J. Pharm. Sci. 2020, 109, 3134–3144. [Google Scholar] [CrossRef]
  96. Takata, F.; Dohgu, S.; Matsumoto, J.; Takahashi, H.; Machida, T.; Wakigawa, T.; Harada, E.; Miyaji, H.; Koga, M.; Nishioku, T.; et al. Brain Pericytes among Cells Constituting the Blood-Brain Barrier Are Highly Sensitive to Tumor Necrosis Factor-α, Releasing Matrix Metalloproteinase-9 and Migrating in Vitro. J. Neuroinflamm. 2011, 8, 106. [Google Scholar] [CrossRef] [PubMed]
  97. Polavarapu, R.; Gongora, M.C.; Winkles, J.A.; Yepes, M. Tumor Necrosis Factor-like Weak Inducer of Apoptosis Increases the Permeability of the Neurovascular Unit through Nuclear Factor-Kappa B Pathway Activation. J. Neurosci. 2005, 25, 10094–10100. [Google Scholar] [CrossRef] [PubMed]
  98. Qin, W.; Li, J.; Zhu, R.; Gao, S.; Fan, J.; Xia, M.; Zhao, R.C.; Zhang, J. Melatonin Protects Blood-Brain Barrier Integrity and Permeability by Inhibiting Matrix Metalloproteinase-9 via the NOTCH3/NF-κB Pathway. Aging 2019, 11, 11391–11415. [Google Scholar] [CrossRef]
  99. Machida, T.; Dohgu, S.; Takata, F.; Matsumoto, J.; Kimura, I.; Koga, M.; Nakamoto, K.; Yamauchi, A.; Kataoka, Y. Role of Thrombin-PAR1-PKCθ/δ Axis in Brain Pericytes in Thrombin-Induced MMP-9 Production and Blood-Brain Barrier Dysfunction in Vitro. Neuroscience 2017, 350, 146–157. [Google Scholar] [CrossRef]
  100. Chen, J.-T.; Chen, T.-G.; Chang, Y.-C.; Chen, C.-Y.; Chen, R.-M. Roles of NMDARs in Maintenance of the Mouse Cerebrovascular Endothelial Cell-Constructed Tight Junction Barrier. Toxicology 2016, 339, 40–50. [Google Scholar] [CrossRef] [PubMed]
  101. Chen, H.-S.; Chen, X.; Li, W.-T.; Shen, J.-G. Targeting RNS/Caveolin-1/MMP Signaling Cascades to Protect against Cerebral Ischemia-Reperfusion Injuries: Potential Application for Drug Discovery. Acta Pharmacol. Sin. 2018, 39, 669–682. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, C.; Zhang, D.; Lu, Z.; Man, J.; Zhang, Z.; Fu, X.; Cui, K.; Wang, J. Metformin Protects against Pericyte Apoptosis and Promotes Neurogenesis through Suppressing JNK P38 MAPK Signalling Activation in Ischemia/Reperfusion Injury. Neurosci. Lett. 2022, 783, 136708. [Google Scholar] [CrossRef]
  103. Pan, P.; Zhao, H.; Zhang, X.; Li, Q.; Qu, J.; Zuo, S.; Yang, F.; Liang, G.; Zhang, J.H.; Liu, X.; et al. Cyclophilin a Signaling Induces Pericyte-Associated Blood-Brain Barrier Disruption after Subarachnoid Hemorrhage. J. Neuroinflamm. 2020, 17, 16. [Google Scholar] [CrossRef]
  104. Yang, D.; Baumann, J.M.; Sun, Y.-Y.; Tang, M.; Dunn, R.S.; Akeson, A.L.; Kernie, S.G.; Kallapur, S.; Lindquist, D.M.; Huang, E.J.; et al. Overexpression of Vascular Endothelial Growth Factor in the Germinal Matrix Induces Neurovascular Proteases and Intraventricular Hemorrhage. Sci. Transl. Med. 2013, 5, 193ra90. [Google Scholar] [CrossRef] [PubMed]
  105. Lambert, V.; Munaut, C.; Jost, M.; Noël, A.; Werb, Z.; Foidart, J.-M.; Rakic, J.-M. Matrix Metalloproteinase-9 Contributes to Choroidal Neovascularization. Am. J. Pathol. 2002, 161, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
  106. Navaratna, D.; McGuire, P.G.; Menicucci, G.; Das, A. Proteolytic Degradation of VE-Cadherin Alters the Blood-Retinal Barrier in Diabetes. Diabetes 2007, 56, 2380–2387. [Google Scholar] [CrossRef] [PubMed]
  107. Kowluru, R.A.; Santos, J.M.; Zhong, Q. Sirt1, a Negative Regulator of Matrix Metalloproteinase-9 in Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2014, 55, 5653–5660. [Google Scholar] [CrossRef]
  108. Mishra, M.; Kowluru, R.A. Role of PARP-1 as a Novel Transcriptional Regulator of MMP-9 in Diabetic Retinopathy. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1761–1769. [Google Scholar] [CrossRef]
  109. Kowluru, R.A. Role of Matrix Metalloproteinase-9 in the Development of Diabetic Retinopathy and Its Regulation by H-Ras. Investig. Ophthalmol. Vis. Sci. 2010, 51, 4320–4326. [Google Scholar] [CrossRef]
  110. Lambert, V.; Wielockx, B.; Munaut, C.; Galopin, C.; Jost, M.; Itoh, T.; Werb, Z.; Baker, A.; Libert, C.; Krell, H.-W.; et al. MMP-2 and MMP-9 Synergize in Promoting Choroidal Neovascularization. FASEB J. 2003, 17, 2290–2292. [Google Scholar] [CrossRef] [PubMed]
  111. Kinoshita, S.; Noda, K.; Tagawa, Y.; Inafuku, S.; Dong, Y.; Fukuhara, J.; Dong, Z.; Ando, R.; Kanda, A.; Ishida, S. Genistein Attenuates Choroidal Neovascularization. J. Nutr. Biochem. 2014, 25, 1177–1182. [Google Scholar] [CrossRef] [PubMed]
  112. Ko, H.-M.; Kang, J.-H.; Choi, J.-H.; Park, S.J.; Bai, S.; Im, S.-Y. Platelet-Activating Factor Induces Matrix Metalloproteinase-9 Expression through Ca(2+)- or PI3K-Dependent Signaling Pathway in a Human Vascular Endothelial Cell Line. FEBS Lett. 2005, 579, 6451–6458. [Google Scholar]
  113. Mishra, M.; Flaga, J.; Kowluru, R.A. Molecular Mechanism of Transcriptional Regulation of Matrix Metalloproteinase-9 in Diabetic Retinopathy. J. Cell Physiol. 2016, 231, 1709–1718. [Google Scholar] [CrossRef]
  114. Hsieh, M.-Y.; Chen, W.-Y.; Jiang, M.-J.; Cheng, B.-C.; Huang, T.-Y.; Chang, M.-S. Interleukin-20 Promotes Angiogenesis in a Direct and Indirect Manner. Genes. Immun. 2006, 7, 234–242. [Google Scholar] [CrossRef] [PubMed]
  115. De Lucca Camargo, L.; Babelova, A.; Mieth, A.; Weigert, A.; Mooz, J.; Rajalingam, K.; Heide, H.; Wittig, I.; Lopes, L.R.; Brandes, R.P. Endo-PDI Is Required for TNFα-Induced Angiogenesis. Free Radic. Biol. Med. 2013, 65, 1398–1407. [Google Scholar] [CrossRef]
  116. Miyoshi, A.; Koyama, S.; Sasagawa-Monden, M.; Kadoya, M.; Konishi, K.; Shoji, T.; Inaba, M.; Yamamoto, Y.; Koyama, H. JNK and ATF4 as Two Important Platforms for Tumor Necrosis Factor-α-Stimulated Shedding of Receptor for Advanced Glycation End Products. FASEB J. 2019, 33, 3575–3589. [Google Scholar]
  117. Yang, R.; Zhang, Y.; Huang, D.; Luo, X.; Zhang, L.; Zhu, X.; Zhang, X.; Liu, Z.; Han, J.-Y.; Xiong, J.-W. Miconazole Protects Blood Vessels from MMP9-Dependent Rupture and Hemorrhage. Dis. Model. Mech. 2017, 10, 337–348. [Google Scholar] [PubMed]
  118. Freundt, G.V.; von Samson-Himmelstjerna, F.A.; Nitz, J.-T.; Luedde, M.; Waltenberger, J.; Wieland, T.; Frey, N.; Preusch, M.; Hippe, H.-J. The Orphan Receptor GPRC5B Activates Pro-Inflammatory Signaling in the Vascular Wall via Fyn and NFκB. Biochem. Biophys. Res. Commun. 2022, 592, 60–66. [Google Scholar]
  119. Bou Khzam, L.; Boulahya, R.; Abou-Saleh, H.; Hachem, A.; Zaid, Y.; Merhi, Y. Soluble CD40 Ligand Stimulates the Pro-Angiogenic Function of Peripheral Blood Angiogenic Outgrowth Cells via Increased Release of Matrix Metalloproteinase-9. PLoS ONE 2013, 8, e84289. [Google Scholar] [CrossRef]
  120. Jin, Y.-J.; Park, I.; Hong, I.-K.; Byun, H.-J.; Choi, J.; Kim, Y.-M.; Lee, H. Fibronectin and Vitronectin Induce AP-1-Mediated Matrix Metalloproteinase-9 Expression through Integrin α(5)β(1)/α(v)β(3)-Dependent Akt, ERK and JNK Signaling Pathways in Human Umbilical Vein Endothelial Cells. Cell Signal 2011, 23, 125–134. [Google Scholar] [CrossRef]
  121. Bhowmik, T.; Gomes, A. Down-Regulation of Cyclin-Dependent Kinase-4 and MAPK through Estrogen Receptor Mediated Cell Cycle Arrest in Human Breast Cancer Induced by Gold Nanoparticle Tagged Toxin Protein NKCT1. Chem. Biol. Interact. 2017, 268, 119–128. [Google Scholar] [CrossRef] [PubMed]
  122. Park, M.-J.; Park, I.-C.; Lee, H.-C.; Woo, S.-H.; Lee, J.-Y.; Hong, Y.-J.; Rhee, C.-H.; Lee, Y.-S.; Lee, S.-H.; Shim, B.-S.; et al. Protein Kinase C-Alpha Activation by Phorbol Ester Induces Secretion of Gelatinase B/MMP-9 through ERK 1/2 Pathway in Capillary Endothelial Cells. Int. J. Oncol. 2003, 22, 137–143. [Google Scholar]
  123. Renault, M.-A.; Roncalli, J.; Tongers, J.; Thorne, T.; Klyachko, E.; Misener, S.; Volpert, O.V.; Mehta, S.; Burg, A.; Luedemann, C.; et al. Sonic Hedgehog Induces Angiogenesis via Rho Kinase-Dependent Signaling in Endothelial Cells. J. Mol. Cell Cardiol. 2010, 49, 490–498. [Google Scholar] [CrossRef] [PubMed]
  124. Chatterjee, S.; Patra, D.; Ghosh, P.; Banerjee, S.; Chowdhury, K.D.; Chakraborty, P.; Basu, A.; Sadhukhan, G.C. Activity of ROCKII Not ROCKI Promotes Pulmonary Metastasis of Melanoma Cells via Modulating Smad2/3-MMP9 and FAK-Src-VEGF Signalling. Cell Signal 2022, 97, 110389. [Google Scholar] [CrossRef] [PubMed]
  125. Duraisamy, A.J.; Mishra, M.; Kowluru, R.A. Crosstalk Between Histone and DNA Methylation in Regulation of Retinal Matrix Metalloproteinase-9 in Diabetes. Investig. Ophthalmol. Vis. Sci. 2017, 58, 6440–6448. [Google Scholar] [CrossRef] [PubMed]
  126. Delgado-Olguín, P.; Dang, L.T.; He, D.; Thomas, S.; Chi, L.; Sukonnik, T.; Khyzha, N.; Dobenecker, M.-W.; Fish, J.E.; Bruneau, B.G. Ezh2-Mediated Repression of a Transcriptional Pathway Upstream of Mmp9 Maintains Integrity of the Developing Vasculature. Development 2014, 141, 4610–4617. [Google Scholar] [CrossRef]
  127. Zhou, X.; Yan, T.; Huang, C.; Xu, Z.; Wang, L.; Jiang, E.; Wang, H.; Chen, Y.; Liu, K.; Shao, Z.; et al. Melanoma Cell-Secreted Exosomal miR-155-5p Induce Proangiogenic Switch of Cancer-Associated Fibroblasts via SOCS1/JAK2/STAT3 Signaling Pathway. J. Exp. Clin. Cancer Res. 2018, 37, 242. [Google Scholar] [CrossRef] [PubMed]
  128. Wang, X.; Lee, S.-R.; Arai, K.; Lee, S.-R.; Tsuji, K.; Rebeck, G.W.; Lo, E.H. Lipoprotein Receptor-Mediated Induction of Matrix Metalloproteinase by Tissue Plasminogen Activator. Nat. Med. 2003, 9, 1313–1317. [Google Scholar] [CrossRef] [PubMed]
  129. Guo, B.; Wu, S.; Zhu, X.; Zhang, L.; Deng, J.; Li, F.; Wang, Y.; Zhang, S.; Wu, R.; Lu, J.; et al. Micropeptide CIP2A-BP Encoded by LINC00665 Inhibits Triple-Negative Breast Cancer Progression. EMBO J. 2020, 39, e102190. [Google Scholar] [CrossRef]
  130. Mandel, A.; Larsson, P.; Sarwar, M.; Semenas, J.; Syed Khaja, A.S.; Persson, J.L. The Interplay between AR, EGF Receptor and MMP-9 Signaling Pathways in Invasive Prostate Cancer. Mol. Med. 2018, 24, 34. [Google Scholar] [CrossRef]
  131. Wu, Q.; Zhou, X.; Li, P.; Ding, M.; You, S.; Xu, Z.; Ye, J.; Chen, X.; Tan, M.; Wang, J.; et al. ROC1 Promotes the Malignant Progression of Bladder Cancer by Regulating P-IκBα/NF-κB Signaling. J. Exp. Clin. Cancer Res. 2021, 40, 158. [Google Scholar] [CrossRef]
  132. Wang, C.; Zhang, R.; Wang, X.; Zheng, Y.; Jia, H.; Li, H.; Wang, J.; Wang, N.; Xiang, F.; Li, Y. Silencing of KIF3B Suppresses Breast Cancer Progression by Regulating EMT and Wnt/β-Catenin Signaling. Front. Oncol. 2020, 10, 597464. [Google Scholar] [CrossRef] [PubMed]
  133. Feng, W.; Guan, Z.; Ying, W.-Z.; Xing, D.; Ying, K.E.; Sanders, P.W. Matrix Metalloproteinase-9 Regulates Afferent Arteriolar Remodeling and Function in Hypertension-Induced Kidney Disease. Kidney Int. 2023, 104, 740–753. [Google Scholar] [CrossRef] [PubMed]
  134. Wågsäter, D.; Zhu, C.; Björck, H.M.; Eriksson, P. Effects of PDGF-C and PDGF-D on Monocyte Migration and MMP-2 and MMP-9 Expression. Atherosclerosis 2009, 202, 415–423. [Google Scholar] [CrossRef]
  135. Yamaguchi, T.; Miyamoto, T.; Shikata, E.; Yamaguchi, I.; Shimada, K.; Yagi, K.; Tada, Y.; Korai, M.; Kitazato, K.T.; Kanematsu, Y.; et al. Activation of the NLRP3/IL-1β/MMP-9 Pathway and Intracranial Aneurysm Rupture Associated with the Depletion of ERα and Sirt1 in Oophorectomized Rats. J. Neurosurg. 2023, 138, 191–198. [Google Scholar] [CrossRef] [PubMed]
  136. Fallah, M.; Rakhshan, K.; Nikbakht, F.; Maleki-Ravasan, N.; Tahghighi, A.; Azizi, Y. Cardioprotective Effects of the Aqueous Extract of Echinops Cephalotes on Myocardial Ischemia-Reperfusion in Rats by Modulation of MMP-2, MMP-9, TIMP, and Oxidative Stress. Biomed. Pharmacother. 2024, 176, 116927. [Google Scholar] [CrossRef] [PubMed]
  137. Huang, R.H.; Nayeem, N.; He, Y.; Morales, J.; Graham, D.; Klajn, R.; Contel, M.; O’Brien, S.; Ulijn, R.V. Self-Complementary Zwitterionic Peptides Direct Nanoparticle Assembly and Enable Enzymatic Selection of Endocytic Pathways. Adv. Mater. 2022, 34, e2104962. [Google Scholar] [CrossRef] [PubMed]
  138. Xu, C.; Sun, Y.; Yu, Y.; Hu, M.; Yang, C.; Zhang, Z. A Sequentially Responsive and Structure-Transformable Nanoparticle with a Comprehensively Improved “CAPIR Cascade” for Enhanced Antitumor Effect. Nanoscale 2019, 11, 1177–1194. [Google Scholar] [CrossRef] [PubMed]
  139. Grünwald, B.; Vandooren, J.; Locatelli, E.; Fiten, P.; Opdenakker, G.; Proost, P.; Krüger, A.; Lellouche, J.P.; Israel, L.L.; Shenkman, L.; et al. Matrix Metalloproteinase-9 (MMP-9) as an Activator of Nanosystems for Targeted Drug Delivery in Pancreatic Cancer. J. Control Release 2016, 239, 39–48. [Google Scholar] [CrossRef]
  140. Han, Q.-J.; Lan, X.-T.; Wen, Y.; Zhang, C.-Z.; Cleary, M.; Sayyed, Y.; Huang, G.; Tuo, X.; Yi, L.; Xi, Z.; et al. Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier to Enhance Endocytosis as Efficient Targeted Delivery System for Cancer Diagnosis and Therapy. Adv. Healthc. Mater. 2021, 10, e2002143. [Google Scholar] [CrossRef]
  141. Kulkarni, P.S.; Haldar, M.K.; Nahire, R.R.; Katti, P.; Ambre, A.H.; Muhonen, W.W.; Shabb, J.B.; Padi, S.K.R.; Singh, R.K.; Borowicz, P.P.; et al. Mmp-9 Responsive PEG Cleavable Nanovesicles for Efficient Delivery of Chemotherapeutics to Pancreatic Cancer. Mol. Pharm. 2014, 11, 2390–2399. [Google Scholar] [CrossRef]
  142. Hu, A.; Pu, Y.; Xu, N.; Yang, H.; Hu, X.; Sun, R.; Jin, R.; Nie, Y. Hierarchically Decorated Magnetic Nanoparticles Amplify the Oxidative Stress and Promote the Chemodynamic/Magnetic Hyperthermia/Immune Therapy. Acta Biomater. 2024, 173, 457–469. [Google Scholar] [CrossRef]
  143. Gao, X.; Zhang, J.; Huang, Z.; Zuo, T.; Lu, Q.; Wu, G.; Shen, Q. Reducing Interstitial Fluid Pressure and Inhibiting Pulmonary Metastasis of Breast Cancer by Gelatin Modified Cationic Lipid Nanoparticles. ACS Appl. Mater. Interfaces 2017, 9, 29457–29468. [Google Scholar] [CrossRef] [PubMed]
  144. Ehrsam, D.; Sieber, S.; Oufir, M.; Porta, F.; Hamburger, M.; Huwyler, J.; Meyer Zu Schwabedissen, H.E. Design, Synthesis, and Characterization of a Paclitaxel Formulation Activated by Extracellular MMP9. Bioconjug Chem. 2020, 31, 781–793. [Google Scholar] [CrossRef] [PubMed]
  145. Jiang, J.; Shen, N.; Ci, T.; Tang, Z.; Gu, Z.; Li, G.; Chen, X. Combretastatin A4 Nanodrug-Induced MMP9 Amplification Boosts Tumor-Selective Release of Doxorubicin Prodrug. Adv. Mater. 2019, 31, e1904278. [Google Scholar] [CrossRef]
  146. Yu, H.; Chen, J.; Liu, S.; Lu, Q.; He, J.; Zhou, Z.; Hu, Y. Enzyme Sensitive, Surface Engineered Nanoparticles for Enhanced Delivery of Camptothecin. J. Control Release 2015, 216, 111–120. [Google Scholar] [CrossRef] [PubMed]
  147. Porta, F.; Ehrsam, D.; Lengerke, C.; Meyer Zu Schwabedissen, H.E. Synthesis and Characterization of PDMS-PMOXA-Based Polymersomes Sensitive to MMP-9 for Application in Breast Cancer. Mol. Pharm. 2018, 15, 4884–4897. [Google Scholar] [CrossRef] [PubMed]
  148. van Rijt, S.H.; Bölükbas, D.A.; Argyo, C.; Datz, S.; Lindner, M.; Eickelberg, O.; Königshoff, M.; Bein, T.; Meiners, S. Protease-Mediated Release of Chemotherapeutics from Mesoporous Silica Nanoparticles to Ex Vivo Human and Mouse Lung Tumors. ACS Nano 2015, 9, 2377–2389. [Google Scholar] [CrossRef] [PubMed]
  149. Nossier, A.I.; Mohammed, O.S.; Fakhr El-Deen, R.R.; Zaghloul, A.S.; Eissa, S. Gelatin-Modified Gold Nanoparticles for Direct Detection of Urinary Total Gelatinase Activity: Diagnostic Value in Bladder Cancer. Talanta 2016, 161, 511–519. [Google Scholar] [CrossRef]
  150. Zhang, W.; Gong, C.; Chen, Z.; Li, M.; Li, Y.; Gao, J. Tumor Microenvironment-Activated Cancer Cell Membrane-Liposome Hybrid Nanoparticle-Mediated Synergistic Metabolic Therapy and Chemotherapy for Non-Small Cell Lung Cancer. J. Nanobiotechnol. 2021, 19, 339. [Google Scholar] [CrossRef] [PubMed]
  151. Deng, B.; Ma, B.; Ma, Y.; Cao, P.; Leng, X.; Huang, P.; Zhao, Y.; Ji, T.; Lu, X.; Liu, L. Doxorubicin and CpG Loaded Liposomal Spherical Nucleic Acid for Enhanced Cancer Treatment. J. Nanobiotechnol. 2022, 20, 140. [Google Scholar] [CrossRef]
  152. Battistella, C.; Callmann, C.E.; Thompson, M.P.; Yao, S.; Yeldandi, A.V.; Hayashi, T.; Carson, D.A.; Gianneschi, N.C. Delivery of Immunotherapeutic Nanoparticles to Tumors via Enzyme-Directed Assembly. Adv. Healthc. Mater. 2019, 8, e1901105. [Google Scholar] [CrossRef]
  153. Li, J.; Zhang, J.; Wang, Y.; Liang, X.; Wusiman, Z.; Yin, Y.; Shen, Q. Synergistic Inhibition of Migration and Invasion of Breast Cancer Cells by Dual Docetaxel/Quercetin-Loaded Nanoparticles via Akt/MMP-9 Pathway. Int. J. Pharm. 2017, 523, 300–309. [Google Scholar] [CrossRef]
  154. Xu, H.; Hou, Z.; Zhang, H.; Kong, H.; Li, X.; Wang, H.; Xie, W. An Efficient Trojan Delivery of Tetrandrine by Poly(N-Vinylpyrrolidone)-Block-Poly(ε-Caprolactone) (PVP-b-PCL) Nanoparticles Shows Enhanced Apoptotic Induction of Lung Cancer Cells and Inhibition of Its Migration and Invasion. Int. J. Nanomed. 2014, 9, 231–242. [Google Scholar]
  155. Sánchez-Rodríguez, C.; Palao-Suay, R.; Rodrigáñez, L.; Aguilar, M.R.; Martín-Saldaña, S.; San Román, J.; Sanz-Fernández, R. α-Tocopheryl Succinate-Based Polymeric Nanoparticles for the Treatment of Head and Neck Squamous Cell Carcinoma. Biomolecules 2018, 8, 97. [Google Scholar] [CrossRef] [PubMed]
  156. Khalil, H.H.; Osman, H.A.; Teleb, M.; Darwish, A.I.; Abu-Serie, M.M.; Khattab, S.N.; Haiba, N.S. Engineered S-Triazine-Based Dendrimer-Honokiol Conjugates as Targeted MMP-2/9 Inhibitors for Halting Hepatocellular Carcinoma. ChemMedChem 2021, 16, 3701–3719. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, X.; Wang, X.; Zhong, W.; Ren, X.; Sha, X.; Fang, X. Matrix Metalloproteinases-2/9-Sensitive Peptide-Conjugated Polymer Micelles for Site-Specific Release of Drugs and Enhancing Tumor Accumulation: Preparation and in Vitro and in Vivo Evaluation. Int. J. Nanomed. 2016, 11, 1643–1661. [Google Scholar]
  158. Stapf, M.; Teichgräber, U.; Hilger, I. Methotrexate-Coupled Nanoparticles and Magnetic Nanochemothermia for the Relapse-Free Treatment of T24 Bladder Tumors. Int. J. Nanomed. 2017, 12, 2793–2811. [Google Scholar] [CrossRef]
  159. Deng, G.; Zhou, F.; Wu, Z.; Zhang, F.; Niu, K.; Kang, Y.; Liu, X.; Wang, Q.; Wang, Y.; Wang, Q. Inhibition of Cancer Cell Migration with CuS@ mSiO2-PEG Nanoparticles by Repressing MMP-2/MMP-9 Expression. Int. J. Nanomed. 2018, 13, 103–116. [Google Scholar] [CrossRef] [PubMed]
  160. Chen, Y.-J.; Lee, Y.-C.; Huang, C.-H.; Chang, L.-S. Gallic Acid-Capped Gold Nanoparticles Inhibit EGF-Induced MMP-9 Expression through Suppression of P300 Stabilization and NFκB/c-Jun Activation in Breast Cancer MDA-MB-231 Cells. Toxicol. Appl. Pharmacol. 2016, 310, 98–107. [Google Scholar] [CrossRef] [PubMed]
  161. Zhao, H.; Li, L.; Zhang, J.; Zheng, C.; Ding, K.; Xiao, H.; Wang, L.; Zhang, Z. C-C Chemokine Ligand 2 (CCL2) Recruits Macrophage-Membrane-Camouflaged Hollow Bismuth Selenide Nanoparticles to Facilitate Photothermal Sensitivity and Inhibit Lung Metastasis of Breast Cancer. ACS Appl. Mater. Interfaces 2018, 10, 31124–31135. [Google Scholar] [CrossRef] [PubMed]
  162. Hao, Y.; Hu, J.; Wang, H.; Wang, C. Gold Nanoparticles Regulate the Antitumor Secretome and Have Potent Cytotoxic Effects against Prostate Cancer Cells. J. Appl. Toxicol. 2021, 41, 1286–1303. [Google Scholar] [CrossRef]
  163. Kang, S.; Zhou, G.; Yang, P.; Liu, Y.; Sun, B.; Huynh, T.; Meng, H.; Zhao, L.; Xing, G.; Chen, C.; et al. Molecular Mechanism of Pancreatic Tumor Metastasis Inhibition by Gd@C82(OH)22 and Its Implication for de Novo Design of Nanomedicine. Proc. Natl. Acad. Sci. USA 2012, 109, 15431–15436. [Google Scholar] [CrossRef]
  164. Chen, S.H.; Kang, S.-G.; Luo, J.; Zhou, R. Charging Nanoparticles: Increased Binding of Gd@C82(OH)22 Derivatives to Human MMP-9. Nanoscale 2018, 10, 5667–5677. [Google Scholar] [CrossRef] [PubMed]
  165. Han, H.; Valdepérez, D.; Jin, Q.; Yang, B.; Li, Z.; Wu, Y.; Pelaz, B.; Parak, W.J.; Ji, J. Dual Enzymatic Reaction-Assisted Gemcitabine Delivery Systems for Programmed Pancreatic Cancer Therapy. ACS Nano 2017, 11, 1281–1291. [Google Scholar] [CrossRef]
  166. Chen, D.; Li, B.; Lei, T.; Na, D.; Nie, M.; Yang, Y.; Xie, C.; He, Z.; Wang, J. Selective Mediation of Ovarian Cancer SKOV3 Cells Death by Pristine Carbon Quantum Dots/Cu2O Composite through Targeting Matrix Metalloproteinases, Angiogenic Cytokines and Cytoskeleton. J. Nanobiotechnol. 2021, 19, 68. [Google Scholar] [CrossRef] [PubMed]
  167. Akers, W.J.; Xu, B.; Lee, H.; Sudlow, G.P.; Fields, G.B.; Achilefu, S.; Edwards, W.B. Detection of MMP-2 and MMP-9 Activity in Vivo with a Triple-Helical Peptide Optical Probe. Bioconjugate Chem. 2012, 23, 656–663. [Google Scholar] [CrossRef]
  168. Black, K.C.L.; Akers, W.J.; Sudlow, G.; Xu, B.; Laforest, R.; Achilefu, S. Dual-Radiolabeled Nanoparticle SPECT Probes for Bioimaging. Nanoscale 2015, 7, 440–444. [Google Scholar] [CrossRef] [PubMed]
  169. Kim, J.; Yu, A.M.; Kubelick, K.P.; Emelianov, S.Y. Gold Nanoparticles Conjugated with DNA Aptamer for Photoacoustic Detection of Human Matrix Metalloproteinase-9. Photoacoustics 2022, 25, 100307. [Google Scholar] [CrossRef] [PubMed]
  170. Haffner, M.C.; Zwart, W.; Roudier, M.P.; True, L.D.; Nelson, W.G.; Epstein, J.I.; De Marzo, A.M.; Nelson, P.S.; Yegnasubramanian, S. Genomic and Phenotypic Heterogeneity in Prostate Cancer. Nat. Rev. Urol. 2021, 18, 79–92. [Google Scholar] [CrossRef] [PubMed]
  171. Feng, Y.; Wang, S.; Xie, J.; Ding, B.; Wang, M.; Zhang, P.; Mi, P.; Wang, C.; Liu, R.; Zhang, T.; et al. Spatial Transcriptomics Reveals Heterogeneity of Macrophages in the Tumor Microenvironment of Granulomatous Slack Skin. J. Pathol. 2023, 261, 105–119. [Google Scholar] [CrossRef] [PubMed]
  172. Forte, A.; Rinaldi, B.; Berrino, L.; Rossi, F.; Galderisi, U.; Cipollaro, M. Novel Potential Targets for Prevention of Arterial Restenosis: Insights from the Pre-Clinical Research. Clin. Sci. 2014, 127, 615–634. [Google Scholar] [CrossRef]
  173. Francis, D.J.; Parish, C.R.; McGarry, M.; Santiago, F.S.; Lowe, H.C.; Brown, K.J.; Bingley, J.A.; Hayward, I.P.; Cowden, W.B.; Campbell, J.H.; et al. Blockade of Vascular Smooth Muscle Cell Proliferation and Intimal Thickening after Balloon Injury by the Sulfated Oligosaccharide PI-88: Phosphomannopentaose Sulfate Directly Binds FGF-2, Blocks Cellular Signaling, and Inhibits Proliferation. Circ. Res. 2003, 92, e70–e77. [Google Scholar] [CrossRef]
  174. Meneghini, B.C.; Tavares, E.R.; Guido, M.C.; Tavoni, T.M.; Stefani, H.A.; Kalil-Filho, R.; Maranhão, R.C. Lipid Core Nanoparticles as Vehicle for Docetaxel Reduces Atherosclerotic Lesion, Inflammation, Cell Death and Proliferation in an Atherosclerosis Rabbit Model. Vascul Pharmacol. 2019, 115, 46–54. [Google Scholar] [CrossRef]
  175. Wang, Y.; Zhao, D.; Wei, X.; Ma, L.; Sheng, J.; Lu, P. PEGylated Polyethylenimine Derivative-Mediated Local Delivery of the shSmad3 Inhibits Intimal Thickening after Vascular Injury. BioMed Res. Int. 2019, 2019, 8483765. [Google Scholar] [CrossRef]
  176. Correction to: Endothelial Foxp1 Regulates Neointimal Hyperplasia Via Matrix Metalloproteinase-9/Cyclin Dependent Kinase Inhibitor 1B Signal Pathway. J. Am. Heart Assoc. 2023, 12, e020899. [CrossRef]
  177. Ramirez-Carracedo, R.; Tesoro, L.; Hernandez, I.; Diez-Mata, J.; Filice, M.; Toro, R.; Rodriguez-Piñero, M.; Zamorano, J.L.; Saura, M.; Zaragoza, C. Non-Invasive Detection of Extracellular Matrix Metalloproteinase Inducer EMMPRIN, a New Therapeutic Target against Atherosclerosis, Inhibited by Endothelial Nitric Oxide. Int. J. Mol. Sci. 2018, 19, 3248. [Google Scholar] [CrossRef] [PubMed]
  178. Nguyen, M.M.; Carlini, A.S.; Chien, M.-P.; Sonnenberg, S.; Luo, C.; Braden, R.L.; Osborn, K.G.; Li, Y.; Gianneschi, N.C.; Christman, K.L. Enzyme-Responsive Nanoparticles for Targeted Accumulation and Prolonged Retention in Heart Tissue after Myocardial Infarction. Adv. Mater. 2015, 27, 5547–5552. [Google Scholar] [CrossRef]
  179. Sarami Foroshani, M.; Sobhani, Z.S.; Mohammadi, M.T.; Aryafar, M. Fullerenol Nanoparticles Decrease Blood-Brain Barrier Interruption and Brain Edema during Cerebral Ischemia-Reperfusion Injury Probably by Reduction of Interleukin-6 and Matrix Metalloproteinase-9 Transcription. J. Stroke Cerebrovasc. Dis. 2018, 27, 3053–3065. [Google Scholar] [CrossRef] [PubMed]
  180. Cai, H.; Huang, L.-Y.; Hong, R.; Song, J.-X.; Guo, X.-J.; Zhou, W.; Hu, Z.-L.; Wang, W.; Wang, Y.-L.; Shen, J.-G.; et al. Momordica Charantia Exosome-Like Nanoparticles Exert Neuroprotective Effects Against Ischemic Brain Injury via Inhibiting Matrix Metalloproteinase 9 and Activating the AKT/GSK3β Signaling Pathway. Front. Pharmacol. 2022, 13, 908830. [Google Scholar] [CrossRef]
  181. Miyagawa, T.; Chen, Z.-Y.; Chang, C.-Y.; Chen, K.-H.; Wang, Y.-K.; Liu, G.-S.; Tseng, C.-L. Topical Application of Hyaluronic Acid-RGD Peptide-Coated Gelatin/Epigallocatechin-3 Gallate (EGCG) Nanoparticles Inhibits Corneal Neovascularization Via Inhibition of VEGF Production. Pharmaceutics 2020, 12, 404. [Google Scholar] [CrossRef] [PubMed]
  182. Zeng, L.; Ma, W.; Shi, L.; Chen, X.; Wu, R.; Zhang, Y.; Chen, H.; Chen, H. Poly(Lactic-Co-Glycolic Acid) Nanoparticle-Mediated Interleukin-12 Delivery for the Treatment of Diabetic Retinopathy. Int. J. Nanomed. 2019, 14, 6357–6369. [Google Scholar]
  183. Huang, K.; Liu, X.; Lv, Z.; Zhang, D.; Zhou, Y.; Lin, Z.; Guo, J. MMP9-Responsive Graphene Oxide Quantum Dot-Based Nano-in-Micro Drug Delivery System for Combinatorial Therapy of Choroidal Neovascularization. Small 2023, 19, e2207335. [Google Scholar] [CrossRef] [PubMed]
  184. Bynoe, M.S.; Viret, C.; Yan, A.; Kim, D.-G. Adenosine Receptor Signaling: A Key to Opening the Blood-Brain Door. Fluids Barriers CNS 2015, 12, 20. [Google Scholar] [CrossRef] [PubMed]
  185. Baghirov, H. Receptor-Mediated Transcytosis of Macromolecules across the Blood-Brain Barrier. Expert. Opin. Drug Deliv. 2023, 20, 1699–1711. [Google Scholar] [CrossRef]
  186. Wang, J.; Li, Z.; Pan, M.; Fiaz, M.; Hao, Y.; Yan, Y.; Sun, L.; Yan, F. Ultrasound-Mediated Blood-Brain Barrier Opening: An Effective Drug Delivery System for Theranostics of Brain Diseases. Adv. Drug Deliv. Rev. 2022, 190, 114539. [Google Scholar] [CrossRef] [PubMed]
  187. Zhang, Y.; Zhang, H.; Zhao, F.; Jiang, Z.; Cui, Y.; Ou, M.; Mei, L.; Wang, Q. Mitochondrial-Targeted and ROS-Responsive Nanocarrier via Nose-to-Brain Pathway for Ischemic Stroke Treatment. Acta Pharm. Sin. B 2023, 13, 5107–5120. [Google Scholar] [CrossRef] [PubMed]
  188. Zhu, Z.; Zhai, Y.; Hao, Y.; Wang, Q.; Han, F.; Zheng, W.; Hong, J.; Cui, L.; Jin, W.; Ma, S.; et al. Specific Anti-Glioma Targeted-Delivery Strategy of Engineered Small Extracellular Vesicles Dual-Functionalised by Angiopep-2 and TAT Peptides. J. Extracell. Vesicles 2022, 11, e12255. [Google Scholar]
  189. Moghimi, S.M.; Haroon, H.B.; Yaghmur, A.; Hunter, A.C.; Papini, E.; Farhangrazi, Z.S.; Simberg, D.; Trohopoulos, P.N. Perspectives on Complement and Phagocytic Cell Responses to Nanoparticles: From Fundamentals to Adverse Reactions. J. Control Release 2023, 356, 115–129. [Google Scholar]
  190. Guo, C.; Yuan, H.; Wang, Y.; Feng, Y.; Zhang, Y.; Yin, T.; He, H.; Gou, J.; Tang, X. The Interplay between PEGylated Nanoparticles and Blood Immune System. Adv. Drug Deliv. Rev. 2023, 200, 115044. [Google Scholar] [CrossRef]
  191. Pandey, R.K.; Prajapati, V.K. Molecular and Immunological Toxic Effects of Nanoparticles. Int. J. Biol. Macromol. 2018, 107, 1278–1293. [Google Scholar]
  192. Lang, T.; Liu, Y.; Zheng, Z.; Ran, W.; Zhai, Y.; Yin, Q.; Zhang, P.; Li, Y. Cocktail Strategy Based on Spatio-Temporally Controlled Nano Device Improves Therapy of Breast Cancer. Adv. Mater. 2019, 31, e1806202. [Google Scholar] [CrossRef]
  193. Medici, S.; Peana, M.; Pelucelli, A.; Zoroddu, M.A. An Updated Overview on Metal Nanoparticles Toxicity. Semin. Cancer Biol. 2021, 76, 17–26. [Google Scholar] [PubMed]
  194. Ren, Q.; Ma, J.; Li, X.; Meng, Q.; Wu, S.; Xie, Y.; Qi, Y.; Liu, S.; Chen, R. Intestinal Toxicity of Metal Nanoparticles: Silver Nanoparticles Disorder the Intestinal Immune Microenvironment. ACS Appl. Mater. Interfaces 2023, 15, 27774–27788. [Google Scholar] [CrossRef] [PubMed]
  195. Ibrahim, M.; Ramadan, E.; Elsadek, N.E.; Emam, S.E.; Shimizu, T.; Ando, H.; Ishima, Y.; Elgarhy, O.H.; Sarhan, H.A.; Hussein, A.K.; et al. Polyethylene Glycol (PEG): The Nature, Immunogenicity, and Role in the Hypersensitivity of PEGylated Products. J. Control Release 2022, 351, 215–230. [Google Scholar] [CrossRef] [PubMed]
  196. Kang, M.-J.; Kim, Y.-H.; Chou, M.; Hwang, J.; Cheon, E.-J.; Lee, H.-J.; Chung, S.-H. Evaluation of the Efficacy and Safety of A Novel 0.05% Cyclosporin A Topical Nanoemulsion in Primary Sjögren’s Syndrome Dry Eye. Ocul. Immunol. Inflamm. 2020, 28, 370–378. [Google Scholar] [CrossRef] [PubMed]
  197. Sanapalli, B.K.R.; Yele, V.; Jupudi, S.; Karri, V.V.S.R. Ligand-Based Pharmacophore Modeling and Molecular Dynamic Simulation Approaches to Identify Putative MMP-9 Inhibitors. RSC Adv. 2021, 11, 26820–26831. [Google Scholar] [CrossRef] [PubMed]
  198. Luo, F.; Li, M.; Chen, Y.; Song, S.; Yu, H.; Zhang, P.; Xiao, C.; Lv, G.; Chen, X. Immunosuppressive Enzyme-Responsive Nanoparticles for Enhanced Accumulation in Liver Allograft to Overcome Acute Rejection. Biomaterials 2024, 306, 122476. [Google Scholar] [CrossRef]
  199. Patrick, P.S.; Stuckey, D.J.; Zhu, H.; Kalber, T.L.; Iftikhar, H.; Southern, P.; Bear, J.C.; Lythgoe, M.F.; Hattersley, S.R.; Pankhurst, Q.A. Improved Tumour Delivery of Iron Oxide Nanoparticles for Magnetic Hyperthermia Therapy of Melanoma via Ultrasound Guidance and 111In SPECT Quantification. Nanoscale 2024, 16, 19715–19729. [Google Scholar] [CrossRef]
  200. Liu, P.; Hu, Q. Engineering Cells for Cancer Therapy. Acc. Chem. Res. 2024, 57, 2358–2371. [Google Scholar] [CrossRef]
Figure 1. Nanoparticles recognize and bind to cancer-associated molecules, facilitating enhanced tumor diagnosis. Adapted from Zhang et al. [23].
Figure 1. Nanoparticles recognize and bind to cancer-associated molecules, facilitating enhanced tumor diagnosis. Adapted from Zhang et al. [23].
Pharmaceutics 17 00479 g001
Figure 2. Regulatory network of signaling pathways modulating MMP-9 expression in tumor progression and vascular disease. This illustration describes the complex interplay of signaling pathways that regulate MMP-9 expression and contribute to the development and progression of tumors and vascular diseases.
Figure 2. Regulatory network of signaling pathways modulating MMP-9 expression in tumor progression and vascular disease. This illustration describes the complex interplay of signaling pathways that regulate MMP-9 expression and contribute to the development and progression of tumors and vascular diseases.
Pharmaceutics 17 00479 g002
Table 1. The structural and functional classification of MMP family proteins (adapted from Wang et al. [3]).
Table 1. The structural and functional classification of MMP family proteins (adapted from Wang et al. [3]).
Classification By FunctionTypes of Human MMPsMain SubstrateMain Cell Source
Collagenases1Collagen, GelatinPlatelets, Macrophages, Endothelium, Smooth Muscle Cell (SMCs), Fibroblasts
8Collagen, Gelatin, AggrecanMacrophages, Neutrophils
13Collagen, Gelatin, FibronectinSMCs, Macrophages, Varicose Veins, Breast Cancer
Gelatinases2Platelets, Leukocytes, Endothelium, Vascular Smooth Muscle (VSM), Collagen, GelatinAdventitia, Aortic Aneurysm, Varicose Veins
9Collagen, Gelatin, ElastinMicrovessels, Macrophages, Neutrophils, Endothelium, VSM, Adventitia, Aortic Aneurysm
Stromelysins3Extracellular Matrix (ECM), pro-MMPEndothelium, Intima, VSM, Platelets, Coronary Artery Disease, Synovial Fibroblasts
10ECM, pro-MMPAtherosclerosis, Uterus, Arthritis, Carcinoma Cells
11Insulin-Like Growth Factor Binding Protein, etc.Brain, Uterus, Angiogenesis
Matrilysins26ECM, pro-MMPEndothelium, Intima, VSM, Uterus
Membrane-Anchored14Fibronectin, Laminin, GelatinBreast Cancer, Endometrial Tumors
15Collagen, pro-MMP-2, pro-MMP-13, FibronectinVSM, Fibroblasts, Platelets, Brain, Uterus, Angiogenesis
16pro-MMP-2, FibronectinFibroblasts, Leukocytes
17pro-MMP-2, FibronectinLeukocytes, Angiogenesis
24pro-MMP-2Brain, Breast Cancer
25pro-MMP-2, FibronectinLeukocytes, Lung, Pancreas, Kidney, Brain, Astrocytoma, Glioblastoma
Metalloelastase12Fibronectin, Tenascin-CLeukocytes, Anaplastic Astrocytomas, Glioblastomas
Enamelysin20Elastin, Fibronectin, LamininSMCs, Fibroblasts, Macrophages
Epilysin28Amelogenin, Dentin SialophosphoproteinTooth Enamel
Table 2. Classification of nanoparticles based on material.
Table 2. Classification of nanoparticles based on material.
Classification by MaterialApplications in MedicineReferences
Metal Nanoparticles
Gold NanoparticlesDrug delivery[24]
Cancer treatment[25]
Diagnostics[26]
Silver NanoparticlesAntimicrobial coatings[27]
Wound dressings[28]
Platinum NanoparticlesCancer therapy[29]
Metal Oxide Nanoparticles
Titanium DioxidePhotocatalysis[30]
Zinc OxideAntibacterial agents[31]
Iron OxideMagnetic resonance imaging contrast agents[32]
Drug delivery[33]
Ceramic Nanoparticles
Silica NanoparticlesDrug delivery[34]
Catalysis[35]
Biosensors[36]
Alumina NanoparticlesCoating[37]
Carbon-Based Nanoparticles
Carbon NanotubesElectronics[38]
Conductive materials[39]
Drug delivery[40]
FullerenesAntioxidants[41]
Drug delivery[42]
GrapheneFlexible electronics[43]
Sensors[44]
Polymeric Nanoparticles
Poly(Lactic-Co-Glycolic Acid)
(PLGA) Nanoparticles
Drug delivery[45]
Vaccine delivery[46]
Cancer therapy[47]
Polycaprolactone
(PCL) Nanoparticles
Drug delivery[48]
Tissue engineering[49]
Gene delivery[50]
Polystyrene (PS) NanoparticlesDiagnostics[51]
Research tools[52]
Chitosan NanoparticlesDrug delivery[53]
Gene delivery[54]
Antimicrobial agents[55]
Poly(N-Isopropylacrylamide)
(PNIPAM) Nanoparticles
Drug delivery[56]
Smart materials[57]
Poly(Methyl Methacrylate)
(PMMA) Nanoparticles
Drug delivery[58]
Bone cement and other base material[59]
DendrimersDrug delivery[60]
Gene delivery[61]
Imaging[62]
Polyethylene Glycol
(PEG) Nanoparticles
Drug delivery[63]
Protein delivery[64]
Poly(Alkyl Cyanoacrylate)
(PACA) Nanoparticles
Drug delivery[65]
Hydrogel NanoparticlesDrug delivery[66]
Tissue engineering[67]
Polypeptide-Based NanoparticlesDrug delivery[68]
Theranostics[69]
Table 3. Classification of nanoparticles based on size.
Table 3. Classification of nanoparticles based on size.
Classification by SizeApplications in MedicineReferences
Quantum Dots
(2–10 nm)
Bioimaging[70]
Quantum computing[71]
Photovoltaics[72]
Ultra-Fine Particles
(1–100 nm)
Catalysis[73]
Drug delivery[74]
Imaging[75]
Fine Particles
(100–1000 nm)
Coatings[76]
Table 4. The regulatory effects of signaling pathways on MMP-9 expression.
Table 4. The regulatory effects of signaling pathways on MMP-9 expression.
MoleculesSignaling Pathways and
Mechanisms
Regulatory Effects on MMP-9
Expression
References
NF-κBBinding directly to promoterUpregulation
Platelet-Activating Factor (PAF)Ca2+/PI3K, ERK pathwaysUpregulation[112]
AP-1Binding directly to promoterUpregulation
Poly ADP Ribosyltransferase-1 (PARP-1)Formation of transcription complex in promoterUpregulation[108]
Sirt-1Reducing the binding of AP-1 and PARP-1 to promoterDownregulation[108,113]
IL-20JNK, ERK1/2, P38 MAPK pathwaysUpregulation[114]
TNF-αJNK, ERK1/2, AP-1 pathwaysUpregulation[115,116]
MiconazoleERK pathwayDownregulation[117]
Transient Receptor Potential Vanilloid 4 (TRPV-4)ERK pathwayDownregulation[10]
G-Protein Coupled Receptor 5B (GPRC5B)ERK1/2, NF-κB pathwaysUpregulation[118]
Soluble CD40 LigandsP38 MAPK pathwayUpregulation[119]
Tenascin-C (TNC)AktUpregulation[82]
FibronectinJNK, ERK, PI3K/Akt pathways, AP-1Upregulation[120]
Estrogen ReceptorERK, P38 MAPK, PI3K/Akt pathways, NF-κBDownregulation[121]
Thrombin Receptor (PAR-1)PKCθ/Akt, PKCδ/ERK pathwaysUpregulation[99]
PKC-AERK1/2 pathwaysUpregulation[122]
Sonic Hedgehog (Shh)Rho, ROCKUpregulation[123]
SmadsROCKIIUpregulation[124]
Ezh2Promoting promoter methylation in retinal endothelial cellsUpregulation[125]
Inhibiting activation in mouse embryosDownregulation[126]
miR-155SOCS1/JAK2/STAT3 pathwayUpregulation[127]
Tissue Plasminogen Activator (tPA)Low-density lipoprotein receptor-associated protein (LRP)Upregulation[128]
Table 5. Relationship between MMP-9-related signaling pathways and tumor progression or vascular diseases.
Table 5. Relationship between MMP-9-related signaling pathways and tumor progression or vascular diseases.
MMP-9-Related
Signaling Pathways
Corresponding Biological EffectsReferences
Tumor progression
VEGF/VEGFRStimulation of angiogenesis and tumor growth[83]
TGF-βInitially functions as the tumor suppressor, but later promotes metastasis[129]
EGFREnhances tumor cell proliferation and survival[130]
MAPKEssential for cell proliferation, differentiation, and survival[121]
NF-κBPromotes tumor progression, angiogenesis, and metastasis[131]
PI3K/Akt/mTORCritical for cell growth, survival, and metabolism; frequently dysregulated in cancer[82]
Wnt/β-cateninInfluences cell proliferation, differentiation, and stem cell renewal; dysregulation can drive tumor development[132]
Vascular diseases
TGF-βStimulates ECM production, normally restraining vascular smooth muscle cell proliferation; MMP-9 disrupts balance, leading to vascular remodeling and lesion formation[133]
PDGFPromotes VSMC migration and proliferation, resulting in neointima formation[134]
TNF-αInduces inflammation and ECM restructuring within the vessel wall[118]
InterleukinContributes to the inflammatory response within vascular lesions[135]
Oxidative stressTriggers ECM degradation, contributing to vascular damage[136]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, X.; Xu, Z. Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases. Pharmaceutics 2025, 17, 479. https://doi.org/10.3390/pharmaceutics17040479

AMA Style

Li X, Xu Z. Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases. Pharmaceutics. 2025; 17(4):479. https://doi.org/10.3390/pharmaceutics17040479

Chicago/Turabian Style

Li, Xuying, and Zhuping Xu. 2025. "Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases" Pharmaceutics 17, no. 4: 479. https://doi.org/10.3390/pharmaceutics17040479

APA Style

Li, X., & Xu, Z. (2025). Applications of Matrix Metalloproteinase-9-Related Nanomedicines in Tumors and Vascular Diseases. Pharmaceutics, 17(4), 479. https://doi.org/10.3390/pharmaceutics17040479

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop