Next Article in Journal
The Devil Lies in the Details: Small Structural and Chemical Changes in Iron Oxide Pigments Largely Alter the Biological Outcomes in Macrophages
Previous Article in Journal
Large-Scale Production of Silver Nanoplates via Ultrasonic-Assisted Continuous-Flow Synthesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Nanoscale Metal–Organic Frameworks in Hepatocellular Carcinoma

1
Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz 51666-16659, Iran
2
Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada
*
Authors to whom correspondence should be addressed.
Nanomaterials 2025, 15(23), 1771; https://doi.org/10.3390/nano15231771
Submission received: 11 November 2025 / Revised: 19 November 2025 / Accepted: 21 November 2025 / Published: 26 November 2025
(This article belongs to the Special Issue Nanoscale Metal–Organic Frameworks for Cancer Therapy)

Abstract

Hepatocellular carcinoma (HCC) represents the predominant type of primary liver cancer and remains a major global health concern. Current therapeutic strategies—such as surgical resection, radiation, and chemotherapy—provide clinical benefits but are frequently accompanied by considerable adverse effects. Consequently, identifying alternative treatment modalities and developing strategies that allow the use of lower drug doses without compromising therapeutic outcomes are essential goals in HCC management. Among emerging nanoscale platforms, metal–organic frameworks (MOFs) have attracted exceptional interest as promising candidates for targeted drug delivery in cancer therapy. Their inherent characteristics, including highly ordered porosity, large surface area, tunable cavities, adjustable chemical functionality, and remarkable drug-loading capacity, set them apart from conventional porous nanomaterials. Owing to their hierarchical architecture, MOFs are especially suitable for multimodal and synergistic anti-cancer treatments. MOF-based systems have demonstrated the ability to reinforce the performance of several therapeutic modalities, including photodynamic therapy (PDT), photothermal therapy (PTT), chemodynamic therapy (CDT), and sonodynamic therapy (SDT), while also serving as efficient carriers for targeted drug release. Their structural versatility further enables improved drug stability, enhanced solubility, and controlled-release behavior. This review provides an overview of recent progress in MOF-enabled therapeutic strategies and discusses their potential applications in the treatment of HCC.

1. Introduction

The World Health Organization identifies cancer as one of the most serious global threats, with profound impacts on health systems, societies, and economies. It accounts for approximately 16.8% of all deaths and nearly one-quarter of mortality attributed to noncommunicable diseases worldwide [1]. Among various malignancies, liver cancer ranks sixth in incidence and third in cancer-related mortality, placing a significant burden on healthcare resources across the world [2]. Projections indicate that, if current patterns persist, annual liver cancer diagnoses will rise dramatically—from 0.87 million reported in 2022 to an estimated 1.52 million by 2050.
Hepatocellular carcinoma (HCC), the predominant subtype of primary liver cancer, comprises nearly 80% of cases globally. In 2020 alone, over 900,000 new diagnoses were recorded, corresponding to an age-standardized incidence rate (ASIR) of 7.3 per 100,000 individuals [3,4]. Epidemiological data consistently reveal striking regional disparities in HCC occurrence, with particularly high incidence levels observed in East Asia and sub-Saharan Africa, while lower—yet steadily increasing—rates are reported in North America and much of Europe. These geographic differences largely stem from the complex, multifactorial origins of HCC, especially varying prevalence of key etiological factors such as chronic hepatitis B virus (HBV) and hepatitis C virus (HCV) infections, which significantly shape regional incidence trends [4].
Standard therapeutic options for HCC primarily rely on surgical intervention, radiotherapy, and chemotherapy. While conventional anti-cancer agents can exhibit therapeutic benefits, their use is frequently limited by severe adverse effects, largely because cytotoxic drugs are unable to selectively target malignant cells and inevitably damage surrounding healthy tissues [5,6]. Moreover, early-stage HCC often progresses without noticeable symptoms, resulting in many individuals being diagnosed only after the disease has reached an advanced or unresectable stage, at which point curative procedures such as surgery or liver transplantation are no longer viable. For patients with advanced HCC, treatments such as chemotherapy and radiotherapy may help prolong survival and improve overall quality of life; however, their clinical performance is often hindered by poor tumor responsiveness, intrinsic or acquired drug resistance, and diminished radiotherapeutic sensitivity. Furthermore, the need for high therapeutic doses frequently causes substantial toxicity, which can reduce patient adherence to treatment and worsen clinical outcomes [7,8,9]. As a result, there is an urgent need to explore innovative therapeutic modalities and to develop strategies that allow effective treatment at lower drug doses without compromising efficacy [10,11,12].
In recent years, drug delivery systems (DDSs) have attracted significant interest because of their ability to enhance cancer therapy through controlled drug release, site-specific delivery, and improved overall treatment performance [13,14,15]. Advances in nanoscience and nanomedicine have further accelerated the development of innovative therapeutic platforms for cancer management [16,17,18]. A wide spectrum of nanomaterials has been evaluated for targeted drug administration, ranging from inorganic systems—such as mesoporous silica, graphene derivatives, iron oxide nanoparticles (NPs), and quantum dots—to organic carriers like liposomes and polymeric micelles, as well as hybrid organic–inorganic constructs that merge the strengths of both material classes [19,20,21]. Among these technologies, micro- and nanohydrogels have become particularly attractive candidates for drug delivery owing to their customizable porosity, excellent biocompatibility, high water retention capacity, and sensitivity to physiological stimuli including pH, temperature, and enzymatic activity. These features enable them to deliver therapeutic agents in a sustained, localized, and stimulus-responsive manner during cancer treatment [22,23,24,25,26,27,28,29]. Hydrogels are capable of encapsulating a broad range of drugs—whether hydrophilic or hydrophobic—shielding them from premature degradation and providing efficient loading and controlled-release behavior [30,31,32,33]. Their adaptable structure also allows incorporation with diverse components such as polymers, NPs, or metal–organic frameworks (MOFs), thereby improving therapeutic outcomes and enabling multifunctional treatment modalities [34,35,36]. Despite these benefits, the advancement of nanomaterial-based DDSs still faces several barriers. Organic nanocarriers may struggle to achieve highly precise, predictable release profiles and can lack the distinctive physicochemical characteristics needed for multifunctional cancer therapy. In contrast, inorganic materials often exhibit limited biodegradability, restricted chemical tunability, and potential biocompatibility concerns. Therefore, selecting an optimal material platform remains one of the central challenges in the design of effective drug delivery systems [37,38,39,40].
MOFs have recently emerged as one of the most promising classes of nanomaterials for targeted drug delivery in cancer therapy, largely due to their exceptional structural features. MOFs are crystalline porous materials composed of metal ions or metal-containing clusters (such as Fe, Mn, Co, Al, Cu, or Zn) connected to organic linkers—including carboxylate, phosphonate, borate, tetrazolate, sulfonate, or imidazolate groups—forming one-, two- or three-dimensional architectures through coordination bonds [41,42,43,44,45,46]. Often referred to as porous coordination polymers (PCPs), MOFs have attracted considerable interest in both fundamental research and practical engineering applications because of their highly ordered structures and versatile chemical properties. These frameworks have been applied across numerous fields, such as gas storage and separation, water splitting, water treatment, catalysis, sensing, energy-related technologies, bioimaging, and targeted therapeutic delivery [47,48]. Their hybrid organic–inorganic nature provides a combination of advantages, including biodegradability, biocompatibility, and safety, which are crucial for biomedical use. Furthermore, MOFs offer exceptionally high porosity, large surface areas, adjustable pore dimensions, and modifiable chemical functionalities, making them ideal candidates for high-capacity drug loading and controlled-release applications [49,50].
Compared with conventional porous nanoparticles, the hierarchical architecture of metal–organic frameworks (MOFs) has been shown to significantly improve their performance in multimodal anti-cancer therapies. As a result, MOFs have become central to the design of advanced tumor-targeted drug delivery systems [50,51,52]. These materials can substantially enhance the effectiveness of various therapeutic strategies, including photodynamic therapy (PDT), photothermal therapy (PTT), chemodynamic therapy (CDT), sonodynamic therapy (SDT), as well as precise drug delivery [53,54,55,56]. The tunable structure of MOFs also makes them highly suitable as drug carriers, offering abundant binding sites, increased stability, enhanced solubility, and the ability to release therapeutic agents in a controlled manner [57,58,59,60]. Additionally, MOFs can transport a wide range of therapeutic molecules—including chemotherapeutic drugs, photosensitizers, and radiosensitizers—directly to tumor tissues, thereby improving treatment outcomes while minimizing unintended side effects on healthy cells [61,62,63,64,65].

2. Metal–Organic Frameworks-Based Therapies for Hepatocellular Carcinoma

With their precisely defined architectures, MOFs can induce tumor cell death through SDT, PDT, PTT, or CDT. This section highlights the therapeutic potential of MOFs in HCC, with a particular focus on PDT, PTT, CDT, and SDT.

2.1. Metal–Organic Frameworks for Photodynamic Therapy of Cancer

PDT, which utilizes photosensitizers (PSs) activated by light at specific wavelengths to produce reactive oxygen species (ROS), has recently gained attention as a non-invasive alternative to traditional cancer treatments. PDT is regarded as a precise and safe therapeutic approach, as it selectively targets malignant cells while minimizing damage to healthy tissues [66,67]. In MOF-mediated PDT, PS-loaded MOFs—such as porphyrin-based frameworks—are directed to tumor sites and subsequently exposed to light of the appropriate wavelength to activate the photosensitizer. The activated PS does not directly attack biomolecules; instead, it transfers energy from light to molecular oxygen, generating ROS such as singlet oxygen (1O2), superoxide radicals (O2−•), hydroxyl radicals (HO), and hydrogen peroxide (H2O2), which induce tumor cell death and tissue destruction [68]. Unlike chemotherapy or radiotherapy, which can cause systemic toxicity, the ROS produced during PDT are confined to the target site and do not exert harmful effects on the rest of the body. Due to these advantages—non-invasiveness, localized activity, and minimal side effects—PDT has been widely employed for treating superficial tumors [68,69].
Liu et al. [70] synthesized nanoscale porphyrin-based MOFs (NPMOFs) by combining biocompatible zirconium ions (Zr4+) with meso-tetrakis(4-carboxyl)-21H,23H-porphine (TCPP) using a microemulsion approach. Doxorubicin (DOX) was employed as a model chemotherapeutic agent to evaluate both its loading capacity and pH-responsive release from the NPMOFs. In vitro studies using HepG2 liver cancer cells demonstrated that the extensive pore network of NPMOFs enabled DOX encapsulation levels up to 109%. When combined with 655 nm laser irradiation, DOX@NPMOFs exhibited a low half-maximal inhibitory concentration (IC50 = 67.72 µg mL−1) and induced approximately 90% cell death. The in vivo efficacy of the combined chemotherapy and PDT approach was further investigated in HepG2 tumor-bearing mice. Fluorescence imaging was used to monitor NPMOF accumulation at tumor sites, and once maximal localization was achieved, tumors in PDT groups were irradiated with a 655 nm laser (180 J cm−2 for 15 min). Tumor progression was then compared across groups receiving monotherapy, combination therapy, or saline control. While tumor growth was partially suppressed in groups treated with either chemotherapy or PDT alone, mice receiving DOX@NPMOFs co-therapy exhibited marked tumor regression within two days. Notably, two tumors were completely eradicated, while the remaining two shrank from 62.5 mm3 to 2 mm3 over 10 days. These findings indicate that DOX@NPMOFs were efficiently internalized by cancer cells and exerted potent cytotoxic effects when applied in combination therapy. Importantly, no observable damage to surrounding skin or tissues was reported following PDT treatment.
As illustrated in Figure 1A, Wang et al. [71] developed a multifunctional MOF-based nanoplatform (PS@MOF-199 NPs) for image-guided PDT. They first synthesized MOF-199, a Cu(II) carboxylate-based framework, and incorporated two PSs—a commercial one, chlorin e6 (Ce6), and a synthesized aggregation-induced emission (AIE) compound, 2-[4-(dimethylamino)phenyl]anthracene-9,10-dione (TPAAQ)—via physical adsorption to obtain PS@MOF-199. To improve dispersibility and biocompatibility, the composite was further encapsulated with polyether F127 through a thin-film dispersion process, yielding PS@MOF-199 NPs. As shown in Figure 1B, the nanoplatform remains photosensitization-inactive when outside cells, since the intact MOF-199 framework restricts oxygen access to the encapsulated PSs. After cellular internalization through the enhanced permeability and retention (EPR) effect, intracellular glutathione (GSH) induces the degradation of MOF-199, releasing TPAAQ or Ce6 to react with O2 and generate 1O2 under light irradiation for PDT. Simultaneously, Cu2+ ions released from the decomposed MOF deplete endogenous GSH through redox reactions, thereby amplifying the ROS-mediated cytotoxicity. The resulting PS@MOF-199 NPs displayed potent phototoxicity toward HepG2 cells and significant tumor suppression in a transgenic zebrafish liver tumor model following irradiation. This approach effectively integrates AIE and conventional PSs within a degradable MOF carrier, achieving oxygen-responsive, image-guided PDT with enhanced therapeutic efficacy and minimal nonspecific phototoxicity.
Cai et al. [72] developed photosensitizer-loaded MOF nanoparticles (PS-MOF NPs) designed to eliminate residual tumor cells following PDT by combining PDT with antihypoxic modulation and immunostimulatory agents, effectively functioning as an in situ tumor vaccine to boost long-term anti-tumor immune responses. The MOF NPs, referred to as PCN224 (PCN), were self-assembled from H2TCPP (photosensitizer) and zirconium ions through coordination interactions, incorporating a hypoxia-inducible factor (HIF) signaling inhibitor (ACF) and the immunologic adjuvant CpG, with a hyaluronic acid (HA) coating applied to the surface (PCN-ACF-CpG@HA). The PCN component within the NPs demonstrated strong PDT-mediated cytotoxicity against tumor cells. Meanwhile, ACF effectively suppressed HIF-1α-driven survival and metastatic pathways that are typically activated post-PDT. Importantly, the combination of CpG with tumor-associated antigens (TAAs) released from damaged tumor cells acted as a localized tumor vaccine, further stimulating the host’s immune system to target and eliminate residual malignant cells. Collectively, this integrated approach represents a promising strategy to synergistically enhance PDT outcomes while simultaneously invoking anti-tumor immunity.
Fu et al. [46] developed a MOF-based nanocomposite incorporating Ce6 to enhance its PDT performance under physiological conditions. Although Ce6 is a widely used second-generation photosensitizer, its clinical utility is limited by poor water solubility, tendency to aggregate in biological media, and insufficient accumulation in tumor tissues, which reduce its therapeutic efficiency. To address these limitations, the researchers employed an appropriate drug delivery carrier. They designed a therapeutic agent by incorporating Ce6 into a zeolitic imidazolate framework-8 (ZIF-8) modified with hyaluronic acid (HA), creating the ZIF-8@Ce6–HA nanocomposite via a one-pot self-assembly approach. ZIF-8, a subclass of MOFs composed of zinc ions coordinated with 2-methylimidazole, is well-suited for targeted drug delivery because it can degrade in the acidic tumor microenvironment. The strong electrostatic interactions between Zn2+ ions and Ce6 molecules enabled high loading efficiency of Ce6 within the MOF structure. Surface modification with HA improved the biocompatibility of the nanocarrier and reduced systemic toxicity. Characterization revealed that ZIF-8@Ce6–HA exhibited high encapsulation efficiency, effective cellular uptake, and excellent biocompatibility. Mass spectrometry analysis indicated that HA modification prolonged blood circulation time and minimized systemic side effects. In vitro studies with HepG2 liver cancer cells showed that free Ce6 caused only about 29.5% cell death following irradiation due to ROS generation, limited by aggregation in aqueous media. In contrast, ZIF-8@Ce6–HA significantly enhanced PDT efficacy, resulting in approximately 88.4% cell death after irradiation. These results demonstrate that ZIF-8@Ce6–HA is a promising and effective platform for photodynamic therapy.
Hu et al. [47] engineered a targeted, synergistic therapy combining PDT and chemotherapy using a glyco-modified MOF nanostructure, termed DOX@Gal-PCN-224, administered via percutaneous transperitoneal puncture, to treat HCC in a mouse model of in situ liver tumors. The chemotherapeutic agent DOX was incorporated into the MOF during a one-pot synthesis, enhancing drug loading and enabling pH-responsive release. Furthermore, the cytotoxic effect of DOX was amplified by local hypoxia resulting from PDT-induced oxygen consumption. The NPs were functionalized with galactose to selectively target the asialoglycoprotein receptor (ASGPR), which is highly expressed on hepatocytes. This targeting capability was confirmed in HepG2 and Huh7 cell lines, demonstrating reduced off-target toxicity in healthy tissues. The intrinsic fluorescence of both DOX and the photosensitizer allowed real-time imaging to monitor NP distribution in both subcutaneous and orthotopic tumor models, confirming effective liver-specific delivery. Following a single interventional PDT irradiation, tumor growth in orthotopic HCC-bearing mice was suppressed by approximately 98%, highlighting the strong potential of this combined interventional PDT and chemotherapy strategy for future clinical applications in HCC treatment.
Pan et al. [48] developed a bimetallic MOF nanoplatform, ZMRPC@HA, containing ruthenium (Ru3+) and platinum (Pt4+) ions, designed to modulate the tumor microenvironment and enhance PDT for hypoxic tumors. The MOFs were loaded with the photosensitizer Ce6 and surface-modified with HA to improve biocompatibility and tumor targeting. ZMRPC@HA exhibited catalase- and glutathione oxidase-like activities, primarily due to Ru3+, enabling the generation of oxygen and depletion of glutathione within the tumor, thereby synergistically improving PDT efficacy under hypoxic conditions. The PDT performance of ZMRPC@HA was first evaluated in vitro using Hepatoma 22 (H22) cells, demonstrating significant inhibition of cellular proliferation. Subsequent in vivo studies in BALB/c mice bearing H22 tumor xenografts confirmed that ZMRPC@HA effectively suppressed tumor growth under 660 nm laser irradiation. Both in vitro and in vivo results indicated that this MOF-based PDT approach could efficiently hinder tumor cell differentiation and proliferation even in deep tissue regions, highlighting its potential as a potent strategy for treating hypoxic tumors.

2.2. Metal–Organic Frameworks for Photothermal Therapy of Cancer

PTT is recognized for its non-invasive nature, localized therapeutic effects, high efficiency, and minimal side effects. In PTT, specific MOFs absorb near-infrared (NIR) light and convert it into heat, generating localized hyperthermia that selectively destroys tumor cells. Essentially, PTT can be considered an extension of photodynamic therapy (PDT), where a photosensitizer absorbs light within a specific wavelength range and transforms optical energy into thermal energy, allowing precise ablation of malignant cells while sparing adjacent healthy tissues [49,50]. Furthermore, combining PDT and PTT has been shown to produce synergistic antitumor effects with reduced side effects. Heat generated during PTT can improve local blood circulation, thereby increasing oxygen availability in tumor tissues and enhancing the efficacy of oxygen-dependent PDT. Simultaneously, ROS produced during PDT can disrupt tumor physiology and alter the microenvironment, which further sensitizes cancer cells to hyperthermia and improves the overall therapeutic outcome [51,52,53].
Shi and colleagues [54] developed a novel theranostic nanoparticle system (MINPs) through a straightforward self-assembly approach, incorporating manganese ions (Mn2+) and indocyanine green (ICG), stabilized by poly(vinylpyrrolidone) (PVP). In vitro studies demonstrated that MINPs significantly inhibited the proliferation of HepG2 liver cancer cells when exposed to 808 nm laser irradiation. For in vivo evaluation, MINPs were administered to mice bearing subcutaneous tumors, and the photoacoustic (PA) signal around the tumor site was monitored for 12 h. The PA signal in the MINP-treated group was approximately three times higher than that in the control group receiving free ICG, indicating enhanced accumulation. Similarly, magnetic resonance imaging (MRI) revealed amplified positive tumor signals over time, with quantitative analysis showing that MRI intensity at 12 h post-injection was 1.8 times greater than pre-injection levels. Both in vitro PTT experiments and in vivo imaging confirmed that MINP treatment induced significant necrosis and tumor cell damage in HepG2 tumors, whereas control groups displayed negligible effects. These findings suggest that MINPs represent a promising platform for multimodal imaging-guided PTT of hepatocellular carcinoma, offering potential for future clinical applications.
Zhou et al. [34] developed HCC-targeted NPs by conjugating the SP94 peptide and cyanine 5.5 (Cy5.5) to Prussian blue (PB) NPs, which were loaded with the chemotherapeutic drug sorafenib (SF), forming SP94-PB-SF-Cy5.5 NPs. These FDA-approved nanoparticles were designed for HCC-specific multimodal imaging and combined PTT with targeted chemotherapy. The researchers tested the NPs in both human (HepG2) and mouse (Hepa1–6) HCC cell lines and assessed their imaging and therapeutic efficacy in vitro and in vivo. The SP94-PB-SF-Cy5.5 NPs accumulated preferentially at HCC tumor sites and were suitable for near-infrared fluorescence (NIRF), photoacoustic imaging (PAI), and MRI, enabling triple-modality imaging. The NPs exhibited strong photothermal effects and facilitated controlled release of SF, effectively eradicating tumors with minimal local recurrence and low systemic toxicity. Additionally, the catalase-like activity of PB enhanced oxygen generation, alleviating tumor hypoxia. The combination of PTT and hypoxia mitigation synergistically promoted an immune-stimulating tumor microenvironment. When combined with anti-PD-L1 monoclonal antibody therapy, SP94-PB-SF-Cy5.5 NPs under NIR irradiation produced notable abscopal effects, significantly reducing metastasis and tumor recurrence. This combinational approach also induced long-term antitumor immune memory, preventing tumor regrowth. Overall, these multifunctional and safe NPs offer a promising strategy for effective HCC treatment.

2.3. Metal–Organic Frameworks for Chemodynamic Therapy of Cancer

CDT is an emerging cancer treatment approach that utilizes CDT agents to convert H2O2 into highly reactive HO via Fenton or Fenton-like reactions, thereby inducing tumor cell apoptosis and necrosis. In MOF-catalyzed Fenton reactions, Fe2+ ions within the MOF facilitate the transformation of H2O2 into HO, causing oxidative damage specifically in cancer cells. Additionally, other metal ions such as Cu+, Mn2+, Co2+, and Ti3+ have been reported to mediate similar Fenton-like reactions, generating HO from H2O2 [73]. Combining CDT with PDT can produce synergistic antitumor effects. The oxygen generated during CDT can enhance the effectiveness of PDT, while the potent cytotoxicity of HO directly kills tumor cells, further amplifying tumor suppression. Recent research has systematically explored this combination, demonstrating that the concurrent application of PDT and CDT increases oxidative stress in tumors and achieves superior therapeutic outcomes compared to either monotherapy alone [74,75].
Ding et al. [76] synthesized iron-based MOF nanoparticles (FeMOFs NPs) using TCPP (Fe) and zirconium clusters as structural units. The hydrophobic chemotherapeutic agent camptothecin (CPT) was incorporated, and small gold (Au) NPs were deposited on the surface in situ, resulting in the novel PEG-Au/FeMOF@CPT NPs. The surface Au NPs were further functionalized with 1-dodecanethiol (C12SH) and methoxy polyethylene glycol thiol (PEG-SH) to enhance stability and biocompatibility. The antitumor activity of CDT was assessed in HepG2 cells using the MTT (3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyl tetrazolium bromide) assay. For CPT monotherapy, the IC50 was 206 ± 22 × 10−9 M. Treatment with PEG-Au/FeMOF NPs, combining cascade starvation and CDT, significantly inhibited cell proliferation with an IC50 of 3.51 ± 0.26 µg mL−1. The combination of chemotherapy and CDT using PEG-Au/FeMOF@CPT NPs yielded the lowest IC50 value at 0.31 ± 0.04 µg mL−1. These findings indicate a strong synergistic effect between CDT and CPT, effectively enhancing tumor growth suppression.

2.4. Metal–Organic Frameworks for Sonodynamic Therapy of Cancer

SDT is an emerging ultrasound-based therapeutic strategy that generates ROS through acoustic cavitation and sonochemical reactions. Compared with PDT, SDT offers deeper tissue penetration, non-invasive tumor ablation, and high therapeutic precision, making it particularly suitable for HCC. However, its efficacy is often limited by insufficient ROS production and tumor hypoxia. The incorporation of MOFs as sonosensitizers or catalytic carriers has shown significant potential in overcoming these limitations and improving therapeutic outcomes [77,78,79,80,81,82].
Porphyrinic MOFs such as Mn–porphyrin and Cu–TCPP frameworks possess intrinsic sonosensitizing properties due to their conjugated ligands and redox-active metal centers, enabling efficient ultrasound-triggered ROS generation. In HepG2 and H22 hepatoma models, these nanosystems markedly enhanced oxidative stress and mitochondrial damage, resulting in significant antitumor effects [80,83,84]. Similarly, ZIF-8- and Fe-MOF-based nanocomposites loaded with Ce6 or hematoporphyrin have exhibited pH-responsive degradation and strong ROS formation under ultrasound, achieving high cytotoxicity against HCC cells in vitro and tumor suppression in vivo [85,86].
Some MOF platforms designed for PDT, including DOX@Gal-PCN-224 and ZMRPC@HA, have also shown efficient ultrasound-induced ROS generation and oxygen-regulating activity, indicating their potential as dual PDT/SDT nanoplatforms for liver cancer [47,48,87]. Overall, MOF-based nanosonosensitizers integrate oxygen generation, GSH depletion, and ROS amplification, offering a versatile and biocompatible platform for precise, ultrasound-activated therapy of HCC [88,89].

3. Conclusions and Future Directions

The design of functional MOFs for cancer phototherapy has emerged as a rapidly advancing area of research. These phototherapeutic MOFs can be constructed directly by coordinating PSs or photothermal agents with metal ions or clusters. In addition, their tunable pore sizes make MOFs excellent carriers for phototherapeutic agents. The highly ordered frameworks prevent aggregation of PSs and photothermal agents, thereby markedly improving their therapeutic efficiency. MOFs can also be easily functionalized with small molecules, biomacromolecules, or other active materials, enabling targeted delivery, imaging-guided treatment, and synergistic therapeutic strategies [90,91,92]. Nevertheless, despite these advantages, significant challenges must still be addressed before MOFs can advance to clinical trials.
(1)
Currently available phototherapeutic agents face notable limitations, such as restricted tissue penetration of light used in phototherapy, dependence on oxygen for ROS generation in PDT, and non-uniform heat distribution in PTT. Although strategies like upconversion NPs or two-photon-activated photosensitizers can enhance light penetration, targeting deep organs like the liver remains difficult. As a result, imaging-guided approaches have become a critical component in phototherapy. Minimally invasive laparoscopic techniques allow optical fibers to access deeper tissues, activating photosensitizers for effective treatment. Such methods have already advanced to clinical applications and show great promise for precision cancer theranostics. Additionally, light-independent modalities, including chemiluminescence and bioluminescence, offer alternative means to overcome the limitations associated with light penetration [50,93,94].
(2)
To improve the biosafety of MOFs as drug carriers, it is important to use biocompatible ligands and metal ions, such as Fe, Ca, and Zn. Surface modifications using peptides, antibodies, or small molecules have proven effective for enhancing tumor targeting and accumulation. Despite these advances, the pharmacokinetics of many MOF systems remain inadequately studied. Once MOFs reach the tumor site, they should degrade efficiently into small molecules or ions that can be metabolized safely, minimizing adverse effects. Although various stimuli—including pH, biothiols, ATP, and hypoxic conditions—have been investigated to trigger MOF degradation, their effectiveness requires further optimization [95,96,97,98,99,100,101].
In summary, precisely engineered and functional MOFs have demonstrated potential for PDT, PTT, SDT, and CDT in cancer therapy [102]. Table 1 summarizes key MOF-based nanosystems for PDT, PTT, and CDT in hepatocellular carcinoma. While this research area is still emerging, ongoing developments are expected to produce increasingly sophisticated MOFs suitable for clinical translation.

Author Contributions

Conceptualization, H.T. and H.N.; Writing—review and editing, H.T., H.N. and S.R.; Writing—original draft, H.T. and H.N.; Resource, S.R.; Supervision, S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ACF, hypoxia-inducible factor (HIF) signaling inhibitor; ACQ, aggregation-caused quenching; AIE, aggregation-induced emission; ASGPR, asialoglycoprotein receptor; ASIR, age-standardized incidence rate; ATP, adenosine triphosphate; Au, gold; C12SH, 1-dodecanethiol; CDT, chemodynamic therapy; Ce6, chlorin e6; CPT, camptothecin; Cy, cyanine; DDSs, drug delivery systems; DOX, doxorubicin; EPR, enhanced permeability and retention; Fe, iron; FeMOF, iron-based metal–organic framework; F127, polyether F127; GSH, glutathione; HA, hyaluronic acid; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; HIF, hypoxia-inducible factor; HO, hydroxyl radical; H2O2, hydrogen peroxide; IC50, half-maximal inhibitory concentration; ICG, indocyanine green; IGTSs, imaging-guided therapy systems; MRI, magnetic resonance imaging; MINPs, manganese-based indocyanine green nanoparticles; MOF, metal–organic framework; MOFs, metal–organic frameworks; MS, mass spectrometry; NIR, near-infrared; NIRF, near-infrared fluorescence; NP, nanoparticle; NPs, nanoparticles; NPMOFs, nanoscale porphyrinic metal–organic frameworks; PA, photoacoustic; PAI, photoacoustic imaging; PCN, porous coordination network; PEG, polyethylene glycol; PEG-SH, methoxy polyethylene glycol thiol; PDT, photodynamic therapy; PD-L1, programmed death-ligand 1; PHS, photosensitizer; PHS@MOF, photosensitizer-loaded metal–organic framework; PVP, polyvinylpyrrolidone; Pt4+, platinum; PTT, photothermal therapy; ROS, reactive oxygen species; Ru3+, ruthenium; 1O2, singlet oxygen; SF, sorafenib; SDT, sonodynamic therapy; SP94, HCC-targeting peptide SP94; O2−•, superoxide radicals, TAAs, tumor-associated antigens; TCPP, meso-tetrakis(4-carboxyl)-21H,23H-porphine; TPAAQ, 2-[4-(dimethylamino)phenyl]anthracene-9,10-dione; Ti3+, titanium; ZIF-8, zeolitic imidazolate framework-8; ZMRPC, bimetallic MOF nanoplatform; Zr, zirconium.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Tutunchi, H.; Naeini, F.; Mobasseri, M.; Ostadrahimi, A. Triglyceride Glucose (TyG) Index and the Progression of Liver Fibrosis: A Cross-Sectional Study. Clin. Nutr. ESPEN 2021, 44, 483–487. [Google Scholar] [CrossRef] [PubMed]
  3. Tutunchi, H.; Arefhosseini, S.; Nomi-Golzar, S.; Ebrahimi-Mameghani, M. Effects of Hydroxycitric Acid Supplementation on Body Composition, Obesity Indices, Appetite, Leptin, and Adiponectin of Women with NAFLD on a Calorie-Restricted Diet. Int. J. Clin. Pract. 2023, 2023, 6492478. [Google Scholar] [CrossRef] [PubMed]
  4. Venook, A.P.; Papandreou, C.; Furuse, J.; de Guevara, L.L. The Incidence and Epidemiology of Hepatocellular Carcinoma: A Global and Regional Perspective. Oncologist 2010, 15 (Suppl. 4), 5–13. [Google Scholar] [CrossRef]
  5. Zafar, A.; Khatoon, S.; Khan, M.J.; Abu, J.; Naeem, A. Advancements and Limitations in Traditional Anti-Cancer Therapies: A Comprehensive Review of Surgery, Chemotherapy, Radiation Therapy, and Hormonal Therapy. Discov. Oncol. 2025, 16, 607. [Google Scholar] [CrossRef]
  6. Wang, K.; Tepper, J.E. Radiation Therapy-Associated Toxicity: Etiology, Management, and Prevention. CA Cancer J. Clin. 2021, 71, 437–454. [Google Scholar] [CrossRef]
  7. Wang, W.; Wei, C. Advances in the Early Diagnosis of Hepatocellular Carcinoma. Genes Dis. 2020, 7, 308–319. [Google Scholar] [CrossRef]
  8. Feng, Y.; Wu, W.; Li, M. Metal–Organic Frameworks for Hepatocellular Carcinoma Therapy and Mechanism. Front. Pharmacol. 2022, 13, 1025780. [Google Scholar] [CrossRef]
  9. Tutunchi, H.; Ebrahimi-Mameghani, M.; Hosseinzadeh-Attar, M.J.; Roshanravan, N.; Mobasseri, M.; Najafipour, F.; Naeini, F.; Naghshi, S.; Asghari, S.; Akbarzadeh, M.; et al. Effects of Oleoylethanolamide Supplementation on the Expression of Lipid Metabolism-Related Genes and Serum NRG4 Levels in Patients with Non-Alcoholic Fatty Liver Disease: A Randomized Controlled Trial. Clin. Nutr. ESPEN 2023, 58, 311–319. [Google Scholar] [CrossRef] [PubMed]
  10. Kong, J.; Cai, M.; Zhu, R.; Zhang, Y.; Du, Y.; Jing, X.; Sun, Y.; Chang, R.; Qu, C.; Dong, X.; et al. The Utilization of Metal–Organic Frameworks in Tumor-Targeted Drug Delivery Systems. J. Sci. Adv. Mater. 2024, 9, 100770. [Google Scholar] [CrossRef]
  11. Linnane, E.; Haddad, S.; Melle, F.; Mei, Z.; Fairen-Jimenez, D. The Uptake of Metal–Organic Frameworks: A Journey into the Cell. Chem. Soc. Rev. 2022, 51, 6065–6086. [Google Scholar] [CrossRef]
  12. Wang, Y.; Yan, J.; Wen, N.; Xiong, H.; Cai, S.; He, Q.; Hu, Y.; Peng, D.; Liu, Z.; Liu, Y. Metal–Organic Frameworks for Stimuli-Responsive Drug Delivery. Biomaterials 2020, 230, 119619. [Google Scholar] [CrossRef]
  13. Pacheco, C.; Baião, A.; Ding, T.; Cui, W.; Sarmento, B. Recent Advances in Long-Acting Drug Delivery Systems for Anticancer Drug. Adv. Drug Deliv. Rev. 2023, 194, 114724. [Google Scholar] [CrossRef] [PubMed]
  14. Li, K.; Guo, B.; Gu, J.; Ta, N.; Yu, H.; Sun, M.; Han, T. Emerging Advances in Drug Delivery Systems (DDSs) for Optimizing Cancer Complications. Mater. Today Bio 2024, 30, 101375. [Google Scholar] [CrossRef]
  15. Chen, Q.; Yang, Z.; Liu, H.; Man, J.; Oladejo, A.O.; Ibrahim, S.; Wang, S.; Hao, B. Novel Drug Delivery Systems: An Important Direction for Drug Innovation Research and Development. Pharmaceutics 2024, 16, 674. [Google Scholar] [CrossRef] [PubMed]
  16. Singh, K.; Singhal, S.; Pahwa, S.; Arora Sethi, V.; Sharma, S.; Singh, P.; Kale, R.D.; Wazed Ali, S.; Sagadevan, S. Nanomedicine and Drug Delivery: A Comprehensive Review of Applications and Challenges. Nano-Struct. Nano-Objects 2024, 40, 101403. [Google Scholar] [CrossRef]
  17. Malik, S.; Muhammad, K.; Waheed, Y. Emerging Applications of Nanotechnology in Healthcare and Medicine. Molecules 2023, 28, 6624. [Google Scholar] [CrossRef]
  18. Xu, M.; Han, X.; Xiong, H.; Gao, Y.; Xu, B.; Zhu, G.; Li, J. Cancer Nanomedicine: Emerging Strategies and Therapeutic Potentials. Molecules 2023, 28, 5145. [Google Scholar] [CrossRef]
  19. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  20. Basu, S.; Biswas, P.; Anto, M.; Singh, N.; Mukherjee, K. Nanomaterial-Enabled Drug Transport Systems: A Comprehensive Exploration of Current Developments and Future Avenues in Therapeutic Delivery. 3 Biotech 2024, 14, 289. [Google Scholar] [CrossRef]
  21. Egwu, C.O.; Aloke, C.; Onwe, K.T.; Umoke, C.I.; Nwafor, J.; Eyo, R.A.; Chukwu, J.A.; Ufebe, G.O.; Ladokun, J.; Audu, D.T.; et al. Nanomaterials in Drug Delivery: Strengths and Opportunities in Medicine. Molecules 2024, 29, 2584. [Google Scholar] [CrossRef] [PubMed]
  22. Davodabadi, F.; Sargazi, S.; Baino, F. Recent Advances in Hydrogel-Based Drug Delivery Systems for Enhanced Cancer Therapy: A Review. Mater. Today Commun. 2025, 48, 113615. [Google Scholar] [CrossRef]
  23. Baweja, R.; Ravi, R.; Baweja, R.; Gupta, S.; Sachan, A.; Pratyusha, V.; Warsi, M.K.; Purohit, S.D.; Mishra, A.; Ahmad, R. A Comprehensive Review of Hydrogels as Potential Drug Carriers for Anticancer Therapies: Properties, Development and Future Prospects. Next Mater. 2025, 8, 100913. [Google Scholar] [CrossRef]
  24. Omer, A.M.; Abd El-Monaem, E.M.; Eltaweil, A.S.; Tamer, T.M.; Mohy Eldin, M.S.; Ouyang, X.-K.; Heydari, A. Advances in Stimuli-Responsive Polymeric Hydrogels for Anticancer Drug Delivery: A Review. J. Drug Deliv. Sci. Technol. 2024, 102, 106394. [Google Scholar] [CrossRef]
  25. Delgado-Pujol, E.J.; Martínez, G.; Casado-Jurado, D.; Vázquez, J.; León-Barberena, J.; Rodríguez-Lucena, D.; Torres, Y.; Alcudia, A.; Begines, B. Hydrogels and Nanogels: Pioneering the Future of Advanced Drug Delivery Systems. Pharmaceutics 2025, 17, 215. [Google Scholar] [CrossRef]
  26. Segneanu, A.-E.; Bejenaru, L.E.; Bejenaru, C.; Blendea, A.; Mogoșanu, G.D.; Biță, A.; Boia, E.R. Advancements in Hydrogels: A Comprehensive Review of Natural and Synthetic Innovations for Biomedical Applications. Polymers 2025, 17, 2026. [Google Scholar] [CrossRef]
  27. Pandya, I.; Kumar, S.; Aswal, V.K.; El Seoud, O.; Assiri, M.A.; Malek, N. Metal–Organic Framework-Based Polymeric Hydrogel: A Promising Drug Delivery Vehicle for the Treatment of Breast Cancer. Int. J. Pharm. 2024, 658, 124206. [Google Scholar] [CrossRef]
  28. Guo, A.; Cao, Q.; Fang, H.; Tian, H. Recent Advances and Challenges of Injectable Hydrogels in Drug Delivery. J. Control. Release 2025, 385, 114021. [Google Scholar] [CrossRef]
  29. Narayanaswamy, R.; Torchilin, V.P. Hydrogels and Their Applications in Targeted Drug Delivery. Molecules 2019, 24, 603. [Google Scholar] [CrossRef] [PubMed]
  30. Gressler, S.; Hipfinger, C.; Part, F.; Pavlicek, A.; Zafiu, C.; Giese, B. A Systematic Review of Nanocarriers Used in Medicine and Beyond: Definition and Categorization Framework. J. Nanobiotechnol. 2025, 23, 90. [Google Scholar] [CrossRef]
  31. Fan, Y.; Han, Q.; Li, H.; Cai, X.; Dyett, B.; Qiao, R.; Drummond, C.J.; Thang, S.H.; Zhai, J. Recent Developments in Nanoparticle–Hydrogel Hybrid Materials for Controlled Release. Adv. Sci. 2025, 12, e07209. [Google Scholar] [CrossRef]
  32. Chen, J.; Zhu, Y.; Kaskel, S. Porphyrin-Based Metal–Organic Frameworks for Biomedical Applications. Angew. Chem. Int. Ed. 2020, 60, 5010–5035. [Google Scholar] [CrossRef]
  33. Cai, M.; Yang, Y.; Kong, J.; Zhang, Y.; Li, S.; Zhang, H.; Xv, X.; Ni, J.; Yin, X. Nanoscale Metal–Organic Frameworks as a Versatile Platform for Synergistic Combination Tumor Therapy. J. Nanobiotechnol. 2025, 23, 601. [Google Scholar] [CrossRef]
  34. Zhou, T.; Liang, X.; Wang, P.; Hu, Y.; Qi, Y.; Jin, Y.; Du, Y.; Fang, C.; Tian, J. A Hepatocellular Carcinoma Targeting Nanostrategy with Hypoxia-Ameliorating and Photothermal Abilities That, Combined with Immunotherapy, Inhibits Metastasis and Recurrence. ACS Nano 2020, 14, 12679–12696. [Google Scholar] [CrossRef] [PubMed]
  35. Hawthorne, D.; Pannala, A.; Sandeman, S.; Lloyd, A. Sustained and Targeted Delivery of Hydrophilic Drug Compounds: A Review of Existing and Novel Technologies from Bench to Bedside. J. Drug Deliv. Sci. Technol. 2022, 78, 103936. [Google Scholar] [CrossRef]
  36. Wang, X.; Zhong, X.; Liu, Z.; Cheng, L. Recent Progress of Chemodynamic Therapy-Induced Combination Cancer Therapy. Nano Today 2020, 35, 100946. [Google Scholar] [CrossRef]
  37. Li, D.; Yadav, A.; Zhou, H.; Roy, K.; Thanasekaran, P.; Lee, C. Advances and Applications of Metal–Organic Frameworks (MOFs) in Emerging Technologies: A Comprehensive Review. Glob. Chall. 2023, 8, 2300244. [Google Scholar] [CrossRef]
  38. Nabipour, H.; Rohani, S. Metal–Organic Frameworks for Overcoming the Blood–Brain Barrier in the Treatment of Brain Diseases: A Review. Nanomaterials 2024, 14, 1379. [Google Scholar] [CrossRef]
  39. Qiu, M.; Singh, A.K.; Fan, Q.; Wu, Z.; He, C.; Zhao, Y.; He, S.; Prasad, P.N. Biocompatible and Biodegradable Inorganic Nanostructures for Nanomedicine: Silicon and Black Phosphorus. Nano Today 2019, 25, 135–155. [Google Scholar] [CrossRef]
  40. Yan, X.; Zhao, X.; Fan, M.; Zheng, W.; Zhu, G.; Li, B.; Wang, L. Acidic Environment-Responsive Metal–Organic Framework-Mediated Dihydroartemisinin Delivery for Triggering Production of Reactive Oxygen Species in Drug-Resistant Lung Cancer. Int. J. Nanomed. 2024, 19, 3847–3859. [Google Scholar] [CrossRef]
  41. Cai, M.; Chen, G.; Qin, L.; Qu, C.; Dong, X.; Ni, J.; Yin, X. Metal–Organic Frameworks as Drug Targeting Delivery Vehicles in the Treatment of Cancer. Pharmaceutics 2020, 12, 232. [Google Scholar] [CrossRef] [PubMed]
  42. Benny, A.; Rajendra Pai, S.D.; Pinheiro, D.; Chundattu, S.J. Metal–Organic Frameworks in Biomedicine: Innovations in Drug Delivery. Results Chem. 2024, 7, 101414. [Google Scholar] [CrossRef]
  43. Khulood, M.T.; Jijith, U.S.; Naseef, P.; Kallungal, S.; Geetha, V.; Pramod, K. Advances in Metal–Organic Framework-Based Drug Delivery Systems. Int. J. Pharm. 2025, 673, 125380. [Google Scholar] [CrossRef]
  44. Filiz, A.; Bayazit, Ş.; Barlas, F. Metal–Organic Frameworks (MOFs) in Drug Delivery: Emerging Trends, Functional Enhancements, and Biocompatibility Challenges. J. Drug Deliv. Sci. Technol. 2025, 12, 107284. [Google Scholar] [CrossRef]
  45. Singh, S.; Sivaram, N.; Nath, B.; Khan, N.A.; Singh, J.; Ramamurthy, P.C. Metal–Organic Frameworks for Wastewater Treatment, Renewable Energy and Circular Economy Contributions. NPJ Clean Water 2024, 7, 124. [Google Scholar] [CrossRef]
  46. Fu, X.; Yang, Z.; Deng, T.; Chen, J.; Wen, Y.; Fu, X.; Zhou, L.; Zhu, Z.; Yu, C. A Natural Polysaccharide-Mediated MOF-Based Ce6 Delivery System with Improved Biological Properties for Photodynamic Therapy. J. Mater. Chem. B 2020, 8, 1481–1488. [Google Scholar] [CrossRef] [PubMed]
  47. Hu, J.; Wu, W.; Qin, Y.; Liu, C.; Wei, P.; Hu, J.; Seeberger, P.H.; Yin, J. Fabrication of Glyco–Metal–Organic Frameworks for Targeted Interventional Photodynamic/Chemotherapy for Hepatocellular Carcinoma through Percutaneous Transperitoneal Puncture. Adv. Funct. Mater. 2020, 30, 1910084. [Google Scholar] [CrossRef]
  48. Pan, M.-M.; Li, P.; Yu, Y.-P.; Jiang, M.; Yang, X.; Zhang, P.; Nie, J.; Hu, J.; Yu, X.; Xu, L. Bimetallic Ions Functionalized Metal–Organic-Framework Nanozyme for Tumor Microenvironment Regulating and Enhanced Photodynamic Therapy for Hypoxic Tumor. Adv. Healthc. Mater. 2023, 12, 2300821. [Google Scholar] [CrossRef]
  49. Shang, Y.; Yi, X.; Xiang, D.; Zhou, L. Nanoagent-Mediated Photothermal Therapy: From Delivery System Design to Synergistic Theranostic Applications. Int. J. Nanomed. 2025, 20, 6891–6927. [Google Scholar] [CrossRef]
  50. Tong, P.-H.; Yang, J.-J.; Zhou, Y.-F.; Tang, Y.-F.; Tang, M.-T.; Zang, Y.; Pan, Y.-F.; Dong, L.-W.; Tan, Y.-X.; Nam, K.T.; et al. Metal–Organic Frameworks (MOFs) for Phototherapy and Synergistic Phototherapy of Cancer. Coord. Chem. Rev. 2025, 526, 216381. [Google Scholar] [CrossRef]
  51. Xu, R.; Wang, S.; Guo, Q.; Zhong, R.; Chen, X.; Xia, X. Anti-Tumor Strategies of Photothermal Therapy Combined with Other Therapies Using Nanoplatforms. Pharmaceutics 2025, 17, 306. [Google Scholar] [CrossRef] [PubMed]
  52. Huang, H.; Liu, Q.; Zhu, J.; Tong, Y.; Huang, D.; Zhou, D.; Long, S.; Wang, L.; Li, M.; Chen, X.; et al. Dimerized Pentamethine Cyanines for Synergistically Boosting Photodynamic/Photothermal Therapy. Aggregate 2025, 6, e706. [Google Scholar] [CrossRef]
  53. Han, H.S.; Choi, K.Y. Advances in Nanomaterial-Mediated Photothermal Cancer Therapies: Toward Clinical Applications. Biomedicines 2021, 9, 305. [Google Scholar] [CrossRef] [PubMed]
  54. Shi, Z.; Chu, C.; Zhang, Y.; Su, Z.; Lin, H.; Pang, X.; Wang, X.; Liu, G.; Li, W. Self-Assembled Metal–Organic Nanoparticles for Multimodal Imaging-Guided Photothermal Therapy of Hepatocellular Carcinoma. J. Biomed. Nanotechnol. 2018, 14, 1934–1943. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, B.; Hu, S.; Teng, Y.; Chen, J.; Wang, H.; Xu, Y.; Wang, K.; Xu, J.; Cheng, Y.; Gao, X. Current Advance of Nanotechnology in Diagnosis and Treatment for Malignant Tumors. Signal Transduct. Target. Ther. 2024, 9, 200. [Google Scholar] [CrossRef]
  56. Lafi, Z.; Matalqah, S.; Abu-Saleem, E.; Asha, N.; Mhaidat, H.; Asha, S.; Al-Nashash, L.; Janabi, H.S. Metal–Organic Frameworks as Nanoplatforms for Combination Therapy in Cancer Treatment. Med. Oncol. 2025, 42, 26. [Google Scholar] [CrossRef]
  57. Sampaio, P.A.; Pereira, E.C.V.; Sá, P.G.S.; Alencar Filho, J.M.T.; Ferraz, L.R.M.; Nishimura, R.H.V.; Ferreira, A.S.; Rolim Neto, P.J.; Araújo, E.S.; Rolim, L.A. A Review on Metal–Organic Frameworks as Technological Excipients: Synthesis, Characterization, Toxicity, and Application in Drug Delivery Systems. Compounds 2025, 5, 1. [Google Scholar] [CrossRef]
  58. Lawson, H.D.; Walton, S.P.; Chan, C. Metal–Organic Frameworks for Drug Delivery: A Design Perspective. ACS Appl. Mater. Interfaces 2021, 13, 7004–7020. [Google Scholar] [CrossRef]
  59. Kush, P.; Singh, R.; Kumar, P. Recent Advances in Metal–Organic Framework-Based Anticancer Hydrogels. Gels 2025, 11, 76. [Google Scholar] [CrossRef]
  60. Xie, Y.; Liu, M.; Cai, C.; Ye, C.; Guo, T.; Yang, K.; Xiao, H.; Tang, X.; Liu, H. Recent Progress of Hydrogel-Based Local Drug Delivery Systems for Postoperative Radiotherapy. Front. Oncol. 2023, 13, 1027254. [Google Scholar] [CrossRef]
  61. Tran, V.A.; Le, V.T.; Doan, V.D.; Vo, G.N.L. Utilization of Functionalized Metal–Organic Framework Nanoparticle as Targeted Drug Delivery System for Cancer Therapy. Pharmaceutics 2023, 15, 931. [Google Scholar] [CrossRef]
  62. Khan, M.U.; Alissa, M.; Inam, M.; Alsuwat, M.A.; Abdulaziz, O.; Mostafa, Y.S.; Hussain, T.; ur Rehman, K.; Zaman, U.; Khan, D. Comprehensive Overview of Utilizing Metal–Organic Frameworks (MOFs) for Precise Cancer Drug Delivery. Microchem. J. 2024, 204, 111056. [Google Scholar]
  63. Yang, N.; He, Z.; Lang, T. Drug Delivery Systems Based on Metal–Organic Frameworks for Tumor Immunotherapy. Pharmaceutics 2025, 17, 225. [Google Scholar] [CrossRef] [PubMed]
  64. Soman, S.; Kulkarni, S.; Kulkarni, J.; Dhas, N.; Roy, A.A.; Pokale, R.; Mukharya, A.; Mutalik, S. Metal–Organic Frameworks: A Biomimetic Odyssey in Cancer Theranostics. Nanoscale 2025, 17, 12620–12647. [Google Scholar] [CrossRef]
  65. Saboorizadeh, B.; Zare-Dorabei, R.; Safavi, M.; Safarifard, V. Applications of Metal–Organic Frameworks (MOFs) in Drug Delivery, Biosensing, and Therapy: A Comprehensive Review. Langmuir 2024, 40, 22477–22503. [Google Scholar] [CrossRef] [PubMed]
  66. Ye, Y.; Zhao, Y.; Sun, Y.; Cao, J. Recent Progress of Metal–Organic Framework-Based Photodynamic Therapy for Cancer Treatment. Int. J. Nanomed. 2022, 17, 2367–2395. [Google Scholar] [CrossRef]
  67. Abodunrin, T.; Egharevba, G.; Owoeye, F.; Omojola, J.; Adeyemi, O. Metal–Organic Frameworks in Photodynamic Therapy of Cancer: Functional Roles of Active Metal Centres and Integrated and Loaded Photosensitizers in the Framework. Mater. Adv. 2025, 6, 1554–1577. [Google Scholar] [CrossRef]
  68. Correia, J.H.; Rodrigues, J.A.; Pimenta, S.; Dong, T.; Yang, Z. Photodynamic Therapy Review: Principles, Photosensitizers, Applications, and Future Directions. Pharmaceutics 2021, 13, 1332. [Google Scholar] [CrossRef]
  69. Al-Jamal, A.N.; Al-Hussainy, A.F.; Abdulrazzaq Mohammed, B.; Hussein Abbas, H.; Mohammed Kadhim, I.; Hassan Ward, Z.; Kar Mahapatra, D.; Muringayil Joseph, T.; Kianfar, E.; Thomas, S. Photodynamic Therapy (PDT) in Drug Delivery: Nano-Innovations Enhancing Treatment Outcomes. Health Sci. Rev. 2025, 14, 100218. [Google Scholar] [CrossRef]
  70. Liu, W.; Wang, Y.; Li, Y.; Cai, S.; Yin, X.; He, X.; Zhang, Y. Fluorescent Imaging-Guided Chemotherapy-and-Photodynamic Dual Therapy with Nanoscale Porphyrin Metal–Organic Framework. Small 2017, 13, 1603459. [Google Scholar] [CrossRef]
  71. Wang, Y.; Wu, W.; Liu, J.; Manghnani, P.N.; Hu, F.; Ma, D.; Teh, C.; Wang, B.; Liu, B. Cancer-Cell-Activated Photodynamic Therapy Assisted by Cu(II)-Based Metal–Organic Framework. ACS Nano 2019, 13, 6879–6890. [Google Scholar] [CrossRef]
  72. Cai, Z.; Xin, F.; Wei, Z.; Wu, M.; Lin, X.; Du, X.; Chen, G.; Zhang, D.; Zhang, Z.; Liu, X.; et al. Photodynamic Therapy Combined with Antihypoxic Signaling and CpG Adjuvant as an In Situ Tumor Vaccine Based on Metal–Organic Framework Nanoparticles to Boost Cancer Immunotherapy. Adv. Healthc. Mater. 2020, 9, e1900996. [Google Scholar] [CrossRef] [PubMed]
  73. Di, X.; Pei, Z.; Pei, Y.; James, T. Tumor Microenvironment-Oriented MOFs for Chemodynamic Therapy. Coord. Chem. Rev. 2023, 484, 215098. [Google Scholar] [CrossRef]
  74. Li, M.; Zhang, W.; Xu, X.; Liu, G.; Dong, M.; Sun, K.; Zhang, P. Nanosystems for Chemodynamic-Based Combination Therapy: Strategies and Recent Advances. Front. Pharmacol. 2022, 13, 1065438. [Google Scholar] [CrossRef] [PubMed]
  75. Meng, X.; Sun, Z.; Chu, H.; Wang, Y. Self-assembled Nanoplatforms for Chemodynamic Therapy. Chem. Eng. J. 2024, 479, 147702. [Google Scholar] [CrossRef]
  76. Ding, Y.; Xu, H.; Xu, C.; Tong, Z.; Zhang, S.; Bai, Y.; Chen, Y.; Xu, Q.; Zhou, L.; Ding, H.; et al. A Nanomedicine Fabricated from Gold Nanoparticles-Decorated Metal–Organic Framework for Cascade Chemo/Chemodynamic Cancer Therapy. Adv. Sci. 2020, 7, 2001060. [Google Scholar] [CrossRef]
  77. Nowak, K.M.; Schwartz, M.R.; Breza, V.R.; Price, R.J. Sonodynamic Therapy: Rapid Progress and New Opportunities for Non-Invasive Tumor Cell Killing with Sound. Cancer Lett. 2022, 532, 215592. [Google Scholar] [CrossRef]
  78. Pan, X.; Wang, H.; Wang, S.; Sun, X.; Wang, L.; Wang, W.; Shen, H.; Liu, H. Sonodynamic Therapy (SDT): A Novel Strategy for Cancer Nanotheranostics. Sci. China Life Sci. 2018, 61, 415–426. [Google Scholar] [CrossRef]
  79. Bai, Y.; Wang, J.; Yan, R.; Zhao, Y.; Han, S.; Zhang, Z.; Zhao, Y. Progress of Metal–Organic Frameworks in Improving the Effect of Sonodynamic Therapy. Biomed. Technol. 2025, 9, 100070. [Google Scholar] [CrossRef]
  80. Yang, F.; Dong, J.; Li, Z.; Wang, Z. Metal–Organic Frameworks (MOF)-Assisted Sonodynamic Therapy in Anticancer Applications. ACS Nano 2023, 17, 4102–4133. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, Y.; Wang, N.; Yan, F.; Shi, Z.; Feng, S. Metal–Organic Frameworks as Candidates for Tumor Sonodynamic Therapy: Designable Structures for Targeted Multifunctional Transformation. Acta Biomater. 2024, 181, 67–97. [Google Scholar] [CrossRef]
  82. Xiao, Z.; Chen, Q.; Yang, Y.; Tu, S.; Wang, B.; Qiu, Y.; Jiang, Y.; Huang, Q.; Ai, K. State of the Art Advancements in Sonodynamic Therapy (SDT): Metal–Organic Frameworks for SDT. Chem. Eng. J. 2022, 449, 137889. [Google Scholar] [CrossRef]
  83. Xu, Q.; Zhan, G.; Zhang, Z.; Yong, T.; Yang, X.; Gan, L. Manganese Porphyrin-Based Metal–Organic Framework for Synergistic Sonodynamic Therapy and Ferroptosis in Hypoxic Tumors. Theranostics 2021, 11, 1937–1952. [Google Scholar] [CrossRef] [PubMed]
  84. Duan, W.; Li, B.; Zhang, W.; Li, J.; Yao, X.; Tian, Y.; Zheng, J.; Li, D. Two-Photon Responsive Porphyrinic Metal–Organic Framework Involving Fenton-Like Reaction for Enhanced Photodynamic and Sonodynamic Therapy. J. Nanobiotechnol. 2022, 20, 217. [Google Scholar] [CrossRef]
  85. Liao, D.; Huang, J.; Jiang, C.; Zhou, L.; Zheng, M.; Nezamzadeh-Ejhieh, A.; Qi, N.; Lu, C.; Liu, J. A Novel Platform of MOF for Sonodynamic Therapy-Advanced Therapies. Pharmaceutics 2023, 15, 2071. [Google Scholar] [CrossRef] [PubMed]
  86. Zhong, L.; Yang, T.; Li, P.; Shi, L.; Lai, J.; Gu, L. Metal–Organic Framework-Based Nanotherapeutics with Tumor Hypoxia-Relieving Ability for Synergistic Sonodynamic/Chemo-Therapy. Front. Mater. 2022, 9, 841503. [Google Scholar] [CrossRef]
  87. Yang, H.; Li, R.; Jin, S.; Tian, Y.; Wang, C.; Sun, Y.; Dai, Z.; Cheng, W. Targeted Nanosensitizer-Augmented Sono-Immunotherapy with STING Agonist to Remodel the Immune Microenvironment in Hepatocellular Carcinoma. Acta Biomater. 2025, 199, 387–397. [Google Scholar] [CrossRef]
  88. Hu, C.; Hou, B.; Xie, S. Application of Nanosonosensitizer Materials in Cancer Sonodynamic Therapy. RSC Adv. 2022, 12, 22722–22747. [Google Scholar] [CrossRef]
  89. Chen, P.; Zhang, P.; Shah, N.H.; Cui, Y.; Wang, Y. A Comprehensive Review of Inorganic Sonosensitizers for Sonodynamic Therapy. Int. J. Mol. Sci. 2023, 24, 12001. [Google Scholar] [CrossRef] [PubMed]
  90. Jiao, L.; Seow, J.Y.R.; Skinner, W.S.; Wang, Z.U.; Jiang, H.-L. Metal–Organic Frameworks: Structures and Functional Applications. Mater. Today 2019, 27, 43–68. [Google Scholar] [CrossRef]
  91. Li, B.; Ashrafizadeh, M.; Jiao, T. Biomedical Application of Metal–Organic Frameworks (MOFs) in Cancer Therapy: Stimuli-Responsive and Biomimetic Nanocomposites in Targeted Delivery, Phototherapy and Diagnosis. Int. J. Biol. Macromol. 2024, 260 Pt 2, 129391. [Google Scholar] [CrossRef]
  92. Liu, J.; Huang, J.; Zhang, L.; Lei, J. Multifunctional Metal–Organic Framework Heterostructures for Enhanced Cancer Therapy. Chem. Soc. Rev. 2021, 50, 1188–1218. [Google Scholar] [CrossRef]
  93. Zou, H.; Wang, F.; Zhou, J.-J.; Liu, X.; He, Q.; Wang, C.; Zheng, Y.-W.; Wen, Y.; Xiong, L. Application of Photodynamic Therapy for Liver Malignancies. J. Gastrointest. Oncol. 2020, 11, 431–442. [Google Scholar] [CrossRef] [PubMed]
  94. Mallidi, S.; Anbil, S.; Bulin, A.-L.; Obaid, G.; Ichikawa, M.; Hasan, T. Beyond the Barriers of Light Penetration: Strategies, Perspectives, and Possibilities for Photodynamic Therapy. Theranostics 2016, 6, 2458–2487. [Google Scholar] [CrossRef] [PubMed]
  95. Guo, Z.; Xiao, Y.; Wu, W.; Zhe, M.; Yu, P.; Shakya, S.; Li, Z.; Xing, F. Metal–Organic Framework-Based Smart Stimuli-Responsive Drug Delivery Systems for Cancer Therapy: Advances, Challenges, and Future Perspectives. J. Nanobiotechnol. 2025, 23, 157. [Google Scholar] [CrossRef] [PubMed]
  96. Khafaga, D.S.R.; El-Morsy, M.T.; Faried, H.; Diab, A.H.; Shehab, S.; Saleh, A.M.; Al, G.A.M. Metal–Organic Frameworks in Drug Delivery: Engineering Versatile Platforms for Therapeutic Applications. RSC Adv. 2024, 14, 30201–30229. [Google Scholar] [CrossRef]
  97. Singh, N.; Qutuba, S.; Khashab, N.M. Biocompatibility and Biodegradability of Metal–Organic Frameworks for Biomedical Applications. J. Mater. Chem. B 2021, 9, 5925–5934. [Google Scholar] [CrossRef]
  98. Mhettar, P.; Kale, N.; Pantwalawalkar, J.; Nangare, S.; Jadhav, N. Metal–Organic Frameworks: Drug Delivery Applications and Future Prospects. ADMET DMPK 2023, 12, 27–62. [Google Scholar] [CrossRef]
  99. Hou, Y.; Zhu, C.; Ban, G.; Shen, Z.; Liang, Y.; Chen, K.; Wang, C.; Shi, H. Advancements and Challenges in the Application of Metal–Organic Framework (MOF) Nanocomposites for Tumor Diagnosis and Treatment. Int. J. Nanomed. 2024, 19, 6295–6317. [Google Scholar] [CrossRef]
  100. Zhang, J.; Li, M.; Liu, M.; Yu, Q.; Ge, D.; Zhang, J. Metal–Organic Framework Nanomaterials as a Medicine for Catalytic Tumor Therapy: Recent Advances. Nanomaterials 2024, 14, 797. [Google Scholar] [CrossRef] [PubMed]
  101. Abánades Lázaro, I.; Chen, X.; Ding, M.; Eskandari, A.; Fairen-Jimenez, D.; Giménez-Marqués, M.; Gref, R.; Lin, W.; Luo, T.; Forgan, R.S. Metal–Organic Frameworks for Biological Applications. Nat. Rev. Methods Primers 2024, 4, 42. [Google Scholar] [CrossRef]
  102. Chai, W.; Chen, X.; Liu, J.; Zhang, L.; Liu, C.; Li, L.; Honiball, J.R.; Pan, H.; Cui, X.; Wang, D. Recent progress in functional metal–organic frameworks for biomedical applications. Regen. Biomater. 2024, 11, rbad115. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) Stepwise synthesis of PS@MOF-199 and F127-coated PS@MOF-199 nanoparticles (PS@MOF-199 NPs). (B) Illustration of the photosensitization quenching and activation behavior of PS@MOF-199 NPs within the tumor microenvironment. Process I: Framework collapse of MOF-199 initiated by intracellular GSH. Process II: ROS production under light irradiation. Reproduced with permission from ref. [71] (copyright 2019, Wang).
Figure 1. (A) Stepwise synthesis of PS@MOF-199 and F127-coated PS@MOF-199 nanoparticles (PS@MOF-199 NPs). (B) Illustration of the photosensitization quenching and activation behavior of PS@MOF-199 NPs within the tumor microenvironment. Process I: Framework collapse of MOF-199 initiated by intracellular GSH. Process II: ROS production under light irradiation. Reproduced with permission from ref. [71] (copyright 2019, Wang).
Nanomaterials 15 01771 g001
Table 1. Representative MOF-based nanosystems for PDT, PTT, and CDT therapies in hepatocellular carcinoma.
Table 1. Representative MOF-based nanosystems for PDT, PTT, and CDT therapies in hepatocellular carcinoma.
Study (Ref)MOF Type/NPPayloadTherapy TypeIn Vitro IC50/EfficacyIn Vivo ModelMain Outcomes
Liu et al. [70]Zr-TCPP NPMOFDOXPDT + Chemo67.72 µg mL−1;
90% cell lethality
HepG2 tumor-bearing miceTwo tumors disappeared, two shrank from 62.5 → 2 mm3; no skin/tissue damage
Wang et al. [71]MOF-199Ce6PDTHigh phototoxicity to HepG2Zebrafish larvaeGSH-responsive PS release; tumor volume reduced; improved imaging-guided PDT
Cai et al. [72]PCN-224@ACF-CpG@HACe6 + ACF + CpGPDT + ImmunotherapyTumor-bearing miceEliminated residual tumor cells; activated host anti-tumor immune response
Fu et al. [46]ZIF-8@Ce6–HACe6PDT88.4% HepG2 cell deathHepG2 cellsEnhanced Ce6 solubility, efficient uptake, mitigated aggregation; improved PDT efficacy
Hu et al. [47]Gal-PCN-224DOX + Ce6PDT + ChemoOrthotopic HCC mouse modelTumor growth inhibited by 98%; ASGPR-targeted delivery; imaging-guided therapy
Pan et al. [48]ZMRPC@HACe6PDTH22 tumor xenograft, BALB/c miceSynergistic ROS and O2 production; effective inhibition under 660 nm laser
Shi et al. [54]Mn2+–ICG MINPsICGPTTSignificant HepG2 inhibition under 808 nm laserSubcutaneous HepG2 tumors in miceTumor necrosis; enhanced MRI and PA imaging; time-dependent accumulation
Zhou et al. [34]SP94-PB-SF-Cy5.5 NPsSorafenibPTT + Targeted therapyEffective tumor ablationHepG2 and Hepa1–6 mouse modelsHigh HCC accumulation; triple-modality imaging; hypoxia relief; immune activation; prevented recurrence
Ding et al. [76]PEG-Au/FeMOF@CPTCPTCDT + Chemo0.31 ± 0.04 µg mL−1HepG2 cellsStrong synergistic tumor suppression: CDT enhanced CPT cytotoxicity
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tutunchi, H.; Nabipour, H.; Rohani, S. Therapeutic Potential of Nanoscale Metal–Organic Frameworks in Hepatocellular Carcinoma. Nanomaterials 2025, 15, 1771. https://doi.org/10.3390/nano15231771

AMA Style

Tutunchi H, Nabipour H, Rohani S. Therapeutic Potential of Nanoscale Metal–Organic Frameworks in Hepatocellular Carcinoma. Nanomaterials. 2025; 15(23):1771. https://doi.org/10.3390/nano15231771

Chicago/Turabian Style

Tutunchi, Helda, Hafezeh Nabipour, and Sohrab Rohani. 2025. "Therapeutic Potential of Nanoscale Metal–Organic Frameworks in Hepatocellular Carcinoma" Nanomaterials 15, no. 23: 1771. https://doi.org/10.3390/nano15231771

APA Style

Tutunchi, H., Nabipour, H., & Rohani, S. (2025). Therapeutic Potential of Nanoscale Metal–Organic Frameworks in Hepatocellular Carcinoma. Nanomaterials, 15(23), 1771. https://doi.org/10.3390/nano15231771

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop