DOI:
10.1039/D3QI00978E
(Review Article)
Inorg. Chem. Front., 2023,
10, 4918-4942
Advances in the application of manganese dioxide and its composites for theranostics
Received
26th May 2023
, Accepted 29th June 2023
First published on 6th July 2023
Abstract
Manganese dioxide (MnO2), a type of redox-active transition-metal dioxide, has found wide applications in catalysts, oxidants, ferrites, achromats and battery materials. In addition, MnO2 is also increasingly being used in the biomedical field for disease diagnosis and treatment. Despite its great potential, there are limited articles that comprehensively review the use of MnO2 in disease diagnosis and treatment. This review provides a concise overview of the latest developments in MnO2 nanomaterials, focusing on their applications as magnetic resonance, photoacoustic and fluorescent contrast agents. We first summarize the preparation methods of MnO2 nanomaterials and the mechanisms by which they serve as magnetic resonance contrast agents. We then briefly introduce their applications as photoacoustic and fluorescent contrast agents. Finally, we provide an overview of the therapeutic applications of MnO2 nanomaterials and discuss the challenges and prospects for their future clinical applications.
 Min Wu | Min Wu is currently an associate professor of the Huaxi MR Research Center (HMRRC), Department of Radiology, at the West China Hospital of Sichuan University in China. She received her PhD degree in biochemistry and molecular biology from the Institute of Nanobiomedical Technology and Membrane Biology, West China Hospital of Sichuan University, in China in 2010 under the supervision of Professor Xiaojun Zhao. Her research focuses on the synthesis of contrast agents/drug delivery systems and their application in small animals to reveal cancer mechanisms and for theranostics. |
1. Introduction
Manganese dioxide (MnO2) is one of the most interesting inorganic compounds due to its excellent physicochemical properties (high oxidation, light absorption capabilities).1,2 Recently, it has been prepared in a variety of forms, such as nanodots,3 nanorods/fibers,4 nanosheets,5 mesopores/molecular sieves,6 branched structures7 and aggregates/flowers,8 and widely used in catalysts,9,10 oxidants,11 ferrites,12,13 achromats14 and battery materials.15,16 It is also used in disease diagnosis and treatment. Molecular imaging techniques (MITs) have garnered significant attention since the early 1990s due to their non-invasive ability to visualize molecular-level processes, shedding light on the underlying biological mechanisms of diseases.17 Molecular imaging is a valuable method in disease diagnosis to assess metabolic changes and detect disease progression. With rapid advancements in nanotechnology, MITs including photoacoustic imaging (PA),18 fluorescence (FL) imaging,19 single-photon emission computed tomography,20 optical microscopy,21 positron emission computed tomography22 and magnetic resonance imaging (MRI),23 have attracted significant attention based on nanometer-scale contrast agents. Among them, MRI contrast agents can provide anatomical structures and pathology with a high spatial resolution,24–26 these agents are used in the fields of inflammation detection,27 fibrosis diagnosis,28 thromboembolic disease imaging29 and cancer therapy.30 Furthermore, the role of photoacoustic imaging and fluorescence imaging in disease diagnosis is paramount.
MnO2 has been extensively investigated as a potential magnetic resonance contrast agent, exhibiting promising results for T1, T2 and T1–T2 dual contrast enhancements. Gadolinium (Gd) chelates31 and superparamagnetic iron oxide (SPIO)32 are the most commonly employed clinical contrast agents for magnetic resonance imaging. However, several investigations have demonstrated that Gd-based contrast agents increase patients’ risk of developing nephrogenic systemic fibrosis and have the potential to deposit in various tissues, including the brain, bone and skin, even in patients with normal kidney function.33,34 In comparison with Gd and SPIO contrast agents, Mn-based contrast agents offer distinct advantages: (1) They come in various forms, such as oxides, nanoparticles and chelates. (2) Even a small amount of Mn2+ has a robust ability to alter the signals in MRI, resulting in substantial contrast enhancements. (3) Mn2+ contrast agents exhibit low toxicity, because manganese is a crucial trace element in the human body and biological regulatory systems effectively maintain its homeostasis.35 Manganese is a naturally occurring element that serves as an essential component of cells and mitochondria, playing a vital role in cellular and mitochondrial functions simultaneously. It has been confirmed that minimal quantities of manganese residues pose lower risks to human health, as the body has the function to maintain metal homeostasis and eliminate excess manganese or incorporate it into the body's manganese pool. (4) Mn2+ contrast agents can be used in neuroimaging. Above all, owing to its excellent biocompatibility and remarkable optical–physical properties,36 biogenic Mn(II) has been extensively investigated as a T1 contrast agent.37 However, there have been several recent studies that have started to investigate MnO2 as a contrast agent for both T2 and T1–T2 dual contrast.
Except for MR contrast agents, studies have demonstrated that MnO2 nanomaterials can also serve as contrast agents for FL imaging38 and PA imaging.39 FL imaging is a technique that takes advantage of the luminescence properties of fluorescent molecules to visualize living organisms. Alternatively, PA imaging is a method that uses the photoacoustic signal generated by biological tissues to provide information about their light absorption properties.40 More specifically, PA imaging is a technique that enables the detection of photoacoustic signals by visualizing the distribution of light absorption in tissues. When combined with MRI, these imaging modalities can offer a more precise diagnosis of diseases and aid in subsequent treatment strategies.41
Due to the significant potential of MnO2 for imaging, extensive articles have been published, with numerous innovative ideas being put forward for subsequent clinical translation. This review provides a comprehensive summary of MnO2 nanomaterial synthesis methods, their principles as MRI contrast agents, their applications in diagnosing various diseases and their therapeutic potential. Additionally, the review briefly introduces the use of MnO2 nanomaterials as photoacoustic and fluorescent contrast agents (Fig. 1). Finally, we present our opinions on potential future directions for research in this field. In summary, MnO2 nanomaterials are widely used as contrast agents and therapeutic drugs, offering valuable insights for their incorporation into diagnosis and treatment strategies.
 |
| Fig. 1 A schematic illustration of the imaging principle, preparation methods, imaging applications and treatment strategies for MnO2 nanomaterials (from center to periphery). | |
2. Preparation of MnO2
In recent years, numerous approaches have been developed for the synthesis of MnO2 nanomaterials. These approaches have resulted in the successful production of various forms of MnO2, including nanowires, one-dimensional nanodots, two-dimensional nanosheets, multidimensional nanocubes, flower-like structures and hollow spheres.42–45 In summary, the primary approaches for the synthesis of MnO2 nanomaterials are the biomineralization method, the template method and the redox method.
Biomineralization is a promising technique for combining biomolecules with inorganic substances (Fig. 2a).46–48 Specifically, it produces inorganic minerals by modulating biomolecules.49,50 This process involves the controlled conversion of ions under physicochemical conditions to transform bio-organic materials, such as serum albumin, into solid minerals.51 Then bio-organic materials can function as nucleating agents, co-regulators or mineral ion templates that influence the composition and characteristics of inorganic materials.52 In the preparation of MnO2, bio-organic materials are introduced to guide Mn2+ nucleation, which then spontaneously forms MnO2via oxidation or growth in alkaline solutions. Additionally, MnO2 can be combined with bio-organic materials such as serum albumin (HSA)53 and genetically engineered proteins54 for imaging purposes. Overall, this approach is one of the most effective strategies due to its economical, convenient and environmentally friendly advantages. However, biomineralization processes involve the use of living organisms, such as bacteria or fungi, to precipitate minerals like MnO2. Controlling the reaction conditions to achieve consistent results is challenging due to the complexity of biological systems and these methods may have a lower efficiency and yield compared to conventional chemical synthesis routes.
 |
| Fig. 2 Preparation of MnO2. (a) Synthesis of MnO2 nanomaterials by biomineralization.46 Copyright 2018, American Chemical Society. (b) Synthesis of MnO2 nanoparticles by the template method, including the soft template method and the hard template method.63 Copyright 2020, Royal Society of Chemistry. (c) Synthesis of MnO2 nanomaterials by the redox method. | |
The template method is a versatile technique that can be used to produce nanoparticles with various structures.55 The templates used can be made of any materials with nanostructures.56 The process typically involves three steps: creating templates, preparing MnO2 according to the templates and removing or not removing the template agent as necessary.57 Furthermore, the template method can be categorized into two types based on the type of template used: soft templates and hard templates.58
Soft templates, which include block copolymers, flexible organic compounds and surfactants, function as structure-directing agents.59,60 The soft template method results in micelles with diverse morphologies depending on the concentration of soft templates. The micelle structures enable inorganic materials to exhibit distribution trends driven by van der Waals forces, hydrogen bonds and electrostatic interactions between surfactant molecules and nanomaterials. This method is advantageous due to its simplicity of operation and the ability to produce materials with various forms. However, the soft template method has limited control over the shape, size and homogeneity of the nanoparticles.
In contrast, the majority of hard templates maintained by covalent bonding are polymer61 and metal templates.62 The hard template method is effective at reducing interference since it does not involve surfactants. Furthermore, this method enables precise control over the size and morphology of the nanomaterials, resulting in a high degree of stability. However, the nanomaterials produced by the hard template method have a simple structure with fewer morphological varieties. Consequently, this method is the preferred approach for producing shell-like MnO2 nanomaterials (Fig. 2b).63 Certainly, the template method also has certain limitations. The template method relies on the use of a specific template material to guide the growth of MnO2, which limits the structural diversity. The resulting MnO2 structure tends to replicate the morphology of the template, making it challenging to obtain a wide range of structures using this method.
Additionally, the redox method has become a common approach for synthesizing MnO2 due to its efficiency and convenience.64 The Mn7+ (KMnO4) redox method is particularly popular among redox techniques because of its effectiveness. Potassium permanganate (KMnO4), a powerful oxidizing agent, can react with certain reducing agents to produce MnO2, similar to small organic molecules, polymers and proteins.65,66 Another redox method is the Mn2+ (MnCl2) oxidation method. For instance, Zhang et al.67 employed a reaction between KMnO4 and H2O2 to generate ultra-small MnO2 (sMnO2) nanosheets (Fig. 2c). Overall, the redox method offers benefits such as a rapid process, low cost, mild synthesis conditions, energy efficiency and diverse applications in the biomedical fields.63 Nevertheless, the redox method can result in inhomogeneous formations and the possibility of the formation of impurities or side products of MnO2, which makes it difficult to mass produce.
3. MnO2 for magnetic resonance imaging
3.1 The mechanism of MnO2 as a contrast agent
MRI contrast agents alter the relaxation of tissues to increase the sensitivity and specificity of diagnostic images.68 Recently, numerous studies have shown that manganese contrast agents could serve as T1, T2 and T1–T2 dual contrast agents, and their mechanisms are briefly summarized as follows.
3.1.1 The mechanism of MnO2 as a T1 contrast agent.
Brownian motion of a paramagnetic substance generates a fluctuating magnetic field that accelerates the relaxation of protons in the surrounding tissue. The time required for the magnetization of protons to return to their initial state after excitation is referred to as the T1 relaxation time constant. The changes in T1 relaxation time are due to excess energy released by the excited proton being transferred to surrounding tissues. T1-Based MRI contrast agents could reduce the T1 relaxation time, resulting in higher signal intensities in T1-weighted MR images and thus T1 contrast agents are called “positive contrast agents”.69 Mn(II)70 and Gd(III)71 are the most common MR contrast agents, thus improving proton relaxation and leading to high signals in T1-weighted imaging.70 The T1 relaxation rate depends on the spin quantum number (S) and is inversely proportional to the distance between the metal ion and the water proton.72
Mn(II) is the critical element in MnO2 nanoparticles for magnetic resonance enhancement imaging. Coordinating Mn atoms with six oxygen atoms in an octahedral geometry while keeping them isolated from an aqueous environment can induce changes in the longitudinal or transverse relaxation of protons. MnO2 nanoparticles can be reduced to Mn(II) by intracellular reducing substances, such as glutathione (GSH) and hydrogen peroxide (H2O2). Two properties of Mn(II) determine its ability to act as a MRI contrast agent: (1) with five unpaired electrons in the ground state, it has a considerable degree of paramagnetism (Fig. 3a);70 (2) the inner layer and the outer layer of water molecules undergo rapid dissociative exchange (Fig. 3b and c).73 One crucial point to consider is that one or more water molecules can be located inside the Mn(II) inner coordination sphere in aqueous biological media. The coordination water molecules are unstable in this context since the Mn–O bond weakens in the high-spin Mn(II) ligand field caused by the interaction with two three-dimensional sigma antibonding electrons.74 The findings demonstrate that the inner sphere in Mn undergoes hydrolytic dissociation with relative ease and the exchange of water between the inner sphere and the medium happens very quickly, in the submicrosecond range.75 As a result, MRI shows high spin Mn(II). In light of this, many Mn-based contrast agents have been developed and are particularly effective at enhancing the contrast of T1-weighted images.76–78
 |
| Fig. 3 Manganese properties. (a) Diagram of the ligand field energy levels for the [Mn(H2O)6]2+ electronic structure. (b) The inner coordination sphere contains six water molecules in an octahedral configuration. (c) Aquo complex water exchange rate constants, unchelated Gd3+ (red writing) has a somewhat greater rate of water exchange than Mn2+ (arrow), which is in the submicrosecond range. Water exchange for chelated Mn(II) ranges from 106 to 107 s−1.73 Copyright 2010, Royal Society of Chemistry. | |
3.1.2 The mechanism of MnO2 as a T2 contrast agent.
T
2 relaxation, also known as spin–spin relaxation, refers to the decay of transverse magnetization of tissue after it has been excited by an external radiofrequency.79T2 relaxation originates from interactions between spins within tissues. These interactions can arise from a variety of sources, including dipole–dipole interactions, chemical shift interactions and interactions with other nearby spins.80 These interactions cause the individual spins to experience slightly different magnetic fields, leading to a spread in their precession frequencies. As a result of this frequency spread, the phase coherence among the spins is gradually lost over time. The loss of phase coherence causes the transverse magnetization to decay exponentially with a characteristic time constant called T2. The T2 relaxation time is influenced by various factors, such as molecular motion, molecular interactions and magnetic field inhomogeneities.
The T2 contrast agent reduces the signal by dephasing the surrounding protons’ spin in bulk water, which leads to a decrease in signal intensities.81 The rate constant linked to the T2 relaxation time is defined as 1/T2. MnO2 can be used as a T2 contrast agent in MRI. T2 contrast agents are substances that alter the relaxation times of nearby water protons, leading to contrast enhancement of T2-weighted MR images.82 The manganese ions included in MnO2 nanoparticles have paramagnetism originating from unpaired electrons. Paramagnetic substances generate local magnetic fields, which could dephase the spins of water protons and thus accelerate their T2 relaxation. Furthermore, MnO2 nanoparticles have a porous structure, which could contribute to an additional mechanism to accelerate the T2 relaxation of water protons. Specifically, pores on the nanoparticles could restrict the diffusion of water molecules, which would lead to increased interactions between water protons and the MnO2 nanoparticles and thus further enhance the T2 relaxation.83
K. Deka et al.84 investigated manganese oxide nanoparticles infused in mesoporous three-dimensional carbon frames as MRI contrast agents. Their research revealed a remarkable increase in T2 contrast by controlling the interaction between magnetic ions and water (Fig. 4a). Meanwhile, Zhu et al.85 developed a hydrogel-based drug-carrying system to prevent postoperative tumor recurrence. KMnO4 was used as the oxidizing agent to prepare the CMC-DAMnO2/MnO hydrogel. In the hydrogel, KMnO4 is not only used as an oxidant to trigger the polymerization and gelation of CMC-DA but is also transformed into MnO2 and MnO to afford the T2-weighted MRI agent to realize tumor visualization. In addition, researchers have used the T2 attenuation of MnO2 to design a nanoprobe for directly determining alkaline phosphatase (ALP) in blood.86 In the presence of ALP, 2-phospho-L-ascorbic acid trisodium salt was catalyzed to produce ascorbic acid, which reduced MnO2 to Mn2+, causing T2 decay. This research is another application of MnO2 as a T2 contrast agent.
 |
| Fig. 4 (a) T2-Weighted in vitro MR images of aqueous dispersions of the Mn2O3@CF nanosystem with various Mn concentrations.84 Copyright 2020, Elsevier. (b) PBMn can be used as a PAI/T1/T2 triple mode contrast agent, in image-guided PTT, and in oxygen regulation of breast cancer grafts in vitro.95 Copyright 2017, American Chemical Society. (c) MnO2 is metabolized by the kidneys. | |
3.1.3 The mechanism of MnO2 as a T1–T2 contrast agent.
MnO2 is traditionally used in T1 magnetic resonance enhanced imaging.87 Moreover, researchers have demonstrated that MnO2 can serve as a T1–T2 dual contrast agent for tumor MRI.88 MnO2 has been found to act as a catalyst for H2O2, which produces oxygen (O2) and releases manganese ions (Mn2+), making it a H2O2-responsive T1 contrast agent. Furthermore, generated O2 can rejuvenate the oxygen supply to the tumor, which enhances its hypoxic environment, a crucial factor in inhibiting tumor growth and metastasis. The conversion of deoxyhemoglobin (Hb) to oxygenated hemoglobin (HbO2) occurs due to generated O2.89–92 Hb is superparamagnetic and HbO2 is diamagnetic. The transformation from Hb to HbO2 leads to the reduction of the transverse relaxation rate (r2) contributing to the high signal intensity of the tumor tissues present in T2-weighted images.93,94 Thus, MnO2 proves to be an efficient oxygen producer and serves as a viable dual T1–T2 contrast agent for MRI.
Based on the mechanism, MnO2 has been extensively studied as a T1–T2 dual contrast agent. For instance, Peng et al.95 synthesized hybrid nanoparticles of Prussian blue (PB) and MnO2 (PBMn) for imaging purposes using T1–T2-PAI. The presence of MnO2 in PBMn led to a reduction in both the r2 of PB and the signal intensity of artifacts inherent in T2-weighted MR images. Additionally, PB in PBMn improved the H2O2 oxygen generation effect and oxygenated deoxygenated Hb to HbO2. HbO2 is diamagnetic and enhances the T2-weighted signal intensity. The experimental results established that PBMn nanomaterials could serve as T2-weighted magnetic contrast agents. Furthermore, the MnO2 nanomaterials served as traditional T1 MR and fluorescence imaging contrast agents, allowing them to be used as three-modality contrast agents for clinical disease diagnosis (Fig. 4b).
Here, more researchers are exploring the use of MnO2 as a T1–T2 dual contrast agent. The discovery of a novel bidirectional magnetic resonance tuned nanoprobe that features bidirectional magnetic resonance tuned with dual-activated T1 and T2 magnetic resonance signals coupled with dual contrast enhances subtraction imaging.96 When injected into patient-derived xenograft models, the nanoprobe demonstrated a high sensitivity in detecting microscopic intracranial brain tumors, while simultaneously displaying a significant increase in contrast enhancements with minimal background signals. Moreover, it can also be employed for the quantitative imaging of molecular targets in tumors. In another example, Zhao et al.97 proposed the construction of yolk–shell nanohybrids (Fe3O4@C/MnO2-PGEA) through a simple process. Making use of the unique yolk–shell structure, the nanohybrids offer accurate T1–T2 dual-mode magnetic resonance imaging by adequately exposing both the Fe3O4 core and MnO2 shell.
Most importantly, Mn(II) released from nanoparticles of MnO2 is easily metabolized by the kidneys. The biological mechanism lies in the semipermeable properties of the glomerular capillary wall, which consists of endothelial cells, the basement membrane and epithelial cells of the renal proximal tubules. The pore size of endothelial cells is approximately 100 nm, and that of the basement membrane is about 3 nm. The filter hole of the podocyte is around 32 nm. All solutions and molecules with a molecular weight lower than that of serum protein (68 kDa) and a hydrodynamic diameter smaller than 5–7 nm can cross the barrier. However, some small nanoparticles are retained and accumulate at the basement membrane to achieve MRI.98 In contrast, MnO2 nanoparticles are generally less than 10 nm in diameter.99 Therefore, their basic properties determine their capabilities in T1, T2 and T1–T2 dual-modal magnetic resonance imaging. They can be cleared by the kidneys quickly and have a fast circulation time in the body, reducing the risk of toxic side effects and ensuring biological safety (Fig. 4c). In conclusion, MnO2 nanoparticles have good biocompatibility and can serve as a strong foundation for subsequent applications as contrast agents.
3.2 The applications of MnO2 nanomaterials in disease diagnosis
According to the imaging mechanism described above, MnO2 nanoparticles play a key role in MRI disease diagnosis. Different disease states produce significantly different intra-cellular environments, including oxidative mediators with high concentrations of H2O2 in mitochondria and reducing mediators with increased glutathione or cysteine concentrations in the endosomes and cytoplasm.70 This results in differences in various cytoplasmic metabolites and biomolecules. Based on these differences in disease states, manganese dioxide nanoparticles which are highly sensitive to weakly acidic and reducing mediators, can decompose and release Mn(II).75 As a result, the MRI signal of Mn(II) in the diseased state differs from the signal in the surrounding normal state, enabling disease diagnosis.
3.2.1 Antimicrobial detection and inflammation imaging.
Enzymatic processes play crucial roles in inflammatory responses. The reactants and biomarker molecules involved in these processes have significant potential for imaging. Inflammation can cause an upregulation of specific enzymes that could potentially serve as imaging targets. However, the concentrations of most enzymes are too low to be directly detected by MR probes.70 Nevertheless, Mn-based MR probes have been designed to detect pathogens by producing detectable changes in signal.100 These probes can chemically react with enzymes and be retained at active enzyme sites in the tissue, allowing for high-precision imaging of inflammatory sites.
Bacterial biofilms present a microenvironment of bacterial infections with characteristics such as weak acidity, hypoxia and overproduction that are distinct from those of healthy tissues.101–103 MnO2 nanoparticles can decompose in an acidic environment to form water-soluble Mn(II), making them useful for detecting bacterial microenvironments. Xiu et al.104 designed a MnO2-based nanoparticle platform that responded to microenvironments for MRI of biofilms. After intravenous or local injection, the T1-weighted magnetic resonance signals of the biofilm-infected tissues were remarkably enhanced compared with those of healthy tissues.105 This finding was further supported by quantitative analysis, demonstrating the excellent bacteria detection abilities of MnO2 nanoparticles.
MnO2 can be used to detect specific enzymes to enable targeted imaging of changes in the bacterial microenvironment. Myeloperoxidase (MPO) is an extracellular marker of inflammation that is a heme-containing enzyme produced by immune cells, such as neutrophils and macrophages. MPO is widely expressed in tissues as part of the inflammatory response and can be used for disease staging in some cases.106 Furthermore, MnO2 can enrich specifically in inflamed areas with weak acidity and hypoxia, allowing imaging of inflammation via detection of MPO. Qiu et al.107 encapsulated budesonide in hollow mesoporous MnO2 nanoparticles and dextran sulfate sodium (Fig. 5a). The nanoparticles could release anti-inflammatory medication, remove reactive oxygen species (ROS) and target active macrophages to reduce MPO levels. This method could be used for the diagnosis and treatment of inflammatory enteritis.
 |
| Fig. 5 (a) MnO2 nanomaterials are used in the diagnosis and treatment of inflammatory enteritis.107 Copyright 2022, Royal Society of Chemistry. (b) BMUIG preparation and (c) BMUIG uses in biomodal imaging and in combination with photobiologic therapy for osteomyelitis. (d) Pre- and post-BMIG injection T1-weighted MR images of post-traumatic osteomyelitis mice, with red arrows indicating infected areas.108 Copyright 2019, Elsevier. | |
Meanwhile, the study found that MnO2 could be utilized for diagnosing and treating osteomyelitis. Lu et al.108 synthesized a bacterial inflammation-specific multifunctional preparation known as bovine serum albumin (BSA)-MnO2-ubiquitin-indocyanine green-gentamicin (BMUIG). This preparation was combined with high-resolution bimodal imaging and antibiotic and photodynamic therapy for osteomyelitis treatment (Fig. 5b and c). To evaluate the imaging effect of BMUIG, they intravenously injected BMUIG into a mouse model of acute osteomyelitis and used photoacoustic imaging and MRI (Fig. 5d). The results showed that BMUIG could quickly and precisely detect the infected area, presenting a new technique for osteomyelitis diagnosis. In conclusion, MnO2 shows great potential for detecting antimicrobial activity and inflammation and awaits further research by future researchers.
3.2.2 Neurodegenerative disease imaging.
Manganese-based contrast agents show great potential for neurodegenerative disease imaging. In contrast to Gd(III) contrast agents, Mn(II) can enter active neurons through voltage-gated Ca(II) channels, enabling the mapping of neuronal activity and projection tracing.70 Despite Mn(II) entry via the decisive metal transporter (DMT), it does not affect the generation of Mn(II)-based signals in the brain. Mn(II) enhances MR contrasts at low doses, permitting longitudinal imaging of the same individual over short or long scan intervals.73 Further, Mn(II) is a micronutrient that participates in several biological processes, and it has good biological safety. In conclusion, MnO2 nanomaterials offer unique advantages for addressing neurodegenerative diseases.
Manganese-enhanced MRI (MEMRI) has the potential for preclinical research of the brain physiology in awake behaving animals. MRI can monitor behavior during Mn(II) take up and capture the high-intensity signal retrospectively, which provides a basis for detecting neuronal diseases. Mn(II) has unique physiological properties that make it an appealing nerve contrast agent, including the ability to monitor cell transport kinetics and neuronal projection anatomy in the brain,109 progress of Alzheimer's disease,110–112 emotion management,113 Parkinson's disease,114etc. Remarkably, MnO2 can be reduced to Mn(II) in vivo, which plays a crucial role in imaging neurodegenerative diseases. Lopes et al.115 embedded a combination of hyaluronic acid (HA) and acrylic gellant (GG-MA) into MnO2 nanoparticles. The results indicate that the nanoparticles have MEMRI properties and can be used for intrathecal injection, promoting localized healing of spinal cord tissues during neurodegenerative diseases.
In addition, some studies have confirmed the use of manganese dioxide for treating Alzheimer's disease. E. Park et al.116 utilized multitargeted bioactive nanoparticles to modify the brain microenvironment to achieve imaging and therapeutic benefits in a well-characterized mouse model of Alzheimer's disease (Fig. 6a). Brain-penetrating MnO2 nanoparticles successfully decreased hypoxia, neuroinflammation and oxidative stress. In addition, MnO2 nanoparticles reduced amyloid protein in the cerebral cortex. MnO2 nanoparticles could have positive impacts by enhancing microvascular integrity, cerebral blood flow and cerebral lymphatic clearance of amyloids. Collectively, these changes modify the brain microenvironment to create favorable conditions for sustained neural function.
 |
| Fig. 6 (a) FAIR-FISP magnetic resonance imaging was used to image blood flow in the cerebral cortex and subcortex in the brains of mice with advanced AD.116 Copyright 2023, Wiley-VCH. (b) Schematic of BM NPs for in vivo MR imaging of BBB permeability and for forecasting the transition of bleeding in the MCAO rat model.122 Copyright 2021, Royal Society of Chemistry. (c) Scheme of the preparation process for Ma@(MnO2 + FTY) nanoparticles. (d) An illustration of Ma@(MnO2 + FTY) therapy for the treatment of ischemic penumbra.127 Copyright 2021, Wiley-VCH. | |
3.2.3 Acute ischemic stroke imaging.
Furthermore, MnO2 can effectively cross the blood–brain barrier (BBB) and be used to evaluate the prognostic impact of acute ischemic stroke treatment. Acute ischemic stroke, occurring when a blood clot abruptly occludes a cerebral blood artery, is a leading global contributor to disability and mortality.117 MRI is a traditional clinical tool in acute ischemic stroke diagnosis, being of paramount importance for ultra-early diagnosis and treatment evaluation.118 The primary objective of acute ischemic stroke therapy is to achieve reperfusion and save compromised neurons in the ischemic penumbra. However, reperfusion usually damages the blood–brain barrier, resulting in an often fatal complication, hemorrhagic transformation (HT).119 Predicting HT outcome in stroke patients now involves quantitative imaging of BBB permeability.120,121 Hou et al.122 employed BSA MnO2 nanoparticles (BM NPs) for imaging BBB permeability in stroke. BM NPs display a high T1 relaxation ability (r1 = 5.9 mM−1 s−1), exceptional MR contrast enhancement capability and notable biocompatibility. Thus, these NPs can non-invasively and promptly display BBB permeability in middle cerebral artery occlusion (MCAO) rats and effectively predict HT occurrence in MCAO rats (Fig. 6b). These studies are pivotal for enhancing the basic understanding of various nervous system pathologies and the advancement of clinical treatments for stroke patients.
Thrombolytic drugs administered in the treatment of ischemic strokes can lead to reperfusion injury, significantly hampering neurological function restoration. Furthermore, the process of reoxygenation after hypoxia can cause oxidative stress leading to immune cell activation, which causes a response that can damage normal brain tissue.123 Moreover, immune cells activated in response cause damage to normal tissue in the brain. Thus, accurate assessment of reperfusion injury is critical to determining the cure rate of ischemic stroke patients.
Moreover, researchers aim to attain reperfusion in the ischemic penumbra in order to salvage damaged neurons, which is the principal purpose of most interventions in acute ischemic stroke therapy.124 However, the main obstacle to neuronal survival is reperfusion injury in acute ischemic stroke.125,126 Li et al.127 employed macrophage-camouflaged honeycomb MnO2 nanospheres loaded with fingolimod (FTY) to rescue the ischemic penumbra. The MnO2 nanospheres facilitate the accumulation of cell adhesion molecules that are over-expressed on the damaged vascular endothelial cells at the site of brain damage through recognition mediated by macrophage membrane proteins, thereby enabling T1 MRI (Fig. 6c and d). Additionally, the MnO2 nanospheres possess the ability to consume excess H2O2 and transform it into dehydrogenated O2. It can be decomposed and released in acidic lysosomes, thereby reducing oxidative stress, increasing the survival of injured neurons and reversing the proinflammatory microenvironment.
3.2.4 Tumor imaging.
Malignant tumors, which originate from human epithelial tissues, pose an increasing threat to human health.128 In 2020 alone, about 19.3 million people worldwide were diagnosed with such tumors, and a staggering 10 million lost their lives to these malignancies, according to the World Health Organization. Notably, mortality rates associated with these tumors are on the rise due to several factors, including the social environment, poor living habits and work-related stress. This ailment now ranks second globally in terms of common causes of death, in large part because symptoms lack clarity in the early stages, resulting in the majority of patients already finding themselves grappling with advanced stages of the disease when they are diagnosed. By this point, the cancer cells have already infiltrated various parts of their bodies, making it challenging to treat them with complete effectiveness through chemotherapy and surgery.129 It has proved impossible thus far to diagnose early-stage tumors with a single diagnostic imaging technique in clinical practice. As researchers seek to address this issue, various drugs with applications in early tumor imaging have emerged as subjects of investigation.
In recent years, there have been several studies regarding the use of MnO2 nanomaterials for tumor imaging via MRI. Due to their rapid growth and abundant blood supply, tumor cells exhibit wider gaps around the blood vessel walls than other cells, allowing the MnO2 nanomaterials to enter cancerous cells through such openings. The MnO2 nanomaterials are introduced into the blood, and using the abnormal tumor vascular gap, they leak into and accumulate in the cancerous tissue. As a result, there is a significant increase in MRI signals in the tumor area, but no significant signal enhancement in important organs, muscles and tissues with normal vascular gaps.130
It is well-established that the tumor microenvironment (TME) is acidic, and MnO2 can be reduced to Mn2+ in this acidic environment, enabling MRI enhancement at the tumor site. For instance, Gao et al.131 produced nanoprobes by coating MnO2 on the black P25 titanium dioxide (b-P25) surface (b-P25@MnO2) (Fig. 7a and b). These nanoprobes exhibit GSH-responsive MRI activity. In the TME, excess GSH reacts with MnO2 to produce Mn2+, resulting in a longitudinal relaxation rate of up to 30.44 mM−1 s−1 for b-P25@MnO2. This led to good intratumoral and cellular MRI findings. Similarly, Huang et al.132 employed oxidizable MnO2 nanosheets to create nanoparticles for dual-activated MRI/fluorescence bimodal tumor imaging. The oxidizable MnO2 nanosheets served as intracellular GSH-activated MRI contrast agents and DNA nanocarriers for imaging and targeted transport.
 |
| Fig. 7 (a) Schematic diagram of the b-P25@MnO2 synthesis process and its GSH response and application in (b) MR imaging and improved PTT.131 Copyright 2021, Royal Society of Chemistry. (c) Schematic illustration of the development of a tumor-targeting MRI contrast agent based on melanin nanoparticles that chelate Mn2+ and have extremely high efficiency clearance in vivo.133 Copyright 2021, Royal Society of Chemistry. | |
Also, MnO2 has high biosafety as a MRI contrast agent. Xu et al.133 proposed an innovative Mn2+-chelated ultrafine water-soluble melanin nanoparticle-based tumor-targeted MRI contrast agent (MNP-PEG-Mn) (Fig. 7c). MRI experiments in vivo demonstrated a high tumor targeting specificity after an intravenous injection of MNP-PEG-Mn into tumor bearing mice. Furthermore, MNP-PEG-Mn is associated with little toxicity to the body's tissues since it can be eliminated via hepatobiliary and renal pathways.
MnO2 is capable of carrying various materials, including chemotherapeutic agents, photosensitizers and other therapeutic components, making it useful for MRI and optical imaging, as well as for loading purposes.134 In the current study, amorphous MnO2 was coated with polydopamine (PDA) and polyethylene glycol (PEG) to generate polydopamine@MnO2-PEG (PDA@MnO2-PEG) core–shell nanoparticles. These particles had uniform morphology and size and were employed for MRI-guided tumor photothermal therapy, taking advantage of their acid-sensitive properties. The study findings reveal that amorphous MnO2 shells are acid-sensitive and effective T1-weighted MRI agents. In the tumor models of mice, the MRI signals showed significant enhancements following injection compared to pre-injection measurements. In addition, PDA@MnO2-PEG was used to perform effective photothermal treatment of tumors, as shown in in vivo experiments.134 Furthermore, Laurie J. Rich et al.135 used nano-crystalline NaYF4:Nd3+/NaGdF4 coated with MnO2 to improve hyperthermia in head and neck squamous cell carcinoma (HNSCC) tumors (Fig. 8a). Mn2+ release in vasculature was observed through T1-weighted MRI and photoacoustic imaging. The nanoparticles accumulated locally and systematically within tumors, significantly improving oxygenation in the tumor microenvironment in patient derived HNSCC xenograft models (Fig. 8b).
 |
| Fig. 8 (a) Synthesis of nano-sized NaYF4:Nd3+/NaGdF4 nanocrystals coated with nano-sized MnO2. (b) Photoacoustic and MR imaging was performed to assess the imaging properties and the ability of usNP-MnO2 to modulate hypoxia in vitro.135 Copyright 2020, MDPI. | |
Recently published research has proved the potential of MnO2 nanoparticles for the early detection of triple-negative breast cancer (TNBC). TNBC lacks the expression of human epidermal growth factor receptor 2, progesterone and estrogen receptors, which leads to a higher mortality rate than other types of breast cancer.136–138 Chemotherapy often has a bad outcome for TNBC patients, resulting in a high tumor recurrence rate and the median survival is less than 12 months.139,140 Therefore, early diagnosis and targeted therapy before tumor metastasis are essential for the effective treatment of TNBC patients.141 Hence, Peng et al.95 synthesized biodegradable PB/MnO2 hybrid nanocellulites for MRI and image-guided photodynamic therapy of breast cancers. Liang et al.142 developed core–shell gold nanocage MnO2 (AM) nanoparticles using the template method. AM nanoparticles indicated excellent breast cancer MRI ability, which made the breast cancer targeting and in situ oxygenation monitoring of AM nanoparticles more accurate (Fig. 9a).
 |
| Fig. 9 (a) Preparation of core–shell AM nanoparticles and Mn2+ release due to acid/H2O2 response for TNBC enhanced MRI.142 Copyright 2018, Elsevier. (b) In vivo MRI T1WI of the intracranial glioma after the intravenous injection of HA-PAH-MnO2 NPs and HA-MnO2 NPs at different time points.146 Copyright 2019, Wiley. (c) Schematic diagram of GSH-responsive MnO2 for NSCLC MRI and enhanced radiotherapy.149 Copyright 2017 Cho, Choi, Kim, Goh and Choi. (d) MR imaging of acute myocardium infarction in rabbits contrasted by MnO2@BSA nanocomposites.157 Copyright 2020, Elsevier. | |
MnO2 nanomaterials offer the significant likelihood for early detection of gliomas through MRI. A glioma, a primary central nervous system tumor caused by astrocytes,143 is considered to be an extremely aggressive and common brain tumor.144 Its rapid proliferation is due to the release of glutamate, causing excitotoxic cell death around the core and quick tumor invasion. Due to its high propensity for medication resistance and metastasis, the glioma is among the most fatal and threatening malignancies.145 Fu et al.146 synthesized multifunctional hyaluronic acid–MnO2 nanoparticles (HA-MnO2 NPs) by mixing sodium permanganate with aqueous hyaluronic acid (Fig. 9b). In the MRI detection of intracranial gliomas in rats, HA-MnO2 NPs provided high imaging sensitivity with a duration of up to 3 days after intravenous injection. The study demonstrated that using HA-MnO2 NPs reduced tumor hypoxia and was effective at detecting gliomas through MRI. Furthermore, Zhang et al.147 developed MnO2@BSA nanoparticles (NPs) for glioma imaging in the brain, producing high MR signals under acidic conditions in the tumor microenvironment due to the release of Mn2+. As a result, MnO2@BSA NPs serve as favorable contrast agents for brain glioma imaging. In addition, Jiang et al.148 developed tLyP-1 modified dopamine-β-cyclodextrin nanoparticles, which encapsulated paclitaxel and MnO2, to improve the efficiency of chemotherapy and image contrast enhancement in gliomas. The nanoparticles accumulated at the tumor sites and crossed the BBB. The nanomaterials disintegrated under weak acidity and high H2O2 concentrations, causing the system to collapse, leading to the generation of Mn2+ and O2. At the same time, these nanoparticles can be used to monitor tumor size through MRI in real time, providing a reliable basis for treatment.
MnO2 was unexpectedly found to be specific for lung tumors in addition to imaging well-known tumors. Although MRI is unavailable for lung cancer due to the absence of protons, MnO2 nanomaterials have exceptional advantages as lung magnetic resonance contrast agents. MnO2 nanoparticles (MnO2 NPs) synthesized by Mi Hyeon Cho et al.149 using biocompatible polymers (polyvinylpyrrolidone and polyacrylic acid) demonstrated GSH responsive lysis and signal enhancement in MRI with or without gefitinib resistance in vitro for non-small cell lung cancer (Fig. 9c). MnO2 NPs showed cytotoxic effects on cells and additional dose-dependent therapeutic effects when exposed to X-ray irradiation, opening up possibilities for the diagnosis and treatment of NSCLC. In the study, Cao et al.150 proposed the combination of MnO2 with the photosensitizer chlorin e6 (Ce6) and mesenchymal stem cells (MSCs) (MnO2@Ce6-MSCs) as a potential nanoparticle for the diagnosis and treatment of lung cancer, reducing mortality rates. Therefore, MnO2 proves to be highly promising in the field of tumor imaging, providing a basis for the subsequent integration of diagnosis and treatment.
3.2.5 Myocardial infarction imaging.
In addition to its use in tumor imaging, MnO2 can also be utilized in the diagnosis of myocardial infarction (MI) which is the leading cause of death globally. Despite drug therapy, patients with MI continue to have high morbidity and mortality rates due to irreversible myocardial remodeling at present.151 Notably, inadequate myocardial blood supply causes acute myocardial infarction (AMI), which poses a significant challenge to public health. Early and precise detection of MI, particularly AMI, is necessary for improving the quality and efficiency of care. Gd-chelated contrast agents have been employed in MRI scans of MI to provide higher contrast enhancements at the infarcted sites approximately 10 to 15 minutes after administration since they clear more slowly from the infarcted area than from the normal myocardium.152,153 Nevertheless, Gd-chelated contrast agents cannot distinguish between MI sites and edema areas,154 and require high dose injections, increasing the risk of nephrogenic fibrosis.155,156 Thus, new contrast agents for the diagnosis of AMI are called for. Wang et al.157 designed a low pH-sensitive albumin nanocomposite with MnO2 (MnO2@BSA) for T1-weighted MRI of myocardial infarction (Fig. 9d). The MnO2@BSA nanocomposites can accumulate progressively in the areas of AMI and be taken up by inflammatory macrophages due to the low pH microenvironment. The released Mn2+ ions can interact with ambient albumin, leading to enhanced imaging contrasts. Importantly, MnO2@BSA shows rapid evacuation kinetics after systemic injection and exhibits superior performance in MRI contrast enhancement in rabbit models with AMI, demonstrating its potential as a promising diagnostic agent for the early detection of AMI.
After conducting research on MnO2 nanomaterials for disease imaging, it is evident that there has been a surge in research interest to develop MnO2 nanomaterials with improved properties, such as subtle design, high imaging efficiency, good biosafety and clinical efficiency. This has resulted in a plethora of new MnO2 nanomaterials being reported, offering an array of promising approaches for the clinical application of manganese-based contrast agents in disease diagnosis and treatment. We anticipate that these recent advancements will accelerate the translation of manganese-based contrast agents for clinical disease imaging in the future.
4. MnO2 for fluorescence imaging
Fluorescence is a phenomenon resulting from excitation with light, during which a fluorophore reaches a high-energy state and then releases energy as emission light.158 Therefore, fluorescence imaging takes advantage of the luminescence properties of fluorescence to image biological tissues. In comparison with traditional imaging methods, fluorescence imaging is characterized by high sensitivity, simultaneous imaging of multiple groups of samples, stability and biological safety.159 By selecting specific fluorescent proteins or dyes, fluorescence imaging is helpful for research and diagnosis of diseases in clinical practice.137
GSH is the most prevalent non-protein thiol species in mammalian cells and is essential for defending against damage caused by toxins and free radicals.160 It functions as an endogenous antioxidant and is present in a reduced state under normal conditions. Many diseases including Alzheimer's disease, osteoporosis, acquired immune deficiency syndrome and atherosclerosis have been linked to GSH levels.161–164 Therefore, detecting glutathione levels is an essential method for clinical processing and diagnosis of relevant diseases. However, current fluorescent probes are often subject to the external environment, which reduces their sensitivity and photostability due to photobleaching and premature leakage.165
Due to its light absorption ability, MnO2 can rapidly and accurately activate various functions after reduction triggered by GSH, including quenching fluorescence, targeting specific aptamer carriers and activating an MRI imaging agent in the presence of intracellular GSH. Therefore, MnO2 can be used for detecting glutathione.166 A new nanoprobe was reported that coated negatively charged MnO2 nanosheets with positively charged polyethylenimine (PEI) polymer and then coupled them to fluorescein isothiocyanate (FITC) using thiourea ligation (MnO2-PEI-FITC) (Fig. 10a).167 The MnO2-PEI-FITC nanoprobes exhibited weak fluorescence, which was absorbed by FITC, but FITC fluorescence was excited in the presence of GSH. At the same time, MnO2 was decomposed to Mn2+, enabling detection of low background levels in yeast cells and the inner epidermal tissue of onions. In addition, Yao et al.168 developed a NIR activatable fluorescence nanoprobe for the imaging and determination of intracellular GSH based on a core–shell nanoparticle, consisting of NIR emitting gold nanocluster doped silica as the fluorescent core and manganese dioxide as the GSH-responsive shell (AuNCs@MnO2). The MnO2 shell preferentially absorbed the excitation light, thus leading to fluorescence quenching. Upon addition of GSH, the fluorescence of the nanoprobe was restored along with the reduction of MnO2 to Mn2+, and could specifically detect GSH signals in cells. Similarly, Du et al.169 also confirmed the potential of MnO2 nanomaterials as glutathione detection agents.
 |
| Fig. 10 (a) The steps involved in creating the MnO2-PEI-FITC nanoprobe and its response mechanism for detecting GSH.167 Copyright 2021, Royal Society of Chemistry. (b) Schematic illustration of the preparation of AuNCs@MnO2 core–shell nanoprobes and their application in the NIR fluorescence imaging of intracellular GSH.168 Copyright 2021, Royal Society of Chemistry. | |
Simultaneously, MnO2 can be paired with near-infrared fluorescent (NIR) nanomaterials to develop responsive fluorescent nanoprobes.170 NIR fluorescent nanomaterials have exceptional properties, including good membrane permeability, photostability, deep tissue penetration and low autofluorescence.171 These nanomaterials can also avoid renal filtration and improve the signal-to-noise ratio, enabling long-term detection in live cells.172 For example, Song et al.172 illustrated a novel fluorescence nanoprobe constructed using graphene quantum dots (GQDs) as near-infrared emitters and MnO2 as the fluorescence quencher (MnO2–GQDs). The MnO2–GQD nanoprobes were degraded in the presence of GSH, releasing Mn2+ and GQDs separately and causing the quenched fluorescence to be recovered. Similarly, MnO2 was paired with the photosensitizer Ce6, which displayed good fluorescence imaging both in vivo and in vitro.173 Furthermore, MnO2 nanomaterials can enable dual-mode MRI and fluorescence imaging by reacting with GSH to produce Mn2+. A study reported a dual-mode MRI/fluorescence platform for tumor cell imaging that detected GSH.174 Upon endocytosis, MnO2 nanosheets and intracellular GSH can interact, resulting in the production of a large amount of Mn2+, which can be amplified by the circle amplification (RCA) reaction (Fig. 11a). Meanwhile, GSH treatment restores most of the fluorescence signal. MnO2 grown in situ on the surface of oleic acid-coated UCNPs can enable multimodal MRI and fluorescence imaging.175 This technique enables improved imaging sensitivity, tissue penetration and can guide subsequent clinical practice by detecting glutathione.
 |
| Fig. 11 (a) The MnO2–GQD nanoprobe for the detection of GSH.174 Copyright 2018, Elsevier. (b) Based on DNAzyme–MnO2 nanoprobes, an experimental method for identifying and visualizing miRNA-155 in living cells has been developed.181 Copyright 2021, Royal Society of Chemistry. | |
MnO2 nanomaterials with the ability to detect GSH can also be used for cancer cell-targeted imaging. MnO2 can rapidly degrade in the presence of reductive agents such as an increase of glutathione in cancer cells.176 Using host–guest interactions and self-assembly between fluorescent polymers and MnO2 2D materials, Wang et al.177 developed a 2D probe for cancer cell-targeted fluorescence imaging in liver and TNBC cells based on biothiol responses.
Moreover, MnO2 can be targeted to release fluorescent signals for accurately detecting microRNAs, which can be utilized as biomarkers for clinical diagnoses and play a crucial role in many biological processes.178–180 For example, to introduce DNAzyme probes into cells through endocytosis, MnO2 nanosheets were used as carriers and were reduced to Mn2+ by intracellular GSH, thereby releasing the probes (Fig. 11b).181 The produced Mn2+ activated the DNAzyme probes, causing fluorescence signal changes. These fluorescence imaging techniques effectively detected miRNA-155 in HeLa and HepG2 cells, this is useful for cancer detection and investigating biological events involving miRNAs. In summary, MnO2 can interact with various substances, producing fluorescence to investigate biological process.
5. MnO2 for photoacoustic imaging
Photoacoustic (PA) imaging is a recently developed non-invasive and non-ionizing biomedical imaging technique that has gained popularity in recent years. Pulsed laser irradiation of biological tissues causes the tissues’ optical absorption domain to produce ultrasonic signals. These signals carry characteristic information and can reconstruct an image of optical absorption in the tissues. PA imaging has gained significant attention due to its advantages of high optical resolution in both space and time.182,183 Nevertheless, the diagnosis of diseases using PA imaging relies on the development of imaging contrast agents that are capable of converting the thermal energy produced by pulsed laser irradiation into ultrasound signals.184–186 Consequently, a variety of materials such as semiconductors, noble metals and polymers have been utilized as PA imaging agents.187 In their study, Yan and colleagues18 synthesized a polymer of gold nanorods (Au NRs) and manganese dioxide nanosheets (Au NR–MnO2). Based on their findings, Au NR–MnO2 exhibits a stronger absorption in the NIR region compared to Au NRs. Additionally, its PA intensity is 2.7 times greater than that of Au NRs when dispersed in water. Importantly, Au NR–MnO2 was successfully employed for in vivo tumor imaging. Therefore, Au NR–MnO2 polymers offer an effective strategy for improving PA imaging (Fig. 12a and b).
 |
| Fig. 12 (a) A glutathione (GSH)-triggered gold@manganese dioxide (Au@MnO2) smart theranostic agent for photoacoustic and MR dual-imaging-guided photothermal-enhanced chemodynamic therapy.18 Copyright 2021, Elsevier. (b) PA imaging of different materials (MnO2, Au NRs, Au NR–MnO2). (c) PA imaging of Au NR–MnO2 at different concentrations.195 Copyright 2021, Royal Society of Chemistry. | |
Gold–manganese dioxide nanoparticles have shown potential for improving the effectiveness of cancer diagnosis and treatment through multimodal imaging.188–190 One study shows the result of the combination of photoacoustic and MRI imaging for more accurate diagnosis.191–194 Wang et al.195 demonstrated that gold–manganese dioxide core–shell nanostructures could be used for dual photoacoustic and MR imaging, showing high stability under physiological conditions (Fig. 12c). When accumulated at the tumor site, Au@MnO2 induced a series of reactions whereby MnO2 was converted into Mn2+ due to high levels of GSH present, which enhanced the contrast of photoacoustic imaging and MRI.
Combining different functional materials with MnO2 can enable multimodal imaging effects. Various approaches have been developed to generate MnO2/PB nanostructures for dual-mode MRI/PA imaging.95,196,197 For example, Xu et al.198 described a novel imaging nanomaterial composed of MnO2-coated porous Pt@CeO2 core–shell nanostructures (Pt@CeO2@MnO2). The porous Pt core gave the core–shell nanostructure a high photothermal conversion efficiency in the NIR, which led to good PA imaging effect for tumor imaging. Additionally, photoacoustic imaging is frequently combined with therapy and exhibits significant potential for therapeutic integration.
6. Treatment
To date, MnO2 nanomaterials have been applied in various biomedical domains. In particular, MnO2 nanomaterials exhibit significant potential regarding cancer treatment owing to their unique nanostructures and physicochemical characteristics.199 The primary application of MnO2 nanomaterials consists of various anti-cancer therapies, including photodynamic therapy (PDT), radiation therapy (RIT), photothermal therapy (PTT) and chemodynamic therapy (CDT).
Photodynamic therapy (PDT) is a therapeutic technique that employs photosensitizing agents and laser activation (Fig. 13a).200–202 Photosensitizers absorb light at specific wavelengths and activate oxygen to produce cytotoxic reactive oxygen species (ROS), which can destroy tumor tissues.203–206 However, the uncontrolled proliferation of tumor cells, in combination with PDT, results in oxygen deprivation due to oxygen consumption, reducing the efficiency of PDT while generating harmful metabolites (e.g., H2O2) during hypoxia.207 Nanoscale MnO2 can mitigate tumor hypoxia by interacting with harmful substances such as H+, H2O2, etc., thus enhancing the therapeutic effects of PDT, while concurrently transforming into Mn2+ to amplify MRI contrast.208–211 Additionally, MnO2 nanostructures, characterized by a large surface area and inner cavity, can accommodate photosensitizers effectively, facilitating fluorescence imaging.
 |
| Fig. 13 (a) Schematic diagram of the PDT effect. (b) Schematic of the effects of other treatments. | |
Numerous MnO2 nanomaterials are loaded with photosensitizers for disease therapy. Sun et al.212 designed and constructed a delivery system of a hydrophobic photosensitizer (BSA-MnO2/Ce6@ZIF-8) based on a metal–organic framework for MRI-guided PDT. With BSA-MnO2 modification of the Ce6@ZIF-8 surface, BSA-MnO2/Ce6@ZIF-8 can generate oxygen in the TME, consequently enhancing the photodynamic activity of cancer treatment. Yang et al.173 assembled glucose oxidase (GOx)-coated mesoporous silica nanoparticles (MONs), MnO2 nanosheets and the photosensitizer Ce6, producing a smart nanoprobe (MONsGOx@MnO2-Ce6), thus greatly improving the efficacy of starvation and photodynamic cancer combination therapy. Ji et al.213 designed a novel multifunctional nanosystem, CaO2/MnO2@polydopamine-methylene blue (MB) nanosheet (CMP-MB), which had the same PDT and MRI effects. Additionally, Liu et al.214 proposed a new idea for the combination of inorganic nanomaterials with MnO2, confirming its role in PDT.
Radiation therapy (RIT) is a prevalent method for the eradication of tumors (Fig. 13b).215 Ionizing radiation is responsible for the eradication of cancerous cells in radio-therapy. However, the primary impediment to radiotherapy is the hypoxic environment within tumors. MnO2 can promptly modify the hypoxic microenvironment by breaking down H2O2 into oxygen, augmenting RIT efficiency greatly. Therefore, MnO2 nanomaterials can be utilized as a radiosensitizer to improve the efficiency of radiotherapy.216 Recently, Tian et al.217 created a novel nanoplatform for RIT via the combination of 131I-labeled HSA with MnO2 (131I-HSA-MnO2). MnO2 can induce the degradation of endogenous H2O2, leading to the production of oxygen that mitigates tumor hypoxia-related RIT resistance. The experiments demonstrated that the nanoparticles could be employed as effective RIT agents for anti-cancer treatment.
Photothermal therapy (PTT) is another promising method for tumor killing (Fig. 13b).218–220 Under the influence of radiation, photothermal nanoparticles convert light into heat. To enhance the efficacy of photothermal nanoparticles in tumor ablation, Liu et al.221 developed a highly sensitive 2D-MnO2 nanosheet for PTT. As anticipated, the MnO2 nanosheet exhibited a high photothermal conversion rate, ensuring more efficient and effective tumor ablation.
Chemodynamic therapy (CDT) is a new therapeutic strategy that generates hydroxyl radicals (˙OH) to kill tumor cells in situ (Fig. 13b).222 It does not require the application of external energy and can generate irreversible damage to tumor cells’ lipids, mitochondria, DNA and proteins. Thus, this approach avoids the side effects and tissue penetration depth restrictions.223 The presence of MnO2 can deplete GSH and facilitate ˙OH production, enhancing CDT's effectiveness. Ding et al.224 synthesized a hybrid nanoplatform (UCMN) that coupled upconversion nanoparticles (UCNP) and MnO2. Its complex structure promotes ˙OH production through GSH depletion and cisplatin activation, significantly improving the efficiency of CDT. Additionally, the reaction of MnO2 with GSH to produce Mn2+ facilitates MR image contrast.
In recent years, an increasing number of researchers have investigated the combination of multiple treatment modes. Multiple treatment modes offer advantages over single treatment modes, thus improving the effectiveness of cancer treatment. PDT is effective at treating tumors, but its applicability is constrained by the tumor microenvironment with diverse properties and low tumor permeability of the materials.225 In the context of PTT, the upregulation of heat-shock proteins affects the efficacy of this treatment. A previous study demonstrated that PDT-produced ROS were capable of inhibiting the production of heat shock proteins, which, in turn, enhanced the photothermal effects.226,227 Furthermore, the increase in temperature caused by the photothermal effect itself is also likely to improve PDT efficacy.214 Therefore, the combination of PDT and PTT is expected to produce a synergistic therapeutic effect. To this end, Sun et al.228 developed a multifunctional therapeutic platform (MnO2–SiO2–APTES&Ce6 (MSA&C)) based on MnO2 nanoflowers, which synergistically augmented PDT and PTT, demonstrating promising therapeutic effects. Similarly, Li et al.229 successfully developed excellent nanotherapeutic Bi2S3–MnO2@BSA-Ce6 (BMBC) NPs, which displayed promising in vitro synergistic effects of PTT and PDT in cell experiments. Additionally, studies by Zhang et al.230 and Xu et al.198 simultaneously confirmed the advantages of MnO2 nanomaterials as a combination of PTT and PDT.
MnO2 nanomaterials exhibit a high specific surface area, potent adsorption capability and biocompatibility, making them amenable for loading various anticancer drugs into MnO2 nanocarriers.231 It is well-established that the degradation of MnO2 in low-acidity and high-GSH environments coincides with drug release. Tang et al.232 recently reported the successful design of a MnO2 nanoparticle loaded with the in vitro anticancer drug Adriamycin and an aza-bodipy photosensitizer. The resultant nanocarriers were shown to respond well to the TME and exhibit degradability. The products were found to effectively aggregate at the tumor site and underwent time-dependent clearance mainly through the liver and kidneys. The exceptional performance of MnO2 in recent years presents an opportunity to enhance chemotherapeutic precision.
7. Conclusions and perspectives
MnO2 nanomaterials possess numerous superior properties that have garnered significant attention in the field of imaging. This review presents the comprehensive synthesis of MnO2 nanomaterials and their biomedical applications, with an emphasis on recent applications of MnO2 in MRI. MnO2 nanomaterials exhibit paramagnetism and rapid exchange with hydrogen protons, resulting in enhanced T1-weighted signals. Accordingly, MnO2 nanomaterials have been utilized for inflammation imaging, brain imaging, tumor imaging and myocardial infarction imaging. MnO2 nanomaterials are also straightforward to synthesize, eco-friendly, low-cost, highly biocompatible and possess excellent photothermal properties. Furthermore, they excel in therapeutic applications. MnO2 also demonstrates great potential in fluorescence imaging and photoacoustic imaging, which together produce excellent results when combined with other therapeutic strategies, thereby providing robust support for subsequent clinical transformations. While numerous studies have demonstrated the excellent performance of MnO2 nano-materials, several challenges remain.
(1) To facilitate the early diagnosis of diseases, imaging contrast agents must exhibit enhanced sensitivity and specificity. However, in the case of molecular changes caused by the disease, morphological structures remain predominantly unaltered, thereby making it challenging for MnO2 nanomaterials to achieve precise molecular imaging. Consequently, the precise imaging of molecular changes necessitates the alteration of the shape and size of MnO2 nanomaterials, thereby enhancing sensitivity and enabling the rapid assessment of benign and malignant properties.
(2) The clinical translation and application of MnO2 are crucial aspects of its usefulness. MnO2 nanomaterials possess outstanding properties, leading to various potential applications. However, most of the research findings remain in the domain of basic research and have not been successfully implemented in clinical practice. Hence, it is imperative that multifunctional nanomaterials based on MnO2 are further developed, thereby facilitating their utilization in the clinical milieu.
(3) Despite several studies showing that MnO2 nanomaterials possess an exceptional level of biosafety, the underlying biological toxicity, potential risks and metabolic processes have not been extensively studied. An in-depth understanding of biosafety is essential. Therefore, future studies should focus on exploring the long-term biosafety of MnO2 nanomaterials and demonstrating their biocompatibility advantages as a novel type of magnetic resonance contrast agent.
(4) Several studies have established a significant correlation between manganese exposure and neurodegenerative diseases.233 Interestingly, the relationship between Mn over-exposure and Alzheimer's disease seems to be converse.234,235 It is widely acknowledged that Mn neurotoxicity can significantly impact neurodevelopment of the brain. However, Mn ions can be modified by various strategies to reduce toxicity.
In this review, we introduced a variety of modification strategies, including assembly with proteins, the preparation of hydrogels or surface modification, etc. Experimental results showed that all showed good biocompatibility in vivo and in vitro, demonstrating the reduction of Mn ion toxicity. Therefore, we need to summarize the results and continue the research, so that MnO2 contrast agents can be applied in clinical practice to solve real issues. In conclusion, the application of MnO2 nanomaterials for medical diagnosis and treatment is a pressing concern. The increased utilization of these nanomaterials could aid with enhancing the cure rates of diseases such as cancer, contributing significantly to human health.
Conflicts of interest
There are no conflicts to declare.
Acknowledgements
This work was supported by the Sichuan Foundation for Distinguished Young Scholars (2022JDJQ0049); the Chengdu International Science and Technology Cooperation Fund (Grant 2019-GH02-00074-HZ); the Chengdu Science and Technology Bureau (Grant 2021-YF05-00698-SN); and the Scientific and Technological Achievements Transformation Fund of West China Hospital, Sichuan University (Grant CGZH21002).
References
- W. Guo, C. Yu, S. F. Li, Z. Wang, J. H. Yu, H. W. Huang and J. S. Qiu, Strategies and insights towards the intrinsic capacitive properties of MnO2 for supercapacitors: challenges and perspectives, Nano Energy, 2019, 57, 459–472 CrossRef CAS.
- Q. Z. Zhang, D. Zhang, Z. C. Miao, X. L. Zhang and S. L. Chou, Research progress in MnO2-carbon based supercapacitor electrode materials, Small, 2018, 14, 1702883 CrossRef PubMed.
- J. Duan, T. Liao, X. Xu, Y. Liu, Y. Kuang and C. Li, Metal-polyphenol nanodots loaded hollow MnO2 nanoparticles with a “dynamic protection” property for enhanced cancer chemodynamic therapy, J. Colloid Interface Sci., 2023, 15, 836–851 CrossRef PubMed.
- D. Gangwar and C. Rath, Structural, optical and magnetic properties of alpha- and beta-MnO2 nanorods, Appl. Surf. Sci., 2021, 557, 149693 CrossRef CAS.
- C. Tu, H. Lu, T. Zhou, W. Zhang, L. Deng, W. Cao, Z. Yang, Z. Wang, X. Wu, J. Ding, F. Xu and C. Gao, Promoting the healing of infected diabetic wound by an anti-bacterial and nano-enzyme-containing hydrogel with inflammation-suppressing, ROS-scavenging, oxygen and nitric oxide-generating properties, Biomaterials, 2022, 286, 121597 CrossRef CAS PubMed.
- M. C. Zhao, Y. F. Luo, L. Z. Zhu, D. P. Cai, Y. X. Zhuang, Q. D. Chen and H. B. Zhan, Ultrathin-MnO2 nanosheets branched onto N-doped carbon nanotubes as binder-free cathode electrodes for aqueous zinc-ion batteries with a high areal capacity, J. Alloys Compd., 2022, 913, 165124 CrossRef CAS.
- A. A. Voskanyan, C. K. Ho and K. Y. Chan, 3D delta-MnO2 nanostructure with ultralarge mesopores as high-performance lithium-ion battery anode fabricated via colloidal solution combustion synthesis, J. Power Sources, 2019, 421, 162–168 CrossRef CAS.
- K. Ahmad and S. M. Mobin, Design and fabrication of cost-effective and sensitive non-enzymatic hydrogen peroxide sensor using co-doped delta-MnO2 flowers as electrode modifier, Anal. Bioanal. Chem., 2021, 413, 789–798 CrossRef CAS PubMed.
- Y. F. Xu, J. G. Liu, J. Wang, G. Y. Ma, J. H. Lin, Y. Yang, Y. W. Li, C. H. Zhang and M. Y. Ding, Selective conversion of syngas to aromatics over Fe3O4@MnO2 and hollow HZSM-5 bifunctional catalysts, ACS Catal., 2019, 9, 5147–5156 CrossRef CAS.
- H. Q. Wang, S. Chen, Z. Wang, Y. Zhou and Z. B. Wu, A novel hybrid Bi2MoO6-MnO2 catalysts with the superior plasma induced pseudo photocatalytic-catalytic performance for ethyl acetate degradation, Appl. Catal., B, 2019, 254, 339–350 CrossRef CAS.
- M. A. Dallaston, C. J. Bettencourt, S. Chow, J. Gebhardt, J. Spangler, M. R. Johnston, C. Wall, J. S. Brusnahan and C. M. Williams, Ranking oxidant sensitiveness: a guide for synthetic utility, Chem. – Eur. J., 2019, 25, 9614–9618 CrossRef CAS PubMed.
- B. B. Liu, Y. B. Zhang, J. Wang, J. Wang, Z. J. Su, G. H. Li and T. Jiang, A further investigation on the MnO2-Fe2O3 system roasted under Co-CO2 atmosphere, Adv. Powder Technol., 2019, 30, 302–310 CrossRef CAS.
- Y. Yang, J. Li, H. W. Zhang, L. C. Jin, F. Xu, G. W. Gan, G. Wang and D. D. Wen, Enhanced gyromagnetic properties of NiCuZn ferrite ceramics for lTCC applications by adjusting MnO2-Bi2O3 substitution, Ceram. Int., 2018, 44, 19370–19376 CrossRef CAS.
- S. S. Danewalia, S. Kaur, N. Bansal, S. Khan and K. Singh, Influence of TiO2 and thermal processing on morphological, structural and magnetic properties of Fe2O3/MnO2 modified glass-ceramics, J. Non-Cryst. Solids, 2019, 513, 64–69 CrossRef CAS.
- S. Su, Y. Xu, Y. Wang, X. Wang, L. Shi, D. Wu, P. Zou, A. Nairan, Z. Lin, F. Kang and C. Yang, Holey nickel nanotube reticular network scaffold for high-performance flexible rechargeable Zn/MnO2 batteries, Chem. Eng. J., 2019, 370, 330–336 CrossRef CAS.
- Y. Liu, Z. Jiang and J. Xu, Self-standing metallic mesh with MnO2 multiscale microstructures for high-capacity flexible transparent energy storage, ACS Appl. Mater. Interfaces, 2019, 11, 24047–24056 CrossRef CAS PubMed.
- H. Zhang, Y. Guo, J. Jiao, Y. Qiu, Y. Miao, Y. He, Z. Li, C. Xia, L. Li, J. Cai, K. Xu, X. Liu, C. Zhang, B. H. Bay, S. Song, Y. Yang, M. Peng, Y. Wang and H. Fan, A hepatocyte-targeting nanoparticle for enhanced hepatobiliary magnetic resonance imaging, Nat. Biomed. Eng., 2023, 7, 221–235 CrossRef CAS PubMed.
- G. Yan, M. J. Liu, Y. Y. Cheng, F. F. Xue, L. An and Q. W. Tian, Small Au nanorods-MnO2 sheet aggregation with enhanced photoacoustic imaging for tumor, Mater. Lett., 2021, 304, 130592 CrossRef CAS.
- N. Y. Yu, Y. Wang, J. D. Xia, J. Abashkin, L. Z. Zhao and J. C. Li, Transferrin-conjugated manganese dioxide/semiconducting polymer hybrid nanoparticles for targeted dual-model imaging of orthotopic glioblastoma, ACS Appl. Polym. Mater., 2023, 5, 4596–4603 CrossRef CAS.
- U. Bhure, J. E. Roos, M. D. Perez-Lago and K. Strobel, Single-photon emission computed tomography/computed tomography arthrography of wrist, ankle, and knee joints, Nucl. Med. Commun., 2020, 41, 182–188 CrossRef PubMed.
- Y. Zhou and M. H. Hong, Realization of noncontact confocal optical microsphere imaging microscope, Microsc. Res. Tech., 2021, 84, 2381–2387 CrossRef PubMed.
- M. Scholte, J. O. Barentsz, J. Sedelaar, M. Gotthardt, J. Grutters and M. M. Rovers, Modelling study with an interactive model assessing the cost-effectiveness of Ga-68 prostate-specific membrane antigen positron emission tomography/computed tomography and nano magnetic resonance imaging for the detection of pelvic lymph node metastases in patients with primary prostate cancer, Eur. Urol. Focus, 2020, 6, 967–974 CrossRef PubMed.
- A. Boeren, E. Niemantsverdriet, M. Verstappen, F. Wouters, J. L. Bloem, M. Reijnierse and A. van der Helm-Van Mil, Towards a simplified fluid-sensitive MRI protocol in small joints of the hand in early arthritis patients: reliability between modified dixon and regular gadolinium enhanced TSE fat saturated MRI-sequences, Skeletal Radiol., 2022, 52, 1193–1202 CrossRef PubMed.
- M. Wiart, C. Tavakoli, V. Hubert, I. Hristovska, C. Dumot, S. Parola, F. Lerouge, F. Chauveau, E. Canet-Soulas, O. Pascual, D. P. Cormode, E. Brun and H. Elleaume, Use of metal-based contrast agents for in vivo MR and CT imaging of phagocytic cells in neurological pathologies, J. Neurosci. Methods, 2023, 383, 120729 CrossRef PubMed.
- D. Foster and J. Larsen, Polymeric metal contrast agents for T1-weighted magnetic resonance imaging of the brain, ACS Biomater. Sci. Eng., 2023, 9, 1224–1242 CrossRef CAS PubMed.
- S. Shuvaev, E. Akam and P. Caravan, Molecular MR contrast agents, Invest. Radiol., 2021, 56, 20–34 CrossRef CAS PubMed.
- J. Luo, Y. Zhang, S. Zhu, Y. Tong, L. Ji, W. Zhang, Q. Zhang and Q. Bi, The application prospect of metal/metal oxide nanoparticles in the treatment of osteoarthritis, Naunyn-Schmiedeberg's Arch. Pharmacol., 2021, 394, 1991–2002 CrossRef CAS PubMed.
- M. D. Burgio, R. Sartoris, A. Beaufrere, J. Gregory, B. Guiu, C. Guillot, P. E. Rautou, L. Castera, M. Bouattour, V. Paradis, V. Vilgrain and M. Ronot, Liver surface nodularity on non-contrast MRI identifies advanced fibrosis in patients with NAFLD, Eur. Radiol., 2022, 32, 1781–1791 CrossRef PubMed.
- K. Maruyama, R. Kawasaki, Y. Koide, H. Horinouchi, K. Sasaki, T. Gentsu, T. Okada, M. Yamaguchi and K. Sugimoto, Preoperative magnetic resonance imaging (MRI) for predicting thromboembolic complications during endovascular treatment for iliac artery occlusions, Ann. Vasc. Surg., 2021, 73, 211–221 CrossRef PubMed.
- J. J. Li, Q. Li, M. J. He, F. Ding, L. L. Cai, M. M. Zhao, L. N. Dong, Q. Wang and K. Xu, AS1411 aptamer-modified theranostic liposomes co-encapsulating manganese oxide nano-contrast agent and paclitaxel for MRI and therapy of cancer, RSC Adv., 2019, 9, 34837–34846 RSC.
- M. Yon, S. Gineste, G. Parigi, B. Lonetti, L. Gibot, D. R. Talham, J. D. Marty and C. Mingotaud, Hybrid polymeric nanostructures stabilized by zirconium and gadolinium ions for use as magnetic resonance imaging contrast agents, ACS Appl. Nano Mater., 2021, 4, 4974–4982 CrossRef CAS.
- J. Qin, S. Laurent, Y. S. Jo, A. Roch, M. Mikhaylova, Z. M. Bhujwalla, R. N. Muller and M. Muhammed, A high-performance magnetic resonance imaging T2 contrast agent, Adv. Mater., 2007, 19, 1874–1878 CrossRef CAS.
- H. Li, D. Luo, C. N. Yuan, X. N. Wang, J. Wang, J. P. Basilion and T. J. Meade, Magnetic resonance imaging of PSMA-positive prostate cancer by a targeted and activatable Gd(III) MR contrast agent, J. Am. Chem. Soc., 2021, 143, 17097–17108 CrossRef CAS PubMed.
- J. Wen, K. Yang and S. G. Sun, MnO2-based nanosystems for cancer therapy, Chem. Commun., 2020, 56, 7065–7079 RSC.
- Z. B. Yin, Q. Ji, D. Wu, Z. W. Li, M. F. Fan, H. X. Zhang, X. L. Zhao, A. G. Wu, L. Cheng and L. Y. Zeng, H2O2-responsive gold nanoclusters@mesoporous silica@manganese dioxide nanozyme for “off/on” modulation and enhancement of magnetic resonance imaging and photodynamic therapy, ACS Appl. Mater. Interfaces, 2021, 13, 14928–14937 CrossRef CAS PubMed.
- H. L. Yu and L. Zheng, Manganese dioxide nanosheets as an optical probe for photometric determination of free chlorine, Microchim. Acta, 2016, 183, 2229–2234 CrossRef CAS.
- H. B. Na, J. H. Lee, K. J. An, Y. I. Park, M. Park, I. S. Lee, D. H. Nam, S. T. Kim, S. H. Kim, S. W. Kim, K. H. Lim, K. S. Kim, S. O. Kim and T. Hyeon, Development of a T-1 contrast agent for magnetic resonance imaging using MnO nanoparticles, Angew. Chem., Int. Ed., 2007, 46, 5397–5401 CrossRef CAS PubMed.
- B. Shen, Z. Zhang, X. Shi, C. Cao, Z. Zhang, Z. Hu, N. Ji and J. Tian, Real-time intraoperative glioma diagnosis using fluorescence imaging and deep convolutional neural networks, Eur. J. Nucl. Med. Mol. Imaging, 2021, 48, 3482–3492 CrossRef PubMed.
- I. Steinberg, D. M. Huland, O. Vermesh, H. E. Frostig, W. S. Tummers and S. S. Gambhir, Photoacoustic clinical imaging, Photoacoustics, 2019, 14, 77–98 CrossRef PubMed.
- S. Schmitz-Valckenberg, M. Pfau, M. Fleckenstein, G. Staurenghi, J. R. Sparrow, A. Bindewald-Wittich, R. F. Spaide, S. Wolf, S. R. Sadda and F. G. Holz, Fundus autofluorescence imaging, Prog. Retinal Eye Res., 2021, 81, 100893 CrossRef CAS PubMed.
- S. M. Zhu, S. C. Wang, M. M. Xia, B. J. Wang, Y. Huang, D. X. Zhang, X. J. Zhang and G. F. Wang, Intracellular imaging of glutathione with MnO2 nanosheet@Ru(bpy)(3)(2+)-UiO-66 nanocomposites, ACS Appl. Mater. Interfaces, 2019, 11, 31693–31699 CrossRef CAS PubMed.
- R. Zhai, J. G. Hu, X. X. Chen, Z. X. Xu, Z. Q. Wen and M. J. Jin, Facile synthesis of manganese oxide modified lignin nanocomposites from lignocellulosic biorefinery wastes for dye removal, Bioresour. Technol., 2020, 315, 123846 CrossRef CAS PubMed.
- C. L. Browning, A. Green, E. P. Gray, R. Hurt and A. B. Kane, Manganese dioxide nanosheets induce mitochondrial toxicity in fish gill epithelial cells, Nanotoxicology, 2021, 15, 400–417 CrossRef CAS PubMed.
- Z. W. Qu, Y. Wang, P. A. Yang, W. Zheng, N. Li, J. Y. Bai, Y. W. Zhang, K. L. Li, D. S. Wang, Z. H. Liu, K. X. Yao, R. Li and Y. X. Zhang, Enhanced electromagnetic wave absorption properties of ultrathin MnO2 nanosheet-decorated spherical flower-shaped carbonyl iron powder, Molecules, 2022, 27, 135 CrossRef CAS PubMed.
- W. C. Feng, G. Liu, P. P. Wang, J. W. Zhou, L. X. Gu, L. Z. Chen, X. Y. Li, Y. Y. Dan and X. F. Cheng, Template synthesis of a heterostructured MnO2@SnO2 hollow sphere composite for high asymmetric supercapacitor performance, ACS Appl. Energy Mater., 2020, 3, 7284–7293 CrossRef CAS.
- B. Xiao, X. Zhou, H. Xu, B. Wu, D. Hu, H. Hu, K. Pu, Z. Zhou, X. Liu, J. Tang and Y. Shen, Integration of polymerization and biomineralization as a strategy to facilely synthesize nanotheranostic agents, ACS Nano, 2018, 12, 12682–12691 CrossRef CAS PubMed.
- J. R. Wu, G. R. Williams, S. W. Niu, Y. B. Yang, Y. Li, X. J. Zhang and L. M. Zhu, Biomineralized bimetallic oxide nanotheranostics for multimodal imaging-guided combination therapy, Theranostics, 2022, 12, 7642 CrossRef PubMed.
- S. Dutz, A. Weidner, M. von der Luhe, C. Grafe, P. Biehl, J. Demut, P. Warncke, S. Jungmann, D. Fischer, F. H. Schacher and J. H. Clement, Hybrid nanomaterials of biomolecule corona coated magnetic nanoparticles and their interaction with biological systems, Phys. Sci. Rev., 2022, 7, 1311–1344 Search PubMed.
- C. C. Chu, M. Su, J. Zhu, D. S. Li, H. W. Cheng, X. Y. Chen and G. Liu, Metal-organic framework nanoparticle-based biomineralization: a new strategy toward cancer treatment, Theranostics, 2019, 9, 3134–3149 CrossRef CAS PubMed.
- Y. Wen, M. Q. He, Y. L. Yu and J. H. Wang, Biomolecule-mediated chiral nanostructures: a review of chiral mechanism and application, Adv. Colloid Interface Sci., 2021, 289, 102376 CrossRef CAS PubMed.
- Z. Y. Yang, S. L. Luo, Y. P. Zeng, Z. J. Yang, C. M. Shi and R. Li, Albumin-mediated biomineralization of shape-controllable and biocompatible ceria nanomaterials, ACS Appl. Mater. Interfaces, 2017, 9, 6839–6848 CrossRef CAS PubMed.
- W. Zhu, L. Zhang, Z. Yang, P. Liu, J. Wang, J. Cao, A. Shen and Z. Xu, An efficient tumor-inducible nanotheranostics for magnetic resonance imaging and enhanced photodynamic therapy, Chem. Eng. J., 2019, 358, 969–979 CrossRef CAS.
- Z. Y. Yan, X. M. Zhang, Y. F. Liu, Y. M. Shen, N. Li, Q. Jia, Y. H. Ji, P. T. Zhang, L. Zhao and Z. W. Meng, HSA-MnO2-131I combined imaging and treatment of anaplastic thyroid carcinoma, Technol. Cancer Res. Treat., 2022, 21, 15330338221106557 Search PubMed.
- T. S. Lin, X. Z. Zhao, S. Zhao, H. Yu, W. M. Cao, W. Chen, H. Wei and H. Q. Guo, O-2-generating MnO2 nanoparticles for enhanced photodynamic therapy of bladder cancer by ameliorating hypoxia, Theranostics, 2018, 8, 990–1004 CrossRef CAS PubMed.
- W. Zhang, R. R. Cheng, H. H. Bi, Y. H. Lu, L. B. Ma and X. J. He, A review of porous carbons produced by template methods for supercapacitor applications, New Carbon Mater., 2021, 36, 69–78 CrossRef CAS.
- Q. Q. Wang, R. Zhou, S. M. Liu, S. Sethupathy, J. Liu, J. Z. Sun, L. H. Zou and Q. Q. Zhu, Templated synthesis and assembly with sustainable cellulose nanomaterial for functional nanostructure, Cellulose, 2022, 29, 4287–4321 CrossRef CAS.
- N. D. Petkovich and A. Stein, Controlling macro- and mesostructures with hierarchical porosity through combined hard and soft templating, Chem. Soc. Rev., 2013, 42, 3721–3739 RSC.
- B. Y. Bai, J. H. Li and J. M. Hao, 1D-MnO2, 2D-MnO2 and 3D-MnO2 for low-temperature oxidation of ethanol, Appl. Catal., B, 2015, 164, 241–250 CrossRef CAS.
- R. Ghosh, B. Sunny, M. Babu and A. Thirumurugan, Role of crystal nucleation and growth conditions on the soft-templated preparation of HKUST-1 MOFs with hierarchical porosity involving gemini surfactants, Microporous Mesoporous Mater., 2022, 334, 111758 CrossRef CAS.
- J. W. Chen, Q. Chen, C. Liang, Z. J. Yang, L. Zhang, X. Yi, Z. L. Dong, Y. Chao, Y. G. Chen and Z. Liu, Albumin-templated biomineralizing growth of composite nanoparticles as smart nanotheranostics for enhanced radiotherapy of tumors, Nanoscale, 2017, 9, 14826–14835 RSC.
- Y. Y. Zhang, F. Lv, Y. R. Cheng, Z. P. Yuan, F. Yang, C. H. Liu, Y. Cao, K. Zhang, H. T. Lu, S. Zada, S. J. Guo, H. F. Dong and X. J. Zhang, Pd@Au bimetallic nanoplates decorated mesoporous MnO2 for synergistic nucleus-targeted NIR-II photothermal and hypoxia-relieved photodynamic therapy, Adv. Healthcare Mater., 2020, 9, 1901528 CrossRef CAS PubMed.
- E. Doustkhah, R. Hassandoost, A. Khataee, R. Luque and M. Assadi, Hard-templated metal-organic frameworks for advanced applications, Chem. Soc. Rev., 2021, 50, 2927–2953 RSC.
- Z. Zhang and Y. H. Ji, Nanostructured manganese dioxide for anticancer applications: preparation, diagnosis, and therapy, Nanoscale, 2020, 12, 17982–18003 RSC.
- G. N. Zhang, J. H. Zhang, H. J. Yang and X. F. Li,
In situ redox reaction derived porous nanosheets of MnO2 for supercapacitors, Mater. Lett., 2022, 306, 130858 CrossRef CAS.
- A. A. Egorova, T. M. Bushkova, I. V. Kolesnik, A. D. Yapryntsev, S. Y. Kottsov and A. E. Baranchikov, Selective synthesis of manganese dioxide polymorphs by the hydrothermal treatment of aqueous KMnO4 solutions, Russ. J. Inorg. Chem., 2021, 66, 146–152 CrossRef CAS.
- K. Fan, J. Su, Z. H. Zeng, J. Hu, H. J. Yang and Z. C. Hou, Anti-fouling and protein separation of PVDF-g-PMAA@MnO2 filtration membrane with in situ grown MnO2 nanorods, Chemosphere, 2022, 286, 131756 CrossRef CAS PubMed.
- S. C. Zhang, Q. Z. Li, N. Yang, Y. H. Shi, W. Ge, W. J. Wang, W. Huang, X. J. Song and X. C. Dong, Phase-change materials-based nanoparticles for controlled hypoxia modulation and enhanced phototherapy, Adv. Funct. Mater., 2019, 29, 1903850 CrossRef.
- W. R. Nitz and P. Reimer, Contrast mechanisms in MR imaging, Eur. Radiol., 1999, 9, 1032–1046 CrossRef CAS PubMed.
- A. Merbach, L. Helm and É. Tóth, The chemistry of contrast agents in medical, Magn. Reson. Imaging, 2013, 1–512 Search PubMed.
- T. W. Uselman, C. S. Medina, H. B. Gray, R. E. Jacobs and E. L. Bearer, Longitudinal manganese-enhanced magnetic resonance imaging of neural projections and activity, NMR Biomed., 2022, 35, e4675 CrossRef CAS PubMed.
- X. Li, Z. H. Xue, J. F. Xia, G. H. Zhou, D. Y. Jiang, M. T. Dai, W. H. Wang, J. Y. Miu, Y. R. Heng, C. Y. Yu and Q. Li, Gd/Y hydroxide nanosheets as highly efficient T1/T2 MRI contrast agents, Nanomaterials, 2021, 11, 17 CrossRef CAS PubMed.
- P. Marckmann, L. Skov, K. Rossen, A. Dupont, M. B. Damholt, J. G. Heaf and H. S. Thomsen, Nephrogenic systemic fibrosis: suspected causative role of gadodiamide used for contrast-enhanced magnetic resonance imaging, J. Am. Soc. Nephrol., 2006, 17, 2359–2362 CrossRef PubMed.
- A. Budimir, K. Jozsef, F. Istvan, G. Lente, B. Istvan, I. Batinic-Haberle and M. Birus, Water exchange rates of water-soluble manganese(III) porphyrins of therapeutical potential, Dalton Trans., 2010, 39, 4405–4410 RSC.
- S. F. Lincoln, Mechanistic studies of metal aqua ions: a semi-historical perspective, Helv. Chim. Acta, 2005, 88, 523–545 CrossRef CAS.
- T. W. Uselman, D. R. Barto, R. E. Jacobs and E. L. Bearer, Evolution of brain-wide activity in the awake behaving mouse after acute fear by longitudinal manganese-enhanced MRI, Neuroimage, 2020, 222, 1–22 CrossRef PubMed.
- J. Y. Hong, Y. G. Lim, Y. J. Song and K. Park, Tumor microenvironment-responsive histidine modified-hyaluronic acid-based MnO2 as in vivo MRI contrast agent, Int. J. Biol. Macromol., 2023, 226, 121–131 CrossRef CAS PubMed.
- W. Bai, K. Zhang, S. Yu, J. Zhang and L. Jin, The preparation of MnO2/BSA/CdTe quantum dots complex for ratiometric fluorescence/T1-weighted MRI detection of H2O2, Talanta, 2023, 252, 123774 CrossRef CAS PubMed.
- Y. Chen, H. L. Cong, Y. Q. Shen and B. Yu, Biomedical application of manganese dioxide nanomaterials, Nanotechnology, 2020, 31, 202001 CrossRef CAS PubMed.
- B. Shah, S. W. Anderson, J. Scalera, H. Jara and J. A. Soto, Quantitative MR imaging: physical principles and sequence design in abdominal imaging, Radiographics, 2011, 31, 867–880 CrossRef PubMed.
- D. Serai, Basics of magnetic resonance imaging and quantitative parameters T1, T2, T2*, T1rho and diffusion-weighted imaging, Pediatr. Radiol., 2022, 52, 217–227 CrossRef PubMed.
- J. Estelrich, M. J. Sanchez-Martin and M. A. Busquets, Nanoparticles in magnetic resonance imaging: from simple to dual contrast agents, Int. J. Nanomed., 2015, 10, 1727–1741 CAS.
- S. Marasini, H. Yue, S. L. Ho, H. Cha, J. A. Park, K. H. Jung, A. Ghazanfari, M. Y. Ahmad, S. Liu, K. S. Chae, Y. Chang and G. H. Lee, A novel paramagnetic nanoparticle T2 magnetic resonance imaging contrast agent with high colloidal stability: polyacrylicacid-coated ultrafine dysprosium oxide nanoparticles, Bull. Korean Chem. Soc., 2020, 41, 829–836 CrossRef CAS.
- M. J. Cho, J. Villanova, D. M. Ines, J. Chen, S. S. Lee, Z. Xiao, X. Guo, J. A. Dunn, D. D. Stueber, P. Decuzzi and V. L. Colvin, Sensitive T2 MRI contrast agents from the rational design of iron oxide nanoparticle surface coatings, J. Phys. Chem. C, 2023, 127, 1057–1070 CrossRef CAS.
- K. Deka, A. Guleria, D. Kumar, J. Biswas, S. Lodha, S. D. Kaushik, S. Dasgupta and P. Deb, Exclusive T2 MRI contrast enhancement by mesoporous carbon framework encapsulated manganese oxide nanoparticles, Curr. Appl. Phys., 2020, 20, 89–95 CrossRef.
- Y. B. Zhu, Y. Zhang, H. Wu, S. Wang and X. Li, An oxidative polymerized carboxymethyl cellulose hydrogel for the combined anti-tumor recurrence, J. Mater. Sci., 2023, 58, 369–382 CrossRef CAS.
- Q. Luo, Z. P. Gao, Q. N. Xiao and E. Song, One-step determination of alkaline phosphatase in human serum based on manganese(IV) dioxide/manganese(II)-mediated nuclear magnetic resonance (NMR) relaxation, Anal. Lett., 2022, 55, 2876–2885 CrossRef CAS.
- X. Zhu, H. Xiong, Q. Zhou, Z. Zhao, Y. Zhang, Y. Li, S. Wang and S. Shi, A pH-activatable MnCO3 nanoparticle for improved magnetic resonance imaging of tumor malignancy and metastasis, ACS Appl. Mater. Interfaces, 2021, 13, 18462–18471 CrossRef CAS PubMed.
- H. W. Lu, A. Chen, X. D. Zhang, Z. X. Wei, R. Cao, Y. Zhu, J. X. Lu, Z. L. Wang and L. L. Tian, A pH-responsive T1-T2 dual-modal MRI contrast agent for cancer imaging, Nat. Commun., 2022, 13, 7948 CrossRef CAS PubMed.
- B. S. Akhmadeev, I. R. Nizameev, K. V. Kholin, A. D. Voloshina, T. P. Gerasimova, A. T. Gubaidullin, M. K. Kadirov, I. E. Ismaev, K. A. Brylev, R. R. Zairov and A. R. Mustafina, Molecular and nano-structural optimization of nanoparticulate Mn2+-hexarhenium cluster complexes for optimal balance of high T1/T2-weighted contrast ability with low hemoagglutination and cytotoxicity, Pharmaceutics, 2022, 14, 1508 CrossRef CAS PubMed.
- G. X. Jiang, D. Fan, J. S. Tian, Z. C. Xiang and Q. J. Fang, Self-confirming magnetosomes for tumor-targeted T1/T2 dual-mode MRI and MRI-guided photothermal therapy, Adv. Healthcare Mater., 2022, 11, e2200841 CrossRef PubMed.
- C. X. Li, Y. Wang, H. Y. Nong, X. X. Hu, Y. Wu, Y. J. Zhang, C. M. Liang, K. G. Chen and S. L. Li, Manganese and dysprosium codoped carbon quantum dots as a potential fluorescent/T1/T2/CT quadri-modal imaging nanoprobe, Nanotechnology, 2022, 33, 025101 CAS.
- R. Mallik, M. Saha and C. Mukherjee, Porous silica nanospheres with a confined mono(aquated) Mn(II)-complex: a potential T1-T2 dual contrast agent for magnetic resonance imaging, ACS Appl. Bio Mater., 2021, 4, 8356–8367 CrossRef CAS PubMed.
- X. M. Cao, L. Y. Gu, S. K. Hu, A. Mukhtar and K. M. Wu, Fe/Mn multilayer nanowires as high-performance T1-T2 dual modal MRI contrast agents, Materials, 2021, 14, 2238 CrossRef CAS PubMed.
- E. A. Kras, E. M. Snyder, G. E. Sokolow and J. R. Morrow, Distinct coordination chemistry of Fe(III)-based MRI probes, Acc. Chem. Res., 2022, 55, 1435–1444 CrossRef CAS PubMed.
- J. R. Peng, M. L. Dong, B. Ran, W. T. Li, Y. Hao, Q. Yang, L. W. Tan, K. Shi and Z. Y. Qian, “one-for-all”-type, biodegradable prussian blue/manganese dioxide hybrid nanocrystal for trimodal imaging-guided photothermal therapy and oxygen regulation of breast cancer, ACS Appl. Mater. Interfaces, 2017, 9, 13875–13886 CrossRef CAS PubMed.
- Z. L. Wang, X. D. Xue, H. W. Lu, Y. X. He, Z. W. Lu, Z. J. Chen, Y. Yuan, N. Tang, C. A. Dreyer, L. Quigley, N. Curro, K. S. Lam, J. H. Walton, Z. Y. Lin, A. Y. Louie, D. A. Gilbert, K. Liu, K. W. Ferrara and Y. P. Li, Two-way magnetic resonance tuning
and enhanced subtraction imaging for non-invasive and quantitative biological imaging, Nat. Nanotechnol., 2020, 15, 482 CrossRef CAS PubMed.
- X. Zhao, K. Guo, K. Zhang, S. Duan, M. Chen, N. Zhao and F. J. Xu, Orchestrated yolk-shell nanohybrids regulate macrophage polarization and dendritic cell maturation for oncotherapy with augmented antitumor immunity, Adv. Mater., 2022, 34, e2108263 CrossRef PubMed.
- B. B. Ding, P. Zheng, F. Jiang, Y. J. Zhao, M. F. Wang, M. Y. Chang, P. A. Ma and J. Lin, MnO(x) nanospikes as nanoadjuvants and immunogenic cell death drugs with enhanced antitumor immunity and antimetastatic effect, Angew. Chem., Int. Ed., 2020, 59, 16381–16384 CrossRef CAS PubMed.
- S. Dawadi, A. Gupta, M. Khatri, B. Budhathoki, G. Lamichhane and N. Parajuli, Manganese dioxide nanoparticles: synthesis, application and challenges, Bull. Mater. Sci., 2020, 43, 277 CrossRef CAS.
- W. Xu, X. Qing, S. Liu, Z. Chen and Y. Zhang, Manganese oxide nanomaterials for bacterial infection detection and therapy, J. Mater. Chem. B, 2022, 10, 1343–1358 RSC.
- S. Wang, Y. Fang, Z. Zhang, Q. Jin and J. Ji, Bacterial infection microenvironment sensitive prodrug micelles with enhanced photodynamic activities for infection control, Colloid Interface Sci. Commun., 2021, 40, 100354 CrossRef CAS.
- W. Xiu, L. Wan, K. Yang, X. Li, L. Yuwen, H. Dong, Y. Mou, D. Yang and L. Wang, Potentiating hypoxic microenvironment for antibiotic activation by photodynamic therapy to combat bacterial biofilm infections, Nat. Commun., 2022, 13, 3875 CrossRef CAS PubMed.
- H. Dong, W. Xiu, L. Wan, Q. Li, Y. Zhang, M. Ding, J. Shan, K. Yang, Z. Teng, L. Yuwen and Y. Mou, Biofilm microenvironment response nanoplatform synergistically degrades biofilm structure and relieves hypoxia for efficient sonodynamic therapy, Chem. Eng. J., 2023, 453, 139839 CrossRef CAS.
- W. Xiu, S. Gan, Q. Wen, Q. Qiu, S. Dai, H. Dong, Q. Li, L. Yuwen, L. Weng, Z. Teng, Y. Mou and L. Wang, Biofilm microenvironment-responsive nanotheranostics for dual-mode imaging and hypoxia-relief-enhanced photodynamic therapy of bacterial infections, Research, 2020, 2020, 9426453 CrossRef CAS PubMed.
- B. Ding, P. Zheng, P. Ma and J. Lin, Manganese oxide nanomaterials: synthesis, properties, and theranostic applications, Adv. Mater., 2020, 32, e1905823 CrossRef PubMed.
- A. G. Siraki, The many roles of myeloperoxidase: from inflammation and immunity to biomarkers, drug metabolism and drug discovery, Redox Biol., 2021, 46, 102109 CrossRef CAS PubMed.
- H. Qiu, H. Gong, Y. Bao, H. Jiang and W. Tong, Reactive oxygen species-scavenging hollow MnO2 nanozymes as carriers to deliver budesonide for synergistic inflammatory bowel disease therapy, Biomater. Sci., 2022, 10, 457–466 RSC.
- X. Lu, R. Chen, J. Lv, W. Xu, H. Chen, Z. Ma, S. Huang, S. Li, H. Liu, J. Hu and L. Nie, High-resolution bimodal imaging and potent antibiotic/photodynamic synergistic therapy for osteomyelitis with a bacterial inflammation-specific versatile agent, Acta Biomater., 2019, 99, 363–372 CrossRef CAS PubMed.
- X. Li, J. Xie, L. Lu, L. Zhang, L. Zhang, Y. Zou, Q. Wang, X. Luo and S. Li, Kinetics of manganese transport and gene expressions of manganese transport carriers in CACO-2 cell monolayers, BioMetals, 2013, 26, 941–953 CrossRef CAS PubMed.
- L. Wu, X. Zhou, Y. Cao, S. H. Mak, L. Zha, N. Li, Z. Su, Y. Han, Y. Wang, M. P. Man Hoi, Y. Sun, G. Zhang, Z. Zhang and X. Yang, Therapeutic efficacy of novel memantine nitrate Mn-08 in animal models of Alzheimer's disease, Aging Cell, 2021, 20, e13371 CrossRef CAS PubMed.
- B. D. Spitznagel, R. A. Buchanan, D. C. Consoli, M. K. Thibert, A. B. Bowman, W. P. Nobis and F. E. Harrison, Acute manganese exposure impairs glutamatergic function in a young mouse model of Alzheimer's disease, Neurotoxicology, 2023, 95, 1–11 CrossRef CAS PubMed.
- Y. Yang, J. Zhang, X. Yang, Z. Li, J. Wang, C. Lu, A. Nan and Y. Zou, Dysregulated APP expression and alpha-secretase processing of APP is involved in manganese-induced cognitive impairment, Ecotoxicol. Environ. Saf., 2021, 220, 112365 CrossRef CAS PubMed.
- B. A. Racette, G. Nelson, W. W. Dlamini, T. Hershey, P. Prathibha, J. R. Turner, H. Checkoway, L. Sheppard and S. S. Nielsen, Depression and anxiety in a manganese-exposed community, Neurotoxicology, 2021, 85, 222–233 CrossRef CAS PubMed.
- Y. Xu, L. Wei, S. Tang, Q. Shi, B. Wu, X. Yang, Y. Zou, X. Wang, Q. Ao, L. Meng, X. Wei, N. Zhang, Y. Li, C. Lan, M. Chen, X. Li and C. Lu, Regulation PP2AC methylation ameliorating autophagy dysfunction caused by Mn is associated with mTORC1/ULK1 pathway, Food Chem. Toxicol., 2021, 156, 112441 CrossRef CAS PubMed.
- S. V. Lopes, P. Walczak, M. Janowski, R. L. Reis, J. Silva-Correia and J. M. Oliveira, Cytocompatible manganese dioxide-based hydrogel nanoreactors for MRI imaging, Biomater. Adv., 2022, 134, 112575 CrossRef PubMed.
- E. Park, L. Y. Li, C. He, A. Z. Abbasi, T. Ahmed, W. D. Foltz, R. O'Flaherty, M. Zain, R. P. Bonin, A. M. Rauth, P. E. Fraser, J. T. Henderson and X. Y. Wu, Brain–penetrating and disease site–targeting manganese dioxide–polymer–lipid hybrid nanoparticles remodel microenvironment of Alzheimer's disease by regulating multiple pathological pathways, Adv. Sci., 2023, 2207238 CrossRef CAS PubMed.
- J. P. Ho, Acute ischemic stroke: emergency department management after the 3-hour window, Emerg. Med. Pract., 2021, 23, 1–33 Search PubMed.
- B. J. Kim, H. G. Kang, H. Kim, S. Ahn, N. Y. Kim, S. Warach and D. Kang, Magnetic resonance imaging in acute ischemic stroke treatment, J. Stroke, 2014, 16, 131–145 CrossRef PubMed.
- J. M. Hong, D. S. Kim and M. Kim, Hemorrhagic transformation after ischemic stroke: mechanisms and management, Front. Neurol., 2021, 12, 703258 CrossRef PubMed.
- S. Bernardo-Castro, H. Donato, L. Ferreira and J. Sargento-Freitas, Permeability of the blood-brain barrier through the phases of ischaemic stroke and relation with clinical outcome: protocol for a systematic review, BMJ Open, 2020, 10, e039280 CrossRef PubMed.
- L. Hong, T. Hsu, Y. Zhang and X. Cheng, Neuroimaging prediction of hemorrhagic transformation for acute ischemic stroke, Cerebrovasc. Dis., 2022, 51, 542–552 CrossRef CAS PubMed.
- W. Hou, Y. Jiang, G. Xie, L. Zhao, F. Zhao, X. Zhang, S. K. Sun, C. Yu and J. Pan, Biocompatible BSA-MnO2 nanoparticles for in vivo timely permeability imaging of blood-brain barrier and prediction of hemorrhage transformation in acute ischemic stroke, Nanoscale, 2021, 13, 8531–8542 RSC.
- E. H. Tedyanto, K. Tini and N. Pramana, Magnetic resonance imaging in acute ischemic stroke, Cureus, 2022, 14, e27224 Search PubMed.
- M. S. Phipps and C. A. Cronin, Management of acute ischemic stroke, Br. Med. J., 2020, 368, l6983 CrossRef PubMed.
- J. Peng, O. M. Salami, O. Habimana, Y. Xie, H. Yao and G. Yi, Targeted mitochondrial drugs for treatment of ischemia-reperfusion injury, Curr. Drug Targets, 2022, 23, 1526–1536 CrossRef CAS PubMed.
- G. Kapanova, G. Tashenova, A. Akhenbekova, A. Tokpinar and S. Yilmaz, Cerebral ischemia reperfusion injury: from public health perspectives to mechanisms, Folia Neuropathol., 2022, 60, 384–389 CrossRef PubMed.
- C. Li, Z. H. Zhao, Y. F. Luo, T. T. Ning, P. X. Liu, Q. J. Chen, Y. C. Chu, Q. Guo, Y. W. Zhang, W. X. Zhou, H. Y. Chen, Z. Zhou, Y. Wang, B. Y. Su, H. Y. You, T. Y. Zhang, X. W. Li, H. L. Song, C. F. Li, T. Sun and C. Jiang, Macrophage-disguised manganese dioxide nanoparticles for neuroprotection by reducing oxidative stress and modulating inflammatory microenvironment in acute ischemic stroke, Adv. Sci., 2021, 8, e2101526 CrossRef PubMed.
- R. L. Siegel, K. D. Miller, H. E. Fuchs and A. Jemal, Cancer statistics, 2022, CA Cancer J. Clin., 2022, 72, 7–33 CrossRef PubMed.
- B. B. Ding, J. Yue, P. Zheng, P. A. Ma and J. Lin, Manganese oxide nanomaterials boost cancer immunotherapy, J. Mater. Chem. B, 2021, 9, 7117–7131 RSC.
- Y. Shi, Y. Shi, Z. Wang, J. Zhang, R. Hao, G. Zhang, X. Zhao and L. Zeng, Glucose-responsive mesoporous prussian blue nanoprobes coated with ultrasmall gold and manganese dioxide for magnetic resonance imaging and enhanced antitumor therapy, Chem. Eng. J., 2023, 453, 139885 CrossRef CAS.
- Y. Gao, Z. B. Yin, Q. Ji, J. B. Jiang, Z. Z. Tao, X. L. Zhao, S. J. Sun, A. G. Wu and L. Y. Zeng, Black titanium dioxide@manganese dioxide for glutathione-responsive MR imaging and enhanced photothermal therapy, J. Mater. Chem. B, 2021, 9, 314–321 RSC.
- X. Huang, Y. Zhang, J. Chen, L. Zhang, Y. Xu, W. Yin, Y. Shi, X. Zou, S. Liu and Z. Dai, Dual-locked DNAzyme platform for in vitro and in vivo discrimination of cancer cells, Anal. Chem., 2022, 94, 12221–12230 CrossRef CAS PubMed.
- W. Xu, J. H. Sun, L. P. Li, X. Y. Peng, R. Zhang and B. Wang, Melanin-manganese nanoparticles with ultrahigh efficient clearance in vivo for tumor-targeting t-1 magnetic resonance imaging contrast agent, Biomater. Sci., 2017, 6, 207–215 RSC.
- Y. Liu, J. Xu, L. Liu, J. Tan, L. Gao, J. Wang, Y. Wang, Z. Yan and L. Yu, Amorphous manganese dioxide coated polydopamine nanoparticles for acid-sensitive magnetic resonance imaging-guided tumor photothermal therapy, J. Biomed. Nanotechnol., 2019, 15, 1771–1780 CrossRef CAS PubMed.
- L. J. Rich, J. A. Damasco, J. C. Bulmahn, H. L. Kutscher, P. N. Prasad and M. Seshadri, Photoacoustic and magnetic resonance imaging of hybrid manganese dioxide-coated ultra-small NaGdF4 nanoparticles for spatiotemporal modulation of hypoxia in head and neck cancer, Cancers, 2020, 12, 3294 CrossRef CAS PubMed.
- H. Zheng, S. Siddharth, S. Parida, X. Wu and D. Sharma, Tumor microenvironment: key players in triple negative breast cancer immunomodulation, Cancers, 2021, 13, 3357 CrossRef CAS PubMed.
- S. Loizides and A. Constantinidou, Triple negative breast cancer: immunogenicity, tumor microenvironment, and immunotherapy, Front. Genet., 2022, 13, 1095839 CrossRef CAS PubMed.
- S. A. Dass, K. L. Tan, R. Selva Rajan, N. F. Mokhtar, E. R. Mohd Adzmi, W. F. Wan Abdul Rahman, T. A. D. A. Tengku Din and V. Balakrishnan, Triple negative breast cancer: a review of present and future diagnostic modalities, Medicina, 2021, 57, 62 CrossRef PubMed.
- N. Vidula, L. W. Ellisen and A. Bardia, Novel agents for metastatic triple-negative breast cancer: finding the positive in the negative, J. Natl. Compr. Cancer Network, 2021, 19, 109–117 CAS.
- R. Yang, Y. Li, H. Wang, T. Qin, X. Yin and X. Ma, Therapeutic progress and challenges for triple negative breast cancer: targeted therapy and immunotherapy, Mol. Biomed., 2022, 3, 8 CrossRef PubMed.
- X. L. Hu, Q. Y. Ca, J. Gao, R. A. Field, G. R. Chen, N. Y. Jia, Y. Zang, J. Li and X. P. He, Self-assembled 2D glycoclusters for the targeted delivery of theranostic agents to triple-negative breast cancer cells, ACS Appl. Mater. Interfaces, 2019, 11, 22181–22187 CrossRef CAS PubMed.
- R. J. Liang, L. L. Liu, H. M. He, Z. K. Chen, Z. Q. Han, Z. Y. Luo, Z. H. Wu, M. B. Zheng, Y. F. Ma and L. T. Cai, Oxygen-boosted immunogenic photodynamic therapy with gold nanocages@manganese dioxide to inhibit tumor growth and metastases, Biomaterials, 2018, 177, 149–160 CrossRef CAS PubMed.
- L. Qiao, H. Yang, X. Shao, Q. Yin, X. Fu and Q. Wei, Research progress on nanoplatforms and nanotherapeutic strategies in treating glioma, Mol. Pharm., 2022, 19, 1927–1951 CrossRef CAS PubMed.
- X. Li, X. Geng, Z. Chen and Z. Yuan, Recent advances in glioma microenvironment-response nanoplatforms for phototherapy and sonotherapy, Pharmacol. Res., 2022, 179, 106218 CrossRef CAS PubMed.
- V. Di Nunno, E. Franceschi, A. Tosoni, L. Gatto, S. Bartolini and A. A. Brandes, Tumor-associated microenvironment of adult gliomas: a review, Front. Oncol., 2022, 12, 891543 CrossRef CAS PubMed.
- C. P. Fu, X. H. Duan, M. H. Cao, S. Q. Jiang, X. H. Ban, N. Guo, F. Zhang, J. J. Mao, T. Huyan, J. Shen and L. M. Zhang, Targeted magnetic resonance imaging and modulation of hypoxia with multifunctional hyaluronic acid-MnO2 nanoparticles in glioma, Adv. Healthcare Mater., 2019, 8, e1900047 CrossRef PubMed.
- H. Zhang, T. J. Wang, Y. Y. Zheng, C. X. Yan, W. Gu and L. Ye, Comparative toxicity and contrast enhancing assessments of Gd2O3@BSA and MnO2@BSA nanoparticles for MR imaging of brain glioma, Biochem. Biophys. Res. Commun., 2018, 499, 488–492 CrossRef CAS PubMed.
- S. Q. Jiang, X. H. Li, F. Zhang, J. J. Mao, M. H. Cao, X. N. Zhang, S. M. Huang, X. H. Duan and J. Shen, Manganese dioxide-based nanocarrier delivers paclitaxel to enhance chemotherapy against orthotopic glioma through hypoxia relief, Small Methods, 2022, 6, e2101531 CrossRef PubMed.
- M. H. Cho, E. S. Choi, S. Kim, S. H. Goh and Y. Choi, Redox-responsive manganese dioxide nanoparticles for enhanced MR imaging and radiotherapy of lung cancer, Front. Chem., 2017, 5, 109 CrossRef PubMed.
- W. Cao, B. Liu, F. F. Xia, M. Duan, Y. P. Hong, J. Q. Niu, L. R. Wang, Y. L. Liu, C. Li and D. X. Cui, MnO2@Ce6-loaded mesenchymal stem cells as an “oxygen-laden guided-missile” for the enhanced photodynamic therapy on lung cancer, Nanoscale, 2020, 12, 3090–3102 RSC.
- E. Braunwald, The war against heart failure: the lancet lecture, Lancet, 2015, 385, 812–824 CrossRef PubMed.
- A. Khan, A. Lahmar, M. Riasat, M. Ehtesham, H. Asif, W. Khan, M. Haseeb and H. Boricha, Myocardial infarction with non-obstructive coronary arteries: an updated overview of pathophysiology, diagnosis, and management, Cureus, 2022, 14, e23602 Search PubMed.
- K. Liang, E. Nakou, M. G. Del Buono, R. A. Montone, D. D'Amario and C. Bucciarelli-Ducci, The role of cardiac magnetic resonance in myocardial infarction and non-obstructive coronary arteries, Front. Cardiovasc. Med., 2022, 8, 821067 CrossRef PubMed.
- Y. L. Wu, Cardiac MRI assessment of mouse myocardial infarction and regeneration, Methods Mol. Biol., 2021, 2158, 81–106 CrossRef CAS PubMed.
- C. L. Wolfe, M. E. Moseley, M. G. Wikstrom, R. E. Sievers, M. F. Wendland, J. W. Dupon, W. E. Finkbeiner, M. J. Lipton, W. W. Parmley and R. C. Brasch, Assessment of myocardial salvage after ischemia and reperfusion using magnetic-resonance imaging and spectroscopy, Circulation, 1989, 80, 969–982 CrossRef CAS PubMed.
- L. Feng, Z. Yang, S. Chen and J. Wan, Diagnostic value of myocardial stress detection based on feature tracking MRI in patients with acute myocardial infarction, J. Thorac. Dis., 2022, 14, 3454–3461 CrossRef PubMed.
- F. Wang, L. Wen, J. Liu, W. Peng, Z. Meng, Q. Chen, Y. Wang, B. Ke, Y. Guo and P. Mi, Albumin nanocomposites with MnO2/Gd2O3 motifs for precise MR imaging of acute myocardial infarction in rabbit models, Biomaterials, 2020, 230, 119614 CrossRef CAS PubMed.
- L. L. Feng, Y. X. Wu, D. L. Zhang, X. X. Hu, J. Zhang, P. Wang, Z. L. Song, X. B. Zhang and W. H. Tan, Near infrared graphene quantum dots-based two-photon nanoprobe for direct bioimaging of endogenous ascorbic acid in living cells, Anal. Chem., 2017, 89, 4077–4084 CrossRef CAS PubMed.
- X. H. Sun, F. Guo, Q. Y. Ye, J. F. Zhou, J. Han and R. Guo, Fluorescent sensing of glutathione and related bio-applications, Biosensors, 2023, 13, 16 CrossRef CAS PubMed.
- T. Liu, L. Sun, Y. B. Zhang, Y. L. Wang and J. Zheng, Imbalanced GSH/ROS and sequential cell death, J. Biochem. Mol. Toxicol., 2022, 36, e22942 CrossRef CAS PubMed.
- Y. Yuan, J. Yang, A. X. Zhuge, L. J. Li and S. Ni, Gut microbiota modulates osteoclast glutathione synthesis and mitochondrial biogenesis in mice subjected to ovariectomy, Cell Proliferation, 2022, 55, e13194 CAS.
- F. Bottino, M. Lucignani, A. Napolitano, F. Dellepiane, E. Visconti, M. Espagnet and L. Pasquini, In vivo brain GSH: MRS methods and clinical applications, Antioxidants, 2021, 10, 1407 CrossRef CAS PubMed.
- O. G. Hamadama, M. Javeres, N. Nyemb, M. M. Fabrice and P. Elsa, Effect of azadirachta indica and senna siamea decoction on CD4+ and CD8+ level, toxicological, and antioxidant profile in HIV/AIDS positive persons, J. Toxicol., 2021, 2021, 5594505 Search PubMed.
- M. Haddad, V. Herve, M. R. Ben Khedher, J. M. Rabanel and C. Ramassamy, Glutathione: an old and small molecule with great functions and new applications in the brain and in Alzheimer's disease, Antioxid. Redox Signal., 2021, 35, 270–292 CrossRef CAS PubMed.
- Y. X. Xiong, C. Xiao, Z. F. Li and X. L. Yang, Engineering nanomedicine for glutathione depletion-augmented cancer therapy, Chem. Soc. Rev., 2021, 50, 6013–6041 RSC.
- H. H. Fan, G. B. Yan, Z. L. Zhao, X. X. Hu, W. H. Zhang, H. Liu, X. Y. Fu, T. Fu, X. B. Zhang and W. H. Tan, A smart photosensitizer-manganese dioxide nanosystem for enhanced photodynamic therapy by reducing glutathione levels in cancer cells, Angew. Chem., Int. Ed., 2016, 55, 5477–5482 CrossRef CAS PubMed.
- A. Qaitoon, J. X. Yong, Z. X. Zhang, J. Liu, Z. P. Xu and R. Zhang, Development of manganese dioxide-based nanoprobes for fluorescence detection and imaging of glutathione, New J. Chem., 2021, 45, 12377–12383 RSC.
- H. Yao, D. Jiang, G. Dong, J. Sun, S. Sun, L. Li, F. Zheng and W. Xiong, Near infrared imaging of intracellular GSH by AuNCs@MnO2 core-shell nanoparticles based on the absorption competition mechanism, Analyst, 2021, 146, 5115–5123 RSC.
- Y. L. Du, H. Liu, J. M. Liang, D. Y. Zheng, J. Li, S. B. Lan, M. Wu, A. X. Zheng and X. L. Liu, Protein-assisted formation of gold clusters-MnO2 nanocomposite for fluorescence imaging of intracellular glutathione, Talanta, 2020, 209, 120524 CrossRef CAS PubMed.
- H. J. Zhang, W. W. Zeng, C. Pan, L. W. Feng, M. T. Ou, X. W. Zeng, X. Liang, M. Y. Wu, X. Y. Ji and L. Mei, SnTe@MnO2-sp nanosheet-based intelligent nanoplatform for second near-infrared light-mediated cancer theranostics, Adv. Funct. Mater., 2019, 29, 1903791 CrossRef.
- T. He, C. Jiang, J. He, Y. F. Zhang, G. He, J. Wu, J. Lin, X. Zhou and P. Huang, Manganese-dioxide-coating-instructed plasmonic modulation of gold nanorods for activatable duplex-imaging-guided NIR-ll photothermal-chemodynamic therapy, Adv. Mater., 2021, 33, e2008540 CrossRef PubMed.
- Z. L. Song, X. Dai, M. R. Li, H. Teng, Z. Song, D. X. Xie and X. L. Luo, Biodegradable nanoprobe based on MnO2 nanoflowers and graphene quantum dots for near infrared fluorescence imaging of glutathione in living cells, Microchim. Acta, 2018, 185, 485 CrossRef PubMed.
- C. L. Yang, Y. Z. Liu, S. Su, N. Gao, J. Jing and X. L. Zhang, A multifunctional oxygen-producing MnO2-based nanoplatform for tumor microenvironment-activated imaging and combination therapy in vitro, J. Mater. Chem. B, 2020, 8, 9943–9950 RSC.
- D. D. Yuan, L. R. Ding, Z. M. Sun and X. M. Li, MRI/fluorescence bimodal amplification system for cellular GSH detection and tumor cell imaging based on manganese dioxide nanosheet, Sci. Rep., 2018, 8, 1747 CrossRef PubMed.
- Y. Wu, D. Li, F. Zhou, H. Liang, Y. Liu, W. J. Hou, Q. Yuan, X. B. Zhang and W. H. Tan, Versatile in situ synthesis of MnO2 nanolayers on upconversion nanoparticles and their application inactivatable fluorescence and MRI imaging, Chem. Sci., 2018, 9, 5427–5434 RSC.
- H. H. Fan, Z. L. Zhao, G. B. Yan, X. B. Zhang, C. Yang, H. M. Meng, Z. Chen, H. Liu and W. H. Tan, A smart dnazyme-MnO2 nanosystem for efficient gene silencing, Angew. Chem., Int. Ed., 2015, 54, 4801–4805 CrossRef CAS PubMed.
- H. Wang, Y. Liu, C. Xu, X. Wang, G. R. Chen, T. D. James, Y. Zang, J. Li, X. Ma and X. P. He, Supramolecular glyco-poly-cyclodextrin functionalized thin-layer manganese dioxide for targeted stimulus-responsive bioimaging, Chem. Commun., 2018, 54, 4037–4040 RSC.
- A. Menon, N. Abd-Aziz, K. Khalid, C. L. Poh and R. Naidu, MiRNA: a promising therapeutic target in cancer, Int. J. Mol. Sci., 2022, 23, 11502 CrossRef CAS PubMed.
- A. N. Saiyed, A. R. Vasavada and S. Johar, Recent trends in miRNA therapeutics and the application of plant miRNA for prevention and treatment of human diseases, Future J. Pharm. Sci., 2022, 8, 24 CrossRef PubMed.
- M. Hill and N. Tran, MiRNA interplay: mechanisms and consequences in cancer, Dis. Models Mech., 2021, 14, 047662 Search PubMed.
- Y. Zhang, C. M. Ma, C. Ma, Y. C. Xiang, S. Mu, Z. X. Zheng, X. Y. Liu and H. X. Zhang, Ratiometric fluorescent detection and imaging of microRNA in living cells with manganese dioxide nanosheet-active DNAzyme, Talanta, 2021, 233, 122518 CrossRef CAS PubMed.
- Y. Zeng, T. Dou, L. Ma and J. Ma, Biomedical photoacoustic imaging for molecular detection and disease diagnosis: “always-on” and “turn-on” probes, Adv. Sci., 2022, 9, e2202384 CrossRef PubMed.
- X. G. Dai, X. Y. Zhao, Y. J. Liu, B. B. Chen, X. K. Ding, N. N. Zhao and F. J. Xu, Controlled synthesis and surface engineering of janus chitosan-gold nanoparticles for photoacoustic imaging-guided synergistic gene/photothermal therapy, Small, 2021, 17, 14 Search PubMed.
- L. Xin, J. F. Gao, X. H. Zhao, W. W. Cai, C. Y. Guo and R. P. Zhang, Facile fabrication of biocompatible metal-phenolic based nanoplatform for magnetic resonance-photoacoustic dual-mode imaging, Mater. Lett., 2021, 287, 129259 CrossRef CAS.
- S. Chen and C. Tian, Review recent developments in photoacoustic imaging and sensing for nondestructive testing and evaluation, Vis. Comput. Ind. Biomed. Art., 2021, 4, 6 CrossRef PubMed.
- M. Zhou, X. Y. Wang, S. C. Lin, Y. F. Liu, J. S. Lin, B. Jiang, X. Z. Zhao and H. Wei, Combining photothermal therapy-induced immunogenic cell death and hypoxia relief-benefited M1-phenotype macrophage polarization for cancer immunotherapy, Adv. Ther., 2021, 4, 2000191 CrossRef CAS.
- J. Jung, Y. Jang, M. Kim and H. Kim, Types/applications of photoacoustic contrast agents: a review, Photonics, 2021, 8, 287 CrossRef CAS.
- P. Liu, Y. R. Wang, L. An, Q. W. Tian, J. M. Lin and S. P. Yang, Ultrasmall WO3−x@gamma-poly-L-glutamic acid nanoparticles as a photoacoustic imaging and effective photothermal-enhanced chemodynamic therapy agent for cancer, ACS Appl. Mater. Interfaces, 2018, 10, 38833–38844 CrossRef CAS PubMed.
- C. Pohling, J. L. Campbell, T. A. Larson, D. Van de Sompel, J. Levi, M. H. Bachmann, S. E. Bohndiek, J. V. Jokerst and S. S. Gambhir, Smart-dust-nanorice for enhancement of endogenous raman signal, contrast in photoacoustic imaging, and T2-shortening in magnetic resonance imaging, Small, 2018, 14, e1703683 CrossRef PubMed.
- C. X. Yang, G. Lin, C. Q. Zhu, X. Pang, Y. Zhang, X. Y. Wang, X. L. Li, B. Wang, H. P. Xia and G. Liu, Metalla-aromatic loaded magnetic nanoparticles for MRI/photoacoustic imaging-guided cancer phototherapy, J. Mater. Chem. B, 2018, 6, 2528–2535 RSC.
- V. Gujrati, A. Mishra and V. Ntziachristos, Molecular imaging probes for multi-spectral optoacoustic tomography, Chem. Commun., 2017, 53, 4653–4672 RSC.
- K. Y. Pu, A. J. Shuhendler, J. V. Jokerst, J. G. Mei, S. S. Gambhir, Z. N. Bao and J. H. Rao, Semiconducting polymer nanoparticles as photoacoustic molecular imaging probes in living mice, Nat. Nanotechnol., 2014, 9, 233–239 CrossRef CAS PubMed.
- M. Kataoka, M. Iima, K. K. Miyake and Y. Matsumoto, Multiparametric imaging of breast cancer: an update of current applications, Diagn. Interv. Imaging, 2022, 103, 574–583 CrossRef PubMed.
- R. F. Liu, L. J. Jing, D. Peng, Y. Li, J. Tian and Z. F. Dai, Manganese(II) chelate functionalized copper sulfide nanoparticles
for efficient magnetic resonance/photoacoustic dual-modal imaging guided photothermal therapy, Theranostics, 2015, 5, 1144–1153 CrossRef CAS PubMed.
- H. Wang, L. An, C. Tao, Z. Ling, J. Lin, Q. Tian and S. Yang, A smart theranostic platform for photoacoustic and magnetic resonance dual-imaging-guided photothermal-enhanced chemodynamic therapy, Nanoscale, 2020, 12, 5139–5150 RSC.
- X. J. Cai, W. Gao, M. Ma, M. Y. Wu, L. L. Zhang, Y. Y. Zheng, H. R. Chen and J. L. Shi, A prussian blue-based core-shell hollow-structured mesoporous nanoparticle as a smart theranostic agent with ultrahigh pH-responsive longitudinal relaxivity, Adv. Mater., 2015, 27, 6382 CrossRef CAS PubMed.
- D. Gu, Z. Liu, H. Wu, P. An, X. Zhi, Y. Yin, W. Liu and B. Sun, Dual catalytic cascaded nanoplatform for photo/chemodynamic/starvation synergistic therapy, Colloids Surf., B, 2021, 199, 111538 CrossRef CAS PubMed.
- Q. Xu, D. Y. Li, H. J. Zhou, B. Q. Chen, J. L. Wang, S. B. Wang, A. Z. Chen and N. N. Jiang, MnO2-coated porous Pt@CeO2 core-shell nanostructures for photoacoustic imaging-guided tri-modal cancer therapy, Nanoscale, 2021, 13, 16499–16508 RSC.
- A. M. Itoo, M. Paul, S. G. Padaga, B. Ghosh and S. Biswas, Nanotherapeutic intervention in photodynamic therapy for cancer, ACS Omega, 2022, 7, 45882–45909 CrossRef CAS PubMed.
- K. Rhew, Y. Chae and J. Chang, Progress and recent trends in photodynamic therapy with nanoparticles, J. Pharm. Invest., 2022, 52, 587–599 CrossRef.
- H. Correia, J. A. Rodrigues, S. Pimenta, T. Dong and Z. Yang, Photodynamic therapy review: principles, photosensitizers, applications, and future directions, Pharmaceutics, 2021, 13, 1332 CrossRef PubMed.
- H. Wang, M. P. Ewetse, C. Ma, W. Pu, B. Xu, P. He, Y. Wang, J. Zhu and H. Chen, The “light knife” for gastric cancer: photodynamic therapy, Pharmaceutics, 2022, 15, 101 CrossRef PubMed.
- C. Chen, C. Wu, J. Yu, X. Zhu, Y. Wu, J. Liu and Y. Zhang, Photodynamic-based combinatorial cancer therapy strategies: tuning the properties of nanoplatform according to oncotherapy needs, Coord. Chem. Rev., 2022, 461, e1703683 CrossRef.
- T. Kubrak, M. Karakula, M. Czop, A. Kawczyk-Krupka and D. Aebisher, Advances in management of bladder cancer-the role of photodynamic therapy, Molecules, 2022, 27, 731 CrossRef CAS PubMed.
- K. Wang, B. Yu and J. L. Pathak, An update in clinical utilization of photodynamic therapy for lung cancer, J. Cancer, 2021, 12, 1154–1160 CrossRef CAS PubMed.
- Y. Zhang, O. Eltayeb, Y. T. Meng, G. M. Zhang, Y. Zhang, S. M. Shuang and C. Dong, Tumor microenvironment responsive mesoporous silica nanoparticles for dual delivery of doxorubicin and chemodynamic therapy (CDT) agent, New J. Chem., 2020, 44, 2578–2586 RSC.
- Y. Yang, S. Shen, P. Wang and X. Wang, Efficacy of photodynamic therapy in actinic cheilitis: a systematic review, Photodiagn. Photodyn. Ther., 2022, 38, 102782 CrossRef CAS PubMed.
- Y. Sun, D. Zhao, G. Wang, Y. Wang, L. Cao, J. Sun, Q. Jiang and Z. He, Recent progress of hypoxia-modulated multifunctional nanomedicines to enhance photodynamic therapy: opportunities, challenges, and future development, Acta Pharm. Sin. B, 2020, 10, 1382–1396 CrossRef CAS PubMed.
- D. Thankachan, R. Anbazhagan, R. Krishnamoorthi, H. C. Tsai, H. T. Gebrie, H. F. Darge, C. Lu and J. Chen, MnO2 nanoparticle encapsulated in polyelectrolytic hybrids from alkyl functionalized carboxymethyl cellulose and azide functionalized gelatin to treat tumors by photodynamic therapy and photothermal therapy, J. Taiwan Inst. Chem. Eng., 2022, 139, 104503 CrossRef CAS.
- S. Liang, G. Liao, W. Zhu and L. Zhang, Manganese-based hollow nanoplatforms for MR imaging-guided cancer therapies, Biomater. Res., 2022, 26, 32 CrossRef CAS PubMed.
- B. Li, X. Wang, S. Hong, Q. Wang, L. Li, O. Eltayeb, C. Dong and S. Shuang, MnO2 nanosheets anchored with polypyrrole nanoparticles as a multifunctional platform for combined photothermal/photodynamic therapy of tumors, Food Funct., 2021, 12, 6334–6347 RSC.
- Q. Q. Sun, H. T. Bi, Z. Wang, C. X. Li, C. Wang, J. T. Xu, D. Yang, F. He, S. L. Gai and P. P. Yang, O2-generating metal-organic framework-based hydrophobic photosensitizer delivery system for enhanced photodynamic therapy, ACS Appl. Mater. Interfaces, 2019, 11, 36347–36358 CrossRef CAS PubMed.
- C. H. Ji, Z. Z. Lu, Y. H. Xu, B. B. Shen, S. Y. Yu and D. Shi, Self-production of oxygen system CaO2/MnO2@PDA-MB for the photodynamic therapy research and switch-control tumor cell imaging, J. Biomed. Mater. Res., Part B, 2018, 106, 2544–2552 CrossRef CAS PubMed.
- Y. Liu, C. S. Gong, L. S. Lin, Z. J. Zhou, Y. J. Liu, Z. Yang, Z. Y. Shen, G. C. Yu, Z. T. Wang, S. Wang, Y. Ma, W. P. Fan, L. C. He, G. Niu, Y. L. Dai and X. Y. Chen, Core-shell metal-organic frameworks with fluorescence switch to trigger an enhanced photodynamic therapy, Theranostics, 2019, 9, 2791–2799 CrossRef CAS PubMed.
- F. Cicone, G. Santo, C. Bodet-Milin, G. L. Cascini, F. Kraeber-Bodere, C. Stokke and A. Kolstad, Radioimmunotherapy of non-hodgkin b-cell lymphoma: an update, Semin. Nucl. Med., 2023, 53, 413–425 CrossRef PubMed.
- A. Rondon, J. Rouanet and F. Degoul, Radioimmunotherapy in oncology: overview of the last decade clinical trials, Cancers, 2021, 13, 5570 CrossRef CAS PubMed.
- L. L. Tian, Q. Chen, X. Yi, J. W. Chen, C. Liang, Y. Chao, K. Yang and Z. Liu, Albumin-templated manganese dioxide nanoparticles for enhanced radioisotope therapy, Small, 2017, 13, 1700640 CrossRef PubMed.
- P. J. An, Z. G. Gao, K. Sun, D. H. Gu, H. S. Wu, C. Q. You, Y. J. Li, K. W. Cheng, Y. Zhang, Z. F. Wang and B. W. Sun, Photothermal-enhanced inactivation of glutathione peroxidase for ferroptosis sensitized by an autophagy promotor, ACS Appl. Mater. Interfaces, 2019, 11, 42988–42997 CrossRef CAS PubMed.
- Y. D. Wang, S. Z. Song, T. Lu, Y. Cheng, Y. L. Song, S. Y. Wang, F. P. Tan, J. Li and N. Li, Oxygen-supplementing mesoporous polydopamine nanosponges with WS2 QDS-embedded for CT/MSOT/MR imaging and thermoradiotherapy of hypoxic cancer, Biomaterials, 2019, 220, 119405 CrossRef CAS PubMed.
- Y. Z. Liu, J. Jing, F. Jia, S. Su, Y. Tian, N. Gao, C. L. Yang, R. B. Zhang, W. Z. Wang and X. L. Zhang, Tumor microenvironment-responsive theranostic nanoplatform for in situ self-boosting combined phototherapy through intracellular reassembly, ACS Appl. Mater. Interfaces, 2020, 12, 6966–6977 CrossRef CAS PubMed.
- Z. Liu, S. J. Zhang, H. Lin, M. L. Zhao, H. L. Yao, L. L. Zhang, W. J. Peng and Y. Chen, Theranostic 2D
ultrathin MnO2 nanosheets with fast responsibility to endogenous tumor microenvironment and exogenous NIR irradiation, Biomaterials, 2018, 155, 54–63 CrossRef CAS PubMed.
- F. Yu, S. Wu and C. Chen, Predictors of long-term outcomes after catheter-directed thrombolysis combined with stent implantation in acute deep vein thrombosis secondary to iliac vein compression, Medicine, 2023, 102, e32646 CrossRef PubMed.
- S. C. Zhang, C. Y. Cao, X. Y. Lv, H. M. Dai, Z. H. Zhong, C. Liang, W. J. Wang, W. Huang, X. J. Song and X. C. Dong, A H2O2 self-sufficient nanoplatform with domino effects for thermal-responsive enhanced chemodynamic therapy, Chem. Sci., 2020, 11, 1926–1934 RSC.
- B. B. Ding, S. Shao, F. Jiang, P. P. Dang, C. Q. Sun, S. S. Huang, P. A. Ma, D. Y. Jin, A. A. Al Kheraif and J. Lin, MnO2-disguised upconversion hybrid nanocomposite: an ideal architecture for tumor microenvironment-triggered UCL/MR bioimaging and enhanced chemodynamic therapy, Chem. Mater., 2019, 31, 2651–2660 CrossRef CAS.
- X. Yi, L. Chen, J. Chen, D. Maiti, Z. F. Chai, Z. Liu and K. Yang, Biomimetic copper sulfide for chemo-radiotherapy: enhanced uptake and reduced efflux of nanoparticles for tumor cells under ionizing radiation, Adv. Funct. Mater., 2018, 28, 150–160 Search PubMed.
- W. W. Ying, Y. Zhang, W. Gao, X. J. Cai, G. Wang, X. F. Wu, L. Chen, Z. Y. Meng, Y. Y. Zheng, B. Hu and X. F. Lin, Hollow magnetic nanocatalysts drive starvation-chemodynamic-hyperthermia synergistic therapy for tumor, ACS Nano, 2020, 14, 9662–9674 CrossRef CAS PubMed.
- D. L. Sheng, T. Z. Liu, L. M. Deng, L. Zhang, X. L. Li, J. Xu, L. Hao, P. Li, H. T. Ran, H. R. Chen and Z. G. Wang, Perfluorooctyl bromide & indocyanine green Co-loaded nanoliposomes for enhanced multimodal imaging-guided phototherapy, Biomaterials, 2018, 165, 1–13 CrossRef CAS PubMed.
- W. R. Sun, H. Yu, D. Q. Wang, Y. J. Li, B. C. Tian, S. Zhu, P. Y. Wang, S. Y. Xie and R. R. Wang, MnO2 nanoflowers as a multifunctional nano-platform for enhanced photothermal/photodynamic therapy and MR imaging, Biomater. Sci., 2021, 9, 3662–3674 RSC.
- T. H. Li, S. Q. Shi, X. S. Lu, L. L. Shi, S. S. Wei, H. Guo, X. Y. Zhang, H. Y. Zhang and G. Y. Sun, A versatile Bi2S3/MnO2 based nano-theranostic agent for triple-modal imaging guided photothermal/photodynamic synergistic therapy, Chin. J. Anal. Chem., 2021, 49, 19–27 Search PubMed.
- Q. C. Zhang, W. T. Wang, M. Zhang, F. Wu, T. Zheng, B. L. Sheng, Y. H. Liu, J. Shen, N. L. Zhou and Y. Sun, A theranostic nanocomposite with integrated black phosphorus nanosheet, Fe3O4@MnO2-doped upconversion nanoparticles and chlorin for simultaneous multimodal imaging, highly efficient photodynamic and photothermal therapy, Chem. Eng. J., 2020, 391, 123525 CrossRef CAS.
- Y. Yao, N. Li, X. Zhang, J. O. Machuki, D. Z. Yang, Y. Y. Yu, J. J. Li, D. Q. Tang, J. W. Tian and F. L. Gao, DNA-templated silver nanocluster/porphyrin/MnO2 platform for label-free intracellular Zn2+ imaging and fluorescence-/magnetic resonance imaging-guided photodynamic therapy, ACS Appl. Mater. Interfaces, 2019, 11, 13991–14003 CrossRef CAS PubMed.
- Q. Y. Tang, Z. J. Cheng, N. Yang, Q. Z. Li, P. Wang, D. P. Chen, W. J. Wang, X. J. Song and X. C. Dong, Hydrangea-structured tumor microenvironment responsive degradable nanoplatform for hypoxic tumor multimodal imaging and therapy, Biomaterials, 2019, 205, 1–10 CrossRef CAS PubMed.
- A. B. Bowman, G. F. Kwakye, E. H. Hernandez and M. Aschner, Role of manganese in neurodegenerative diseases, J. Trace Elem. Med. Biol., 2011, 25, 191–203 CrossRef CAS PubMed.
- K. Du, M. Y. Liu, Y. Z. Pan, X. Zhong and M. J. Wei, Association of circulating manganese levels with Parkinson's disease: a meta-analysis, Neurosci. Lett., 2018, 665, 92–98 CrossRef CAS PubMed.
- K. Du, M. Y. Liu, Y. Z. Pan, X. Zhong and M. J. Wei, Association of serum manganese levels with Alzheimer's disease and mild cognitive impairment: a systematic review and meta-analysis, Nutrients, 2017, 9, 231 CrossRef PubMed.
Footnote |
† Both authors contributed equally. |
|
This journal is © the Partner Organisations 2023 |
Click here to see how this site uses Cookies. View our privacy policy here.