Amir
Kashtiaray
,
Mahdi
Karimi
,
Mostafa
Ghafori-Gorab
and
Ali
Maleki
*
Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology (IUST), Tehran 16846-13114, Iran. E-mail: maleki@iust.ac.ir
First published on 4th April 2025
Nanozymes have been developed as engineered nanomaterials that mimic the catalytic functions of natural enzymes. This review systematically evaluates the potential of nanozymes for detecting and treating breast cancer. The limitations of natural enzymes, which are associated with high cost, poor stability, and limited modifiability, are overcome by nanozymes through enhanced stability, lower expense, and tunable properties. Various nanozyme systems, including bimetallic catalysts, metal nanoclusters, MXene-based materials, metal–organic frameworks (MOFs), and carbon-based platforms, are examined. Advanced synthesis methods, such as hydrothermal, solvothermal, and biogenic approaches, are employed to produce nanozymes with well-defined structures and high catalytic activity. Therapeutic strategies are classified into catalytic therapy, sonodynamic therapy (ST), radiotherapy (RT), phototherapy, immunotherapy (IMT), and starvation therapy (ST), while diagnostic techniques are based on colorimetric, electrochemical, photothermal, and photoelectrochemical detection. The relationship between material composition and catalytic performance is analyzed, and challenges associated with drug resistance, tumor heterogeneity, and toxicity are addressed. It is demonstrated that nanozyme-based theranostic approaches are offered as promising alternatives to conventional treatments. Future clinical applications are expected to be improved by integrating these multifunctional platforms, and the need for safe, efficient, and cost-effective cancer treatment is emphasized. This study provides a clear basis for future clinical research.
Nanozymes have obtained significant attention in cancer treatment for their ability to target tumors’ unique pathological features. Cancer is fundamentally characterised by uncontrolled cell proliferation, evasion of apoptosis, and a distinct microenvironment marked by hypoxia, acidity, and elevated levels of reactive oxygen species (ROS).7 Nanozymes can be engineered to exploit these vulnerabilities by catalysing the localised production of cytotoxic species, such as hydroxyl radicals, which induce oxidative stress and trigger apoptosis in cancerous cells.8 Moreover, their intrinsic multifunctionality enables nanozymes to serve as both therapeutic agents and diagnostic tools—facilitating real-time imaging and monitoring of treatment efficacy. This theranostic approach enhances targeted drug delivery, minimises systemic side effects, and complements conventional therapies, paving the way for more precise and effective cancer interventions.9
Capitalizing on these versatile catalytic functions in cancer therapy, researchers have extended nanozyme applications to target the unique microenvironment and treatment challenges of particular cancers such as breast cancer.10,11 Breast cancer represents a diverse group of malignancies affecting the breast tissue and stands as one of the most prevalent cancers worldwide, especially among women.12,13 Its heterogeneity is evident in the distinct molecular subtypes—such as hormone receptor-positive, HER2-positive, and triple-negative breast cancers (TNBC) —each with unique biological behaviors and treatment responses.14,15 These differences influence prognosis and dictate the need for personalized therapeutic approaches. In recent years, innovative treatment strategies have emerged, including targeted therapies, immunotherapies, and advanced nanotechnology-based platforms. Among these, nanozyme-based interventions are showing promise by exploiting the unique tumor microenvironment (TME) of breast cancer. By harnessing the catalytic capabilities of nanozymes to generate ROS or enhance drug delivery, these systems offer a dual function—serving both as therapeutic agents and diagnostic tools—thereby paving the way for more precise and effective management of breast cancer.16,17
Building on these advances, a broad spectrum of nanozyme strategies has emerged to tackle breast cancer treatment and detection. Researchers have harnessed bimetallic nanozymes, metal nanoclusters, MXene-based constructs, MOF-based systems, and carbon-based platforms, each offering unique enzyme-like activities—such as peroxidase (POD)-like, oxidase (OXD)-like, catalase (CAT)-like, and Fenton-like reactions—to amplify ROS generation for effective tumor ablation.18–20 These mechanisms enhance catalytic therapy via chemodynamic (CDT), photothermal (PTT), and photodynamic (PDT) modalities and extend to sonodynamic (SDT), radiotherapy-enhancing (RT), and starvation (ST) strategies that target tumor metabolism and microenvironment vulnerabilities.21 In parallel, innovative detection approaches employing colorimetric, electrochemical, and photothermal sensors have been developed for sensitive and specific identification of breast cancer biomarkers—including HER2, MUC1, and miRNA-21—thereby integrating therapeutic and diagnostic (theranostic) functionalities.22,23 This multifaceted nanozyme-based paradigm is set to revolutionize personalized breast cancer management by combining targeted treatment with real-time therapeutic efficacy monitoring.
This review is the first comprehensive work that bridges the gap between nanozyme-based detection and treatment strategies for breast cancer, offering an unparalleled synthesis of knowledge that spans the full spectrum of innovative catalytic nanotechnologies. By systematically analyzing diverse nanozyme platforms this article reveals how these multifaceted tools harness enzyme-like activities to not only ablate tumors through enhanced ROS generation and metabolic disruption but also to provide ultra-sensitive, real-time detection of key biomarkers such as HER2, MUC1, and miRNA-21. Readers will discover detailed discussions on the evolution of synthesis methodologies, structural designs, and multi-modal therapeutic approaches—ranging from catalytic, PTT, and SDT to advanced radiotherapy and starvation techniques—that address the complex challenges posed by the breast TME. Moreover, the review meticulously highlights all emerging trends in each application section, including innovations in material composition, synthesis precision, and integration of diagnostic and therapeutic functionalities, thereby mapping the transformative potential of nanozyme-based theranostics in personalized cancer care. Explore the subsequent sections, where every aspect of this cutting-edge field is examined in depth, setting the stage for future breakthroughs and redefining the paradigm of breast cancer management.
![]() | ||
Fig. 1 TEM image of (a) PdPt3 NPs31 [reproduced from ref. 31 with permission from Elsevier, copyright 2022]. SEM images of (b) Ag79Au21/GO33 [reproduced from ref. 33 with permission from American Chemical Society, copyright 2021], (c) Cu1.5Mn1.5O4 CFNSs28 [reproduced from ref. 28 with permission from Elsevier, copyright 2022]. TEM image of (d) Pd@Ir NSs29 [reproduced from ref. 29 with permission from American Chemical Society, copyright 2022], and (e) FeCu–GOx PNzyme35 [reproduced from ref. 35 with permission from Elsevier, copyright 2022], (f) AgPd@BSA/DOX37 [reproduced from ref. 37 with permission from Elsevier, copyright 2020]. In this series of images, you can observe different morphologies with varying metals that directly contribute to their catalytic capabilities. |
The development of nanozyme-based therapies for breast cancer has significantly evolved in recent years, driven by structural design and synthesis techniques advancements. Early nanozymes, such as boron oxynitride (BON), were synthesized through high-temperature pyrolysis, providing POD-like activity for generating ROS (Fig. 2(d)–(f)).67 Over time, more sophisticated structures have emerged, incorporating multiple functionalities to enhance therapeutic effects. Hydrothermal and solvothermal methods have enabled the creation of advanced nanozymes, such as cerium oxide (CeO2) nanoparticles and carbon-encapsulated magnetite nano-doughnuts (CEMNDs), which exhibit both POD-like and SOD-like activities (Fig. 2(a)–(c)).68,69 This progression reflects a shift from single-function systems to multi-functional, synergistic designs that can address the complex needs of breast cancer therapy.
![]() | ||
Fig. 2 (a)–(c) SEM images of CeO2/Cu–W, CeO2/Cu–C, and CeO2–C are examples of various types of morphologies used for catalytic therapy68 [reproduced from ref. 68 with permission from John Wiley and Sons, copyright 2023]. TEM images of BON (d), BON1000 (e), and BON1400 which demonstrate that physical treatment can alter the characteristics of the nanozyme (f)67 [reproduced from ref. 67 with permission from John Wiley and Sons, copyright 2021]. (g)–(k) Fluorescence imaging, utilized for real-time monitoring, is extensively employed in cancer therapy and nanozyme technology70 [reproduced from ref. 70 with permission from John Wiley and Sons, copyright 2022]. (h) Diagram illustrating the mechanism by which Co-PN3 SA/CHO functions as a therapeutic agent. It depicts the catalytic therapy process and describes the synergistic effects of PTT71 [reproduced from ref. 71 with permission from John Wiley and Sons, copyright 2023]. (l)–(o) TEM images are used to examine the ultrastructure68 [reproduced from ref. 68 with permission from John Wiley and Sons, copyright 2023]. |
The mechanisms of action employed by these nanozymes center on their ability to catalyze critical reactions within the TME. A predominant approach involves POD-like activities, where nanozymes catalyze the decomposition of H2O2 into hydroxyl radicals, effectively inducing oxidative stress in cancer cells. Advanced nanozymes, such as 2D-CuPd nanosheets, combine OXD-like and POD-like activities to generate ROS, promoting apoptosis and cell cycle arrest in tamoxifen-resistant breast cancer cells.72 Additionally, CDT, a subtype of catalytic therapy, has gained prominence for using Fenton and Fenton-like reactions to enhance ROS generation. Nanozymes like MoS2-PEG and Cu3−xP@HNTs leverage these reactions, catalyzing H2O2 in acidic environments to maximize localized oxidative damage while minimizing systemic side effects.73,74
The targeted specificity of nanozymes in this category is achieved by engineering them to interact with specific substrates and cell types. Typical targets include molecules abundant in the TME, such as H2O2, glutathione (GSH), and cholesterol. For example, CEMNDs oxidize GSH, reducing its antioxidant protection in cancer cells, while Co–PN3 SA/CHO nanozymes deplete cholesterol, disrupting cellular integrity (Fig. 2(h)).69,71 These nanozymes have demonstrated efficacy in various breast cancer models, including murine 4T1 cells, human TNBC cells, and tamoxifen-resistant (MCF-7-TamR) lines, underscoring their adaptability to diverse cancer types.
In this category, nanozymes have also been integrated into various therapeutic strategies to enhance their clinical utility. Catalytic therapy remains the cornerstone, utilizing ROS production to target tumor cells selectively. CDT completes this by amplifying oxidative damage through Fenton-like reactions. Phototherapy, including PTT and PDT, has further augmented the therapeutic arsenal. Systems like CD47@CCM-Lap-CuS NPs combine PTT and CDT, achieving synergistic effects that improve therapeutic outcomes.71 Additionally, immunotherapy applications, such as IR780@BSA@SPIO, demonstrate the potential of nanozymes to stimulate immune responses in traditionally resistant “immune-cold” breast cancers.75
Beyond therapy, nanozymes in this category are being explored for their diagnostic capabilities, offering dual roles in treatment and detection. As seen with aptamer-modified gold nanoparticles (pA-AuNPs), colorimetric detection methods enable sensitive visual diagnostics alongside therapeutic applications.76 Photoelectrochemical systems, like MS-ICG@MnO2, provide imaging-guided therapeutic capabilities, bridging the gap between diagnostics and treatment.77 Self-powered electrosensitive detection devices, such as triboelectric nano-generators, integrate diagnostic and therapeutic functionalities into a single wearable system, representing a step forward in personalized cancer care (Fig. 2(g)–(k)).70
The collective progress in catalytic therapy by nanozyme has significantly impacted breast cancer treatment by addressing key challenges, including drug resistance, tumor heterogeneity, and systemic toxicity (Fig. 2(i)–(o)).68 The ability of nanozymes to leverage the unique properties of the TME, such as acidic pH and elevated ROS levels, allows for precise, localized action with minimal off-target effects. Moreover, their integration into multi-modal therapies enhances the overall efficacy of treatment, leading to reduced tumor growth, suppression of metastasis, and improved survival rates. As the field progresses, nanozymes are poised to be increasingly central in breast cancer's precise and effective management. Table 1 provides a review of recent studies on the catalytic therapy of breast cancer using nanozymes.
Nanozyme | Synthesis method | Activity | Substrate | Cell type | Therapeutic approach | Ref. |
---|---|---|---|---|---|---|
BON | High-temperature pyrolysis | POD-like | H2O2 | 4T1 | Catalytic | 67 |
CeO2/Cu | Hydrothermal | SOD-like, POD-like | ROS | MDA-MB-231 | Catalytic | 68 |
pA-AuNPs | Co-precipitation | POD-like, OXD-like | Dopamine, glucose | MDA-MB-231 | Catalytic | 76 |
COF-CNT | Hydrothermal | POD-like | H2O2 | 4T1 | Catalytic | 70 |
2D-CuPd nanosheets | Co-precipitation | OXD-like, POD-like | H2O2 | MCF-7-TamR | Catalytic | 72 |
CD47@CCM-Lap-CuS NPs | Template sacrificial method | POD-like | TMB, H2O2 | 4T1 | PTT, CDT | 78 |
PEG-RLS/Fe@CDs | Solvothermal | POD-like | TMB, H2O2, methylene blue (MB) | 4T1 | CDT, PTT, photothermal detection | 79 |
CEMNDs | Solvothermal | POD-like, GSH-OXD-like | H2O2, GSH | 4T1 | CDT, PTT | 69 |
MS-ICG@MnO2@PEG | Co-precipitation | POD-like, CAT-like | H2O2, GSH | 4T1 | CDT, PDT | 77 |
Cu3−xP@HNTs | Co-precipitation | POD-like | H2O2 | 4T1 | CDT, PDT | 74 |
MoS2-PEG | Hydrothermal | POD-like | H2O2 | 4T1, GL261 | Catalytic | 73 |
Co–PN3 SA/CHO | Solvothermal | OXD-like, CAT-like, Fenton-like | O2, H2O2, cholesterol | 4T1, Hepa 1–6 | Catalytic, PTT | 71 |
Supramolecular magnetonanohybrids | Co-precipitation | POD-like, CAT-like | ROS | TNBC | Catalytic, hyperthermia treatment, chemotherapy | 80 |
IR780@BSA@SPIO | Self-assembly | POD-like | H2O2 | 4T1 | CDT, PTT, MRI, fluorescence Imaging | 75 |
The use of nanozymes in breast cancer IMT has evolved significantly, marked by advancements in structural complexity, synthesis methods, and therapeutic strategies. Early studies focused on relatively simple designs, such as the Zr–CeO nanozyme, synthesized via co-precipitation to scavenge ROS and improve oxygenation in TME. These nanozymes demonstrated SOD-like and CAT-like activities, enabling the conversion of superoxide anion and hydrogen peroxide into oxygen and water. By reducing ROS levels, they reprogrammed immunosuppressive cells, such as myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs), enhancing the effectiveness of PD-1 blockade therapy. This foundational work laid the groundwork for more intricate and targeted designs.83,84
Building on these initial successes, subsequent research introduced structurally sophisticated nanozymes with dual or multifunctional catalytic capabilities. For instance, the trilobal PtNi structures of PPTNS nanozymes, synthesized through organic solution-heat injection, demonstrated enhanced ROS generation and magnetocaloric oscillation. These nanozymes activated the caspase-1-NLRP3 pathway, inducing pyroptosis—a form of programmed cell death critical for stimulating cytokine recruitment and anti-tumor immune responses. The transition from basic ROS scavenging to advanced ROS-driven cell death highlights the growing complexity of nanozyme mechanisms.85
The evolution of synthesis methods also contributed to increased functionality. As seen in CuCH-NCs, biomineralization techniques enabled the development of pH-sensitive, tumor-targeting nanozymes capable of catalyzing hydrogen peroxide into hydroxyl radicals. These structures, enclosed in albumin nanocages, combined hemodynamic and chemotherapy for TNBC, a challenging subtype of breast cancer. The specificity and efficacy of these designs underscored the importance of tailored synthesis methods in advancing nanozyme applications.86
A turning point in nanozyme development came with the integration of dual-modality therapies. For example, PFB nanozymes, designed for cold exposure (CE) therapy, combined glucose starvation with ROS generation. Their platelet membrane biomimetic coating ensured targeted delivery to cancer cells, while the CE treatment synergized with nanozyme activity to reduce intracellular glucose and ATP levels. This innovation enhanced ROS cytotoxicity and boosted immune responses, inhibiting tumor growth and metastasis. The ability to combine metabolic and immunotherapeutic mechanisms marked a significant advancement.87
Recent advancements, including the FeCu-DA and MDPH nanozymes, have refined therapeutic strategies by addressing tumor heterogeneity and cancer stem cells (CSCs). The FeCu-DA nanozyme, synthesized through pyrolysis, possesses dual-atom catalytic sites that enhance POD-like and CAT-like activities. Its ability to oxidize GSH and sustain hydroxyl radical generation made it particularly effective for inducing immunogenic cell death (ICD).88 Similarly, MDPH nanozymes, designed to target CSCs, combined ferroptosis induction with immune checkpoint blockade, effectively suppressing tumor recurrence and metastasis. These advanced designs demonstrate how nanozymes can tackle complex challenges within the TME.89
The continuous evolution of nanozyme technology has significantly impacted breast cancer therapy by improving treatment precision, reducing off-target effects, and enhancing therapeutic outcomes. Innovations in biocompatibility, such as citrate coatings and platelet biomimicry, have reduced toxicity while ensuring effective delivery to tumor sites. By targeting key pathways like ROS regulation, metabolic disruption, and immune activation, nanozymes have achieved remarkable success in reshaping the TME and boosting the efficacy of immunotherapies.
In conclusion, the progression of nanozyme-based therapies reflects a concerted effort to address the multifaceted nature of breast cancer. These advancements have transformed the landscape of cancer immunotherapy from simple ROS scavenging to sophisticated dual-modality approaches. As research continues, integrating novel mechanisms and targeted delivery strategies promises even more significant potential for nanozymes in combating breast cancer and other challenging diseases. Table 2 presents a summary of recent studies on breast cancer treatment using nanozymes via IMT.
Nanozyme | Synthesis method | Activity | Substrate | Cell type | Therapeutic approach | Ref. |
---|---|---|---|---|---|---|
Zr–CeO | Co-precipitation | SOD-like, CAT-like | ROS | MDSCs, TAMs | IMT | 83 |
PPTNS | Organic solution-heat injection method | POD-like, CAT-like | H2O2, TMB | HUVECs | IMT, catalytic | 85 |
CuCH-NCs | Biomineralization | POD-like | H2O2 | TNBCs | IMT, catalytic | 86 |
PFB | Adsorption-calcination strategy | POD-like | H2O2 | 4T1 | IMT, catalytic, ST | 87 |
MDPH | Co-precipitation | CAT-like | CSCs | 4T1 | IMT, catalytic | 89 |
FeCu-DA | Pyrolysis | POD-like, CAT-like, GSH-OXD-like | H2O2, GSH | 4T1 | IMT, photothermal detection, catalytic | 88 |
The structural evolution of nanozymes marks a notable trend, moving from simple cubic configurations to more intricate and multifunctional designs. Early systems like CaF2 nanozymes, synthesized via direct precipitation, demonstrated POD-like activity, decomposing H2O2 to ROS in acidic tumor microenvironments (TME) (Fig. 3(f)–(h)).91 These structures provided the foundation for SDT, offering minimal toxicity and straightforward synthesis. However, the field soon advanced to more sophisticated systems like LaFeO3 (LFO) nanocrystals, which incorporate glucose oxidase (GOx) to form cascade-reactive nanoreactors. These perovskite nanozymes, with multiple enzyme-mimicking properties, represented a leap forward, enhancing ROS production through the synergistic activation of oxidative stress pathways.92 Further innovations, such as 2D NiCoOx nanosheets and magnetic hydrogel nanozymes (MHZ), integrated multifunctionality, including GSH depletion, hyperthermia generation, and catalysis.93,94 These developments underscore a clear trend toward increasing structural complexity to address TME challenges.
![]() | ||
Fig. 3 (a) After the specified treatments, there were variations in the tumor volume of the mice. (n = 5), mean ± SD, which shows that using nanozyme diminished the tumor volume95 [reproduced from ref. 95 with permission from Elsevier, copyright 2023]. (b) CAT-like propertie of P-RuCu: O2 generation in a 1 mM H2O2 solution at various concentrations of P-RuCu at pH 6.596 [reproduced from ref. 96 with permission from Elsevier, copyright 2022]. (c) and (d) The illustration shows the structure and use of MHZ at a micro level, and it also shows the schematic mechanisms that are used in SDT. (c) Procedure for creating synthetic MHZ and its composition. (d) Scheme of the co-operative mechanism of MHZ on the generation of ROS and hyperthermia for cancer therapy94 [reproduced from ref. 94 with permission from American Chemical Society, copyright 2019]. (e) Representative tumor image after different treatments95 [reproduced from ref. 95 with permission from Elsevier, copyright 2023]. (f)–(h) Exploring therapeutic mechanisms on 4T1 cells through high-throughput transcriptome sequencing. (f) The volcano map reveals the US-triggered CaF2 + H2O2 group's downregulated or upregulated genes compared to the control group. (g) Examination of genes with varying expression levels, (h) the circular chart for analyzing GO and KEGG91 [reproduced from ref. 91 with permission from John Wiley and Sons, copyright 2022]. |
The evolution of synthesis methods mirrors this growing complexity. While earlier nanozymes relied on direct precipitation for simplicity, more recent systems utilize advanced techniques like hydrothermal synthesis and hard-template fabrication. For instance, the hydrothermal method of producing PB + Ce6 hydrogel systems enabled precise control over Prussian blue nanoparticles’ crystalline and catalytic properties.97 Similarly, the hard-template approach to fabricating NiCoOx nanosheets created highly organized 2D structures with diverse enzyme-like activities.93 These methodologies improved the catalytic efficiency of nanozymes and facilitated their integration into multimodal therapies. Ligand exchange processes in MHZ synthesis highlight the adaptability of modern techniques in incorporating functional elements for combined hyperthermia and SDT applications.94
The functional diversity of nanozymes has also significantly expanded, moving beyond essential ROS generation to include multiple enzyme-mimicking activities. Nanozymes like CaF2 primarily exhibit POD-like activity, catalyzing the breakdown of H2O2.91 However, systems like LFO and NiCoOx feature additional OXD, CAT, and GPx-like activities, enabling them to address oxidative stress, deplete antioxidants like GSH, and alleviate hypoxia in tumor cells. The integration of these capabilities enhances tumor cell death pathways, such as pyroptosis and apoptosis, as seen in LFO nanozymes, which activate the ROS-TXNIP-NLRP3-GSDMD mechanism.92 This multifunctionality has proven crucial in overcoming the complex defenses of TME, ensuring sustained ROS production and effective tumor suppression.
Another key development is the adaptation of substrates and therapeutic approaches. Most nanozymes target H2O2, leveraging its abundance in tumor tissues. However, modern systems have begun incorporating secondary substrates like glucose, as in MHZ, where glucose oxidase generates additional H2O2 to amplify ROS production.94 This adaptability extends to therapeutic strategies, where the combination of SDT with complementary modalities has gained prominence. Early nanozyme in this category focused solely on SDT, while recent advancements incorporate hyperthermia and PTT. For instance, MHZ nanozymes combine magnetic heating and catalytic activity, while PB + Ce6 hydrogel systems merge PTT with SDT to maximize tumor ablation.97 These synergistic approaches address tumor heterogeneity and improve treatment efficacy, as demonstrated by tumor inhibition rates of over 69% in 4T1 breast cancer models.
The impact of these innovations on breast cancer therapy is profound. In vivo studies reveal modern nanozymes achieve substantial tumor suppression with minimal side effects. For example, LFO@GOx and NiCoOx systems reduce tumor viability to as low as 12.4–12.6% in 4T1 cells.92 Furthermore, systems like MHZ and PB + Ce6 demonstrate better biocompatibility, ensuring that healthy tissues remain unaffected during treatment.97 These advancements highlight the potential of nanozymes to overcome key challenges in cancer therapy, such as tumor resistance, hypoxia, and off-target toxicity. Integrating complexity, multifunctionality, and synergy, nanozymes offer a transformative approach to breast cancer treatment, leading to more effective, personalized therapies.
In conclusion, the continuous advancements in nanozyme design, synthesis, and application underscore their critical role in enhancing SDT for breast cancer. The trend toward more complex, multifunctional, and adaptive systems reflects a growing understanding of TME and the need for targeted multimodal therapies. As research progresses, integrating nanozymes with other emerging technologies promises to refine further and revolutionize cancer treatment strategies. The most recent studies in this field are summarized in Table 3.
Nanozyme | Synthesis method | Activity | Substrate | Cell type | Therapeutic approach | Ref. |
---|---|---|---|---|---|---|
CaF2 | Direct precipitation | POD-like | H2O2 | 4T1, H22 | SDT, catalytic | 91 |
LFO | Hydrothermal | OXD-like, POD-like, GPx-like, CAT-like | ROS, H2O2, GSH, O2 | 4T1 | SDT, catalytic | 92 |
2D NiCoOx | Hard template | OXD-like, POD-like, GPx-like, CAT-like | H2O2, GSH, O2 | Mouse 4T1 | SDT, catalytic | 93 |
MHZ | Ligand exchange | POD-like | H2O2, glucose | Mouse 4T1 | SDT, hyperthermia | 94 |
PB + Ce6@Hy | Hydrothermal | CAT-like | H2O2 | 4T1 | SDT, PTT | 97 |
Beyond structural complexity, the synthesis methods of nanozymes have also evolved, moving from simple co-precipitation techniques, as seen in RuCu NPs and BSA@CeO/Fe2+, to multi-step functionalization approaches such as anchoring-pyrolysis (FeSAE@Au) and in situ polymerization (CuPy-Au@EM).96,101,102 These modifications enabled precise control over particle size, surface functionalization, and enzymatic activity. For example, the one-pot solvothermal synthesis of P-RuCu facilitated high stability and biocompatibility by incorporating polyethylene glycol (PEG).96 Similarly, the co-reduction method used in Au–Ag@HA NPs allowed for the simultaneous integration of multiple elements, enhancing their enzyme-mimetic activities and modulating ROS production.100 Such advancements in synthesis have improved the efficiency of nanozymes in modulating TME and sensitizing cancer cells to radiation.
The mechanism of action of nanozymes has evolved from single-enzyme mimics to multi-enzyme catalytic systems, significantly enhancing their therapeutic effects. Initially, nanozymes exhibited POD-like activity, as seen in FeSAE@Au, where glucose oxidation led to H2O2 accumulation, further catalyzed to generate ˙OH, inducing oxidative damage to cancer cells.95 More recently, multi-functional nanozymes, such as P-RuCu, have combined POD-like and CAT-like activities, enabling dual functionalities to enhance radiation-induced DNA damage (Fig. 3(b)).96 Similarly, BSA@CeO/Fe2+ exhibited a combination of CAT-like, SOD-like, and Fenton-like reactions, sequentially converting O2− into H2O2 and then into highly toxic ˙OH radicals. This cascade reaction mechanism intensified oxidative stress within tumors, overcoming radioresistance and increasing therapeutic efficacy.101
Substrate specificity has also expanded to optimize nanozyme-based RT. Early designs focused on simple oxidation reactions utilizing glucose and H2O2 (GOx), whereas recent nanozymes interact with more complex substrates, including GSH and tumor cell-derived exosomes. For example, CuPy-Au@EM, which mimicked tumor-derived exosomes, leveraged POD, GOx, and GSH oxidase (GSH-OXD) activities to generate ROS and disrupt redox homeostasis in cancer cells.102 This approach enhanced radiosensitization by depleting intracellular GSH, weakening the tumor's antioxidant defense mechanisms. Additionally, nanozymes such as SnFe2O4 incorporated GSH-OXD activity to exploit TME vulnerabilities further, reducing radioresistance by amplifying oxidative stress.103
The efficacy of nanozyme-based RT has been validated in preclinical models, particularly in murine breast cancer models using the 4T1 cell line, known for its aggressive and metastatic nature. Studies have demonstrated significant tumor suppression and improved survival outcomes. For instance, FeSAE@Au combined with RT resulted in a 4.5-fold reduction in tumor weight compared to PBS-treated controls.95 Similarly, BSA@CeO/Fe2+ achieved a tumor suppression rate of 83.07% in 4T1-bearing mice, demonstrating potent radiosensitization effects.101 These results highlight the effectiveness of nanozymes in enhancing radiation-induced cytotoxicity while minimizing damage to normal tissues. Furthermore, nanozymes such as CuP-based hydrogel and SnFe2O4 have introduced additional functionalities, including PTT, further enhancing therapeutic outcomes by inducing apoptosis and mitochondrial disruption.103,104
A key advantage of nanozyme-based RT is its ability to overcome significant limitations of conventional radiotherapy, such as tumor hypoxia, radioresistance, and systemic toxicity. By generating oxygen within the TME, nanozymes like P-RuCu and SnFe2O4 have alleviated hypoxia-induced resistance, leading to increased radiation efficacy. Additionally, the integration of high-Z elements such as gold (Au), ruthenium (Ru), and platinum (Pt) has enhanced X-ray energy deposition, amplifying radiation effects at lower doses. Moreover, hydrogel-based delivery systems, such as CuP-based hydrogel and SnFe2O4, have enabled localized and sustained nanozyme release, minimizing systemic toxicity and improving biocompatibility. These advancements have significantly improved the therapeutic window of RT while reducing side effects.96,103
In conclusion, the field of nanozyme-based RT for breast cancer has evolved into a sophisticated and highly effective therapeutic approach. Advancements in structural complexity, synthesis methodologies, enzymatic activity, and targeted delivery have significantly enhanced treatment precision and efficacy. Integrating multi-modal therapies, such as PTT, has further improved tumor eradication, making nanozymes a promising platform for future clinical applications. Research efforts should focus on optimizing nanozyme formulations for clinical translation, addressing biocompatibility concerns, long-term stability, and regulatory approval to ensure their successful implementation in breast cancer therapy. The most recent studies in this field are summarized in Table 4.
Nanozyme | Synthesis method | Activity | Substrate | Cell type | Therapeutic approach | Ref. |
---|---|---|---|---|---|---|
FeSAE@Au | Anchoring-pyrolysis | POD-like, GOx-like | Glucose, H2O2, TMB | 4T1 | RT, catalytic, colorimetric detection | 95 |
RuCu NPs | Co-precipitation | POD-like, CAT-like | H2O2, TMB | MDA-MB-231 | RT, catalytic, colorimetric detection | 96 |
Au–Ag@HA NPs | Co-reduction | POD-like, CAT-like | H2O2 | 4T1 | RT, catalytic | 100 |
SFO | Hydrothermal | GSH-OXD-like, CAT-like | GSH, H2O2 | 4T1 | RT, catalytic, PTT | 103 |
CuPy-Au@EM | In situ nucleation and chemical deposition | GOx-like, GPx-like, POD-like | Glucose, GSH, H2O2 | 4T1 | RT, catalytic, colorimetric detection | 102 |
BSA@CeO/Fe2+ | Co-precipitation | CAT-like, SOD-like, POD-like | H2O2, O2− | 4T1 | RT, catalytic | 101 |
CuP-based hydrogel | In situ chemical oxidative polymerization | POD-like, GSH-OXD-like | GSH, H2O2 | 4T1 | RT, catalytic, PTT | 104 |
Pt@Alg | Co-precipitation | POD-like | ROS | 4T1 | RT | 105 |
The increasing complexity of nanozymes has been closely linked to advancements in synthesis techniques. Initially, simple wet-chemical methods dominated the field, but recent studies have employed more precise strategies such as co-precipitation, hydrothermal, solvothermal, and thermal decomposition methods. For instance, hydrothermal synthesis was used in MoS2–bPEI–CeFe2O4 nanoflowers, yielding high photothermal conversion efficiency and Fenton-like catalytic activity, leading to 80% tumor cell destruction.106 Similarly, thermal decomposition facilitated the development of Ag2S@Fe2C nanozymes, ensuring monodispersity and enhanced tumor-homing functionality.107 These methodological refinements have significantly improved nanozyme stability, biocompatibility, and catalytic performance, making them more suitable for clinical applications.
Beyond synthesis improvements, nanozymes in this category have evolved from single-function catalytic agents to multi-enzyme mimetic platforms, incorporating POD, CAT-like, glucose oxidase, and SOD-like activities. While early nanozymes primarily mimicked POD-like behavior to catalyze H2O2 decomposition into ˙OH, recent designs have expanded their enzymatic repertoire to address tumor hypoxia and oxidative stress regulation. For example, the Sm-TCPP-Pt/TPP nanozyme has CAT-like activity, improving PDT efficacy and leveraging mitochondria-targeting ligands to enhance ROS accumulation precisely where it is most effective.108 This shift towards multi-functionality has directly translated into increased tumor apoptosis rates and reduced drug resistance, as demonstrated in PdRu-RCE@PCM, which incorporated both POD- and CAT-like functionalities, enhancing photothermal conversion and ROS-driven cytotoxicity.109
In parallel with enzymatic improvements, the variety of substrates utilized by these nanozymes has expanded, broadening their potential applications in breast cancer therapy. Hydrogen peroxide remains the primary substrate, as seen in Sm-TCPP-Pt/TPP, which leveraged H2O2 decomposition to alleviate hypoxia.108 However, recent developments have explored alternative substrates, such as glucose in MnZ@Au, which utilized GOx-like activity to disrupt cancer cell metabolism while simultaneously generating ROS.62 Additionally, some nanozymes, such as PPy@BSA–MnO2, have been designed to target intracellular GSH, further amplifying oxidative stress and enhancing CDT.110 The strategic diversification of substrates has played a crucial role in improving nanozyme selectivity and the ability to manipulate the TME to enhance therapeutic efficacy.
These advancements have been particularly beneficial in treating TNBC, an aggressive subtype with limited treatment options. Most studies in this collection focused on TNBC models, particularly 4T1 and MDA-MB-231 cell lines, due to their high metastatic potential and resistance to conventional treatments.111 Research utilizing HM/D-I-BL nanozymes demonstrated effective tumor oxygenation and enhanced PDT performance, achieving a remarkable 93.5% tumor inhibition rate in 4T1 tumor-bearing mice.112 Similarly, MoS2–bPEI–CeFe2O4 nanoflowers exhibited potent CDT and photothermal synergy, making them effective in MDA-MB-231 cells.106 Some nanozymes, such as PdRu-RCE@PCM, have even been tested across multiple cancer models, including HeLa and A549 cells, confirming their broad-spectrum applicability.109 These findings suggest that nanozymes can be tailored to different breast cancer subtypes, paving the way for more personalized treatment strategies.
One of the most promising trends in nanozyme-based phototherapy has been the integration of multiple treatment modalities, such as PTT, PDT, CDT, and chemotherapy. Several studies highlight the enhanced efficacy of these synergistic approaches (Fig. 4(f) and (g)).113–115 For instance, HMPB@Lip leveraged iron redox reactions alongside photothermal conversion, leading to a 92.2% tumor inhibition rate (Fig. 4(c)–(e)).116 Likewise, I/C@M used MnO2-mediated oxygenation to amplify ROS-dependent cytotoxicity, resulting in 86.3% tumor cell death (Fig. 4(h)).114 Furthermore, as demonstrated in PNC nanozymes, photothermal catalytic therapy achieved 45.06% photothermal conversion efficiency, further improving tumor destruction.117 By combining multiple mechanisms, these advanced nanozyme systems have led to higher treatment efficacy, minimized side effects, and enhanced tumor selectivity.
![]() | ||
Fig. 4 (a) Images of the tumors that have been dissected after undergoing treatments for 14 days. This image shows how nanozyme is effective. (b) The tumor sections were examined using hematoxylin and eosin staining for histological studies after a 14-day treatment. The dashed white lines indicate the presence of necrotic areas62 [reproduced from ref. 62 with permission from American Chemical Society, copyright 2023]. (c)–(e) TEM images of solid PB NCs, HMPB@Lip, and HMPB NCs116 [reproduced from ref. 116 with permission from Elsevier, copyright 2021]. (f) and (g) The diagram illustrates how nanoparticles are prepared and the nanozymes trigger cascade reactions. (f) Structures of the Lipo-OGzyme-AIE and OGzymes and. (g) The nanoparticles in tumors are thought to act by being encapsulated into liposomes, which allows for efficient tumor uptake of the OGzymes. Afterward, the OGzymes are believed to penetrate hypoxic tumor tissues and normoxic tumor areas. The OGzymes are thought to possess CAT-like activity, which enables them to generate oxygen intratumorally through a catalytic reaction that responds to TME, particularly in hypoxic conditions115 [reproduced from ref. 115 with permission from Elsevier, copyright 2020]. (h) Quantitative assessment of the necrotic area114 [reproduced from ref. 114 with permission from American Chemical Society, copyright 2023]. (i)–(m) HADDF-STEM and elemental mapping of BDS@MnOx. Scale bars = 100 nm118 [reproduced from ref. 118 with permission from American Chemical Society, copyright 2023]. |
As research continues to refine phototherapy by nanozyme technology, the focus has shifted toward clinical applicability. The structural evolution, advanced synthesis methodologies, multi-enzyme activity, expanded substrate interactions, and hybrid therapeutic strategies collectively represent a significant step toward real-world cancer treatment solutions. Moving forward, key areas of development include personalized nanozyme formulations for specific breast cancer subtypes, hypoxia-responsive and tumor-targeted nanozymes, and the enhancement of biocompatibility for safer human application. Given their capacity for precision-targeted, minimally invasive, and highly effective therapy, nanozymes hold immense promise in revolutionizing breast cancer treatment by phototherapy. If these advancements can be translated into clinical settings, nanozyme-based phototherapy could represent a new frontier in oncology, offering hope for patients with aggressive and treatment-resistant breast cancers. Table 5 summarizes the latest studies in this field.
Nanozyme | Synthesis method | Activity | Substrate | Cell type | Therapeutic approach | Ref. |
---|---|---|---|---|---|---|
MnZ@Au | Ion exchange and in situ reduction | GOx-like, CAT-like | Glucose, H2O2 | 4T1, MCF-7 | PDT, photoacoustic (PA) imaging | 62 |
Sm-TCPP-Pt/TPP | Solvothermal | CAT-like | H2O2 | MCF-7 | PDT, catalytic | 108 |
nGO-hemin-Ce6 | Co-encapsulation | CAT-like | H2O2 | MCF-7 | PDT, catalytic | 63 |
Ce6@HMPB NPs | Chemical etching | CAT-like | H2O2 | 4T1 | PTT, PDT, catalytic | 113 |
OGzyme | Biomimetic synthesis | CAT-like | H2O2 | 4T1 | PDT, catalytic | 115 |
PtCo@Gem-HA-PEG | Co-precipitation | CAT-like, POD-like | H2O2 | 4T1 | PDT, catalytic | 119 |
PdRu-RCE@PCM | Co-precipitation and thermal decomposition | POD-like, CAT-like | H2O2 | 4T1, A549, HeLa, 3T3 | PTT, PDT, catalytic | 109 |
Plasmonic Au NBP@Cu2O | Co-precipitation | POD-like | H2O2 | 4T1 | PTT, Catalytic | 120 |
Ag2S@Fe2C-DSPE-PEG-iRGD | Thermal decomposition | POD-like | ROS | 4T1 | PTT, catalytic, MRI, fluorescence imaging | 107 |
FeS-Dox@bLf NZs | Wet-chemical synthesis | POD-like | H2O2 | 4T1 | PDT, catalytic | 121 |
MoS2–bPEI–CeFe2O4 NFs | Hydrothermal | POD-like | H2O2, TMB | MDA-MB-231 | PTT, catalytic, colorimetric detection | 106 |
PNC | Thermal decomposition | POD-like | H2O2 | 4T1 | PTT, catalytic | 117 |
HMPB | Hydrothermal | POD-like | Unsaturated lipids | 4T1 | PTT, catalytic | 116 |
PPy@BSA–MnO2 | Chemical oxidation polymerization | GSH-OXD-like, POD-like | GSH, H2O2 | 4T1 murine | PTT, catalytic, MRI | 110 |
HM/D-I-BL | Co-precipitation, hydrothermal | GSH-OXD-like, POD-like | GSH, H2O2 | Mouse 4T1 | PDT, PTT, catalytic, MRI | 112 |
Fe–N–C SAzyme | Co-precipitation | POD-like | H2O2 | TNBC | PTT, catalytic | 111 |
I/C@M | Co-precipitation | GSH-OXD-like, POD-like, CAT-like | GSH, H2O2 | 4T1, MDA-MB-231 | PDT, PTT, catalytic | 114 |
AFH | Co-precipitation | GSH-OXD-like | GSH | 4T1 | PTT, catalytic | 122 |
The advancement of nanozyme-based ST in breast cancer has led to increasingly complex nanoplatforms with improved catalytic mechanisms and therapeutic efficacy. Initially, simple enzyme-mimicking nanoparticles were used to catalyze glucose oxidation, depriving tumor cells of nutrients. Over time, these systems evolved into multifunctional architectures incorporating synergistic treatments such as PTT, CDT, and IMT.127 Structural innovations, such as hierarchical core–shell architectures in Fe3O4@ZIF-8/GOx@MnO2, have enhanced catalytic efficiency, stability, and TME responsiveness.127 Additionally, advances in synthesis techniques, including thermal decomposition and wet-chemical methods, have enabled precise control over nanozyme properties, ensuring better biocompatibility and therapeutic effectiveness.128,129
A key development in nanozyme therapy has been integrating multiple catalytic activities to enhance tumor starvation and oxidative stress-induced apoptosis. While early nanozymes relied on GOx-like activity to deplete glucose and generate hydrogen peroxide,128 newer designs incorporate POD-like, OXD-like, and glutathione-oxidase (GSH-OXD)-like functions (Fig. 4(i)–(m)).118,129 These advanced nanozymes, such as Fe2O3/Au hybrids and BDS-GOx@MnOx, exploit metabolic vulnerabilities in cancer cells by generating ROS and disrupting antioxidant defenses. Furthermore, targeting specific tumor substrates, such as H2O2 and GSH, has expanded the effectiveness of nanozyme therapy beyond simple starvation, ensuring multi-pathway tumor destruction. The superior performance of these platforms has been demonstrated in aggressive breast cancer subtypes like TNBC, where nanozymes have achieved substantial tumor reduction compared to conventional therapies.126
The integration of combination therapies has further amplified the impact of nanozyme-based ST. These platforms have achieved superior tumor suppression and immune activation by merging catalytic reactions with PTT, CDT, and IMT.127 For instance, Fe2O3/Au hybrid nanozymes leverage mild PTT and ferroptosis induction, while Fe3O4@ZIF-8/GOx@MnO2 promotes macrophage polarization and immune modulation.127,129 These strategies enhance systemic anti-cancer immunity, as evidenced by increased M1 macrophage activation and reduced regulatory T cells (Tregs). Tumor inhibition data confirm the superior efficacy of nanozyme therapy, with platforms like BP@Au@MnO2-PEG achieving a 64.4% reduction in cancer cell viability. Moving forward, research will likely focus on tumor-targeted, biodegradable nanozymes with enhanced immune system activation, ensuring long-term suppression and minimal side effects.127 Nanozyme-based ST is thus emerging as a highly efficient, minimally invasive approach to overcoming aggressive breast cancer. The latest research in this field is summarized in Table 6.
Nanozyme | Synthesis method | Activity | Substrate | Cell type | Therapeutic approach | Ref. |
---|---|---|---|---|---|---|
BP@Au@MnO2-PEG | In situ reduction | CAT-like, GOx | H2O2, glucose | 4T1 | ST, catalytic, PDT, PTT | 127 |
IrRu-GOx@PEG | Thermal decomposition | CAT-like, GOx, POD-like | H2O2, glucose | 4T1, U87 | ST, catalytic | 128 |
Fe2O3/Au hybrid | Thermal decomposition | GOx, POD-like | H2O2, glucose | TNBC | ST, catalytic, PTT | 126 |
BDS-GOx@MnOx | Wet-chemical synthesis | GOx, GSH-OXD-like | Glucose, GSH | 4T1 | ST, CDT | 118 |
Fe3O4@ZIF-8/GOx@MnO2 | Solvothermal | OXD-like, CAT-like, GOx | H2O2, glucose | 4T1 | ST, catalytic, PTT | 129 |
Building on this foundation, recent advancements in nanozyme development have expanded the applications of colorimetric methods, particularly in the realm of breast cancer detection;132 The development of nanozymes for breast cancer detection through colorimetric methods has seen remarkable progress, marked by innovations in material composition, catalytic mechanisms, target specificity, and clinical applications.133 These advancements are pushing the boundaries of cancer diagnostics and laying the groundwork for sustainable and accessible healthcare technologies.
The synthesis methods for nanozymes demonstrate a trend toward eco-friendliness and precision. Liquid-phase exfoliation and ultrasonication are frequently employed to produce nanoscale materials with enhanced catalytic properties, as seen in FeOCl nanosheets.134 Hydrothermal synthesis, used for blood-derived nanoparticles (BDNPs) and cauliflower-derived carbon dots (CFCDs), emphasizes sustainability by utilizing natural precursors.135,136 Acid oxidation and rolling circle amplification, as applied in graphene quantum dot nanozymes (TMB-GQDzymes), highlight advanced functionalization techniques for hybrid systems.137 Additionally, chemical co-precipitation with subsequent surface modification (e.g., Fe3O4@MnO2) underscores integrating multi-functional capabilities (Fig. 5(f)–(i)).137 These approaches prioritize simplicity, biocompatibility, and adaptability for clinical translation.
![]() | ||
Fig. 5 (a) The selectivity of the electrochemical immunoassay138 [reproduced from ref. 138 with permission from Elsevier, copyright 2023]. (b) and (c) Graphs of electrochemical impedance spectra show Nyquist plots (b) and cyclic voltammograms (c) of bare GCE22 [reproduced from ref. 22 with permission from Elsevier, copyright 2023]. (d) and (e) Fe3O4@MnO2NPs grafted with anti-CD44 mAbs139 reproduced from [ref. 139 with permission from Elsevier, copyright 2023]. (f) and (g) SEM images and (h) and (i) TEM images of DFs and TMB-GQDzymes@DFs with 1 μm and 500 nm scale bars, respectively137 reproduced from [ref. 137 with permission from Elsevier, copyright 2023]. |
Expanding on these synthesis strategies, nanozyme development has diversified significantly in terms of structural design and material composition. The field now incorporates many nanostructures, ranging from simple metallic oxides to more complex hybrid and carbon-based materials. For instance, BDNPs synthesized from hemoglobin showcase a sustainable and biocompatible approach to material sourcing.135 Similarly, graphene quantum dots (GQDs) and CFCDs are emerging as promising carbon-based materials due to their strong surface chemistry, electron conductivity, and environmental friendliness.136,137 Hybrid structures, such as manganese dioxide-modified magnetite nanoparticles (Fe3O4@MnO2), integrate multiple functionalities, enabling simultaneous magnetic separation and catalytic activity.
These structural advancements facilitate the customization of nanozymes to meet specific diagnostic needs, such as enhancing stability, sensitivity, and selectivity under complex biological conditions. This adaptability makes nanozymes versatile tools in addressing the diverse challenges of cancer detection.
Complementing these advancements in structural design, the catalytic mechanisms employed by nanozymes have expanded beyond traditional POD-like activity. For instance, FeOCl nanosheets and BDNPs use H2O2 to generate ROS, whereas Fe3O4@MnO2 nanozymes utilize molecular oxygen, allowing catalytic reactions without relying on H2O2.134,135,139 Furthermore, CFCDs improve enzymatic efficiency through competitive activation, which differs from traditional Michaelis–Menten kinetics by enhancing substrate binding affinity.136 Hybrid systems like TMB-GQDzymes integrate catalytic oxidation with photothermal effects, introducing dual-modality detection capabilities.137
Complementing these advancements in structural design and catalytic mechanisms, nanozymes have demonstrated exceptional sensitivity in detecting clinically relevant biomarkers, which are crucial for early breast cancer diagnosis. For instance, FeOCl nanosheets achieve detection limits as low as ∼2.23–2.76 μM for biothiols such as glutathione (GSH) and cysteine (Cys). Tumor-derived exosomes, biomarkers critical for early cancer diagnosis, are targeted using dual-aptamer recognition strategies with LOD of 1027 particles per μL.134 Similarly, CFCD-enhanced assays reduce the detection threshold for alkaline phosphatase (ALP), a breast cancer marker, from 0.1 U mL−1 to 0.01 U mL−1, while Fe3O4@MnO2 nanozymes achieve cellular detection limits as low as 186 MDA-MB-231 cells.136
Leveraging the exceptional sensitivity and precision of nanozyme-based colorimetric diagnostics, integrating multimodal detection systems is a significant trend in the field. Nanozymes like TMB-GQDzymes exemplify hybrid platforms that combine colorimetric detection with photothermal effects under near-infrared (NIR) laser irradiation.137 This dual-mode approach enhances diagnostic reliability by providing orthogonal validation of results. Similarly, Fe3O4@MnO2 nanozymes enable magnetic separation of cancer cells from biological fluids, followed by colorimetric detection, showcasing their utility in handling complex sample matrices (Fig. 5(d) and (e)).139
The incorporation of hybrid systems reflects a shift toward strong diagnostic platforms that can operate in diverse clinical and research settings. These versatile systems improve the reliability of cancer diagnostics and broaden the scope of nanozyme applications to include multi-disease detection and real-time monitoring.
Alongside these advancements, the increasing focus on eco-friendly materials and scalable synthesis methods highlights the commitment to developing sustainable, cost-effective solutions that improve the accessibility and global impact of nanozyme technologies. Using eco-friendly materials and scalable synthesis methods is another emerging trend in nanozyme research. For example, BDNPs derived from hemoglobin and CFCDs synthesized from cauliflower leverage natural, renewable resources, aligning with green chemistry principles. Scalable methods such as hydrothermal synthesis further reduce production costs, making these materials viable for widespread use.135,136
While sustainability and scalability drive the global adoption of nanozyme technologies, ongoing research must address critical challenges in specificity, biocompatibility, and integration with cutting-edge diagnostic platforms to unlock their full potential. Despite these advancements, challenges remain in achieving absolute specificity and eliminating background interference. Strategies such as dual-aptamer designs and selective surface functionalization have shown promise but require further optimization. Future research should also focus on improving the biocompatibility of nanozymes to minimize cytotoxicity and immune responses in vivo. Additionally, integrating nanozyme-based platforms with wearable devices and multiplexed systems could revolutionize cancer diagnostics by enabling real-time and multi-biomarker detection.
Building on efforts to overcome existing challenges, the transformative potential of nanozymes in cancer detection—particularly in breast cancer—becomes apparent through their structural versatility and innovative applications in diagnostics. Nanozymes are emerging as a powerful tool for detecting breast cancer using colorimetric methods. Their structural diversity, multifunctional catalytic mechanisms, and clinical applicability indicate a field poised for significant impact. By addressing current challenges and focusing on sustainable, scalable designs, nanozyme research has the potential to redefine the landscape of cancer diagnostics, making early detection more accessible, reliable, and cost-effective. The ongoing convergence of nanotechnology, materials science, and clinical medicine will propel this promising field to new heights.
Nanozyme-based electrochemical detection methods have emerged as powerful tools for biomarker identification, offering exceptional sensitivity, LOD, and dynamic applicability in disease diagnosis, particularly cancer. These systems leverage the catalytic properties of nanozymes to catalyse key reactions like the decomposition of H2O2 into measurable electrochemical signals.142
Innovations in nanozyme synthesis have paralleled these advancements in detection, optimizing their catalytic performance for specific applications. Synthesis methods have evolved to create defect-rich, high-surface-area nanozymes tailored for specific applications. Hydrothermal and sol–gel methods are prevalent, as seen in FeMn-NC and FeNC nanozymes, where controlled doping with metals like Fe, Mn, or Cu optimizes catalytic performance (Fig. 5(a)).23,138 As in Mn3O4/Pd@Pt nanostructures and DNA-functionalized nanozymes, self-assembly techniques enable precise structural configurations, enhancing active site exposure and stability (Fig. 5(b) and (c)).22,143,144 Bioconjugation strategies, such as incorporating Cu2+ ions into BSA nanoparticles, exemplify efforts to enhance reactivity through surface functionalisation.145 Hybrid nanozymes, integrating multi-functional materials like MOFs and metallic nanoparticles, are becoming increasingly sophisticated. For instance, the MOF@Pt@MOF structure provides a synergistic catalytic framework combining conductivity, porosity, and enzyme-like activity.20
These advancements in nanozyme synthesis have directly influenced detection mechanisms, where innovative catalytic and signal amplification strategies are driving improvements in sensitivity, stability, and specificity. The detection mechanisms often center on POD-like activity, where nanozymes catalyse H2O2 decomposition, triggering substrate oxidation (e.g., TMB, dopamine) and producing detectable signals. This enzymatic mimicry, augmented by nanostructural engineering, enhances signal intensity and stability.146,147 Advanced systems incorporate signal amplification strategies, such as DNAzyme walker cleavage cycles and CRISPR/Cas12a-mediated amplification, significantly boosting detection thresholds.144,148 Tetrahedral DNA nanostructures and aptamer-functionalized interfaces enable selective target recognition, particularly for biomarkers like HER2 and MUC1, ensuring high specificity.143 As seen in binanozyme cytosensors, magnetic separation techniques enhance detection by seamlessly isolating CTCs under a magnetic field, combining enrichment and electrochemical analysis.149
Integrating these advanced detection mechanisms has enabled nanozyme systems to achieve remarkable sensitivity and specificity, with detection limits at fM levels and dynamic adaptability for clinical applications. The miRNA-21 ratiometric biosensor achieved a LOD of 0.16 fM, while HER2 and MUC1 biosensors exhibited LODs of 4.5 pg mL−1 and 0.085 pg mL−1, respectively.22,146 These ultra-sensitive detection capabilities are crucial for early diagnosis and monitoring of low-abundance biomarkers in complex biological samples. The dynamic detection ranges, spanning from fM to nM concentrations, highlight the adaptability of these systems across varying analyte levels, ensuring their reliability for clinical applications.
Building on their ultra-sensitive detection capabilities, integrating hybrid materials in nanozyme design has further enhanced performance, leveraging properties like conductivity, catalytic synergy, and multi-functionality. The integration of hybrid materials marks a significant trend in nanozyme design. Carbon-based nanozymes like FeNC and rGO/MoS2 are favored for their electrical conductivity and modifiable surfaces, while dual-metal nanozymes such as FeMn-NC and CH–Cu@J-Cu2O capitalize on synergistic effects for enhanced catalysis. Multifunctional hybrids like MOF@Pt@MOF and TCPP-Fe@HMUiO@Au-ABEI incorporate additional features such as signal amplification and structural flexibility, achieving superior performance compared to single-component systems.20,144,147,149
The growing focus on genetic biomarkers, particularly miRNA detection, reflects the ongoing evolution of nanozyme-based systems, with hybrid materials and advanced techniques paving the way for more versatile and clinically applicable diagnostic tools. A notable focus on miRNA detection underscores the shift toward genetic biomarkers, with miRNA-21 and miRNA-155 prime targets for breast and cervical cancer diagnostics. Techniques combining nanomotors, DNAzyme amplification, and electrochemiluminescence (ECL) offer promising real-time and multiplexed analysis opportunities.150 As the field advances, emphasis on hybrid materials, miniaturized detection systems, and integration with portable devices could further expand the clinical and point-of-care applications of nanozyme-based sensors.
Interdisciplinary innovations driving enzyme-based electrochemical detection forward highlighted. These innovations merge nanotechnology, materials science, and biomedical engineering to tackle critical challenges in diagnostics. The versatility and sensitivity of these systems hold significant promise for future developments in healthcare and disease management.
Recent studies have significantly advanced the synthesis of nanozymes, incorporating innovative materials and catalytic properties to enhance their functionality. For instance, heterostructured Ag3PO4/Ag/TiO2 nanorod arrays, PtCo@Prussian Blue nanozymes, and IrWOx nanoparticles have been synthesized using tailored methods to improve photothermal and enzymatic activities.148,152,153 These nanozymes facilitate versatile detection mechanisms, such as bifunctional photoelectrochemical sensing, DNAzyme walker processes, and ROS scavenging, enhancing sensitivity and specificity. Combining photothermal properties with catalytic activity, as seen in Ti3C2@Au@Pt nanozymes, amplifies thermal signals under NIR exposure, enabling highly sensitive detection of biomarkers like CTCs and miR-155.52 These approaches demonstrate the versatility and efficacy of nanozyme-enabled technologies in both diagnostics and therapy.
Targeting critical breast cancer biomarkers, including MUC1, CEA, and miR-155, underscores the diagnostic precision of nanozyme-based platforms. Detection limits have reached remarkable sensitivity, with values as low as 0.1 ng mL−1 for CEA and consistent reproducibility across assays. Additionally, integrating photothermal and imaging-guided systems addresses the dual goals of diagnosis and treatment, especially in advanced cancer stages like metastasis. Emerging trends highlight using multi-functional materials, biocompatibility enhancements, and imaging modalities to ensure clinical relevance. As the field evolves, addressing scalability and cost will be critical to translating these technologies from the lab to broader clinical applications, paving the way for improved outcomes in breast cancer management. Table 7 presents an overview of the nanozymes used in breast cancer detection.
Nanozymes | Mechanism | Type of sensor | Target | LOD | Ref. |
---|---|---|---|---|---|
FeOCl nanosheets | POD-like | Colorimetric | GSH and Cys | 2.23 μM and 2.76 μM | 134 |
TMB-GQDzymes | POD-like | Colorimetric/photothermal dual-mode biosensor | MCF-7 cell-derived exosomes | 1027 particles per μL, 2170 particles per μL | 137 |
CFCDs | Phosphatase | Colorimetric | Alkaline phosphatase (ALP) | 0.01 U mL−1 | 136 |
CD44FM nanozymes | OXD-like | Colorimetric | TNBC, MDA-MB-231 cells | 186 cells | 139 |
BDNP-100 | POD-like | Colorimetric | H2O2 and glucose | 40 μM | 135 |
MNPs | POD-like | Colorimetric | Melanoma CTCs | 13 cells per mL | 154 |
Au@PtOs | POD-like | Colorimetric, SERS (surface-enhanced Raman scattering), photothermal mode | Breast cancer exosomes | 2.6 × 103 exosomes per μL, 4.1 × 101 exosomes per μL, 4.6 × 102 exosomes per μL | 19 |
FeMn-NCetch/SAC | POD-like | Electrochemical and photothermal | HER2 | 3.9 pg, 7.5 pg mL−1 | 23 |
FeMn-NCedge | POD-like | Electrochemical immunoassay | HER2 | 5.4 pg mL−1 | 138 |
CuS@Pt-SA | POD-like | Electrosensitive | miRNA-21 | 0.16 fM | 22 |
Mn3O4/Pd@Pt/HRP nanoprobe | POD-like | Electrosensitive | HER2 | 0.08 ng mL−1 | 143 |
rGO/MoS2 | POD-like | Electrosensitive | CTCs | 6 cells mL−1 | 149 |
CuO nanozyme | POD-like | Electrosensitive | CTCs | 27 cells mL−1 | 155 |
FeNC | POD-like, GOx | Electrosensitive | HER2 | 4.5 pg mL−1 | 146 |
MOF@Pt@MOF | POD-like | Electrosensitive | Exosomal miRNA-21 | 0.29 fM | 20 |
CH–Cu@J-Cu2O | CAT-like, POD-like | Electrosensitive | MUC1 | 0.085 pg·mL−1 | 147 |
PtCo@rGO | POD-like | Electrochemiluminescence (ECL) cytosensor | MCF-7 | 1 cells mL−1 | 156 |
sponge-like Au@Ru | POD-like | Electrochemical immunosensor | HER2 | 0.15 pg mL−1 | 157 |
TCPP-Fe@HMUiO@Au-ABEI | POD-like | ECL detection | exomiR-155 | 273.20 aM | 144 |
TiO2@Ag | POD-like | ECL detection | miRNA-155 | 0.45 fM | 150 |
Ag3PO4/Ag/TiO2 | POD-like, alkaline phosphatase | Photothermal and photoelectrochemical detection | MUC1, CEA | 0.430 ng mL−1, 0.058 ng mL−1 | 152 |
PB | POD-like | Photoelectrochemical detection | miR-155 | 1.2 fM | 148 |
IrWOx-PEG | CAT-like | Photothermal and photoelectrochemical detection | Metastatic breast cancer cells | — | 153 |
SPIO@NC | POD-like, CAT-like | Dual-modality imaging | TNBC cells | — | 158 |
Another critical avenue is the exploration and integration of hybrid nanostructures. By merging carbon-based materials’ robust, biocompatible characteristics (e.g., graphene, carbon dots) with metal nanoparticles’ or MOFs’ high catalytic efficiency, researchers can create multifunctional systems that overcome current limitations. For instance, such hybrid nanozymes could harness carbonaceous materials’ superior conductivity and chemical stability alongside metallic components’ tailored enzyme-mimetic activities. This synergy is expected to yield platforms that perform highly efficient catalysis and offer enhanced optical, magnetic, and electrical properties, paving the way for real-time imaging and precise therapeutic interventions.
In addition to structural innovations, significant progress is anticipated in targeted delivery and controlled activation. The future development of smart nanozymes will involve surface modifications that grant immune resistance and active targeting capabilities. Techniques like PEG conjugation, cell membrane cloaking, and aptamer functionalization will be instrumental in extending circulation time, reducing off-target toxicity, and ensuring that these catalysts home in on specific tumor markers or pH gradients characteristic of the TME. Next-generation nanozymes can be activated “on demand,” providing precise temporal and spatial control over therapeutic action by responding to endogenous stimuli such as hypoxia, oxidative stress, or enzymatic cues.
Furthermore, integrating nanozymes with state-of-the-art imaging modalities is critical to realizing accurate theranostic systems. Incorporating functionalities for magnetic resonance imaging (MRI), positron emission tomography (PET), or near-infrared (NIR) fluorescence into nanozyme platforms will allow clinicians to monitor drug distribution, assess treatment efficacy, and dynamically adjust therapeutic regimens in real time. Such multifunctional platforms can simultaneously deliver therapy and provide diagnostic feedback, enhancing personalized treatment strategies for various breast cancer subtypes.
Scaling up production without compromising nanozyme quality is another challenge for future research. Developing green, cost-effective, reproducible synthesis methods is vital for clinical translation. Emphasis on eco-friendly precursors and scalable processes—such as liquid-phase exfoliation or hydrothermal synthesis using natural templates—will facilitate the manufacture of nanozymes that meet rigorous safety and performance standards. In parallel, comprehensive studies addressing pharmacokinetics, biodistribution, and long-term biocompatibility in relevant animal models must be undertaken to build a robust preclinical foundation.
Moreover, converging nanozyme research with emerging fields like artificial intelligence and machine learning is anticipated to accelerate discovery and optimization. Data-driven approaches can analyze vast experimental datasets to identify trends and predict the behavior of new nanozyme compositions. Such insights will streamline the development process and enable real-time customization of nanozyme properties to suit individual patient profiles, thus moving closer to genuinely personalized cancer therapy.
Lastly, the future of nanozyme-based breast cancer therapy will likely benefit from combinatorial treatment strategies. Integrating nanozymes with established modalities—such as chemotherapy, radiotherapy, immunotherapy, and phototherapy—can exploit synergistic mechanisms to overcome drug resistance and tumor heterogeneity. By concurrently disrupting metabolic pathways, inducing oxidative stress, and modulating immune responses, these multimodal platforms promise to enhance overall therapeutic efficacy and patient outcomes.
In summary, its interdisciplinary approach will define the next generation of nanozyme research—merging advanced material science, precision engineering, computational modeling, and clinical insights. With ongoing innovations focused on enhancing catalytic efficiency, targeting specificity, and multifunctionality, nanozymes are set to become a cornerstone in the future landscape of breast cancer theranostics, offering a highly precise, adaptable, and scalable solution for improved patient care.
The accumulated evidence demonstrates that nanozymes are effective in tumor ablation through enhanced ROS production and early diagnosis by detecting breast cancer biomarkers with high sensitivity. This dual functionality promises to revolutionize personalized cancer care by providing precise therapy and diagnostic capabilities. However, several challenges remain. Future research must optimize biocompatibility and minimize systemic toxicity while ensuring long-term stability and reproducibility of nanozyme formulations. Addressing issues of scalability, tumor heterogeneity, and targeted delivery is crucial for advancing clinical translation. Furthermore, comprehensive regulatory and safety evaluations will be essential to integrate nanozyme technology into standard treatment protocols. Overcoming these bottlenecks will pave the way for nanozymes to become a cornerstone in next-generation breast cancer management, offering more effective, minimally invasive, and personalized therapeutic solutions.
Overall, continued interdisciplinary collaboration and innovative nanomaterial engineering are imperative to fully realize the clinical potential of nanozyme-based therapies effectively.
This journal is © The Royal Society of Chemistry 2025 |