Back to Journals » International Journal of Nanomedicine » Volume 20
Advancements in Nanocarrier Delivery Systems for Photodynamic Therapy in Lung Cancer
Authors Wang YY, Li T, Zhou H, Liu C
Received 8 February 2025
Accepted for publication 7 May 2025
Published 29 May 2025 Volume 2025:20 Pages 6853—6874
DOI https://doi.org/10.2147/IJN.S521444
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Yan Shen
Yuanyuan Wang, Tao Li, Hui Zhou, Chunyu Liu
Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
Correspondence: Chunyu Liu, Email [email protected]
Abstract: Photodynamic therapy (PDT), as a non-invasive treatment modality, has shown potential as an alternative to traditional therapies in lung cancer treatment. However, its clinical application is still limited by key challenges, including low photosensitizer (PS) delivery efficiency, tumor microenvironment hypoxia, and the restricted diffusion of reactive oxygen species (ROS). This review systematically discusses innovative strategies employed by nanocarrier delivery systems to overcome these bottlenecks in PDT. The first section focuses on the application and challenges of PDT in lung cancer treatment: although PDT induces localized tumor cell death through ROS generation mediated by PSs, its efficacy is hindered by the delivery barriers of hydrophobic PSs to the lung, insufficient ROS generation due to tumor hypoxia, and PS self-quenching. The second section presents nanotechnology-driven solutions: 1) using nanocarriers equipped with catalase or hemoglobin and perfluorocarbon to alleviate tumor hypoxia and enhance ROS production; 2) modifying the surface of nanocarriers with targeting ligands to improve PS accumulation in tumor tissues and reduce self-quenching effects; and 3) combining PS-loaded nanocarriers with immune checkpoint inhibitors and chemotherapy agents to synergistically enhance anti-tumor efficacy and suppress metastasis. Future research should focus on further optimizing the biocompatibility, clinical translation potential, and multimodal synergistic mechanisms of nanocarriers, to promote the widespread application of PDT in precise lung cancer treatment.
Keywords: lung cancer treatment, nanocarrier delivery systems, photodynamic therapy, photosensitizers, fighting hypoxia
Introduction
A recent report by the International Agency for Research on Cancer estimated that approximately 19.3 million new cancer cases and 9.9 million cancer-related deaths occurred globally in 2020.1 The report also predicts a 56.4% increase in these numbers over the next 20 years.1 Lung cancer is one of the most prevalent malignancies, accounting for 11.4% of all cases, and it remains the leading cause of cancer-related deaths, contributing to 18% of total cancer mortality.2 Although breast cancer has surpassed lung cancer in incidence, lung cancer remains the second most frequently diagnosed cancer globally.3 Advances in diagnostic techniques, particularly high-resolution CT imaging, have facilitated the detection of early-stage lung cancer. However, the prognosis for late-stage lung cancer remains dismal, with five-year survival rates significantly reduced due to delayed diagnoses and limited treatment efficacy. For early-stage lung cancer, surgical resection is the preferred treatment modality.4–6 However, patients with underlying pulmonary conditions may not be suitable candidates for radical surgery, necessitating alternative therapies such as chemotherapy and radiotherapy.7 Conventional chemotherapy and radiotherapy are associated with significant systemic toxicity and may compromise organ function,8,9 while stereotactic ablative radiotherapy poses risks of severe radiation pneumonitis in patients with compromised respiratory function.10 Despite progress in targeted therapies, fewer than 25% of lung cancer patients benefit from such treatments, with resistance emerging almost universally over time.11
Photodynamic therapy (PDT) has emerged as a promising treatment option for lung cancer.12–14 PDT utilizes photosensitizers (PS) (Table 1), oxygen, and specific wavelengths of light to generate reactive oxygen species (ROS), which induce tumor cell apoptosis or necrosis (Figure 1).15–18 Compared to traditional therapies, PDT offers advantages such as reduced invasiveness, enhanced selectivity, and minimized damage to surrounding healthy tissues.19 It is particularly effective in reducing the size of inoperable tumors and improving patient quality of life.
![]() |
Table 1 Photosensitizers Commonly Used in Lung Cancer Treatment |
![]() |
Figure 1 Schematic Illustration of Photochemical Reaction Mechanisms in PDT.15–18 (a) The PS absorbs light at specific wavelengths, transitioning from the ground state (S0) to the first singlet excited state (S1) or the second singlet excited state (S2). The S2 state rapidly decays to S1 through internal conversion (IC). From S1, the PS can return to S0 by emitting fluorescence (F) to release excess energy or undergo intersystem crossing (ISC) to form the more stable triplet excited state (T1). The T1 state can relax back to S0 via phosphorescence (P) or transfer energy to molecular oxygen (O2), generating singlet oxygen (1O2) in the Type II PDT process. Additionally, the T1 state can engage in electron transfer with intracellular biomolecules (such as nucleic acids, proteins, and lipids), producing superoxide anion radicals. These radicals further react with H2O2 or O2 to generate other ROS, including hydroxyl radicals (OH), defining the Type I PDT process. Currently, most PSs predominantly operate through the Type II PDT mechanism. (b) Six types of cell death induced by PDT. Created with BioGDP.com.49 |
However, the efficacy of PDT is limited by challenges such as poor solubility, low bioavailability, and non-specific distribution of PS, alongside the hypoxic nature of the tumor microenvironment (TME), which restricts oxygen availability for ROS generation. Nanocarrier delivery systems (NDSs) provide a potential solution to these limitations by enhancing the solubility, stability, and tumor-targeting capability of PS, as well as improving oxygenation at tumor sites through various mechanisms (Table 2). Strategies such as enzyme-catalyzed oxygen generation, direct oxygen delivery, and stimuli-responsive materials have been developed to overcome hypoxia and enhance the therapeutic efficacy of PDT. This review explores the clinical applications and limitations of PDT in lung cancer and discusses recent advancements in NDSs as a means to address these challenges. By synthesizing cutting-edge research, we aim to provide meaningful insights and references for advancing PDT in lung cancer therapy.
![]() |
Table 2 Nanocarrier Delivery Systems for PDT |
Applications and Constraints of PDT in Lung Cancer Therapy
PDT as an Alternative Therapeutic Approach
In the 1970s, PDT gradually emerged as a novel technique for cancer treatment. In 1994, Japan approved the use of Sodium Porfimer for PDT in central early-stage lung cancer.65 In 2002, Japanese lung cancer treatment guidelines recommended PDT as an option for early-stage central lung cancer.66 The 2013 guidelines from the American College of Chest Physicians recommended PDT for curative treatment of central-type lung cancer patients with mucosal layer-restricted lesions (Grade 1C) and early-stage squamous cell carcinoma, with regular bronchoscopic monitoring advise.67 The 2019 guidelines from the National Comprehensive Cancer Network recommended PDT for treating non-small cell lung cancer (NSCLC) patients experiencing airway obstruction or severe hemoptysis due to local symptoms or recurrence.3
PDT in clinical applications for lung cancer primarily targets superficial NSCLC that has not spread beyond the lungs and early-stage central lung cancer.68 However, most lung cancer cases in clinical practice involve larger, peripheral, and advanced-stage tumors, which limits the standalone use of PDT.12,69 Consequently, PDT is often integrated into multimodal treatment regimens to restore airway patency obstructed by tumors or convert initially inoperable tumors into resectable ones, thus improving therapeutic outcomes. Kato et al reported a representative case involving a 79-year-old patient with squamous cell carcinoma of the lung, presenting with an obstruction of the right B1 bronchus.66 The carcinoma exhibited proximal invasion extending to the carina and distal extension reaching the intermediate bronchus. After PDT, the patient underwent only a right upper sleeve lobectomy, and histological examination of the bronchial stump showed no evidence of residual tumor cells. Jung et al applied intraoperative PDT to both the lumen and outer wall of the bronchi and distal trachea to achieve local tumor control in a patient for whom complete anatomical resection by pneumonectomy was not feasible.70 At 13 months of follow-up, the patient remains alive with no evidence of disease recurrence. A similar improvement was reported by the Ohio State University Medical Center in 2006, where 41 patients with locally advanced, non-metastatic central-type non-small cell lung cancer (NSCLC) were treated with preoperative PDT.71 In this study, 60% of patients were initially considered inoperable due to extensive invasion of the carina or severe endobronchial obstruction. However, after preoperative PDT combined with chemotherapy and/or radiation, 50% of these patients became eligible for surgery. Furthermore, 27% of patients who were initially scheduled for pneumonectomy were able to undergo lobectomy instead. Among the 22 patients who underwent surgery, 14 showed a lower pathological stage than their preoperative clinical staging, with 4 patients having no residual tumor at the bronchial margin.71 In a small retrospective study by Mehta et al, 10 out of 11 patients did not experience local recurrence at the irradiation site within two years after undergoing post-operative bronchoscopic PDT. In another study evaluating the effectiveness of bronchial PDT in preventing NSCLC recurrence, it was found that the 5-year survival rate for patients with positive bronchial margins who received post-operative PDT was 12 out of 17, with only one case of margin recurrence.72 Chhatre et al analyzed data from the National Cancer Database (2004–2016) to examine the relationship between PDT and mortality in patients with stage III–IV NSCLC.73 Their study found that, compared to the radiation therapy alone group, the PDT group had a 50% reduction in mortality rates. Kimura et al also demonstrated that PDT combined with chemotherapy significantly improved airway stenosis in patients with advanced NSCLC as early as one week post-treatment, with the improvements largely maintained after one month.74 Additionally, the oxidative stress induced by PDT and some PS may generate novel and distinct tumor antigens. This unique mechanism renders PDT-treated cancer cells more immunogenic compared to cells eliminated through other treatments.75,76 While PDT alone has a limited ability to induce systemic antitumor immunity, combining it with conventional immune checkpoint inhibitors or immunoadjuvants can significantly enhance its efficacy, offering unique advantages in managing metastatic lung cancer.77,78 The study by Gao et al demonstrated that PDT combined with immune checkpoint inhibitors effectively treats both primary tumors and metastatic lesions.79
Persistent Challenges in PDT
PDT induces apoptosis by generating ROS.80 Compared to other lung cancer treatments, PDT is less invasive than traditional surgical resection, exhibits stronger targeting capabilities than chemotherapeutic agents, and has minimal risk of resistance.81,82 Moreover, PDT can be performed on an outpatient basis and leaves no scars after healing.83,84 However, conventional PDT faces several limitations. The limited tissue penetration of PS reduces their efficiency in inducing tumor cell apoptosis, while their low selectivity for tumor cells further compromises efficacy. Patients undergoing PDT must also avoid sunlight exposure until the PS is fully eliminated to prevent phototoxicity.85 Additionally, oxygen consumption and vascular damage during the PDT process may exacerbate tumor hypoxia, potentially shifting its effect from pro-apoptotic to anti-apoptotic.86,87 Bush et al demonstrated that the spatial distribution of hypoxia during PDT significantly impacts tumor destruction and vascular damage.88 Therefore, it is crucial to utilize n NDSs to target PS specifically to tumor sites, minimizing phototoxicity in other organs.89,90 These systems can also increase oxygen levels at the tumor site, reverse the hypoxic microenvironment, inhibit tumor metastasis and recurrence, and enhance the overall therapeutic efficacy of PDT.91
Nanotechnology-Driven Strategies to Overcome Key Barriers
Over the past few decades, NDSs have gained widespread use as platform carriers in cancer treatment due to three key physicochemical properties: (1) a high surface-to-volume ratio, which enhances therapeutic efficacy while reducing biotoxicity;92,93 (2) excellent multi-functionalization capabilities, particularly through the incorporation of disease-specific targeting ligands, thereby increasing the targeting precision and functionality of NDSs;94,95 and (3) the ability to protect drugs from enzymatic or chemical degradation, preventing premature clearance and ensuring sufficient drug doses reach the tumor site (Figure 2).96–98 These properties enable NDSs to overcome significant limitations of traditional delivery systems, providing controlled and localized drug release while minimizing systemic side effects. Oxygen-dependent NDSs can enhance oxygen levels in tumor regions, enabling PS to generate ROS under specific wavelengths of light. This process directly induces oxidative stress in tumor cells or sensitizes cancer tissues to achieve desired therapeutic outcomes.99–101 NDSs are designed to deliver PS to tumor sites in a controlled and targeted manner, ensuring the stability, plasma half-life, and bioavailability of the PS. These systems protect PS from enzymatic or chemical degradation before reaching the target tumor tissues. Furthermore, NDSs enhance the solubility, improve stability, and increase tumor-targeting efficiency of PS, as demonstrated by functionalized nanomaterials like liposomes, polymeric nanoparticles, and metal-based nanozymes.102,103
![]() |
Figure 2 Advantages of Nanocarrier Delivery Systems in PDT for Lung Cancer.49,96–98 |
Fighting Hypoxia with Nanocarrier Delivery Systems to Enhance PDT
Hemoglobin-and Perfluorocarbon-Based Oxygen Delivery
Hemoglobin (Hb) serves as the primary carrier for oxygen transport and exchange in the body,50 and perfluorocarbons (PFCs) are commonly used artificial oxygen carriers, boasting an oxygen solubility approximately twice that of normal blood.104 However, free Hb has a short circulation time in the bloodstream, is highly unstable, and tends to form dimers that cause renal toxicity.105 Similarly, PFCs may prematurely release oxygen, triggering allergic reactions, making both inadequate for optimal PDT outcomes.106 NDSs have been developed to address these challenges by delivering oxygen-loaded PFCs and Hb to target sites, thereby enhancing Hb stability and reducing premature oxygen release from PFCs.107 Xavierselvan et al fabricated nanodroplets using perfluoropentan, benzoporphyrin derivatives, and indocyanine green(ICG), controlling particle size to approximately 200 nm. Upon intravenous injection and subsequent laser irradiation at 690 nm, these nanodroplets selectively accumulated at tumor sites. The perfluoropentan and ICG enabled synchronized oxygen release during PDT, ensuring effective intratumoral oxygen supply.108 Guo et al engineered liposomal NDSs by encapsulating oxygen-loaded Hb and ICG complexes. This system effectively extended the circulation half-life of Hb in vivo. Post-injection, oxygen and PS effectively accumulated in deep tumor regions. T2-weighted MRI and immunohistochemical analyses revealed significantly enhanced intratumoral oxygen levels, along with a marked downregulation of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor, leading to improved PDT efficacy (Figure 3a).109 To further enhance tumor specificity and pH responsiveness, Ma et al developed a pH-sensitive nanoplatform by incorporating hydrophobic IR780 dye, functionalized fluorocarbon nanoparticles, and tumor-penetrating peptide iRGD. This system selectively accumulated in deep tumor regions, where pH-triggered drug release occurred. Encapsulating oxygen and IR780 in a single core accelerated singlet oxygen generation, substantially boosting PDT efficiency.110 To mitigate immune responses triggered by nanomaterials, biomimetic delivery systems, such as those utilizing red blood cell (RBC) membranes or exosomes, have been employed. Ding et al constructed an RBC membrane-coated nanovesicle, integrating Hb and semiconducting polymer nanoparticles. This system enhanced photostability, PS targeting, and oxygen self-supply, achieving prolonged circulation and efficient tumor accumulation through immune evasion.111
![]() |
Figure 3 Fighting Hypoxia with Nanocarrier Delivery Systems to Enhance PDT. (a) Schematic illustration of alleviation of tumor hypoxia and enhanced PDT based on PS and hemoglobin co-loaded liposomes.109 (b) Schematic illustration of alleviating tumor hypoxia and enhancing PDT using a catalase-based NDSs. (c) The Type I-dominated PS with small ΔEST. It has a low enough reduction potential in the ground state and a high enough reduction potential in the triplet excited state.112 (d) hyperbaric oxygenation (HBO) and interval-based illumination for tissue reoxygenation, have been explored to counteract the hypoxic conditions exacerbated by PDT itself. Created with BioGDP.com.49 |
Catalase-Mediated Oxygen Generation
Due to the abnormal metabolism of tumor cells, the intracellular hydrogen peroxide (H₂O₂) levels are higher than those in normal tissues. In the presence of catalase (CAT), H₂O₂ can be catalyzed to produce oxygen, alleviating tumor hypoxia. Several studies have focused on employing NDSs loaded with catalysts to promote the decomposition of H₂O₂ and generate oxygen (Figure 3b). For example, a pH-responsive smart drug delivery system was developed using silica nanoparticles loaded with CAT, chlorin e6 (Ce6), and an anti-cancer drug. In vivo photoacoustic imaging experiments demonstrated that this system increased tumor oxygenation levels from 1.5% to 12.6%, significantly improving the hypoxic microenvironment.54 To enhance the targeting specificity of NDSs, researchers have designed hyaluronic acid (HA)-modified systems, where IR820 and CAT were incorporated into biocompatible PLGA nanoparticles. This system leverages CD44 receptor-mediated endocytosis to facilitate tumor cell uptake and catalyze endogenous H2O2 conversion to oxygen.113 To further improve the stability of CAT, Wang et al performed vinylation of the primary amines on the lysine residues of CAT, followed by in situ free-radical polymerization with acrylamide monomers and crosslinking with the Ce6. The resulting protein nano-capsules significantly enhanced the catalytic oxygen production capacity of CAT under physiological conditions.114 Manganese dioxide (MnO2) nanoparticles can also catalyze the decomposition of H2O2 to generate oxygen, with the reaction byproducts being harmless Mn2+ ions that are water-soluble and easily excreted via the kidneys. However, MnO2 nanoparticles can damage normal tissues. To improve colloidal stability, prolong blood circulation time, and reduce toxicity, polyelectrolytes synthesized from alkynyl-modified carboxymethyl cellulose and azide-modified gelatin were employed for the in-situ growth of MnO2 nanoparticles. Alternatively, MnO2 has been assembled into nanozymes that exhibit excellent PDT efficacy under white-light illumination.115
Designing Hypoxia-Adaptable PS for ROS Generation
Most existing PDT approaches rely on Type II photoreactions, which are highly dependent on oxygen levels and involve significant oxygen consumption. This dependence on oxygen can exacerbate tumor hypoxia during PDT, especially due to oxygen consumption and microvascular damage, resulting in suboptimal therapeutic outcomes. In contrast, Type I PSs work differently by transferring electrons to molecular oxygen to produce superoxide anions. These anions undergo further cascade reactions mediated by superoxide dismutase (SOD), generating highly cytotoxic hydroxyl radicals that synergistically damage intracellular lysosomes and induce apoptosis in tumor cells (Figure 3c).112 For example, Zhao et al developed a silicon phthalocyanine derivative (NanoPcAF), which demonstrates efficient Type I photoreaction capabilities. Unlike conventional Type II PSs, NanoPcAF generates a significant amount of superoxide anions even under hypoxic conditions. Their research demonstrated that NanoPcAF achieved 3.4 times higher superoxide anion production compared to conventional methylene blue under similar hypoxic conditions, while its photothermal conversion efficiency was 2.4 times higher than ICG. Preclinical trials revealed excellent tumor accumulation and no significant systemic toxicity.116 Similarly, Tamoxifen (TAM), commonly known as an anti-estrogen drug, was observed to alleviate intrinsic tumor hypoxia by modulating mitochondrial electron transport and reducing oxygen consumption rates. TAM also downregulated HIF-1α, further enhancing the therapeutic potential of PDT in hypoxic environments.117
Additional Approaches to Mitigate Tumor Hypoxia
PDT effectiveness is significantly influenced by the availability of molecular oxygen in the TME. Strategies to increase oxygen supply, such as hyperbaric oxygenation (HBO) and interval-based illumination for tissue reoxygenation, have been explored to counteract the hypoxic conditions exacerbated by PDT itself (Figure 3d). Tomaselli et al conducted a study involving 30 patients (22 males, 8 females; mean age 68.8 years) with inoperable NSCLC and endobronchial stenosis, assessing the acute effects of combined PDT and HBO. Patients were sensitized with hematoporphyrin derivative (2 mg/kg BW) 48 hours before treatment, and a light dose of 300 J/cm was delivered. At one and four weeks after the treatment, patients reported significant improvements in dyspnea and hemoptysis. Objective measures, such as reductions in tumor stenosis and resolution of post-stenotic pneumonia, supported these findings. The combined therapy achieved notable tumor stenosis reduction (p < 0.05) and an increase in Karnofsky performance status without treatment-related complications.118 In addition to HBO, pulsed illumination has been proposed to enhance oxygenation during PDT. By delivering light in intervals, tissue reoxygenation can occur between exposures, mitigating hypoxia-induced therapy resistance. Such methods offer a promising approach to improve PDT outcomes by sustaining oxygen availability during treatment.86
Improving PS with Nanocarrier Delivery Systems to Enhance PDT
Traditional Unmodified Nanocarrier Delivery Systems
Nanomedicine offers substantial advantages in improving the solubility of hydrophobic drugs, enhancing biodistribution and pharmacokinetics, and enabling preferential accumulation at target sites. Commonly used nanocarriers include lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles.
Constructed from phospholipid bilayers, lipid nanoparticles are widely applied for the targeted delivery of PSs due to their ease of preparation, excellent biocompatibility, minimal side effects, and ability to extend circulation time. This enhances drug accumulation in tumors. Liu et al encapsulated Ce6 and SB-3CT within nanoliposomes, which significantly improved the stability of Ce6 and showed efficient tumor cell apoptosis upon light irradiation.119 Similarly, Visudyne®, a clinically approved PS, addresses the aggregation issue commonly faced by PS, improving their tissue penetration capacity.120
Polymeric nanoparticles, synthesized from natural or synthetic polymers such as dextran, chitosan, or polyethylene glycol (PEG), offer diverse structural variations for controlled drug delivery. These systems can encapsulate drugs in polymer reservoirs, embed them within polymer matrices, or use polymer-drug conjugates for targeted delivery. Ma et al developed folate-modified PEG-b-PAsp micelles that encapsulate PS, enhancing their accumulation in tumors through folate receptor-mediated uptake and acid-triggered release.121 Similarly, Zhang et al constructed tetraphenyl porphyrin loaded polymeric nanoparticles, achieving a high drug-loading capacity, reducing aggregation-induced quenching, and significantly improving antitumor efficacy.122
Inorganic nanoparticles, such as gold and silica-based systems, are known for their superior stability, tunable size, and surface modification capabilities. These properties make them suitable for designing environmentally responsive delivery systems that improve PS targeting and release control.103 Haimov et al conjugated hydrophobic Tetrahydroxyphenylchlorin with gold nanoparticles, resulting in enhanced solubility, stable colloidal properties, and increased tumor cell uptake.123 Similarly, mesoporous silica nanoparticles conjugated with Visudyne® significantly improved the PS’s bioavailability, leading to reduced tumor lymphangiogenesis and enhanced photodynamic efficacy.124
Targeted Nanocarrier Delivery Systems
While the enhanced permeability and retention (EPR) effect facilitates the accumulation of PS nanoparticles in tumors, it lacks tumor cell specificity. This limitation often results in insufficient uptake by tumor cells, thereby reducing the therapeutic efficacy.125 Tumor cells overexpress specific antigens or receptors, such as epidermal growth factor receptors, folate receptors, and low-density lipoprotein receptors. To enhance tumor-specific PS accumulation, corresponding antibodies or ligands including small molecule ligands, peptides, or lipid-based ligands can be conjugated to the surface of nanoparticles. These ligand-receptor interactions enhance the tumor-targeting capability of nanoparticles, improving PDT outcomes.126–128 For instance, Karges et al encapsulated a Ru(II)-based polypyridyl complex in a polymer functionalized with terminal folate groups. This system mitigated the inherent cytotoxicity of the Ru(II) complex in the dark and exhibited over an 8-fold higher selectivity toward folate receptor-overexpressing tumor cells compared to healthy cells.129 Similarly, Sun et al employed porous Fe3O4 nanoparticles to load ICG, modifying the nanoparticle surface with HA. This HA-functionalized system demonstrated a significant increase in tumor uptake through CD44 receptor-mediated endocytosis. Moreover, under 808 nm laser irradiation, the system effectively suppressed tumor growth in animal models, showcasing the synergy between photodynamic and photothermal therapies.130
Beyond nanoparticle-based delivery systems, targeted PS molecules also exhibit promise. For example, biotin-a ligand for receptors overexpressed in many cancer types-has been used to enhance PS targeting.131 Li et al synthesized a biotin-functionalized PS (ENBS-B), which demonstrated an 87-fold higher uptake in cancer cells compared to normal cells. Remarkably, ENBS-B retained its efficacy under hypoxic conditions, utilizing a Type I photodynamic mechanism to generate superoxide anions, thus overcoming the challenges posed by tumor hypoxia.132 Similarly, mannose receptors overexpressed in certain cancer cells have been targeted using mannose-functionalized PS molecules. Zhang et al conjugated a PS with mannose and assembled it into nanoparticles, which achieved improved tumor accumulation via both the EPR effect and mannose receptor-mediated uptake, significantly enhancing PDT efficacy.133
Multifunctional Nanocarrier Delivery Systems
In the complex microenvironment of tumors, diverse mechanisms drive both proliferation and metastasis. These mechanisms not only include the activation of bypass signaling pathways but also involve the inactivation of downstream apoptotic signals. In recent years, combination therapies have emerged as a promising strategy to suppress tumor proliferation and metastasis. Huang et al introduced a polymeric system capable of co-delivering three therapeutic agents, including Yes-associated protein (YAP) siRNA and gefitinib (Gef). Upon internalization into NSCLC cells, this polymeric nanoplatform selectively cleaves disulfide bonds, releasing YAP-siRNA and Gef. Gef effectively blocks the epidermal growth factor receptor signaling pathway, while YAP-siRNA suppresses the bypass pathways associated with gefitinib resistance. Additionally, this system facilitates PDT, enhancing ROS production to promote tumor apoptosis. Importantly, the introduction of YAP-siRNA and Gef not only alleviates PDT-induced hypoxia but also inhibits glycolysis and downregulates HIF-1α, synergistically triggering apoptosis.12 Guo et al developed a nanoplatform integrating PDT and immunotherapy by encapsulating ICG into hollow MnO2 nanospheres, further encapsulated within PD-L1 antibody-modified exosomes. This system, termed ICG@MnO2@Exo-anti-PD-L1, precisely targets PD-L1-overexpressing tumor cells, delivering anti-PD-L1 antibodies to the acidic TME to activate T-cell responses. The MnO2 catalyzes the conversion of H2O2 into oxygen, mitigating hypoxia within the tumor. Under 808 nm near-infrared (NIR) irradiation, ICG utilizes the generated oxygen to produce singlet oxygen (¹O2), effectively inducing tumor cell apoptosis.134 Furthermore, the Mn2+ released during the reaction polarizes macrophages from the M2 phenotype to the tumor-suppressing M1 phenotype, enhancing immune activation and supporting T-cell responses.
Carrier-Free Nanocarrier Delivery Systems
In recent years, NDSs have emerged as a potent solution to address the challenges in anticancer drug delivery.135 However, traditional NDSs often rely on encapsulating drugs within carrier materials through intermolecular interactions, such as hydrophobic forces. This approach presents several drawbacks, including premature drug leakage,136 low drug loading, increased toxicity,137,138 and challenges in manufacturing and clinical translation.139
Pure drug nano-assemblies (PANAs), which self-assemble or co-assemble directly from unmodified drug molecules, have garnered significant attention. Unlike conventional drug delivery systems, PANAs eliminate the need for carrier materials, simplifying preparation and improving drug loading to ultra-high levels-exceeding 60% or even 100%.140,141 These assemblies can be fabricated using straightforward methods like nanoprecipitation, which offers uniform particle formation, high reproducibility, and scalability.142 Acting as both carriers and cargos, PANAs minimize carrier-related toxicity and enhance therapeutic outcome. Zhang et al mentioned that a Förster Resonance Energy Transfer (FRET)-based PS system has been proposed, integrating Ce6 as the FRET donor and 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide (DiR) as the FRET acceptor. In this design, Ce6’s photodynamic activity is “switched off” until DiR is photobleached by NIR laser irradiation, ensuring controlled activation and minimizing systemic phototoxicity (Figure 4).140 This cascaded activation mechanism not only enhances PDT outcomes but also improves patient safety by preventing unwanted side effects.
![]() |
Figure 4 Schematic representation of the preparation of erythrocyte camouflaged Ce6@DiR NPs and its programmed cascade-activatable photothermal-photodynamic therapy for TNBC with low phototoxicity in normal tissues. Reprinted from Zhang X, Xiong J, Wang K, et al. Erythrocyte membrane-camouflaged carrier-free nanoassembly of FRET photosensitizer pairs with high therapeutic efficiency and high security for programmed cancer synergistic phototherapy. Bioact Mater. 2021;6(8):2291–2302.140 |
Biomimetic Nanocarrier Delivery Systems
Biomimetic NDSs (BM-NDSs) are innovative platforms capable of mimicking biological functions, enabling them to bypass immune recognition, extend circulation time, and enhance drug accumulation at target sites.143–145 Among the widely utilized technologies in BM-NDSs are extracellular vesicle-based formulations and cell membrane-coating strategies.146 RBC membranes are attracting attention as stealth barriers in drug delivery systems due to their low immunogenicity, high biocompatibility, and ability to prolong systemic circulation.147,148 By leveraging the inherent biological interfaces and self-recognition properties of RBC membranes, nanoparticles can interact effectively with their surrounding environments, enhancing retention in vivo.149 Yang et al developed RBC membrane-modified nanoparticles loaded with NIR-II photothermal agents (IR1061) and an IDO-1 inhibitor.150 These multifunctional nanoparticles showed targeted tumor accumulation and controlled drug release under laser irradiation, as evidenced by in vitro and in vivo results. Despite their promise, issues like ex vivo contamination and blood-type compatibility limit their large-scale use. Cancer cell membranes are another promising biomimetic approach.151 Tumor-derived membranes enable nanoparticles to evade immune detection and exhibit homotypic targeting, owing to surface proteins such as N-cadherin and epithelial adhesion molecules.152 Liu et al demonstrated that nanoparticles coated with HepG-2 cell membranes, encapsulating doxorubicin (DOX), showed prolonged circulation, immune evasion, and increased cellular uptake in homotypic tumor environments.153 Similarly, mesoporous silica nanoparticles wrapped in cancer cell membranes and functionalized with CaCO3 layers exhibited pH-sensitive drug release, leading to enhanced therapeutic efficacy against prostate cancer.154 Despite significant progress, challenges such as scalability, immune reaction control, and maintaining membrane functionality remain. Innovative solutions like the use of macrophage membranes to combine immune evasion with inflammatory targeting are under exploration. The integration of such biomimetic platforms into clinical applications demands further refinement to optimize stability, loading capacity, and biocompatibility while ensuring regulatory compliance.
Nanocarrier-Based PS Delivery Systems Combined with Other Lung Cancer Treatment Modalities
The complex TME is driven by a variety of mechanisms that promote tumor proliferation and metastasis, including the activation of bypass signaling pathways and the suppression of key apoptotic pathways. In recent years, combination therapies have emerged as a promising strategy to effectively inhibit tumor growth and metastasis. By combining PDT with chemotherapy, immunotherapy, and photothermal therapy (PTT), these synergistic strategies achieve precise targeting of lung cancer and promote the reprogramming of the TME.
Combining PDT with Chemotherapy
PDT induces tumor cell death by generating ROS through PSs, while chemotherapy drugs damage DNA or inhibit cell division. Combining both therapies can overcome tumor heterogeneity and enhance the ROS-mediated killing effect. Wang et al developed charge-switchable nano-micelles (NMs) that co-deliver doxorubicin (DOX) and paclitaxel (PTX), facilitating drug delivery via endosomal escape and significantly inhibiting the growth of lung adenocarcinoma.155 Long-term chemotherapy use often leads to multidrug resistance (MDR) in tumor cells; however, ROS generated by PDT can directly kill tumor cells, and its mechanism of action is unaffected by traditional resistance pathways. Additionally, PDT efficacy is often limited by hypoxia in the TME, but chemotherapy can alleviate this by disrupting tumor vasculature or inhibiting antioxidant systems (such as reducing glutathione levels), thereby enhancing PDT efficiency. Zhang et al designed a nanocarrier system that co-loads cisplatin and PSs to sensitize chemotherapy with PDT, reverse drug resistance, and reduce systemic side effects in NSCLC.156 Nanocarriers or responsive drug delivery systems, such as ROS-sensitive liposomes, can precisely control drug release, minimizing systemic toxicity. Liu et al developed a pH/ROS dual-responsive nanocarrier system (ZnP-OC-M), which triggers targeted release of PSs and chemotherapy drugs within the TME, thus enhancing the efficacy of PDT.157
Combining PDT with Immunotherapy
PDT induces immunogenic cell death (ICD), releasing tumor-associated antigens (TAAs) and damage-associated molecular patterns (DAMPs), which activate dendritic cells (DCs) and promote T-cell infiltration (Figure 5).158 This process disrupts the immune tolerance of tumors and creates a more sensitive microenvironment for immunotherapy, such as PD-1/PD-L1 inhibitors. Immunotherapy further amplifies the anti-tumor immune response triggered by PDT by relieving T-cell inhibition, generating systemic immune memory, and inhibiting distant metastasis and recurrence. The response rate to immune checkpoint inhibitors (ICIs) is relatively low (about 20%-30%), largely due to the “cold tumor” microenvironment. PDT can convert “cold tumors” into “hot tumors” by inducing ICD, significantly enhancing the efficacy of ICIs. Zheng et al designed a near-infrared light-responsive nanocarrier system (such as NIR-715) that induces endoplasmic reticulum (ER) stress via PDT, enhancing antigen presentation. When combined with anti-PD-1 antibodies, this system significantly inhibits the progression of “cold tumors”.159 However, it is important to note that when combining PDT with immunotherapy, it is crucial to optimize the co-delivery ratio of PSs and immunomodulators to avoid excessive immune activation, which could lead to a cytokine storm.160,161
![]() |
Figure 5 Illustration of near-infrared region II (NIR-II) photo-accelerated polymer nanoparticle (PPNP) enhancing tumor immunotherapy via Programmed Death-Ligand 1 (PD-L1) silencing and immunogenic cell death (ICD). (a) The preparation of PPNP/siRNA. (b) Upon activation by 808 nm laser and overexpressed GSH, PPNP/siRNA will undergo cleavage, releasing both the NIR-II PS and siPD-L1. ROS production will be stimulated, leading to tumor ICD, the release of damage-associated molecular patterns (DAMPs), recruitment of T cells to lymph nodes, activation of T cells, and initiation of T cell immune responses. Simultaneously, the released siPD-L1 will inhibit PD-L1 activity on tumor cells, enhance phagocytosis of tumor cells by T cells, activate systemic anti-tumor immune responses, and ultimately reshape the immune microenvironment. This photo-accelerated immunotherapy strategy facilitates tumor-specific immune responses and remodeling of the TME. Reprinted from Zhang T, Tang D, Wu P, et al. NIR-II photo-accelerated polymer nanoparticles boost tumor immunotherapy via PD-L1 silencing andimmunogenic cell death. Bioact Mater. 2025;46:285–300.158 Abbreviations: iDC, Immature Dendritic Cells; DC, Dendritic Cells; CTLs, Cytotoxic T Lymphocytes. |
Synergistic PDT and PTT
The photothermal effect can enhance tumor vascular permeability, promoting the accumulation of nanocarriers, and accelerate the diffusion of ROS through localized heating, thus expanding the area of tumor cell destruction. Copper nanoclusters (CuNCs) loaded with the PS Ce6 and hyaluronic acid (HA) simultaneously generate ROS and localized high temperatures under near-infrared light, disrupting tumor blood vessels and inducing apoptosis.162 Traditional PDT/PTT combination therapies require lasers of two different wavelengths, but novel nanomaterials, such as SPAuNCs and Ce6-PEG-AuNR, can activate both PDT and PTT with a single wavelength laser. This reduces treatment time and minimizes the risk of skin burns. For instance, gold nanoparticles (AuNPs) can serve as carriers for both PDT and PTT, achieving dual-mode killing with a single laser exposure, thereby causing more thorough destruction of lung cancer cells.163
Multifunctional Nanocarrier Delivery Systems
The hypoxic TME limits the generation of ROS in PDT. Nanocarriers equipped with catalase or oxygen carriers (such as ICG@ZIF-82) can provide in situ oxygenation during PDT, while simultaneously activating hypoxia-activated chemotherapy drugs (eg, tirapazamine). This enables the synergistic effect of PDT and hypoxia-activated chemotherapy.164,165 PDT’s ability to penetrate deep tumors is often limited, but smart nanocarriers (such as mPEG-azo-HA-Ce6) can dissociate in the acidic TME, releasing the PS Ce6 and enhancing ROS accumulation. When combined with chemotherapy, this can improve penetration into deep tumor tissues.166,167 Additionally, targeted nanocarriers can enhance tumor targeting via receptor-mediated endocytosis, reducing damage to normal tissues.168
Conclusion and Future Prospects
In the field of lung cancer treatment, PDT has gradually emerged as an important alternative or adjunctive treatment due to its non-invasive nature, precise targeting, and low systemic toxicity. However, traditional PDT still faces key challenges such as low PS delivery efficiency, TME hypoxia, and limited diffusion range of ROS. The introduction of nanocarrier technology has provided innovative solutions to overcome these bottlenecks, significantly advancing the clinical application of PDT. Despite the immense potential of nanocarriers in lung cancer PDT, there are still challenges in their clinical translation: 1) the stability and biocompatibility of certain nanomaterials need further verification; and 2) existing research predominantly focuses on in vitro or small animal models, necessitating large-scale clinical trials to confirm their safety and efficacy.
In conclusion, nanocarrier technology offers a novel approach to optimize lung cancer PDT through precise delivery, oxygen enhancement, and multimodal synergistic strategies. Future research should focus on addressing clinical translation barriers and promoting the development of personalized nanomedicines to achieve more efficient and precise lung cancer treatment.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA a Cancer J Clinicians. 2021;71(3):209–249. doi:10.3322/caac.21660
2. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA a Cancer J Clinicians. 2024;74(1):12–49. doi:10.3322/caac.21820
3. Ettinger DS, Wood DE, Aisner DL, et al. NCCN guidelines insights: non–small cell lung cancer, version 2.2021: featured updates to the NCCN guidelines. J National Compr Cancer Network. 2021;19(3):254–266. doi:10.6004/jnccn.2021.0013
4. Nasim F, Sabath BF, Eapen GA. Lung cancer. Med Clin North Am. 2019;103(3):463–473. doi:10.1016/j.mcna.2018.12.006
5. Howington JA, Blum MG, Chang AC, Balekian AA, Murthy SC. Treatment of stage I and II non-small cell lung cancer. Chest. 2013;143(5):e278S–e313S. doi:10.1378/chest.12-2359
6. Kim J, Kim J, Yoon H, Chae YJ, Rhew K, Chang JE. The in vitro and in vivo anticancer effect of photomed for photodynamic therapy: comparison with photofrin and radachlorin. CIMB. 2023;45(3):2474–2490. doi:10.3390/cimb45030162
7. Akopov A, Papayan G. Photodynamic theranostics of central lung cancer: present state and future prospects. Photodiagn Photodyn Ther. 2021;33:102203. doi:10.1016/j.pdpdt.2021.102203
8. Liu YQ, Wang XL, He DH, Cheng YX. Protection against chemotherapy- and radiotherapy-induced side effects: a review based on the mechanisms and therapeutic opportunities of phytochemicals. Phytomedicine. 2021;80:153402. doi:10.1016/j.phymed.2020.153402
9. Sun R, Ma W, Ling M, et al. pH-activated nanoplatform for visualized photodynamic and ferroptosis synergistic therapy of tumors. J Control Release. 2022;350:525–537. doi:10.1016/j.jconrel.2022.08.050
10. Mayne NR, Lin BK, Darling AJ, et al. Stereotactic body radiotherapy versus delayed surgery for early-stage non-small-cell lung cancer. Ann Surg. 2020;272(6):925–929. doi:10.1097/SLA.0000000000004363
11. Wang M, Herbst RS, Boshoff C. Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med. 2021;27(8):1345–1356. doi:10.1038/s41591-021-01450-2
12. Huang J, Zhuang C, Chen J, et al. Targeted drug/gene/photodynamic therapy via a stimuli‐responsive dendritic‐polymer‐based nanococktail for treatment of EGFR‐TKI‐resistant non‐small‐cell lung cancer. Adv Mater. 2022;34(27):2201516. doi:10.1002/adma.202201516
13. Brown SB, Brown EA, Walker I. The present and future role of photodynamic therapy in cancer treatment. Lancet Oncol. 2004;5(8):497–508. doi:10.1016/S1470-2045(04)01529-3
14. Shen J, Xia H, Zhou X, et al. Selenium enhances photodynamic therapy of C-phycocyanin against lung cancer via dual regulation of cytotoxicity and antioxidant activity. ABBS. 2023;55(12):1925–1937. doi:10.3724/abbs.2023159
15. Callaghan S, Senge MO. The good, the bad, and the ugly–controlling singlet oxygen through design of photosensitizers and delivery systems for photodynamic therapy. Photochem Photobiol Sci. 2018;17(11):1490–1514. doi:10.1039/c8pp00008e
16. Olszowy M, Nowak-Perlak M, Woźniak M. Current strategies in photodynamic therapy (PDT) and photodynamic diagnostics (PDD) and the future potential of nanotechnology in cancer treatment. Pharmaceutics. 2023;15(6):1712. doi:10.3390/pharmaceutics15061712
17. Kustov AV, Berezin DB, Zorin VP, et al. Monocationic chlorin as a promising photosensitizer for antitumor and antimicrobial photodynamic therapy. Pharmaceutics. 2022;15(1):61. doi:10.3390/pharmaceutics15010061
18. Chang JE, Cho HJ, Yi E, Kim DD, Jheon S. Hypocrellin B and paclitaxel-encapsulated hyaluronic acid–ceramide nanoparticles for targeted photodynamic therapy in lung cancer. J Photochem Photobiol B Biol. 2016;158:113–121. doi:10.1016/j.jphotobiol.2016.02.035
19. Shafirstein G, Battoo A, Harris K, et al. Photodynamic therapy of non–small cell lung cancer. Narrative Rev Future Directions Annals ATS. 2016;13(2):265–275. doi:10.1513/AnnalsATS.201509-650FR
20. Zhang J, Jiang C, Longo JPF, Azevedo RB, Zhang H, Muehlmann LA. An updated overview on the development of new photosensitizers for anticancer photodynamic therapy. Acta Pharmaceutica Sinica B. 2018;8(2):137–146. doi:10.1016/j.apsb.2017.09.003
21. Shi R, Li C, Jiang Z, Li W, Wang A, Wei J. Preclinical study of antineoplastic sinoporphyrin sodium-PDT via in vitro and in vivo models. Molecules. 2017;22(1):112. doi:10.3390/molecules22010112
22. Hsieh YJ, Chien KY, Yang IF, et al. Oxidation of protein-bound methionine in Photofrin-photodynamic therapy-treated human tumor cells explored by methionine-containing peptide enrichment and quantitative proteomics approach. Sci Rep. 2017;7(1):1370. doi:10.1038/s41598-017-01409-9
23. Wang S, Bromley E, Xu L, Chen JC, Keltner L. Talaporfin sodium. Expert Opin Pharmacother. 2010;11(1):133–140. doi:10.1517/14656560903463893
24. Sasaki M, Tanaka M, Kojima Y, et al. Anti-tumor immunity enhancement by photodynamic therapy with talaporfin sodium and anti-programmed death 1 antibody. Mol Ther. 2023;28:118–131. doi:10.1016/j.omto.2022.12.009
25. Yuan P, Ruan Z, Yan L. Tetraphenylporphine-modified polymeric nanoparticles containing nir photosensitizer for mitochondria-targeting and imaging-guided photodynamic therapy. ACS Biomater Sci Eng. 2020;6(2):1043–1051. doi:10.1021/acsbiomaterials.9b01662
26. Liang HY, Jiang Y, Yan YJ, Wang LQ, Chen PR, Chen ZL. Synthesis of novel meso-tetraphenyltetrabenzoporphyrins and evaluation of their photodynamic anticancer activities. ACS Omega. 2025;10(11):11119–11127. doi:10.1021/acsomega.4c10221
27. Kustov AV, Morshnev PK, Kukushkina NV, et al. Solvation, cancer cell photoinactivation and the interaction of chlorin photosensitizers with a potential passive carrier non-ionic surfactant tween 80. IJMS. 2022;23(10):5294. doi:10.3390/ijms23105294
28. Yang J, Hou M, Sun W, et al. Sequential PDT and PTT using dual‐modal single‐walled carbon nanohorns synergistically promote systemic immune responses against tumor metastasis and relapse. Adv Sci. 2020;7(16):2001088. doi:10.1002/advs.202001088
29. Zhang P, Wu Q, Yang J, et al. Tumor microenvironment-responsive nanohybrid for hypoxia amelioration with photodynamic and near-infrared II photothermal combination therapy. Acta Biomater. 2022;146:450–464. doi:10.1016/j.actbio.2022.04.044
30. Yurt F, Ince M, Colak SG, et al. Investigation of in vitro PDT activities of zinc phthalocyanine immobilised TiO2 nanoparticles. Int J Pharm. 2017;524(1):467–474. doi:10.1016/j.ijpharm.2017.03.050
31. Jing C, Wang R, Ou H, et al. Axial modification inhibited H-aggregation of phthalocyanines in polymeric micelles for enhanced PDT efficacy. Chem Commun. 2018;54(32):3985–3988. doi:10.1039/C7CC09954A
32. Luo T, Zhang Q, Lu QB. Combination of near infrared light-activated photodynamic therapy mediated by indocyanine green with etoposide to treat non-small-cell lung cancer. Cancers. 2017;9(6):63. doi:10.3390/cancers9060063
33. Yang L, Huang B, Hu S, et al. Indocyanine green assembled free oxygen-nanobubbles towards enhanced near-infrared induced photodynamic therapy. Nano Res. 2022;15(5):4285–4293. doi:10.1007/s12274-022-4085-0
34. Obstoy B, Salaun M, Bohn P, Veresezan L, Sesboué R, Thiberville L. Photodynamic therapy using methylene blue in lung adenocarcinoma xenograft and hamster cheek pouch induced squamous cell carcinoma. Photodiagn Photodyn Ther. 2016;15:109–114. doi:10.1016/j.pdpdt.2016.03.003
35. Kirla H, Wu J, Hamzah J, Henry DJ. One-pot synthesis and covalent conjugation of methylene blue in mesoporous silica nanoparticles – a platform for enhanced photodynamic therapy. Colloids Surf B. 2025;245:114195. doi:10.1016/j.colsurfb.2024.114195
36. de Andrade GP, de Souza TFM, Cerchiaro G, da S Pinhal MA, Ribeiro AO, Girão MJBC. Hypericin in photobiological assays: an overview. Photodiagn Photodyn Ther. 2021;35:102343. doi:10.1016/j.pdpdt.2021.102343
37. de Morais FAP, Gonçalves RS, Campanholi KS, et al. Photophysical characterization of Hypericin-loaded in micellar, liposomal and copolymer-lipid nanostructures based F127 and DPPC liposomes. Spectrochimica Acta Part A. 2021;248:119173. doi:10.1016/j.saa.2020.119173
38. Sun M, Zhang Y, He Y, et al. Green synthesis of carrier-free curcumin nanodrugs for light-activated breast cancer photodynamic therapy. Colloids Surf B. 2019;180:313–318. doi:10.1016/j.colsurfb.2019.04.061
39. Amaroli A, Panfoli I, Bozzo M, Ferrando S, Candiani S, Ravera S. The bright side of curcumin: a narrative review of its therapeutic potential in cancer management. Cancers. 2024;16(14):2580. doi:10.3390/cancers16142580
40. Tavakkoli Yaraki M, Liu B, Tan YN. Emerging strategies in enhancing singlet oxygen generation of nano-photosensitizers toward advanced phototherapy. Nano-Micro Lett. 2022;14(1):123. doi:10.1007/s40820-022-00856-y
41. Machuca A, Garcia-Calvo E, Anunciação DS, Luque-Garcia JL. Rhodium nanoparticles as a novel photosensitizing agent in photodynamic therapy against cancer. Chem Eur J. 2020;26(34):7685–7691. doi:10.1002/chem.202001112
42. Mariano S, Carata E, Calcagnile L, Panzarini E. Recent advances in photodynamic therapy: metal-based nanoparticles as tools to improve cancer therapy. Pharmaceutics. 2024;16(7):932. doi:10.3390/pharmaceutics16070932
43. Li Y, Wu Q, Kang M, Song N, Wang D, Tang BZ. Boosting the photodynamic therapy efficiency by using stimuli-responsive and AIE-featured nanoparticles. Biomaterials. 2020;232:119749. doi:10.1016/j.biomaterials.2019.119749
44. Wu K, Liu J, Zhang X, et al. Bovine serum albumin framed activatable NIR AIE photosensitizer for targeted tumor therapy. Biomaterials. 2025;315:122918. doi:10.1016/j.biomaterials.2024.122918
45. Wang DH, Chen LJ, Zhao X, Yan XP. Enhancing near-infrared AIE of photosensitizer with twisted intramolecular charge transfer characteristics via rotor effect for AIE imaging-guided photodynamic ablation of cancer cells. Talanta. 2021;225:122046. doi:10.1016/j.talanta.2020.122046
46. Yu Z, Zhou J, Ji X, et al. Discovery of a monoiodo Aza-BODIPY near-infrared photosensitizer: in vitro and in vivo evaluation for photodynamic therapy. J Med Chem. 2020;63(17):9950–9964. doi:10.1021/acs.jmedchem.0c00882
47. Pinjari D, Patil Y, Misra R. Near-infrared absorbing aza-BODIPY dyes for optoelectronic applications. Chem – Asian J. 2024;19(15):e202400167. doi:10.1002/asia.202400167
48. Ruan X, Wei M, He X, et al. Asymmetric aza-BODIPY photosensitizer for photoacoustic/photothermal imaging-guided synergistic photodynamic/photothermal therapy. Colloids Surf B. 2023;231:113547. doi:10.1016/j.colsurfb.2023.113547
49. Jiang S, Li H, Zhang L, et al. Generic Diagramming Platform (GDP): a comprehensive database of high-quality biomedical graphics. Nucleic Acids Res. 2025;53(D1):D1670–D1676. doi:10.1093/nar/gkae973
50. Luo Z, Zheng M, Zhao P, et al. Self-monitoring artificial red cells with sufficient oxygen supply for enhanced photodynamic therapy. Sci Rep. 2016;6(1):23393. doi:10.1038/srep23393
51. Li L, Rashidi LH, Yao M, et al. CuS nanoagents for photodynamic and photothermal therapies: phenomena and possible mechanisms. Photodiagn Photodyn Ther. 2017;19:5–14. doi:10.1016/j.pdpdt.2017.04.001
52. Varon E, Blumrosen G, Sinvani M, et al. An engineered nanocomplex with photodynamic and photothermal synergistic properties for cancer treatment. Int J Mol Sci. 2022;23(4):2286. doi:10.3390/ijms23042286
53. Adnane F, Soliman SMA, ElZayat E, Abdelsalam EM, Fahmy HM. Evaluation of chlorophyll-loaded mesoporous silica nanoparticles for photodynamic therapy on cancer cell lines. Lasers Med Sci. 2024;39(1):45. doi:10.1007/s10103-024-03988-2
54. Kim J, Cho HR, Jeon H, et al. Continuous O 2 -evolving MnFe 2 O 4 nanoparticle-anchored mesoporous silica nanoparticles for efficient photodynamic therapy in hypoxic cancer. J Am Chem Soc. 2017;139(32):10992–10995. doi:10.1021/jacs.7b05559
55. Zhang M, Liu X, Mao Y, et al. Oxygen-generating hydrogels overcome tumor hypoxia to enhance photodynamic/gas synergistic therapy. ACS Appl Mater Interfaces. 2022;14(24):27551–27563. doi:10.1021/acsami.2c02949
56. Gao M, Fan F, Li D, et al. Tumor acidity-activatable TAT targeted nanomedicine for enlarged fluorescence/magnetic resonance imaging-guided photodynamic therapy. Biomaterials. 2017;133:165–175. doi:10.1016/j.biomaterials.2017.04.013
57. Li T, Yan L. Functional polymer nanocarriers for photodynamic therapy. Pharmaceuticals. 2018;11(4). doi:10.3390/ph11040133
58. Chkair R, Couvez J, Brégier F, et al. Activity of hydrophilic, biocompatible, fluorescent, organic nanoparticles functionalized with purpurin-18 in photodynamic therapy for colorectal cancer. Nanomaterials. 2024;14(19):1557. doi:10.3390/nano14191557
59. Montaseri H, Kruger CA, Abrahamse H. Inorganic nanoparticles applied for active targeted photodynamic therapy of breast cancer. Pharmaceutics. 2021;13(3). doi:10.3390/pharmaceutics13030296
60. Youssef Z, Vanderesse R, Colombeau L, et al. The application of titanium dioxide, zinc oxide, fullerene, and graphene nanoparticles in photodynamic therapy. Cancer Nanotechnol. 2017;8(1):6. doi:10.1186/s12645-017-0032-2
61. Song Y, Li Y, Zhang Y, Wang L, Xie Z. Self-quenching synthesis of coordination polymer pre-drug nanoparticles for selective photodynamic therapy. J Mater Chem B. 2019;7(48):7776–7782. doi:10.1039/C9TB01937E
62. Yao B, Xu F, Tian Z, et al. Strong persistent luminescence NaYF4-based Nanoparticles combined with manipulated hyperfractionated irradiation for X-ray-excited photodynamic therapy enhancement. ACS Appl Mater Interfaces. 2025;17(11):16561–16575. doi:10.1021/acsami.4c20049
63. Zhang W, Zhang X, Shen Y, et al. Ultra-high FRET efficiency NaGdF4: tb3+-Rose Bengal biocompatible nanocomposite for X-ray excited photodynamic therapy application. Biomaterials. 2018;184:31–40. doi:10.1016/j.biomaterials.2018.09.001
64. Sun B, Wang L, Li Q, et al. Bis(pyrene)-doped cationic dipeptide nanoparticles for two-photon-activated photodynamic therapy. Biomacromolecules. 2017;18(11):3506–3513. doi:10.1021/acs.biomac.7b00780
65. Wang K, Yu B, Pathak JL. An update in clinical utilization of photodynamic therapy for lung cancer. J Cancer. 2021;12(4):1154–1160. doi:10.7150/jca.51537
66. Kato H, Harada M, Ichinose S, Usuda J, Tsuchida T, Okunaka T. Photodynamic therapy (PDT) of lung cancer: experience of the Tokyo Medical University. Photodiagn Photodyn Ther. 2004;1(1):49–55. doi:10.1016/S1572-1000(04)00008-0
67. Detterbeck FC, Lewis SZ, Diekemper R, Addrizzo-Harris D, Alberts WM. Executive summary. Chest. 2013;143(5):7S–37S. doi:10.1378/chest.12-2377
68. Tsuchida T, Matsumoto Y, Imabayashi T, Uchimura K, Sasada S. Photodynamic therapy can be safely performed with Talaporfin sodium as a day treatment for central-type early-stage lung cancer. Photodiagn Photodyn Ther. 2022;38:102836. doi:10.1016/j.pdpdt.2022.102836
69. Simone CB, Friedberg JS, Glatstein E, et al. Photodynamic therapy for the treatment of non-small cell lung cancer. J Thorac Dis. 2012;4(1):63–75. doi:10.3978/j.issn.2072-1439.2011.11.05
70. Jung HS, Kim HJ. Definitive surgery and intraoperative photodynamic therapy for locally advanced non-small cell lung cancer: a case report. World J Surg Oncol. 2022;20(1):265. doi:10.1186/s12957-022-02729-5
71. Ross PJ, Grecula J, Bekaii-Saab T, Villalona-Calero M, Otterson G, Magro C. Incorporation of photodynamic therapy as an induction modality in non-small cell lung cancer. Lasers Surg Med. 2006;38(10):881–889. doi:10.1002/lsm.20444
72. Mehta HJ, Biswas A, Fernandez-Bussy S, Pipkin M, Machuca T, Jantz MA. Photodynamic therapy for bronchial microscopic residual disease after resection in lung cancer. J Bronchology Interv Pulmonol. 2019;26(1):49–54. doi:10.1097/LBR.0000000000000510
73. Chhatre S, Vachani A, Allison RR, Jayadevappa R. Survival outcomes with photodynamic therapy, chemotherapy and radiation in patients with stage III or stage IV non-small cell lung cancer. Cancers. 2021;13(4):803. doi:10.3390/cancers13040803
74. Kimura M, Miyajima K, Kojika M, Kono T, Kato H. Photodynamic therapy (PDT) with chemotherapy for advanced lung cancer with airway stenosis. Int J Mol Sci. 2015;16(10):25466–25475. doi:10.3390/ijms161025466
75. Falk‐Mahapatra R, Gollnick SO. Photodynamic therapy‐induced cyclooxygenase 2 expression in tumor‐draining lymph nodes regulates B‐cell expression of interleukin 17 and neutrophil infiltration. Photochem Photobiol. 2022;98(5):1207–1214. doi:10.1111/php.13601
76. Yu TT, Hu J, Li QR, et al. Chlorin e6-induced photodynamic effect facilitates immunogenic cell death of lung cancer as a result of oxidative endoplasmic reticulum stress and DNA damage. Int Immunopharmacol. 2023;115:109661. doi:10.1016/j.intimp.2022.109661
77. Guo R, Wang S, Zhao L, et al. Engineered nanomaterials for synergistic photo-immunotherapy. Biomaterials. 2022;282:121425. doi:10.1016/j.biomaterials.2022.121425
78. Choi J, Sun IC, Sook hwang H, Yeol Yoon H, Kim K. Light-triggered photodynamic nanomedicines for overcoming localized therapeutic efficacy in cancer treatment. Adv Drug Delivery Rev. 2022;186:114344. doi:10.1016/j.addr.2022.114344
79. Gao L, Zhang C, Gao D, et al. Enhanced anti-tumor efficacy through a combination of integrin αvβ6-targeted photodynamic therapy and immune checkpoint inhibition. Theranostics. 2016;6(5):627–637. doi:10.7150/thno.14792
80. Pérez-Hernández M, Del Pino P, Mitchell SG, et al. Dissecting the molecular mechanism of apoptosis during photothermal therapy using gold nanoprisms. ACS Nano. 2015;9(1):52–61. doi:10.1021/nn505468v
81. Petty WJ, Paz-Ares L. Emerging strategies for the treatment of small cell lung cancer: a review. JAMA Oncol. 2023;9(3):419. doi:10.1001/jamaoncol.2022.5631
82. Tang JLY, Moonshi SS, Ta HT. Nanoceria: an innovative strategy for cancer treatment. Cell Mol Life Sci. 2023;80(2):46. doi:10.1007/s00018-023-04694-y
83. Crous A, Abrahamse H. Photodynamic therapy of lung cancer, where are we? Front Pharmacol. 2022;13:932098. doi:10.3389/fphar.2022.932098
84. Laszló IP, Laszló MR, Popescu T, et al. The comparative effects of resveratrol and curcumin in combination with photodynamic therapy. Med Pharm Rep. 2022. doi:10.15386/mpr-2497
85. Verissimo TV, Santos NT, Silva JR, Azevedo RB, Gomes AJ, Lunardi CN. In vitro cytotoxicity and phototoxicity of surface-modified gold nanoparticles associated with neutral red as a potential drug delivery system in phototherapy. Mater Sci Eng C. 2016;65:199–204. doi:10.1016/j.msec.2016.04.030
86. Larue L, Myrzakhmetov B, Ben-Mihoub A, et al. Fighting hypoxia to improve PDT. Pharmaceuticals. 2019;12(4):163. doi:10.3390/ph12040163
87. Kim TE, Chang JE. Recent studies in photodynamic therapy for cancer treatment: from basic research to clinical trials. Pharmaceutics. 2023;15(9):2257. doi:10.3390/pharmaceutics15092257
88. Busch TM. Hypoxia and perfusion labeling during photodynamic therapy. In: Gomer CJ editor. Photodynamic Therapy. Vol 635. Methods in Molecular Biology. Humana Press; 2010:107–120. doi:10.1007/978-1-60761-697-9_8
89. Zheng X, Pan D, Chen M, et al. Tunable hydrophile–lipophile balance for manipulating structural stability and tumor retention of amphiphilic nanoparticles. Adv Mater. 2019;31(35):1901586. doi:10.1002/adma.201901586
90. Li F, Du Y, Liu J, et al. Responsive assembly of upconversion nanoparticles for pH‐activated and near‐infrared‐triggered photodynamic therapy of deep tumors. Adv Mater. 2018;30(35):1802808. doi:10.1002/adma.201802808
91. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer. 2011;11(6):393–410. doi:10.1038/nrc3064
92. Alshehri S, Imam SS, Rizwanullah M, et al. Progress of cancer nanotechnology as diagnostics, therapeutics, and theranostics nanomedicine: preclinical promise and translational challenges. Pharmaceutics. 2020;13(1):24. doi:10.3390/pharmaceutics13010024
93. Navya PN, Kaphle A, Srinivas SP, Bhargava SK, Rotello VM, Daima HK. Current trends and challenges in cancer management and therapy using designer nanomaterials. Nano Convergence. 2019;6(1):23. doi:10.1186/s40580-019-0193-2
94. Zhang Y, Li M, Gao X, Chen Y, Liu T. Nanotechnology in cancer diagnosis: progress, challenges and opportunities. J Hematol Oncol. 2019;12(1):137. doi:10.1186/s13045-019-0833-3
95. Silva CO, Pinho JO, Lopes JM, Almeida AJ, Gaspar MM, Reis C. Current trends in cancer nanotheranostics: metallic, polymeric, and lipid-based systems. Pharmaceutics. 2019;11(1):22. doi:10.3390/pharmaceutics11010022
96. Saleem HM, Ramaiah P, Gupta J, et al. Nanotechnology-empowered lung cancer therapy: from EMT role in cancer metastasis to application of nanoengineered structures for modulating growth and metastasis. Environ Res. 2023;232:115942. doi:10.1016/j.envres.2023.115942
97. Güleryüz B, Işık A, Gülsoy M. Synergistic effect of mesoporous silica nanocarrier-assisted photodynamic therapy and anticancer agent activity on lung cancer cells. Lasers Med Sci. 2024;39(1):91. doi:10.1007/s10103-023-03969-x
98. Ruman U, Fakurazi S, Masarudin MJ, Hussein MZ. Nanocarrier-based therapeutics and theranostics drug delivery systems for next generation of liver cancer nanodrug modalities. IJN. 2020;15:1437–1456. doi:10.2147/IJN.S236927
99. Huo M, Wang L, Chen Y, Shi J. Tumor-selective catalytic nanomedicine by nanocatalyst delivery. Nat Commun. 2017;8(1):357. doi:10.1038/s41467-017-00424-8
100. Hao Y, Chen Y, He X, et al. Polymeric nanoparticles with ROS‐responsive prodrug and platinum nanozyme for enhanced chemophotodynamic therapy of colon cancer. Adv Sci. 2020;7(20):2001853. doi:10.1002/advs.202001853
101. Zhu X, Chen X, Huo D, et al. A hybrid nanozymes in situ oxygen supply synergistic photothermal/chemotherapy of cancer management. Biomater Sci. 2021;9(15):5330–5343. doi:10.1039/D1BM00667C
102. Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol. 2020;17(11):657–674. doi:10.1038/s41571-020-0410-2
103. Hong EJ, Choi DG, Shim MS. Targeted and effective photodynamic therapy for cancer using functionalized nanomaterials. Acta Pharmaceutica Sinica B. 2016;6(4):297–307. doi:10.1016/j.apsb.2016.01.007
104. Krafft MP. Alleviating tumor hypoxia with perfluorocarbon-based oxygen carriers. Curr Opin Pharmacol. 2020;53:117–125. doi:10.1016/j.coph.2020.08.010
105. Li T, Jing X, Huang Y. Polymer/hemoglobin assemblies: biodegradable oxygen carriers for artificial red blood cells. Macromol Biosci. 2011;11(7):865–875. doi:10.1002/mabi.201000469
106. Wan Y, Fu L, Li C, Lin J, Huang P. Conquering the hypoxia limitation for photodynamic therapy. Adv Mater. 2021;33(48):2103978. doi:10.1002/adma.202103978
107. Jin Z, Zhao Q, Yuan S, Jiang W, Hu Y. Strategies of alleviating tumor hypoxia and enhancing tumor therapeutic effect by macromolecular nanomaterials. Macromol Biosci. 2021;21(8):2100092. doi:10.1002/mabi.202100092
108. Xavierselvan M, Cook J, Duong J, Diaz N, Homan K, Mallidi S. Photoacoustic nanodroplets for oxygen enhanced photodynamic therapy of cancer. Photoacoustics. 2022;25:100306. doi:10.1016/j.pacs.2021.100306
109. Guo X, Qu J, Zhu C, et al. Synchronous delivery of oxygen and photosensitizer for alleviation of hypoxia tumor microenvironment and dramatically enhanced photodynamic therapy. Drug Delivery. 2018;25(1):585–599. doi:10.1080/10717544.2018.1435751
110. Ma S, Zhou J, Zhang Y, et al. An oxygen self-sufficient fluorinated nanoplatform for relieved tumor hypoxia and enhanced photodynamic therapy of cancers. ACS Appl Mater Interfaces. 2019;11(8):7731–7742. doi:10.1021/acsami.8b19840
111. Ding L, Wu Y, Wu M, et al. Engineered red blood cell biomimetic nanovesicle with oxygen self-supply for near-infrared-II fluorescence-guided synergetic chemo-photodynamic therapy against hypoxic tumors. ACS Appl Mater Interfaces. 2021;13(44):52435–52449. doi:10.1021/acsami.1c19096
112. Chen W, Wang Z, Tian M, et al. Integration of TADF photosensitizer as “Electron Pump” and BSA as “Electron Reservoir” for boosting type I photodynamic therapy. J Am Chem Soc. 2023;145(14):8130–8140. doi:10.1021/jacs.3c01042
113. Hou X, Tao Y, Li X, et al. CD44-targeting oxygen self-sufficient nanoparticles for enhanced photodynamic therapy against malignant melanoma. IJN. 2020;15:10401–10416. doi:10.2147/IJN.S283515
114. Wang H, Chao Y, Liu J, et al. Photosensitizer-crosslinked in-situ polymerization on catalase for tumor hypoxia modulation & enhanced photodynamic therapy. Biomaterials. 2018;181:310–317. doi:10.1016/j.biomaterials.2018.08.011
115. Chen Y, Cai Y, Yu X, et al. A photo and tumor microenvironment activated nano-enzyme with enhanced ROS generation and hypoxia relief for efficient cancer therapy. J Mater Chem B. 2021;9(39):8253–8262. doi:10.1039/D1TB01437D
116. Zhao YY, Zhang L, Chen Z, et al. Nanostructured phthalocyanine assemblies with efficient synergistic effect of type I photoreaction and photothermal action to overcome tumor hypoxia in photodynamic therapy. J Am Chem Soc. 2021;143(34):13980–13989. doi:10.1021/jacs.1c07479
117. Daurio NA, Tuttle SW, Worth AJ, et al. AMPK activation and metabolic reprogramming by tamoxifen through estrogen receptor–independent mechanisms suggests new uses for this therapeutic modality in cancer treatment. Cancer Res. 2016;76(11):3295–3306. doi:10.1158/0008-5472.CAN-15-2197
118. Tomaselli F, Maier A, Sankin O, et al. Acute effects of combined photodynamic therapy and hyperbaric oxygenation in lung cancer—A clinical pilot study. Lasers Surg Med. 2001;28(5):399–403. doi:10.1002/lsm.1067
119. Liu H, Yao C, Zhang L, Xin J, Zhang Z, Wang S. Nanoliposomes co-encapsulating Ce6 and SB3CT against the proliferation and metastasis of melanoma with the integration of photodynamic therapy and NKG2D-related immunotherapy on A375 cells. Nanotechnology. 2021;32(45):455102. doi:10.1088/1361-6528/ac1afd
120. Md S, Haque S, Madheswaran T, et al. Lipid based nanocarriers system for topical delivery of photosensitizers. Drug Discovery Today. 2017;22(8):1274–1283. doi:10.1016/j.drudis.2017.04.010
121. Ma Z, Hu P, Guo C, et al. Folate-mediated and pH-responsive chidamide-bound micelles encapsulating photosensitizers for tumor-targeting photodynamic therapy. IJN. 2019;14:5527–5540. doi:10.2147/IJN.S208649
122. Zheng N, Zhang Z, Kuang J, et al. Poly(photosensitizer) nanoparticles for enhanced in vivo photodynamic therapy by interrupting the π–π stacking and extending circulation time. ACS Appl Mater Interfaces. 2019;11(20):18224–18232. doi:10.1021/acsami.9b04351
123. Haimov E, Weitman H, Polani S, Schori H, Zitoun D, Shefi O. meso -tetrahydroxyphenylchlorin-conjugated gold nanoparticles as a tool to improve photodynamic therapy. ACS Appl Mater Interfaces. 2018;10(3):2319–2327. doi:10.1021/acsami.7b16455
124. Clemente N, Miletto I, Gianotti E, et al. Verteporfin-loaded mesoporous silica nanoparticles’ topical applications inhibit mouse melanoma lymphangiogenesis and micrometastasis in vivo. IJMS. 2021;22(24):13443. doi:10.3390/ijms222413443
125. Das RP, Gandhi VV, Singh BG, Kunwar A. Passive and active drug targeting: role of nanocarriers in rational design of anticancer formulations. CPD. 2019;25(28):3034–3056. doi:10.2174/1381612825666190830155319
126. Wang Q, Zhang P, Li Z, et al. Evaluation of polymer nanoformulations in hepatoma therapy by established rodent models. Theranostics. 2019;9(5):1426–1452. doi:10.7150/thno.31683
127. Keyvan Rad J, Mahdavian AR, Khoei S, Shirvalilou S. Enhanced photogeneration of reactive oxygen species and targeted photothermal therapy of C6 glioma brain cancer cells by folate-conjugated gold–photoactive polymer nanoparticles. ACS Appl Mater Interfaces. 2018;10(23):19483–19493. doi:10.1021/acsami.8b05252
128. Chen W, Luo G, Zhang X. Recent advances in subcellular targeted cancer therapy based on functional materials. Adv Mater. 2019;31(3):1802725. doi:10.1002/adma.201802725
129. Karges J, Tharaud M, Gasser G. Polymeric encapsulation of a Ru(II)-based photosensitizer for folate targeted photodynamic therapy of drug resistant cancers. J Med Chem. 2020. doi:10.26434/chemrxiv.13258613.v1
130. Sun X, Xu Y, Guo Q, et al. A novel nanoprobe for targeted imaging and photothermal/photodynamic therapy of lung cancer. Langmuir. 2022;38(4):1360–1367. doi:10.1021/acs.langmuir.1c02434
131. Paszko E, Ehrhardt C, Senge MO, Kelleher DP, Reynolds JV. Nanodrug applications in photodynamic therapy. Photodiagn Photodyn Ther. 2011;8(1):14–29. doi:10.1016/j.pdpdt.2010.12.001
132. Li M, Xia J, Tian R, et al. Near-infrared light-initiated molecular superoxide radical generator: rejuvenating photodynamic therapy against hypoxic tumors. J Am Chem Soc. 2018;140(44):14851–14859. doi:10.1021/jacs.8b08658
133. Zhang Q, Cai Y, Li Q, et al. Targeted delivery of a mannose‐conjugated BODIPY photosensitizer by nanomicelles for photodynamic breast cancer therapy. Chemistry a European J. 2017;23(57):14307–14315. doi:10.1002/chem.201702935
134. Guo J, Zhao W, Xiao X, et al. Reprogramming exosomes for immunity-remodeled photodynamic therapy against non-small cell lung cancer. Bioact Mater. 2024;39:206–223. doi:10.1016/j.bioactmat.2024.05.030
135. Sato Y, Nakamura T, Yamada Y, Harashima H. The nanomedicine rush: new strategies for unmet medical needs based on innovative nano DDS. J Control Release. 2021;330:305–316. doi:10.1016/j.jconrel.2020.12.032
136. Zhao Y, Fay F, Hak S, et al. Augmenting drug–carrier compatibility improves tumour nanotherapy efficacy. Nat Commun. 2016;7(1):11221. doi:10.1038/ncomms11221
137. Cai K, He X, Song Z, et al. Dimeric drug polymeric nanoparticles with exceptionally high drug loading and quantitative loading efficiency. J Am Chem Soc. 2015;137(10):3458–3461. doi:10.1021/ja513034e
138. Yang Y, Sun B, Zuo S, et al. Trisulfide bond–mediated doxorubicin dimeric prodrug nanoassemblies with high drug loading, high self-assembly stability, and high tumor selectivity. Sci Adv. 2020;6(45):eabc1725. doi:10.1126/sciadv.abc1725
139. Min Y, Caster JM, Eblan MJ, Wang AZ. Clinical translation of nanomedicine. Chem Rev. 2015;115(19):11147–11190. doi:10.1021/acs.chemrev.5b00116
140. Zhang X, Xiong J, Wang K, et al. Erythrocyte membrane-camouflaged carrier-free nanoassembly of FRET photosensitizer pairs with high therapeutic efficiency and high security for programmed cancer synergistic phototherapy. Bioact Mater. 2021;6(8):2291–2302. doi:10.1016/j.bioactmat.2021.01.004
141. Fu S, Li G, Zang W, Zhou X, Shi K, Zhai Y. Pure drug nano-assemblies: a facile carrier-free nanoplatform for efficient cancer therapy. Acta Pharmaceutica Sinica B. 2022;12(1):92–106. doi:10.1016/j.apsb.2021.08.012
142. Zhang X, Sun B, Zuo S, et al. Self-assembly of a pure photosensitizer as a versatile theragnostic nanoplatform for imaging-guided antitumor photothermal therapy. ACS Appl Mater Interfaces. 2018;10(36):30155–30162. doi:10.1021/acsami.8b10421
143. Kim H, Lee J, Oh C, Park JH. Cooperative tumour cell membrane targeted phototherapy. Nat Commun. 2017;8(1):15880. doi:10.1038/ncomms15880
144. Wu Y, Wan S, Yang S, et al. Macrophage cell membrane-based nanoparticles: a new promising biomimetic platform for targeted delivery and treatment. J Nanobiotechnol. 2022;20(1):542. doi:10.1186/s12951-022-01746-6
145. Cao Z, Liu X, Zhang W, et al. Biomimetic macrophage membrane-camouflaged nanoparticles induce ferroptosis by promoting mitochondrial damage in glioblastoma. ACS Nano. 2023;17(23):23746–23760. doi:10.1021/acsnano.3c07555
146. Mohammad‐Rafiei F, Khojini JY, Ghazvinian F, et al. Cell membrane biomimetic nanoparticles in drug delivery. Biotech App Biochem. 2023;70(6):1843–1859. doi:10.1002/bab.2487
147. Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem Soc Rev. 2012;41(7):2971. doi:10.1039/c2cs15344k
148. Marshall SK, Angsantikul P, Pang Z, Nasongkla N, Hussen RSD, Thamphiwatana SD. Biomimetic Targeted Theranostic Nanoparticles for Breast Cancer Treatment. Molecules. 2022;27(19):6473. doi:10.3390/molecules27196473
149. Han H, Bártolo R, Li J, Shahbazi MA, Santos HA. Biomimetic platelet membrane-coated nanoparticles for targeted therapy. Eur J Pharm Biopharm. 2022;172:1–15. doi:10.1016/j.ejpb.2022.01.004
150. Yang Z, Gao D, Guo X, et al. Fighting immune cold and reprogramming immunosuppressive tumor microenvironment with red blood cell membrane-camouflaged nanobullets. ACS Nano. 2020;14(12):17442–17457. doi:10.1021/acsnano.0c07721
151. Jiang Q, Xie M, Chen R, et al. Cancer cell membrane-wrapped nanoparticles for cancer immunotherapy: a review of current developments. Front Immunol. 2022;13:973601. doi:10.3389/fimmu.2022.973601
152. Harris JC, Scully MA, Day ES. Cancer cell membrane-coated nanoparticles for cancer management. Cancers. 2019;11(12):1836. doi:10.3390/cancers11121836
153. Liu X, Sun Y, Xu S, et al. Homotypic cell membrane-cloaked biomimetic nanocarrier for the targeted chemotherapy of hepatocellular carcinoma. Theranostics. 2019;9(20):5828–5838. doi:10.7150/thno.34837
154. Liu CM, Chen GB, Chen HH, et al. Cancer cell membrane-cloaked mesoporous silica nanoparticles with a pH-sensitive gatekeeper for cancer treatment. Colloids Surf B. 2019;175:477–486. doi:10.1016/j.colsurfb.2018.12.038
155. Wang QY, Hu QH, Huang SH, Lin J, Zhou QH, Zhou Q-H. Surface charge switchable nano-micelle for pH/redox-triggered and endosomal escape mediated co-delivery of doxorubicin and paclitaxel in treatment of lung adenocarcinoma. Colloids Surf B. 2022;216:112588. doi:10.1016/j.colsurfb.2022.112588
156. Zhang P, Zheng L, Zhang X, et al. Mesoporous graphene oxide nanocomposite effective for combined chemo/photo therapy against non-small cell lung cancer. IJN. 2024;19:7493–7508. doi:10.2147/IJN.S460767
157. Liu Y, Li X, Niu X, et al. In situ self-assembled biosupramolecular porphyrin nanofibers for enhancing photodynamic therapy in tumors. Nanoscale. 2020;12(20):11119–11129. doi:10.1039/C9NR10646D
158. Zhang T, Tang D, Wu P, et al. NIR-II photo-accelerated polymer nanoparticles boost tumor immunotherapy via PD-L1 silencing and immunogenic cell death. Bioact Mater. 2025;46:285–300. doi:10.1016/j.bioactmat.2024.12.018
159. Zheng ZY, Lin W, Su JW, et al. NIR-715 photodynamic therapy induces immunogenic cancer cell death by enhancing the endoplasmic reticulum stress response. Cell Death Dis. 2024;15(12):890. doi:10.1038/s41419-024-07283-4
160. Choi J, Kim SY. Multi-shell structured upconversion nanocarriers that combine IDO inhibitor-induced immunotherapy with NIR-triggered photodynamic therapy for deep tumors. Biomater Sci. 2023;11(13):4684–4699. doi:10.1039/D3BM00248A
161. Wang Z, Yang L. Natural-product-based, carrier-free, noncovalent nanoparticles for tumor chemo-photodynamic combination therapy. Pharmacol Res. 2024;203:107150. doi:10.1016/j.phrs.2024.107150
162. Zhang J, Li P, Wang T, et al. A copper nanocluster-based multifunctional nanoplatform for augmented chemo/chemodynamic/photodynamic combination therapy of breast cancer. Pharmacol Res. 2023;187:106632. doi:10.1016/j.phrs.2022.106632
163. Liu B, Qiao G, Han Y, et al. Targeted theranostics of lung cancer: PD-L1-guided delivery of gold nanoprisms with chlorin e6 for enhanced imaging and photothermal/photodynamic therapy. Acta Biomater. 2020;117:361–373. doi:10.1016/j.actbio.2020.09.040
164. Cheng D, Ji Y, Wang B, et al. Enzyme/GSH dual-responsive biodegradable nanohybrid for spatiotemporally specific photodynamic and hypoxia-augmented therapy against tumors. Int J Pharm. 2021;603:120730. doi:10.1016/j.ijpharm.2021.120730
165. Zhu J, Xiao T, Zhang J, et al. Surface-charge-switchable nanoclusters for magnetic resonance imaging-guided and glutathione depletion-enhanced photodynamic therapy. ACS Nano. 2020;14(9):11225–11237. doi:10.1021/acsnano.0c03080
166. Xue Y, Bai S, Wang L, et al. A dual-responsive nanoplatform with feedback amplification improves antitumor efficacy of photodynamic therapy. Nanoscale. 2022;14(7):2758–2770. doi:10.1039/D1NR06875J
167. Liu J, Sun B, Li W, et al. Wireless sequential dual light delivery for programmed PDT in vivo. Light Sci Appl. 2024;13(1):113. doi:10.1038/s41377-024-01437-x
168. Bender L, Ayoub AM, Schulze J, et al. Evaluating the photodynamic efficacy of nebulized curcumin-loaded liposomes prepared by thin-film hydration and dual centrifugation: in vitro and in ovo studies. Biomaterials Adv. 2024;159:213823. doi:10.1016/j.bioadv.2024.213823
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles


Global Trends and Research Progress of Photodynamic Therapy in Skin Cancer: A Bibliometric Analysis and Literature Review
Sun J, Zhao H, Fu L, Cui J, Yang Y
Clinical, Cosmetic and Investigational Dermatology 2023, 16:479-498
Published Date: 21 February 2023
Photodynamic Therapy for Oral Squamous Cell Carcinoma: Current Status, Challenges, and Prospects
Zhang W, Chen S, Bai Z, Gan M, Chen M, Zhang Y, Liu S, Liu D
International Journal of Nanomedicine 2024, 19:10699-10710
Published Date: 22 October 2024