Thursday, January 22, 2026
Economy & Markets
162 min read

Gold Nanoparticles Revolutionize Bladder Cancer Treatment

Dove Medical Press
January 19, 20263 days ago
Gold Nanoparticles in Bladder Cancer Applications: A Paradigm Shift fr

AI-Generated Summary
Auto-generated

Gold nanoparticles (AuNPs) are revolutionizing bladder cancer management by transitioning from simple diagnostic tools to multifunctional theranostic platforms. Their unique properties enable ultra-sensitive detection of biomarkers for early diagnosis and monitoring. In treatment, AuNPs are utilized for precise physical ablation and enhancing immunotherapy, while also overcoming chemoresistance. This integrated approach aims for personalized precision medicine.

Introduction Bladder cancer ranks as the tenth most common malignancy worldwide, with over 570,000 new cases and more than 210,000 deaths annually.1 Among these, NMIBC accounts for 70–80% of initial diagnoses. Although transurethral resection of bladder tumor (TURBT) combined with postoperative intravesical chemotherapy or immunotherapy can achieve favorable outcomes, 50–70% of patients experience recurrence within five years, with 10–30% progressing to muscle-invasive bladder cancer (MIBC).2 This “recurrence-treatment-recurrence” cycle consumes substantial medical resources and imposes significant physical and psychological burdens on patients. Traditional cystoscopy combined with urine cytology remains the gold standard for bladder cancer diagnosis and monitoring. However, cystoscopy is invasive, costly, and may cause complications, while urine cytology exhibits low sensitivity, particularly for low-grade bladder cancer.3 Therefore, developing non-invasive, highly sensitive diagnostic methods and efficient, specific therapeutic strategies has become a critical need in bladder cancer research. In this context, gold nanoparticles (AuNPs) have shown tremendous potential. As a prominent member of the metal nanoparticle family, AuNPs offer a versatile platform compared to other metallic systems due to their superior stability and tunable surface chemistry.4–6 Their application in oncology has paved the way for more complex nanomedicines, though their safety profile requires rigorous evaluation across different physiological environments.7,8 Owing to their unique physicochemical properties such as surface plasmon resonance (SPR), high surface-to-volume ratio, ease of functionalization, and excellent biocompatibility, have shown tremendous potential in biomedical applications.9–14 In bladder cancer management, AuNPs have evolved from initially serving solely as diagnostic tools to multifunctional platforms integrating diagnosis, treatment, and immunomodulation, providing innovative approaches to address key challenges in bladder cancer management. António et al comprehensively summarized gold nanoparticle-based urine biodetection methods, emphasizing their potential for non-invasive diagnosis.15 However, existing reviews primarily focus on specific application areas, lacking a systematic description of AuNPs’ “role evolution” in bladder cancer theranostics. This article systematically reviews recent advances in AuNP applications in bladder cancer diagnosis and treatment, aiming to highlight the development trend of AuNPs transitioning from single applications to integrated theranostic platforms and the underlying driving mechanisms. We will explore how AuNPs overcome limitations of traditional therapeutic approaches through multiple mechanisms, including enhancing diagnostic sensitivity, improving therapeutic targeting, modulating the tumor microenvironment, and overcoming treatment resistance. Additionally, this paper provides in-depth analysis of current translational challenges and future directions, offering a reference for the clinical translation of AuNPs in bladder cancer. Unique Properties of Gold Nanoparticles and Their Precision Adaptation for Bladder Cancer Management Core Physicochemical Properties: The Foundation for Multifunctionality The core physicochemical properties of AuNPs include: Surface Plasmon Resonance (SPR): This is fundamental to their optical and photothermal applications. Upon excitation by light at specific wavelengths, AuNPs can efficiently convert photonic energy into thermal energy, providing the physical basis for photothermal therapy (PTT) and photoacoustic imaging (PAI).16 High Surface-to-Volume Ratio and Ease of Functionalization: The large surface area makes them ideal “multifunctional linking platforms.” Through well-established thiol-gold chemistry, targeting molecules (eg, antibodies, aptamers), therapeutic drugs, or imaging probes can be easily covalently conjugated to their surface, enabling modular and diverse functionality.17 Controllable Size and Morphology: By tuning the size and morphology of AuNPs (eg, nanospheres, nanorods, nanostars), their SPR absorption peak can be precisely adjusted to the near-infrared (NIR) window, which offers better tissue penetration for deep tumor imaging and therapy.18 Excellent Biocompatibility and Stability: The chemical inertness of gold provides good biocompatibility, while surface modifications (eg, PEGylation) can further prolong their circulation time in vivo and confer targeting or responsiveness to specific microenvironments.13 Targeting Strategies Responsive to the Bladder Tumor Microenvironment AuNPs can be functionally designed to target specific features of the bladder tumor microenvironment: Advantages of Local Drug Delivery: The intravesical administration environment provides natural advantages for local application and retention of AuNPs, achieving high local drug concentrations while reducing systemic toxicity.19 Passive and Active Targeting: The enhanced permeability and retention (EPR) effect in bladder tumor tissues facilitates passive targeting of AuNPs.20 More importantly, tumor-specific receptor expression (eg, EGFR, folate receptor) provides a molecular basis for active targeting using antibody- or aptamer-modified AuNPs.21 Microenvironment-Responsive Drug Release: Through sophisticated chemical design, AuNPs can be engineered as “smart” drug carriers that respond to acidic pH in the tumor microenvironment or specifically overexpressed enzymes (eg, hyaluronidase) to achieve on-demand, precise drug release.21 Based on these properties, AuNPs have become ideal carriers for constructing theranostic platforms. They can integrate diagnostic modules (eg, SERS reporters, photoacoustic contrast agents) and therapeutic modules (eg, chemotherapeutics, photothermal conversion agents, radiotherapy sensitizers) into a single nanoparticle. This not only enables “diagnosis-guided treatment” (eg, initiating photothermal therapy after precise lesion localization through imaging) but also allows real-time monitoring of therapeutic effects during treatment and adjustment of treatment regimens based on feedback, ultimately achieving truly personalized precision medicine. These versatile properties are schematically summarized in Figure 1. Cellular Uptake and Intracellular Trafficking The biological efficacy of AuNPs depends heavily on their internalization pathways, which typically involve clathrin-mediated endocytosis, caveolae-mediated endocytosis, or macropinocytosis, depending on particle size and surface charge. Once internalized, the ability of AuNPs to achieve endosomal escape—often through the “proton sponge” effect or membrane fusion facilitated by surface modifications—is critical for delivering therapeutic payloads directly to the cytosol or nucleus.22 Applications of Gold Nanoparticles in Bladder Cancer Diagnosis: Pursuing Non-Invasiveness and Ultra-Sensitivity Ultra-Sensitive Detection Platforms Based on Urinary Biomarkers: Revolutionizing Liquid Biopsies Urine, as the most direct “information source” for bladder cancer, is an ideal sample for non-invasive diagnosis (ie, liquid biopsy). However, urinary biomarkers are present at extremely low concentrations, imposing high demands on detection sensitivity. Leveraging their unique signal amplification mechanisms, such as aggregation-induced color changes, surface-enhanced Raman scattering (SERS) effects, and electrochemical signal enhancement, AuNPs have pushed the detection limits of these biomarkers to new heights. Telomerase Activity Detection: From Laboratory Validation to Clinical Application Telomerase, as a bladder cancer-specific biomarker, is overexpressed in approximately 85–90% of bladder cancer cases but is barely expressed in normal urothelial tissues, making it an ideal diagnostic target.23 Researchers have developed various ingenious detection strategies using AuNPs. Wang et al24 pioneered the combination of strand displacement amplification (SDA) with dynamic light scattering (DLS). Through telomerase extension products triggering SDA reactions, leading to the aggregation of oligonucleotide-modified AuNP probes, telomerase detection could be achieved with only 3 MCF-7 cells, showing a linear range of 5–1000 cells. Li et al25 further developed a DLS-based method for urinary telomerase activity detection. Telomerase extension products triggered hybridization chain reactions, causing DNA-modified gold nanoparticles to aggregate and increase in hydrodynamic diameter. The detection limit was as low as 4 MCF-7 cells, and it was successfully applied to distinguish urine samples from bladder cancer patients. Zou et al26 developed a highly sensitive telomerase activity detection method based on catalytic hairpin assembly-DLS (CHA-DLS), using the change in particle size of AuNP probes as the detection signal: telomerase extension products (TEP) trigger CHA reactions, forming numerous AuNP-H1/H2 nanostructures, resulting in a significant increase in particle size detected by DLS. This technology could detect telomerase activity as low as 6 MCF-7 cells, 10 Huh7 cells, and 35637cells. Although these DLS-based methods are sensitive, they require sophisticated instruments. To promote point-of-care testing (POCT), Duan et al proposed a more intuitive bidirectional colorimetric strategy based on four detection color states of bifunctional gold nanoparticle probes (blue, purple, red, and precipitation), enabling intuitive judgment of telomerase activity and bladder cancer diagnosis. This method could distinguish normal individuals from bladder cancer patients by the naked eye or UV-Vis spectroscopy, showing great potential for POCT in resource-limited areas.27 To balance sensitivity and accuracy, Feng et al28 developed a dual-mode probe based on SERS and colorimetry, providing a more reliable tool for telomerase detection. Hyaluronidase (HAase) Detection: From Colorimetry to Multi-Modal Sensing Hyaluronidase (HAase) is highly expressed in various malignancies, and its activity is closely related to bladder cancer grading and progression, making it another highly promising urinary biomarker.29 Xu et al30 pioneered the use of electrostatic interactions between positively charged AuNPs and negatively charged hyaluronic acid (HA) to construct a colorimetric detection method. After HAase degraded HA, AuNPs aggregated, producing an intuitive “red-blue” color change. Although this method is simple and straightforward, it is susceptible to interference from complex components in urine. To further address issues of sensitivity and specificity and enhance performance, researchers have developed more sophisticated sensing strategies. Cheng et al31 constructed probes based on fluorescence resonance energy transfer (FRET), while Wang, Si, and Chen et al32–34 turned to SERS technology, designing different Raman reporters and internal references to achieve ultra-sensitive and highly precise quantitative detection of HAase, with detection limits as low as 0.32–0.4 mU/mL. These studies mark the transition of HAase detection from qualitative/semi-quantitative to high-precision quantitative analysis. Multi-Biomarker Detection Strategies: Enhancing Diagnostic “Resolution” Given the heterogeneity of bladder cancer, single biomarkers often result in “false positives” or “false negatives,” limiting diagnostic accuracy. Therefore, simultaneous detection of multiple biomarkers using a “combination approach” to enhance diagnostic precision is an inevitable trend. AuNP-based platforms demonstrate significant advantages in multi-biomarker detection due to their ease of functionalization. Electrochemical immunosensors are particularly favored for their high sensitivity, low cost, and ease of miniaturization. For example, Zhang, Zhou, and Wang et al35–37 constructed sensors that can simultaneously or separately detect multiple bladder cancer-related protein biomarkers such as CFHR1, NMP22, and NUMA1, with detection limits reaching the astonishing fg/mL level. Li et al utilized SERS technology to achieve simultaneous quantitative detection of FGFR3 and NMP22 by constructing a unique AgAu@Ag core-shell structure substrate.38 This fully demonstrates the great potential of AuNP platforms in developing high-throughput, high-sensitivity diagnostic tools. Exosomal microRNA Detection: Opening a New Chapter in Liquid Biopsy MicroRNAs (miRNAs) carried by exosomes are considered “messengers” reflecting tumor status, representing a cutting-edge direction in liquid biopsy. However, exosomal miRNAs in plasma or urine are present in extremely minute quantities, making their precise capture and signal amplification major technical challenges. Zhang et al39 ingeniously combined inorganic nanoscintillators with DNAzyme walkers to detect miRNAs at the femtomolar level. Meanwhile, Liyanage, Hati, and colleagues40,41 utilized the unique localized surface plasmon resonance (LSPR) effects of structures like gold triangular nanoprisms to further push detection sensitivity to the attomolar level and successfully distinguished between early and late-stage bladder cancer patients. In addition to pursuing ultimate sensitivity, developing detection methods suitable for different scenarios is also crucial. Lin et al developed pH-responsive DNA nanoswitches for SPR platform detection;42 Nossier43 and Mollasalehi et al44 focused on developing amplification-free, user-friendly colorimetric or cross-linking-free hybridization techniques, thereby paving the way for point-of-care testing (POCT); Masterson et al combined SERS and plasmon-enhanced fluorescence (PEF) to develop a multimodal analysis method, effectively reducing false positives and false negatives and demonstrating high stability in clinical applications.45 These diverse studies collectively promote the clinical translation of exosomal miRNAs as bladder cancer biomarkers. A comparison of the major AuNP-based urinary biomarker detection methods discussed in this section is provided in Table 1. Other Biomarker Detection Methods In addition to the above biomarkers, AuNP-based detection methods have also been used for various other bladder cancer-related molecules. These include rapid colorimetric detection of total gelatinases (MMP-2/9) activity46 and HURP RNA;47 competitive amperometric immunosensor detection of p53 protein;48 nanobody-coupled gold nanoparticle cluster detection of survivin protein;49 and direct detection of long non-coding RNA UCA1 based on nanoprobe hybridization technology.50 Similarly, Jazayeri et al51 developed a colorimetric method based on antibody-conjugated AuNPs that can rapidly and non-invasively detect survivin protein in urine, showing potential in cancer grading (especially distinguishing low-grade from high-grade), providing new tools for early diagnosis and prognosis of bladder cancer. These studies collectively enrich the “toolbox” for non-invasive bladder cancer diagnosis (Figure 2A). Epigenetic Biomarker Detection: DNA Methylation Emerging research highlights urinary DNA methylation as a highly effective biomarker for early detection and recurrence monitoring. AuNP-based platforms are now being engineered for the ultrasensitive detection of these epigenetic markers. For instance, plasmonic biosensors utilizing functionalized AuNPs can capture methylated DNA fragments with high specificity, offering a non-invasive “liquid biopsy” tool that significantly enhances diagnostic resolution.52 Enhanced Imaging Diagnosis CT/MR Dual-Modal Contrast Agents: High-Resolution Bladder Imaging The high atomic number of AuNPs makes them excellent CT contrast agents, outperforming the clinically used iohexol.53 To combine the high resolution of CT with the excellent soft tissue contrast of magnetic resonance imaging (MRI), Wen et al54 developed dendrimer-entrapped gold nanoparticles combined with Gd(III) chelates, successfully achieving CT/MR dual-modal imaging of rat bladders. Photoacoustic Imaging (PAI): Early Visualization of Tiny Lesions PAI combines the advantages of optics and ultrasound, and AuNPs are ideal contrast agents for it. Jeon et al55 showed that transurethral injection of gold nanocages (GNCs) enhanced the photoacoustic signal in rat bladders by approximately 2240%, demonstrating the enormous potential of AuNPs as basic contrast agents. Building on this, Li et al56 further explored their functional diversity by conjugating different antibodies to gold nanorods with different absorption peaks, successfully achieving simultaneous imaging of multiple targets, breaking through the limitation of traditional PAI being limited to anatomical imaging. The key to advancing this technology to clinical application is the ability to detect tiny lesions. To this end, Alchera and Venegoni et al57,58 developed gold nanorods (GNRs) targeting integrin α5β1, visualizing tiny lesions smaller than 1 mm in animal models, providing a powerful tool to solve the problem of postoperative minimal residual disease monitoring. Canning et al59 combined photoacoustic imaging with photothermal therapy, achieving real-time thermal imaging monitoring and dosimetry during the treatment process, showing great potential for theranostics. Surface-Enhanced Raman Scattering (SERS): Achieving Molecular-Level Diagnosis SERS technology can provide “molecular fingerprint” information of tissues and cells. Jin et al28 used AuNPs as enhancement substrates to successfully distinguish normal and cancerous bladder tissues. Yang et al60 used SERS platforms to screen specific membrane receptors for different grades of bladder cancer cells, providing new targets for precise diagnosis and targeted therapy. Microfluidic Technology and Rapid Diagnostic Platforms Combining AuNP detection technology with microfluidic chips is an important direction to achieve automated, high-throughput, and point-of-care testing (POCT). Thompson et al61 developed a 3D-printed modular microfluidic system for AuNP colorimetric detection of HAase. Sun et al62 combined aptamer biochips with AuNPs, achieving rapid simultaneous detection of bladder cancer-related infection bacteria through electrospray printing technology. Applications of Gold Nanoparticles in Bladder Cancer Treatment: Advancing Toward Precision Synergy and Immune Remodeling Gold nanoparticles represent a robust and sophisticated toolkit for the mechanical and thermal eradication of malignant cells, primarily operating through the complementary avenues of photothermal therapy (PTT) and the targeted enhancement of radiographic sensitivity. By functionalizing AuNPs with highly specific ligands—most notably anti-EGFR antibodies or integrin α5β1—researchers have successfully engineered “precision-guided” agents that selectively localize within the complex bladder tumor microenvironment.63–65 These engineered platforms possess the unique ability to efficiently transform absorbed near-infrared (NIR) light into concentrated hyperthermia, thereby inducing localized cytolysis through thermal stress while effectively minimizing systemic exposure and collateral damage to healthy tissues. While various gold-based architectures have been explored, empirical evidence suggests that specific morphologies, such as silica-cored gold nanoshells, exhibit a superior capacity for photothermal killing, often requiring a significantly lower particle concentration than other nanostructures to achieve comparable therapeutic outcomes66 (Figure 2Bi). One of the most formidable challenges in the clinical implementation of PTT involves the prevention of accidental thermal impairment to the delicate and healthy urothelial lining. To mitigate this risk, recent advancements have introduced armored gold nanostars integrated with real-time photoacoustic thermometry. This integration facilitates meticulous dosimetry control and has resulted in 100% survival rates within preclinical bladder cancer models by allowing clinicians to monitor temperature changes in situ.59 Furthermore, advanced computational and numerical simulations have elucidated that variables such as the precise anatomical position of the tumor and the specific composition of overlying tissues significantly modulate the overall efficiency of PTT. This underscores the critical necessity for individualized optimization of both laser parameters and nanoparticle dimensions in a patient-specific manner.67 Beyond pure thermal ablation, the high atomic number of gold renders these nanoparticles excellent candidates for radiotherapy sensitization. AuNPs tend to accumulate within the tumor-associated stroma, functioning as radiosensitizers that amplify the therapeutic index of X-ray irradiation—a strategy that is particularly vital for developing bladder-preserving protocols in patients with muscle-invasive bladder cancer (MIBC).68 It is important to note, however, that while AuNPs produce significant sensitizing effects under standard clinical X-ray irradiation, their efficiency is markedly reduced when utilizing proton beam therapy. This distinction is a critical consideration for the design of future clinical trials and the selection of radiation modalities.69 To further ensure long-term biosafety, the field is increasingly shifting toward the development of renal-clearable, ultra-small AuNPs with diameters of less than 6 nm, which facilitates rapid clearance and minimizes the risk of chronic organ accumulation.70 Major physical therapy and sensitization strategies are summarized in Table 2. Strategies for Targeted Drug Delivery and the Mitigation of Chemoresistance In the realm of pharmacology, AuNPs function as highly versatile nanocarriers designed to circumvent the intrinsic limitations of conventional intravesical chemotherapy, such as poor drug selectivity and the rapid onset of multidrug resistance (MDR). The successful penetration of these carriers often depends on overcoming structural barriers; in this context, the development of molecular tools to assess collagen denaturation has provided essential methodologies for evaluating the physical impediments within the bladder wall that hinder drug diffusion.76 Beyond their role as passive vehicles, AuNPs themselves can exert direct inhibitory effects on bladder cancer cells by inducing oxidative stress and activating ROS-mediated apoptotic signaling pathways.77,78 Targeted delivery via folate receptor-specific or cell membrane-camouflaged AuNPs significantly elevates intracellular drug concentrations while effectively bypassing traditional efflux pump-mediated resistance71,72 (Figure 2Bi and Table 2). These platforms are also being engineered to combat complex molecular drivers of resistance; for instance, since ac4C modification-mediated DNA damage repair has been identified as a primary engine of cisplatin resistance, future AuNP systems could co-deliver siRNAs or small-molecule inhibitors targeting NAT10 to restore chemosensitivity.79 Furthermore, AuNPs have proven to be superior scaffolds for Antibody-Drug Conjugates (ADCs). Utilizing AuNPs to deliver HER2-targeting ADCs (such as RC48) can facilitate enhanced tissue penetration and achieve more regulated, responsive drug release within the unique bladder microenvironment compared to traditional systemic administration.80,81 Remodeling the Tumor Microenvironment and Enhancing Immunotherapy AuNPs are undergoing a functional transition from passive tumor killers to active immune activators capable of fundamentally remolding the immunosuppressive “tumor soil”. These nanoplatforms can trigger the repolarization of pro-tumor M2-type macrophages into anti-tumor M1-type phenotypes and activate the cGAS-STING signaling pathway to dismantle the mechanisms of local immune evasion74 (Figure 2Bii). By conjugating particles with tumor-specific antigens or utilizing them to directly neutralize immunosuppressive factors like TGF-β1, clinicians can effectively convert immunologically “cold” tumors into “hot” ones characterized by robust T-cell infiltration.82,83 The integration of AuNPs with natural plant extracts, such as EGCG or Abies spectabilis, has also demonstrated synergistic anti-tumor effects that far exceed the performance of single components by activating mitochondrial-dependent apoptosis.84,85 Furthermore, combining AuNPs with ferroptosis-inducing agents like DHA or RSL3 within intelligent hydrogel systems allows for a “divide-and-rule” strategy that promotes immunogenic cell death and fosters durable systemic anti-tumor immunity.73,74,86,87 Relevant immune synergistic mechanisms are detailed in Table 2. Towards Theranostic Integration: A Closed-Loop AuNP Platform The ultimate paradigm shift in bladder cancer management involves the construction of a closed-loop clinical workflow. By utilizing specific molecular targets such as HER2 or FGFR3, functionalized AuNPs enable high-precision patient stratification through ultra-sensitive SERS detection or multimodal imaging techniques.38,57,60 In this integrated model, therapeutic interventions—such as PTT or targeted chemotherapy—are initiated only after precise lesion localization has been confirmed via imaging. Simultaneously, real-time imaging provides a continuous feedback loop on the patient’s therapeutic response, allowing for the dynamic adjustment of treatment regimens based on real-time efficacy.75 Such a closed-loop system ensures that bladder cancer management moves toward a truly personalized precision medicine model. From Laboratory to Clinic: Challenges and Future Prospects Safety and Biocompatibility Considerations The clinical translation of AuNPs is fundamentally dependent on a comprehensive and rigorous understanding of their biological fate and long-term safety. While ultra-small AuNPs (< 1.9 nm) are primarily excreted via the renal-bladder pathway, existing data suggests that renal impairment can significantly correlate with reduced clearance and increased systemic retention. The biological performance of these particles is governed by their intracellular trafficking—including clathrin-mediated endocytosis—and their subsequent ability to achieve endosomal escape through mechanisms like the “proton sponge” effect. Furthermore, the formation of a protein corona upon contact with biological fluids remains a critical hurdle, as it can mask targeting ligands and alter the intended biodistribution. Designing zwitterionic polymer coatings to create immune-silent surfaces is a promising strategy to evade the mononuclear phagocyte system.70,88,89 Large-Scale Production and Quality Control Transitioning from laboratory-scale synthesis to clinical-grade production involves significant technical and economic hurdles. Established laboratory methods, such as green synthesis or chemical reduction, must be strictly standardized to ensure that every production batch meets rigorous requirements for size uniformity, morphology, and surface ligand density.90,91 Establishing manufacturing facilities that comply with global Good Manufacturing Practice (GMP) standards represents a major capital investment. Consequently, the development of low-cost, high-stability synthetic routes is an essential prerequisite for the eventual dissemination of AuNP-based diagnostic and therapeutic tools to primary healthcare centers and regions with limited medical infrastructure. Major translational challenges and potential solutions are summarized in Table 3. Regulatory Pathways and Clinical Translation Prospects The current lack of longitudinal human data remains a primary obstacle to the translation process. Regulatory agencies worldwide face challenges in establishing clear evaluation standards for nanomedicines, particularly those operating under a drug-device combination model for theranostic use.57,59,91 To bridge this gap, there is an urgent need for multi-center, prospective clinical trials designed to empirically validate the superiority of AuNP-based interventions over traditional cystoscopy and conventional intravesical chemotherapy. Addressing these regulatory and clinical gaps through intensive interdisciplinary cooperation will be essential to bring these innovative technologies to the patient’s bedside.27,42–44 Conclusion and Future Prospects Gold nanoparticles exhibit revolutionary potential in bladder cancer management due to their unique surface plasmon resonance effect, ease of functionalization, and excellent biocompatibility. AuNPs have evolved from simple imaging or detection tools into multifunctional theranostic platforms integrating multimodal imaging, targeted delivery, controlled release, and immune modulation. In diagnostics, ultra-sensitive AuNP-based sensing platforms enable femtomolar detection of urinary biomarkers, significantly improving early screening accuracy. In therapy, AuNPs serve as efficient carriers to overcome chemoresistance and achieve precise physical ablation while actively remodeling the tumor immune microenvironment through pathways such as cGAS-STING. This transition from “passive delivery” to “intelligent intervention” is fundamentally reshaping the diagnostic and therapeutic paradigm for bladder cancer. Future breakthroughs will depend on establishing a new paradigm of “intelligent responsive” and “closed-loop” theranostics. Ideal AuNP platforms should dynamically sense and respond to the tumor microenvironment (eg, low pH or specific enzymes) or external stimuli to achieve precise drug “detonation” and “dual-track” immune regulation. By integrating AuNPs with technologies such as NIR-II window imaging and real-time photoacoustic thermometry, urologists can implement closed-loop management from diagnostic stratification to real-time efficacy monitoring, enhancing controllability and preventing damage to the healthy urothelium. Furthermore, combining AuNPs with CRISPR-Cas9 technology targeting markers like HER2 or FGFR3 offers a theoretically promising strategy for simultaneously repairing oncogenic mutations and eradicating residual cancer cells. Achieving truly personalized precision medicine requires the deep integration of Artificial Intelligence (AI) with nanomedicine. By analyzing large-scale multi-omics patient data, AI can guide the “tailor-made” optimization of AuNP morphology and ligand density to precisely predict and improve individual treatment responses. Simultaneously, the development of biomimetic platforms—such as exosome-coated or cell-membrane-camouflaged AuNPs—and microbiome-aware systems will further enhance targeting efficiency and reduce immunogenicity. Regarding clinical safety concerns, the exploration of biodegradable or “self-dissolving” gold nanostructures is an essential pathway to eliminate long-term accumulation toxicity and meet the safety requirements of repeated intravesical administrations. While clinical translation still faces significant challenges in GMP standardization and regulatory approval, the potential of AuNPs in the urinary system extends far beyond bladder cancer, including applications in prostate cancer management and sensitive assessment of acute kidney injury. Beyond oncology, AuNPs show promise in non-cancerous diseases like chemical cystitis and urinary tract infections.92,93 Cutting-edge explorations such as urease-driven nanomotors further illustrate their potential for complex intravesical interventions.94,95 Multidisciplinary collaboration among urologists, materials scientists, and AI experts will be the driving force behind turning these research achievements into clinical reality. We firmly believe that as our understanding of nano-bio interface interactions deepens, these “golden seeds” will bear healthy fruits for bladder cancer patients worldwide. Data Sharing Statement No data was used for the research described in the article. Funding This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. Disclosure The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. References 1. Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F. Bladder Cancer Incidence and Mortality: a Global Overview and Recent Trends. Eur Urol. 2017;71(1):96–108. doi:10.1016/j.eururo.2016.06.010 2. Babjuk M, Burger M, Capoun O, et al. European Association of Urology Guidelines on Non-muscle-invasive Bladder Cancer (Ta, T1, and Carcinoma in Situ). Eur Urol. 2022;81(1):75–94. doi:10.1016/j.eururo.2021.08.010 3. Owens CL, Vandenbussche CJ, Burroughs FH, Rosenthal DL. A review of reporting systems and terminology for urine cytology. Cancer Cytopathol. 2013;121(1):9–14. doi:10.1002/cncy.21253 4. Nie Y, Li D, Peng Y, et al. Metal organic framework coated MnO(2) nanosheets delivering doxorubicin and self-activated DNAzyme for chemo-gene combinatorial treatment of cancer. Int J Pharm. 2020;585:119513. doi:10.1016/j.ijpharm.2020.119513 5. Wang Y, Xu Y, Song J, et al. Tumor Cell-Targeting and Tumor Microenvironment-Responsive Nanoplatforms for the Multimodal Imaging-Guided Photodynamic/Photothermal/Chemodynamic Treatment of Cervical Cancer. Int J Nanomed. 2024;19:5837–5858. doi:10.2147/IJN.S466042 6. Dong L, Ding J, Zhu L, Liu Y, Gao X, Zhou W. Copper carbonate nanoparticles as an effective biomineralized carrier to load macromolecular drugs for multimodal therapy. Chin Chem Lett. 2023;34(9):108192. doi:10.1016/j.cclet.2023.108192 7. Song Y, Huang Y, Zhou F, Ding J, Zhou W. Macrophage-targeted nanomedicine for chronic diseases immunotherapy. Chin Chem Lett. 2022;33(2):597–612. doi:10.1016/j.cclet.2021.08.090 8. Jiang Z, Xiang H, Tang X. Smart Inorganic Nanomaterials for Tumor Microenvironment Modulation. Inorganics. 2025;13(10):337. doi:10.3390/inorganics13100337 9. Dykman LA, Khlebtsov NG. Gold nanoparticles in biology and medicine: recent advances and prospects. Acta Naturae. 2011;3(2):34–55. doi:10.32607/20758251-2011-3-2-34-55 10. Yeh YC, Creran B, Rotello VM. Gold nanoparticles: preparation, properties, and applications in bionanotechnology. Nanoscale. 2012;4(6):1871–1880. doi:10.1039/C1NR11188D 11. Ashraf S, Pelaz B, Del Pino P, et al. Gold-Based Nanomaterials for Applications in Nanomedicine. Top Curr Chem. 2016;370:169–202. 12. Shukla R, Chanda N, Zambre A, et al. Laminin receptor specific therapeutic gold nanoparticles (198AuNP-EGCg) show efficacy in treating prostate cancer. Proc Natl Acad Sci U S A. 2012;109(31):12426–12431. doi:10.1073/pnas.1121174109 13. Delong RK, Reynolds CM, Malcolm Y, Schaeffer A, Severs T, Wanekaya A. Functionalized gold nanoparticles for the binding, stabilization, and delivery of therapeutic DNA, RNA, and other biological macromolecules. Nanotechnol Sci Appl. 2010;3:53–63. doi:10.2147/NSA.S8984 14. Daniel MC, Astruc D. Gold nanoparticles: assembly, supramolecular chemistry, quantum-size-related properties, and applications toward biology, catalysis, and nanotechnology. Chem Rev. 2004;104(1):293–346. doi:10.1021/cr030698 15. António M, Vitorino R, Daniel-da-silva AL. Gold nanoparticles-based assays for biodetection in urine. Talanta. 2021;230:122345. doi:10.1016/j.talanta.2021.122345 16. Eustis S, el-Sayed MA. Why gold nanoparticles are more precious than pretty gold: noble metal surface plasmon resonance and its enhancement of the radiative and nonradiative properties of nanocrystals of different shapes. Chem Soc Rev. 2006;35(3):209–217. doi:10.1039/B514191E 17. Jain PK, Lee KS, El-Sayed IH, El-Sayed MA. Calculated absorption and scattering properties of gold nanoparticles of different size, shape, and composition: applications in biological imaging and biomedicine. J Phys Chem B. 2006;110(14):7238–7248. doi:10.1021/jp057170o 18. Mahan MM, Doiron AL. Gold nanoparticles as X‐ray, CT, and multimodal imaging contrast agents: formulation, targeting, and methodology. J Nanomater. 2018;2018(1):5837276. doi:10.1155/2018/5837276 19. Tyagi P, Hafron J, Kaufman J, Chancellor M. Enhancing Therapeutic Efficacy and Safety of Immune Checkpoint Inhibition for Bladder Cancer: a Comparative Analysis of Injectable vs. Intravesical Administration Int J Mol Sci. 2024;25(9):1. 20. Meher N, Bidkar AP, Wadhwa A, et al. PET Imaging Using 89Zr-Labeled StarPEG Nanocarriers Reveals Heterogeneous Enhanced Permeability and Retention in Prostate Cancer. Mol Cancer Ther. 2025;24(1):141–151. doi:10.1158/1535-7163.MCT-24-0024 21. Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater. 2021;6(7):1973–1987. doi:10.1016/j.bioactmat.2020.12.010 22. Wang X, Sun K, Dong J, et al. Carrier-free nanoparticles based on natural products trigger dual “synergy and attenuation” for enhanced phototherapy of liver cancer. Mater Today Bio. 2025;35:102278. doi:10.1016/j.mtbio.2025.102278 23. Peng L, Li J, Meng C, et al. Diagnostic Value of Telomerase Activity in Patients With Bladder Cancer: a Meta-Analysis of Diagnostic Test. Front Oncol. 2020;10:570127. doi:10.3389/fonc.2020.570127 24. Wang J, Zhang J, Li T, Shen R, Li G, Ling L. Strand displacement amplification-coupled dynamic light scattering method to detect urinary telomerase for non-invasive detection of bladder cancer. Biosens Bioelectron. 2019;131:143–148. doi:10.1016/j.bios.2019.02.014 25. Li T, Zou L, Zhang J, Li G, Ling L. Non-invasive diagnosis of bladder cancer by detecting telomerase activity in human urine using hybridization chain reaction and dynamic light scattering. Anal Chim Acta. 2019;1065:90–97. doi:10.1016/j.aca.2019.03.039 26. Zou L, Li X, Zhang J, Ling L. A Highly Sensitive Catalytic Hairpin Assembly-Based Dynamic Light-Scattering Biosensors for Telomerase Detection in Bladder Cancer Diagnosis. Anal Chem. 2020;92(18):12656–12662. doi:10.1021/acs.analchem.0c02858 27. Duan R, Wang B, Zhang T, et al. Sensitive and bidirectional detection of urine telomerase based on the four detection-color states of difunctional gold nanoparticle probe. Anal Chem. 2014;86(19):9781–9785. doi:10.1021/ac5024364 28. Feng E, Zheng T, Tian Y. Dual-Mode Au Nanoprobe Based on Surface Enhancement Raman Scattering and Colorimetry for Sensitive Determination of Telomerase Activity Both in Cell Extracts and in the Urine of Patients. ACS Sens. 2019;4(1):211–217. 29. Ho SM, Cheng RY. A promising bladder cancer detection assay: the HA-HAase test. J Urol. 2004;172(3):824–825. doi:10.1097/01.ju.0000136503.68531.6b 30. Nossier AI, Eissa S, Ismail MF, Hamdy MA, Azzazy HM. Direct detection of hyaluronidase in urine using cationic gold nanoparticles: a potential diagnostic test for bladder cancer. Biosens Bioelectron. 2014;54:7–14. doi:10.1016/j.bios.2013.10.024 31. Cheng D, Han W, Yang K, Song Y, Jiang M, Song E. One-step facile synthesis of hyaluronic acid functionalized fluorescent gold nanoprobes sensitive to hyaluronidase in urine specimen from bladder cancer patients. Talanta. 2014;130:408–414. doi:10.1016/j.talanta.2014.07.005 32. Wang W, Li D, Zhang Y, et al. One-pot synthesis of hyaluronic acid-coated gold nanoparticles as SERS substrate for the determination of hyaluronidase activity. Mikrochim Acta. 2020;187(11):604. 33. Si Y, Li L, He B, Li J. A novel surface-enhanced Raman scattering-based ratiometric approach for detection of hyaluronidase in urine. Talanta. 2020;215:120915. doi:10.1016/j.talanta.2020.120915 34. Chen X, Huang Q, Ruan S, et al. Self-calibration SERS sensor with “core-satellite” structure for detection of hyaluronidase activity. Anal Chim Acta. 2022;1227:340302. doi:10.1016/j.aca.2022.340302 35. Zhang J, Guo F, Zhu J, et al. Ultrasensitive Electrochemiluminescence Immunosensor for Bladder Marker Human Complement Factor H-Related Protein Detection. Anal Chem. 2023;95(30):11440–11448. doi:10.1021/acs.analchem.3c01786 36. Zhou Z, Tian Y, Zou L, et al. An electrochemical ratiometric immunosensor for the detection of NMP22 based on ZIF-8@MWCNTs@Chit@Fc@AuNPs and AuPt-MB. Anal Methods. 2024;16(31):5467–5474. doi:10.1039/D4AY01066C 37. Wang J, Guo F, Zhang J, Chao J. Potential-resolved electrochemiluminescence for simultaneous determination of multiplex bladder cancer markers. Chem Commun. 2024;60(34):4609–4612. doi:10.1039/D4CC00996G 38. Li HZ, Zhu J, Weng GJ, Li JJ, Li L, Zhao JW. AgAu@Ag core-shell triangular nanonet jointed with composite SERS enhanced substrate for capturing and sensing urine biomarkers FGFR3 and NMP22. Biosens Bioelectron. 2025;286:117635. doi:10.1016/j.bios.2025.117635 39. Zhang X, Wei X, Qi J, et al. Simultaneous Detection of Bladder Cancer Exosomal MicroRNAs Based on Inorganic Nanoflare and DNAzyme Walker. Anal Chem. 2022;94(11):4787–4793. doi:10.1021/acs.analchem.1c05588 40. Liyanage T, Masterson AN, Oyem HH, Kaimakliotis H, Nguyen H, Sardar R. Plasmoelectronic-Based Ultrasensitive Assay of Tumor Suppressor microRNAs Directly in Patient Plasma: design of Highly Specific Early Cancer Diagnostic Technology. Anal Chem. 2019;91(3):1894–1903. doi:10.1021/acs.analchem.8b03768 41. Hati S, Langlais SR, Masterson AN, et al. Photoswitchable Machine-Engineered Plasmonic Nanosystem with High Optical Response for Ultrasensitive Detection of microRNAs and Proteins Adaptively. Anal Chem. 2021;93(41):13935–13944. doi:10.1021/acs.analchem.1c02990 42. Lin PY, Chang YF, Chen CC, Su LC, Willner I, Ho JA. pH-Responsive Triplex DNA Nanoswitches: surface Plasmon Resonance Platform for Bladder Cancer-Associated microRNAs. ACS Nano. 2025;19(7):7140–7153. doi:10.1021/acsnano.4c16396 43. Nossier AI, Abdelzaher H, Matboli M, Eissa S. Dual approach for the colorimetric determination of unamplified microRNAs by using citrate capped gold nanoparticles. Mikrochim Acta. 2018;185(4):236. doi:10.1007/s00604-018-2767-9 44. Mollasalehi H, Hamidi A. Early-phase nano-genosensing of cell-free nucleobiomarkers in the plasma of cancerous patients. Nanomedicine. 2021;32:102344. doi:10.1016/j.nano.2020.102344 45. Masterson AN, Liyanage T, Berman C, Kaimakliotis H, Johnson M, Sardar R. A novel liquid biopsy-based approach for highly specific cancer diagnostics: mitigating false responses in assaying patient plasma-derived circulating microRNAs through combined SERS and plasmon-enhanced fluorescence analyses. Analyst. 2020;145(12):4173–4180. doi:10.1039/D0AN00538J 46. Nossier AI, Mohammed OS, Fakhr El-Deen RR, Zaghloul AS, Eissa S. Gelatin-modified gold nanoparticles for direct detection of urinary total gelatinase activity: diagnostic value in bladder cancer. Talanta. 2016;161:511–519. doi:10.1016/j.talanta.2016.09.015 47. Eissa S, Shawky SM, Matboli M, Mohamed S, Azzazy HM. Direct detection of unamplified hepatoma upregulated protein RNA in urine using gold nanoparticles for bladder cancer diagnosis. Clin Biochem. 2014;47(1–2):104–110. doi:10.1016/j.clinbiochem.2013.10.022 48. Giannetto M, Bianchi MV, Mattarozzi M, Careri M. Competitive amperometric immunosensor for determination of p53 protein in urine with carbon nanotubes/gold nanoparticles screen-printed electrodes: a potential rapid and noninvasive screening tool for early diagnosis of urinary tract carcinoma. Anal Chim Acta. 2017;991:133–141. doi:10.1016/j.aca.2017.09.005 49. Tang Y, Zhao X, Nie Z, He B, Deng D. Nanobody Coupled with Au Nanoparticle Cluster to Detect Survivin Protein by Lateral Flow Immunoassay. ACS Appl Bio Mater. 2025;8(7):6252–6260. doi:10.1021/acsabm.5c00745 50. Eissa S, Matboli M, Essawy NO, Shehta M, Kotb YM. Rapid detection of urinary long non-coding RNA urothelial carcinoma associated one using a PCR-free nanoparticle-based assay. Biomarkers. 2015;20(3):212–217. doi:10.3109/1354750X.2015.1062918 51. Jazayeri MH, Aghaie T, Nedaeinia R, Manian M, Nickho H. Rapid noninvasive detection of bladder cancer using survivin antibody-conjugated gold nanoparticles (GNPs) based on localized surface plasmon resonance (LSPR). Cancer Immunol Immunother. 2020;69(9):1833–1840. doi:10.1007/s00262-020-02559-y 52. Chen X, Zhang J, Ruan W, et al. Urine DNA methylation assay enables early detection and recurrence monitoring for bladder cancer. J Clin Invest. 2020;130(12):6278–6289. doi:10.1172/JCI139597 53. Yu Y, Wu Y, Liu J, Zhan Y, Wu D. Ultrasmall dopamine-coated nanogolds: preparation, characteristics, and CT imaging. J Exp Nanosci. 2016;11(sup1):S1–S11. doi:10.1080/17458080.2015.1102343 54. Wen S, Li K, Cai H, et al. Multifunctional dendrimer-entrapped gold nanoparticles for dual mode CT/MR imaging applications. Biomaterials. 2013;34(5):1570–1580. doi:10.1016/j.biomaterials.2012.11.010 55. Jeon M, Jenkins S, Oh J, et al. Nonionizing photoacoustic cystography with near-infrared absorbing gold nanostructures as optical-opaque tracers. Nanomedicine. 2014;9(9):1377–1388. doi:10.2217/nnm.13.103 56. Li PC, Wei CW, Liao CK, et al. Photoacoustic imaging of multiple targets using gold nanorods. IEEE Trans Ultrason Ferroelectr Freq Control. 2007;54(8):1642–1647. doi:10.1109/TUFFC.2007.435 57. Venegoni C, Tortorella S, Caliendo A, et al. Urine-Stable Aptamer-Conjugated Gold Nanorods for the Early Detection of High-Grade Bladder Cancer Residual Disease. Adv Healthc Mater. 2025;14(10):e2403314. doi:10.1002/adhm.202403314 58. Alchera E, Monieri M, Maturi M, et al. Early diagnosis of bladder cancer by photoacoustic imaging of tumor-targeted gold nanorods. Photoacoustics. 2022;28:100400. doi:10.1016/j.pacs.2022.100400 59. Canning AJ, Vu T, Menozzi L, et al. Advancing precision photothermal therapy by integrating armored gold nanostars with real-time photoacoustic thermometry and imaging. Sci Adv. 2025;11(33):eadx6350. doi:10.1126/sciadv.adx6350 60. Yang YT, Hsu IL, Cheng TY, et al. Off-Resonance SERS Nanoprobe-Targeted Screen of Biomarkers for Antigens Recognition of Bladder Normal and Aggressive Cancer Cells. Anal Chem. 2019;91(13):8213–8220. doi:10.1021/acs.analchem.9b00775 61. Thompson B, Bhargava KC, Czaja AT, Pan B, Samuelsen BT, Malmstadt N. Spectrophotometry in modular microfluidic architectures. Biomicrofluidics. 2019;13(6):064121. doi:10.1063/1.5124303 62. Sun J, Yang H, Zhuang J, et al. Copper single-atom-based flexible aptamer biochip for simultaneous monitoring of bladder cancer-related bacteria. Mikrochim Acta. 2025;192(9):602. doi:10.1007/s00604-025-07421-5 63. Chen CH, Wu YJ, Chen JJ. Photo-thermal therapy of bladder cancer with Anti-EGFR antibody conjugated gold nanoparticles. Front Biosci. 2016;21(6):1211–1221. doi:10.2741/4451 64. Yang X, Su LJ, La Rosa FG, et al. The Antineoplastic Activity of Photothermal Ablative Therapy with Targeted Gold Nanorods in an Orthotopic Urinary Bladder Cancer Model. Bladder Cancer. 2017;3(3):201–210. doi:10.3233/BLC-170096 65. Armanetti P, Locatelli I, Venegoni C, et al. Gold nanorod-assisted theranostic solution for nonvisible residual disease in bladder cancer. Proc Natl Acad Sci U S A. 2024;121(37):e2411583121. doi:10.1073/pnas.2411583121 66. Yang H, Liu A, Wei M, et al. Visual, Label-Free Telomerase Activity Monitor via Enzymatic Etching of Gold Nanorods. Anal Chem. 2017;89(22):12094–12100. doi:10.1021/acs.analchem.7b02608 67. Cheong JK, Popov V, Alchera E, et al. A numerical study to investigate the effects of tumour position on the treatment of bladder cancer in mice using gold nanorods assisted photothermal ablation. Comput Biol Med. 2021;138:104881. doi:10.1016/j.compbiomed.2021.104881 68. Smilowitz HM, Tarmu LJ, Sanders MM, et al. Biodistribution of gold nanoparticles in BBN-induced muscle-invasive bladder cancer in mice. Int J Nanomed. 2017;12:7937–7946. doi:10.2147/IJN.S140977 69. Jeynes JC, Merchant MJ, Spindler A, Wera AC, Kirkby KJ. Investigation of gold nanoparticle radiosensitization mechanisms using a free radical scavenger and protons of different energies. Phys Med Biol. 2014;59(21):6431–6443. doi:10.1088/0031-9155/59/21/6431 70. Ahrari S, Luo Y, Ning X, et al. Blood Retention and Kidney Clearance of Renal-Clearable Gold Nanoparticles Strongly Correlate with Renal Injury Biomarkers. Angew Chem Int Ed Engl. 2025;64(34):e202509978. doi:10.1002/anie.202509978 71. Marangoni VS, Cancino Bernardi J, Reis IB, Fávaro WJ, Zucolotto V. Photothermia and Activated Drug Release of Natural Cell Membrane Coated Plasmonic Gold Nanorods and β-Lapachone. ACS Appl Bio Mater. 2019;2(2):728–736. doi:10.1021/acsabm.8b00603 72. Lv S, Jing R, Liu X, et al. One-Step Microfluidic Fabrication of Multi-Responsive Liposomes for Targeted Delivery of Doxorubicin Synergism with Photothermal Effect. Int J Nanomed. 2021;16:7759–7772. doi:10.2147/IJN.S329621 73. Zhao H, Zhu H, Du Y, He M, Ding M, Cheng F. Gold nanoparticles/Cu decorated metal-organic frameworks for synergistic photodynamic/ferroptosis cancer therapy. Biomed Mater. 2025;20(2):025042. doi:10.1088/1748-605X/adba2e 74. Guo P, Wang L, Shang W, et al. Intravesical In Situ Immunostimulatory Gel for Triple Therapy of Bladder Cancer. ACS Appl Mater Interfaces. 2020;12(49):54367–54377. doi:10.1021/acsami.0c15176 75. Cho SK, Emoto K, Su LJ, Yang X, Flaig TW, Park W. Functionalized gold nanorods for thermal ablation treatment of bladder cancer. J Biomed Nanotechnol. 2014;10(7):1267–1276. doi:10.1166/jbn.2014.1838 76. Hwang J, San BH, Turner NJ, et al. Molecular assessment of collagen denaturation in decellularized tissues using a collagen hybridizing peptide. Acta Biomater. 2017;53:268–278. doi:10.1016/j.actbio.2017.01.079 77. Daei S, Abbasalipourkabir R, Khanaki K, Bahreini F, Ziamajidi N. Effects of gold nanoparticles on oxidative stress status in bladder cancer 5637 cells. Folia Med. 2022;64(4):641–648. doi:10.3897/folmed.64.e66784 78. Daei S, Ziamajidi N, Abbasalipourkabir R, Khanaki K, Bahreini F. Anticancer Effects of Gold Nanoparticles by Inducing Apoptosis in Bladder Cancer 5637 Cells. Biol Trace Elem Res. 2022;200(6):2673–2683. doi:10.1007/s12011-021-02895-9 79. Xie R, Cheng L, Huang M, et al. NAT10 Drives Cisplatin Chemoresistance by Enhancing ac4C-Associated DNA Repair in Bladder Cancer. Cancer Res. 2023;83(10):1666–1683. doi:10.1158/0008-5472.CAN-22-2233 80. Hong X, Chen X, Wang H, et al. A HER2-targeted Antibody-Drug Conjugate, RC48-ADC, Exerted Promising Antitumor Efficacy and Safety with Intravesical Instillation in Preclinical Models of Bladder Cancer. Adv Sci. 2023;10(32):e2302377. doi:10.1002/advs.202302377 81. Chen X, Huang M, Chen Z, et al. Intravesical Disitamab Vedotin (RC48) for HER2-Expressing High-Risk Non-Muscle-Invasive Bladder Cancer: a Single-Arm, Dose-Escalation Phase I Trial Study. MedComm. 2025;6(7):e70288. doi:10.1002/mco2.70288 82. Tsai YS, Chen YH, Cheng PC, et al. TGF-β1 conjugated to gold nanoparticles results in protein conformational changes and attenuates the biological function. Small. 2013;9(12):2119–2128. doi:10.1002/smll.201202755 83. Terán-Navarro H, Zeoli A, Salines-Cuevas D, et al. Gold Glyconanoparticles Combined with 91–99 Peptide of the Bacterial Toxin, Listeriolysin O, Are Efficient Immunotherapies in Experimental Bladder Tumors. Cancers. 2022;14(10):2413. doi:10.3390/cancers14102413 84. Hsieh DS, Wang H, Tan SW, et al. The treatment of bladder cancer in a mouse model by epigallocatechin-3-gallate-gold nanoparticles. Biomaterials. 2011;32(30):7633–7640. doi:10.1016/j.biomaterials.2011.06.073 85. Wu T, Duan X, Hu C, et al. Synthesis and characterization of gold nanoparticles from Abies spectabilis extract and its anticancer activity on bladder cancer T24 cells. Artif Cells Nanomed Biotechnol. 2019;47(1):512–523. doi:10.1080/21691401.2018.1560305 86. Zhang C, Liu S, Zhang J, et al. A Multifunctional Fe-EGCG@RSL3 Nanomedicine Synergizes Ferroptosis Induction and Tumor Microenvironment Remodeling for Enhanced Bladder Cancer Immunotherapy. Research. 2025;8(6774):0735. doi:10.1126/science.adu2836 87. Guo P, Shang W, Peng L, et al. A gel system for single instillation of non-muscle-invasive bladder Cancer: a “divide-and-rule” strategy. J Control Release. 2018;285:46–55. doi:10.1016/j.jconrel.2018.06.036 88. Gessner I, Park JH, Lin HY, Lee H, Weissleder R. Magnetic Gold Nanoparticles with Idealized Coating for Enhanced Point-Of-Care Sensing. Adv Healthc Mater. 2022;11(2):e2102035. doi:10.1002/adhm.202102035 89. Cho SK, Su LJ, Mao C, Wolenski CD, Flaig TW, Park W. Multifunctional nanoclusters of NaYF(4):Yb(3+),Er(3+) upconversion nanoparticle and gold nanorod for simultaneous imaging and targeted chemotherapy of bladder cancer. Mater Sci Eng C Mater Biol Appl. 2019;97:784–792. doi:10.1016/j.msec.2018.12.113 90. Ramos J, Potta T, Scheideler O, Rege K. Parallel synthesis of poly(amino ether)-templated plasmonic nanoparticles for transgene delivery. ACS Appl Mater Interfaces. 2014;6(17):14861–14873. doi:10.1021/am5017073 91. Berce C, Lucan C, Petrushev B, et al. In vivo assessment of bone marrow toxicity by gold nanoparticle-based bioconjugates in Crl:CD1(ICR) mice. Int J Nanomed. 2016;11:4261–4273. doi:10.2147/IJN.S108711 92. Guelfi G, Stefanetti V, Zampini D, et al. Gold nanoparticles approach to detect chondroitin sulphate and hyaluronic acid urothelial coating. Sci Rep. 2017;7(1):10355. doi:10.1038/s41598-017-09872-0 93. Shal B, Amanat S, Khan AU, et al. Potential applications of PEGylated green gold nanoparticles in cyclophosphamide-induced cystitis. Artif Cells Nanomed Biotechnol. 2022;50(1):130–146. doi:10.1080/21691401.2022.2078340 94. Hortelao AC, Simó C, Guix M, et al. Swarming behavior and in vivo monitoring of enzymatic nanomotors within the bladder. Sci Rob. 2021;6(52). doi:10.1126/scirobotics.abd2823. 95. Mou Y, Liu Z, Xu W, et al. Dual-source powered sea urchin-like nanomotors for intravesical photothermal therapy of bladder cancer. J Nanobiotechnology. 2025;23(1):355. doi:10.1186/s12951-025-03446-3

Rate this article

Login to rate this article

Comments

Please login to comment

No comments yet. Be the first to comment!
    Gold Nanoparticles in Bladder Cancer: New Hope