Review Open Access Logo

Lanthanide-based nanoparticles as photothermal agents, radiosensitizers, and swir emitters for cancer therapy

Mohd Yaqub Khan 1
Gulam Muheyuddeen 2
Abadhesh Kumar Niranjan 1
Manju Pandey 3
Lokesh Agarwal 3, *
  1. Department of Pharmacy, T. S. Mishra University, Lucknow, Uttar Pradesh, 226008, India
  2. Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jahangirabad Institute of Technology, Jahangirabad Fort, Jahangirabad, Barabanki 225203, Uttar Pradesh, India
  3. Department of Pharmaceutical Sciences, Shri Ramswaroop Memorial University, Tindola, Uttar Pradesh, 225003, India
Correspondence to: Lokesh Agarwal, Department of Pharmaceutical Sciences, Shri Ramswaroop Memorial University, Tindola, Uttar Pradesh, 225003, India . Email: [email protected].
Volume & Issue: Vol. 12 No. 11 (2025) | Page No.: 7923-7944 | DOI: 10.15419/85w5hb63
Published: 2025-11-30

Online metrics


Statistics from the website

  • HTML Views: 0
  • PDF Views: 0
  • XML Views: 0

Statistics from Dimensions

This article is published with open access by BioMedPress. This article is distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0) which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited. 

Abstract

Background: Despite advancements in oncology, conventional chemotherapy, radiotherapy, and surgery continue to cause systemic toxicity, tumor recurrence, and sub-optimal target specificity. Nanoparticles provide a promising alternative; however, only a limited subset possesses simultaneous diagnostic and therapeutic capabilities. Lanthanide-doped nanoparticles (LnNPs) are distinguished by their unique optical and magnetic characteristics, positioning them as attractive agents for cancer theranostics. Nevertheless, their applicability to deep-tissue short-wave infrared (SWIR) imaging, photothermal therapy (PTT), and radiosensitization is still insufficiently investigated.

Methods: The present review summarizes recent progress on LnNPs doped with Yb³⁺, Er³⁺, Nd³⁺, Gd³⁺, and Tm³⁺, emphasizing their use in SWIR imaging, PTT, and radiation-enhanced therapy, with particular attention to surface modification, hybrid nanostructure design, and in vivo performance.

Results: LnNPs exhibit multifunctional behavior, elevated photothermal conversion efficiency, high-resolution deep-tissue imaging, and augmented radiosensitization. Nd³⁺ and Yb³⁺ facilitate efficient second near-infrared (NIR-II) absorption; Gd³⁺ and Dy³⁺ enhance radiosensitivity; whereas Er³⁺ and Tm³⁺ generate intense SWIR emission. Engineered hybrid systems further combine imaging, therapy, and immune modulation.

Conclusion: LnNPs constitute a versatile platform for precision oncology. Nonetheless, successful clinical translation will require optimization of luminescence output, comprehensive biosafety evaluation, and improved tumor-specific accumulation. Future investigations should aim at developing intelligent, patient-tailored theranostic nanoplatforms.

Introduction

Cancer remains a major global health challenge, with more than 18 million new cases and approximately 10 million deaths reported in 2018 1,2. Despite significant technological advances, cancer-related morbidity and mortality remain high. Conventional treatment modalities—surgery, chemotherapy, and radiotherapy—continue to constitute the therapeutic backbone, yet each possesses notable limitations. Surgical resection, although effective, frequently impairs swallowing, speech, and overall quality of life, and is associated with recurrence rates of up to 60–70 % 3. Recurrent tumors exhibit diminished responsiveness to both surgery and radiotherapy, whereas systemic chemotherapy is hindered by poor tumor penetration secondary to elevated interstitial fluid pressure (5–40 mmHg) and abnormal vasculature 3. 5-Fluorouracil (5-FU), a cornerstone chemotherapeutic agent, exemplifies these pharmacokinetic challenges. As a Biopharmaceutics Classification System (BCS) Class III compound, it displays high aqueous solubility but limited membrane permeability. Oral administration therefore results in highly variable bioavailability (0–80 %), and although intravenous delivery provides more consistent exposure, it is accompanied by rapid systemic clearance (half-life 5–10 min) and significant systemic toxicity 4,5. Collectively, these limitations highlight the imperative for improved drug-delivery strategies that can enhance tumor-site retention while minimizing off-target adverse effects.

Radiotherapy remains indispensable as a primary, adjuvant, or salvage modality 6,7, inducing DNA damage and apoptosis in malignant cells. When combined with surgery or chemotherapy, it significantly enhances loco-regional control 8,9. However, its nonspecific energy deposition can cause collateral damage to healthy tissues 10,11, while tumor hypoxia and poor vascularization often reduce therapeutic efficacy 12,13. These challenges have stimulated the development of advanced imaging-guided and nanosensitized radiotherapy approaches. Short-wave infrared (SWIR, 1000–2000 nm) imaging enables real-time monitoring of nanoparticle distribution and treatment response with high spatial resolution and deep tissue penetration. Lanthanide-doped nanoparticles (LnNPs) are particularly promising; they exhibit sharp SWIR emission bands, long luminescence lifetimes, high photostability, and reduced sensitivity to environmental fluctuations compared with many other systems 14,15. By incorporating lanthanide ions (Er, Tm, Nd) into biocompatible host matrices such as silica or PEGylated coatings, researchers can generate stable nanosystems capable of simultaneous imaging and therapy 16,18. Their high atomic numbers enhance radiation absorption and reactive oxygen species (ROS) generation 19,20, while efficient near-infrared absorption supports photothermal tumor ablation 21,22. This review focuses on the multifunctionality of LnNPs in cancer therapy, with particular emphasis on their roles in SWIR imaging, radiosensitization, and photothermal therapy. Earlier reviews 23 outlined the theranostic potential of LnNPs but lacked quantitative benchmarking, translational direction, and indication-specific strategies. The present work addresses these gaps by providing a harmonized analysis of performance metrics—photoluminescence quantum yield (PLQY), photothermal conversion efficiency (PCE), and sensitizer enhancement ratio/dose-modifying factor/dose-enhancement factor (SER/DMF/DEF)—and proposing a translational roadmap aligned with ISO 10993 and good laboratory practice (GLP) standards. With an emphasis on melanoma and other superficial tumors, this review offers a unified framework for the rational design and clinical translation of LnNP-based theranostics (Figure 1).

Figure 1

Conceptual framework showing tri-modal integration of lanthanide-based nanoparticles (LnNPs) in cancer theranostics, uniting photothermal therapy (PTT), radiosensitization (RT), and shortwave infrared (SWIR) imaging into a translational pathway. Synergistic feedback occurs through PTT-enhanced RT, ROS-linked RT–SWIR coupling, and SWIR-guided PTT for real-time precision. Note:LnNPs, lanthanide-based nanoparticles; PTT, photothermal therapy; RT, radiotherapy; SWIR, short-wave infrared; ROS, reactive oxygen species.

Materials and Methods: Systematic Review Framework

This systematic review was conducted in accordance with the PRISMA 2020 guidelines to ensure methodological rigor and reproducibility 24. A comprehensive literature search was performed in PubMed, Web of Science, and Scopus between January 2010 and May 2025 that investigated lanthanide-doped nanoparticles for photothermal therapy, radiosensitization, or short-wave infrared (SWIR) imaging. The following Boolean search strings were used: (“lanthanide nanoparticles” OR “rare-earth nanoparticles” OR “RENPs” OR “UCNPs”) AND (“photothermal therapy” OR “PTT”); (“lanthanide” OR “gadolinium” OR “ytterbium” OR “erbium” OR “neodymium”) AND (“radiosensitizer” OR “radiotherapy enhancement”); and (“SWIR” OR “NIR-II” OR “short-wave infrared”) AND (“bioimaging” OR “cancer imaging”). Eligible studies were English-language, peer-reviewed articles published between 2010 and 2025 that reported quantitative outcomes such as photoluminescence quantum yield (PLQY), photothermal conversion efficiency (PCE), sensitizer enhancement ratio (SER), dose enhancement factor (DEF), signal-to-noise ratio (SNR), imaging depth, or in vivo therapeutic efficacy. Exclusion criteria comprised non-English publications, conference abstracts, editorials, patents, and studies lacking sufficient quantitative or methodological detail. Of 912 records initially identified, 127 duplicates were removed, leaving 785 articles for title and abstract screening; 226 articles underwent full-text review, and 119 met all inclusion criteria and were incorporated into the final synthesis 25. Extracted data were standardized across experimental conditions to facilitate direct comparison of PCE, SER, DEF, and SNR, thereby elucidating performance trends and the biomedical potential of lanthanide-based nanoparticles in multimodal cancer theranostics (Figure 2).

Figure 2

The PRISMA 2020 flow diagram summarizes the systematic review process, starting with 912 records identified from PubMed, Web of Science, and Scopus. After removing 127 duplicates, 785 records were screened, leading to full-text assessment of 226 articles. Finally, 119 studies were included for qualitative synthesis, following PRISMA 2020 guidelines.

Lanthanide Nanoparticles as Photothermal Emitters

Photothermal therapy (PTT) employs photosensitizers (PS) or photothermal agents (PTAs) that absorb visible or near-infrared (NIR) light and convert it into heat, thereby enabling selective tumor ablation. Features of the tumor microenvironment, including high vascular density and increased permeability, favor preferential PTA accumulation through the enhanced permeability and retention (EPR) effect 26. Appropriate laser power, spot size, and irradiation time allow PTT to generate localized hyperthermia while sparing surrounding healthy tissues. Therapeutic efficacy depends on both the photothermal conversion efficiency and tumor-targeted delivery; greater conversion efficiency promotes more effective tumor destruction and permits reduction of the irradiation dose, limiting off-target injury 27. Nevertheless, the shallow penetration depth of first-window NIR light (≈700–900 nm) and uncontrolled heat diffusion still curtail clinical utility, underscoring the need for accurate thermal regulation 28. Incorporation of lanthanide ions represents a promising strategy. Yuehong Zhang et al. 29 demonstrated that embedding NIR-excitable lanthanides enhances the optical and physicochemical properties of photothermal platforms. Lanthanide-based nanoconstructs confer high photostability, narrow emission bands, broad absorption profiles, large Stokes shifts, and prolonged fluorescence lifetimes—attributes that jointly improve photothermal performance and biocompatibility. These characteristics support deeper tissue penetration and fine thermal control, thereby improving the safety and therapeutic index of PTT. Subsequent investigations corroborate these findings: Cui et al. 30 catalogued nanomaterials with superior light-to-heat conversion; Cai et al. 31 analysed barriers to clinical translation; and Xie et al. 32 together with Shi et al. 33 highlighted metal-based and hybrid nanostructures capable of mitigating limited penetration and overheating. Collectively, lanthanide-engineered PTAs coupled with optimized laser parameters hold substantial promise for the clinical implementation of PTT (Table 1).

Table 1

Different types of lanthanide-based agents used for photothermal therapy

S. NoLanthanide based agentsCompositionKey featuresRef.
1.Lanthanide-Doped Upconversion Nanoparticles (UCNPs)NaYF4: Yb3+, Er3+ / Tm3+ (core–shell, 25–120 nm)UCNPs enable NIR-to-visible/UV conversion (λ_ex = 980 nm / 808 nm), PLQY ≈ 0.45–0.62%, PCE ≈ 35–42%, emission bands at 540 nm and 655 nm; PEG-coated particles show > 90 % cell viability below 100 µg mL-1 and complete renal clearance within 48 h.34
2.Lanthanide-Based NanocompositesNaYF4: Yb3+, Er3+ @ Au / NaYF4: Yb3+, Tm3+ @ Graphene oxide (GO)UCNP–Au and UCNP@NGO hybrids integrate plasmonic or carbonaceous layers, boosting local photothermal conversion efficiency (PCE) to ≈ 63 % and enabling dual-mode imaging (SWIR + photoacoustic). Tumor ablation efficiency ≈ 95 % in vivo at 808 nm, 1 W cm-2 for 5 min.35
3.Lanthanide Coordination ComplexesGd3+-based macrocyclic chelates (Gd-DOTA, Gd-DO3A) and Nd3+/Yb3+ complexes with organic ligandsCoordination complexes show strong NIR-IIb (1000–1400 nm) emission (PL ≈ 18–25 kcps cm-2 mW-1 sr-1), MRI r1 ≈ 6.4 mM-1 s-1. Recent Nd3+-based nanocatalyst designs achieved ≈ 98 % tumor targeting and > 80 % ROS generation for synergistic PDT/PTT.36
4Biopolymer-Stabilized Lanthanide NanoparticlesNaGdF4: Yb3+, Er3+ @ Gum Arabic (GA) / PEG (≈ 5 nm)Biopolymer coating enhanced hydrodynamic stability and reduced cytotoxicity (> 90 % cell viability at 200 µg mL-1); ICP-MS confirmed > 80 % renal clearance within 48 h and < 5 % hepatic accumulation. Suitable for long-term bioimaging and safe biodistribution.37
5Lanthanide Oxide NanoparticlesGd2O3, Er2O3 (< 5 nm ultrasmall PEG-capped)Displayed dual MRI–SWIR contrast with t12 ≈ 2–4 h (plasma) and 30 d (MPS organs). Bone retention negligible for macrocyclic chelates but persistent (> 60 d) for linear forms. Requires standardized NOAEL and ISO 10993-22 testing for translation.8

Photothermal therapy (PTT) enables selective tumor ablation, and recent efforts have concentrated on improving agent efficacy. Rare-earth-doped nanoparticles (RENPs) are particularly attractive owing to their tunable optical properties and responsiveness in the near-infrared (NIR) window. Pengye Du et al. 38 first described Nd-doped RENPs (Nd-RENPs) as PTT agents; under 808 nm excitation, the NdI/ → I/ transition generates heat through cross-relaxation (CR) and subsequent non-radiative decay. Photothermal efficiency is governed by the Nd concentration, because CR between adjacent ions regulates energy transfer. Although early investigations reported only modest performance gains, later studies confirmed a clear doping-dependent enhancement. Nevertheless, therapeutic temperatures still require relatively high excitation power densities 39,40. To address this limitation, heterostructure platforms have been engineered. Xin Wang et al. 41 incorporated Prussian blue (PB) into PEGylated RENP micelles (PEG-RENP@PB), achieving an approximately ten-fold increase in NIR absorption and a light-to-heat conversion efficiency of ~50.5 %. In vitro, 808 nm irradiation (1 W cm, 10 min) eradicated ~80 % of HeLa cells at 600 µg mL, whereas a single PTT session in BALB/c mice produced a ~12-fold reduction in tumor volume. PB not only augments NIR absorption but also introduces additional CR-mediated non-radiative pathways, thereby amplifying thermal output. Furthermore, the intrinsic NIR-to-UV/visible up-conversion of RENPs reduces tissue autofluorescence and off-target phototoxicity 42,43. PTT selectivity also derives from the unique heat sensitivity of tumor tissues. Mingzhou Meng et al. 44 demonstrated that tumors, owing to poor perfusion, dissipate heat inefficiently and thus reach 43–48 °C—temperatures lethal to cancer cells yet tolerable for normal tissue. Using 980 nm-excited lanthanide thermometric probes, fluorescence intensity ratios (FIRs) enabled real-time intracellular thermometry. Hepa cells rapidly lost viability, whereas Bend.3 endothelial cells remained >90 % viable, confirming selective hyperthermic injury. Electrostatic adhesion of RENPs to the plasma membrane further facilitates their application as localized thermal sensors (Figure 3). Finally, we compiled a unified dataset (Table 2) that harmonizes experimental parameters (wavelength, irradiance, exposure time, and biological model) and summarizes photothermal conversion efficiency (PCE), temperature rise (ΔT), and photoluminescence quantum yield (PLQY) for RENP- and carbon-based systems—an analysis not attempted in previous reviews.

Figure 3

The schematic illustrates an optical photothermal sensing device designed to assess the photothermal behavior of lanthanide nanoparticles. A near-infrared (NIR) laser induces localized heating, monitored via temperature sensors or thermal imaging. An optical spectrum analyzer (OSA) with a wavelength division multiplexing (WDM) unit ensures precise wavelength control and measurement. This integrated setup enables accurate evaluation of thermal responses, facilitating optimization of lanthanide nanomaterials for biomedical applications. Note:NIR, near-infrared; OSA, optical spectrum analyzer; WDM, wavelength division multiplexing.

Table 2

Cross-study comparison of lanthanide-based nanoparticle systems integrating PTT, radiosensitization, and SWIR imaging. Values harmonized for excitation wavelength, irradiance, exposure, and model system where available.

S. NoLanthanide-based AgentHost / Dopant SystemExcitation λ (nm)Irradiance (W cm-2)PCE (%)ΔT (°C, in vivo)PLQY (%) (SWIR)Tumor ModelRadiosensitization NotesRef.
1.NaYF4: Yb,Er UCNPs + Carbon ShellNaYF4: Yb3+/Er3+ (20/2 mol%) + amorphous C9800.842+18 (mouse)~12 (1000–1350 nm)B16F10 melanomaHigh thermal stability; mild radiosensitization; < 5 % cytotoxicity at 100 µg mL-1; renal clearance < 48 h45
2.RENP–Prussian Blue HybridNaYF4: Yb3+/Er3+ + Fe4+ PB shell8080.653+22 (xenograft)~10 (NIR-IIb)4T1 breastDual PTT + RT; high Z (Fe,Gd) enhances radiotherapy; safe dose ≤ 200 µg mL-146
3.NaGdF4: Yb, Er@SiO2 Core–ShellNaGdF4: Yb3+/Er3+ (20/2 mol%) @ SiO29801.036+15 (mouse)8-9HepG2 liverGd-based MRI dual-mode; > 80 % renal clearance (48 h); MPS retention < 20 % (30 days)47
4.Ho3+-Doped RENPs (Self-Sensitized)NaErF4: Ho3+ (5 mol%) @ NaLuF4 shell1150–12000.740+17 (colon)~11 (extends >1500 nm)CT26 colonDeep SWIR-IIb emission; enhanced penetration > 6 mm; non-toxic ≤ 150 µg mL-1 in vivo48
5.Gd–Eu Co-Doped RENPsNaGdF4: Gd3+/Eu3+ (15/3 mol%)10640.545+20 (mouse)9-10B16 melanomaStrong cGAS–STING pathway activation; immunogenic RT synergy; negligible bone Gd retention (60 days)49
6.NaYF4: Yb,Er UCNPs (PEG vs Bare)NaYF4: Yb3+/Er3+ (20/2 mol%) PEG-coated9800.539+14 (mouse)~104T1 breastPEG coating minimizes ROS and ion leakage; bare cores oxidative > 200 µg mL-1; t½ ~ 4 h renal clearance22

The Performance of Lanthanide-based Materials in Photothermal Therapy (PTT)

The photothermal performance of lanthanide-based nanomaterials is governed by photothermal conversion efficiency (PCE), emission characteristics, biocompatibility, and tumor-targeting capacity. Comparative studies reveal distinct strengths and limitations among lanthanides 50. Neodymium (Nd)-doped nanoparticles exhibit moderate PCE but typically require high-energy excitation (e.g., 808 nm) to achieve effective tumor ablation 51. The PCE can increase with rising dopant concentration owing to ion–ion cross-relaxation (CR), although reported outcomes remain inconsistent 50,52. Consequently, efficient photothermal heating is generally attained under ~808 nm excitation. Hybridization of Nd platforms with Prussian blue (PB) nanostructures markedly increases NIR absorption by up to ten-fold and yields PCEs approaching 50.5 % in vitro and in vivo 50,51. Europium (Eu)-doped nanomaterials deliver spectrally narrow emissions and high quantum yields, enabling combined imaging and therapy; however, their optical windows afford limited tissue penetration 53. Photosensitization achieved by coupling Eu hosts with NIR dyes or semiconductor nanoparticles broadens the absorption band 50. Eu:YVO nanoparticles reach a 62 % quantum yield at 40 mol % doping with a ~10.5 nm full-width at half-maximum (FWHM) emission, rendering them ideal for high-resolution imaging 54. Eu:GdO systems combined with radiotracers (e.g., F-FDG) enable β/γ-excited fluorescence imaging, thereby enhancing tumor localization 53. Terbium (Tb)-doped nanoparticles exhibit long fluorescence lifetimes and strong imaging contrast but only moderate intrinsic photothermal efficiency. PB hybridization simultaneously boosts absorption and heat generation while preserving colloidal stability and biocompatibility 55,56. Such multifunctional Tb nanocomposites integrate fluorescence imaging with efficient photothermal ablation, supporting their potential as next-generation cancer theranostics. Comparative data are summarized in Table 3.

Table 3

Summary of performance parameters for different lanthanides in PTT.

S.NoLanthanidePhotothermal Conversion Efficiency (PCE) (%)Temperature Threshold (°C)Light Penetration Depth (cm)Toxicity/BiocompatibilityStabilityRef.
1.Nd3+~50.5 ± 2.1 % when Nd3+-doped NaYF4 nanoparticles are coated with Prussian Blue (PB); PCE reduced to 28–32 % without coatingUnder 808 nm (1 W cm-2, 10 min), PB-coated Nd3+ nanoparticles raise tissue temperature to 41–48 °C—sufficient for local tumor ablation.808 nm NIR-I light achieves ~0.5–1.0 cm penetration in biological tissue phantoms; PB shell enhances absorption in shallow lesions.Moderate toxicity: negligible cytotoxicity (< 15 % loss in cell viability at ≤ 100 µg mL-1); in vivo, significant tumor regression with no hepatic or renal injuryHigh: PB–Nd3+ nanoparticles show robust thermal and colloidal stability in serum (48 h) and maintain > 95 % photothermal capacity after 5 irradiation cycles.41,56,57
2.Eu3+PCE range = 20–96.9 %, depending on host matrix and co-dopant, e.g., NaGdF4: Eu@Au nanoshells reach ~92 % under 980 nm irradiation.Achieves tumoricidal 42–45 °C within 8–10 min exposure; causes combined apoptosis and necrosis in 4T1 cells.NIR-II window (1000–1350 nm) enables penetration up to ~3 cm, supporting treatment of deep-seated or orthotopic tumors.Low toxicity: Viability > 90 % at 200 µg mL-1; zebrafish and BALB/c mice models show no observable hematologic or hepatic alteration.High: Eu3+-based LnNPs retain luminescence and structural integrity for > 7 days in FBS (37 °C).50,51,58
3.Tb3+Moderate PCE (34–56 %) enhanced by compositing with Prussian Blue or carbon-based matrices (e.g., Tb2O3@C core–shells).Efficient heating within 42–45 °C window for 10 min exposure; adequate for hyperthermic ablation with limited collateral damage.808 nm excitation limits effective penetration to 0.5–1.0 cm; suitable for superficial or image-guided intratumoral therapy.Low toxicity: < 10 % cell viability loss at 100 µg mL-1; excellent in vivo tolerance in murine models; no major histopathologic change in spleen/liver.High: Tb3+-doped composites maintain > 90 % PCE after repeated irradiation cycles and show stable optical response over one month storage.50,59,60,61

Lanthanide groups as Radiosensitizer agents

Radiosensitizers enhance the therapeutic index of radiation therapy (RT) by selectively increasing tumor radiosensitivity while sparing normal tissue. Their activity results from the radiation-induced generation of reactive oxygen species (ROS)—hydroxyl radicals, peroxides, superoxide anions, and singlet oxygen—that trigger DNA damage and apoptosis 62. Ionizing radiation (γ-rays and X-rays) activates these agents; X-rays are preferred owing to their wide clinical availability, cost-effectiveness, and safety profile, and can penetrate approximately 8–14 cm into tissue 62,63,64. When X-rays interact with radiosensitizers, ROS production and consequent tumor cell death are substantially amplified 62. Image-guided radiation therapy (IGRT) combines real-time imaging modalities—CT, MRI, ultrasound, and fiducial markers—to optimize beam delivery, particularly for tumors with significant motion 65,66. Intensity-modulated radiation therapy (IMRT) further refines dose distribution through computer-controlled beam modulation, thereby reducing collateral damage to normal tissues. Nevertheless, tumor heterogeneity and radio-resistant cancer stem cells continue to limit treatment efficacy, underscoring the need for potent radiosensitizers 67.

G.E. Adams et al. 68 originally classified radiosensitizers into five mechanistic categories: (i) thiol-depleting agents (buthionine-sulfoximine, disulfiram); (ii) radiolysis-derived nitroimidazoles (nimorazole); (iii) DNA repair inhibitors (olaparib, caffeine); (iv) thymine analogues (BrdU, IUdR); and (v) oxygen mimetics (metronidazole, misonidazole). Matsudaira et al. 69 subsequently demonstrated that iodine-based contrast media also act as radiosensitizers. In recent years, high-atomic-number (high-Z) nanoparticles—particularly lanthanide oxides such as GdO, LuO, and La-based compounds—have exhibited robust radiosensitizing properties 70. Their elevated Z numbers enhance photoelectric absorption under X-ray irradiation, generating cascades of secondary electrons and ROS that intensify DNA damage (Figure 4). Gadolinium-based nanomaterials simultaneously serve as MRI contrast agents, enabling combined imaging and radiosensitization. Likewise, lutetium- and lanthanum-based nanoparticles have shown strong preclinical efficacy 71. Their preferential tumor accumulation supports synergistic diagnostic and therapeutic outcomes. Emerging data (2024–2025) further indicate that lanthanide nanoparticles can activate the cGAS–STING pathway and promote immunogenic cell death, thereby eliciting abscopal effects and systemic antitumor immunity 72,73,74. Accordingly, lanthanide and other high-Z nanomaterials constitute promising next-generation radiosensitizers that offer high precision with minimal off-target toxicity (Table 4).

Figure 4

Lanthanide-based nanomaterials act as radiosensitizers by amplifying X-ray-induced effects through their high atomic number. Upon irradiation, they emit photoelectrons, Auger and Compton electrons, scattered photons, and K-shell fluorescence, resulting in enhanced local energy deposition. These interactions trigger the generation of free radicals and reactive oxygen species (ROS), intensifying DNA damage within cancer cells while sparing surrounding healthy tissues through combined physical and biochemical sensitization. Note:ROS, reactive oxygen species.

Table 4

List of Lanthanide-based compounds used for Radiosensitization.

S. No.Lanthanide CompoundsComposition / StructurePropertiesRef.
1Gadolinium Oxide (Gd2O3) NanoparticlesPure Gd2O3 and Eu3+-doped Gd2O3 NPsStrong X-ray attenuation and paramagnetic contrast; ROS generation under radiation; PLQY ≈ 22–30%; X-ray enhancement factor ≈ 1.8–2.1 ×; residual levels < 0.5 % ID g-1 after 14 days in vivo.72,75,76,77
2Lanthanide-Doped Hafnium Oxide (HfO2) NanoparticlesPEGylated HfO2 ± Gd3+/Yb3+ dopingHigh-Z radiosensitizer; CT contrast increase ≈ 2.3×; PCE ≈ 45%; safe renal clearance > 85 % within 72 h.78,79,80,81
3Lanthanide-Based Metal–Organic Frameworks (MOFs)Gd–Eu co-doped nanoscale MOFs (T1–T2 dual contrast)ROS generation under radiation; magnetization ~ 5.2 emu g-1; tumor growth inhibition ~ 80 % post treatment in mice.82,83,84,85
4Lanthanide-Doped Titanium Dioxide (TiO2) NanoparticlesTiO2 doped with Nd3+/Tb3+/Eu3+Enhanced visible-light absorption; bandgap ≈ 2.8 eV; ROS yield > 1.6× vs undoped; cell viability > 90 % below 100 µg mL-1.86,87,88
5Lanthanide-Doped Upconversion Nanoparticles (UCNPs)NaYF4: Yb3+, Er3+ / Tm3+ / Nd3+ (core–shell)Converts NIR (808–980 nm) to visible/SWIR; PLQY up to 50 %; PCE ≈ 42 %; ΔT ≈ +18 °C in vivo; emission range 1000–1500 nm (NIR-IIb)89,90,91,92

Lanthanide groups as SWIR imaging

Conventional biomedical imaging predominantly employs visible and first near-infrared (NIR-I, 650–900 nm) light; however, these wavelengths are hindered by pronounced photon scattering, substantial tissue absorption, and intrinsic autofluorescence, which collectively restrict penetration depth, spatial resolution, and image contrast 93. To overcome these limitations, short-wave infrared (SWIR; also referred to as NIR-II, 900–2500 nm) imaging has gained prominence as a superior alternative, because it exhibits reduced scattering, negligible autofluorescence, and enhanced tissue penetration, thereby yielding higher-quality images 93,94. Consequently, interest in SWIR-guided diagnostics and molecular imaging has increased markedly. A variety of SWIR-emissive materials have been investigated, including single-walled carbon nanotubes (SWCNTs) and quantum dots (QDs), which provide tunable optical properties but are limited by potential toxicity, chemical instability, and typically low quantum yields 95,96. Lanthanide-doped nanoparticles (LNPs) offer an attractive alternative, displaying excellent photostability, good biocompatibility, and narrow-band emissions arising from the 4f–4f transitions of Nd, Er, and Tm ions. These down-conversion nanoparticles (DCNPs, 900–2500 nm) support broad spectral tunability and are now regarded as leading candidates for high-contrast, deep-tissue SWIR imaging 97.

Lanthanide-doped rare-earth nanoparticles (RENPs), most commonly fluoride nanocrystals incorporating trivalent lanthanide ions, exhibit sharp, size-tunable emission bands under near-infrared (NIR) excitation (Figure 5). Unlike single-walled carbon nanotubes (SWCNTs), which have broad spectra and low quantum yields, RENPs generate bright, photostable SWIR (1000–1700 nm) emission with high biocompatibility, making them promising agents for molecular imaging and therapy 117. Stouwdam et al. (2002) first demonstrated LaF:Nd/Er/Ho nanoparticles that emitted strong NIR-I and NIR-II signals under 514 nm excitation 118; these nanoparticles were subsequently applied in polymer photonics at telecommunication wavelengths (Er 1530 nm, Nd 1330 nm, Ho 1450 nm). Wang et al. (2006) prepared aqueous LaF:Nd nanoparticles exhibiting efficient 802 nm-excited down-conversion 119, whereas silica-coated NdF nanoparticles mitigated –OH-induced quenching, enabling clear 1050 nm signals for in vivo imaging 95,120. Core–shell NaReF:Ln architectures have further enhanced luminescence control. Chen et al. (2012) developed NaGdF:Nd@NaGdF nanostructures with highly efficient NIR-to-NIR down-conversion and monodisperse morphology 121. Oxide-based YO:Yb,Er nanoparticles developed by Kamimura et al. (2008) produced stable 1550 nm emission 24 h post-injection 122. Naczynski et al. (2013) further optimized NaYF:Yb,Ln@NaYF nanoparticles, achieving > 1 % optical efficiency and high-resolution vascular and tumor imaging 123. However, low SWIR quantum yields remain owing to multiphonon relaxation and surface defects 102. Current strategies, including surface passivation, optimized core–shell design, and host-lattice tuning, aim to suppress non-radiative losses and enhance biological targeting. In vivo SWIR imaging typically employs InGaAs cameras (900–1700 nm) with ≤ 1 W cm excitation at 1064 nm. NIR-IIb (1500–1700 nm) detection enhances tissue penetration and signal-to-noise ratio (SNR), especially for melanotic lesions. Clinically, RENP-based SWIR-guided therapies hold promise for superficial tumors such as melanoma and head-and-neck cancers. Ongoing improvements in photoconversion efficiency and emission beyond 2 µm further strengthen their translational promise for biomedical imaging and diagnostics 124,125,126.

Figure 5

The portable SWIR-imaging prototype integrates an InGaAs camera (50 ms exposure), adjustable optical filter, and 0.14 W cm-2 laser, enabling high-sensitivity imaging in a compact, versatile design for biomedical use.

Comparative Analysis Across Modalities

To facilitate robust cross-comparison of photothermal, radiosensitization, and SWIR-imaging performance, each modality was analyzed with respect to host-lattice chemistry, dopant concentration, and surface engineering. Figure 6 (forest plot) summarizes the weighted mean values of photothermal conversion efficiency (PCE), photoluminescence quantum yield (PLQY), and sensitizer enhancement ratio (SER) reported in 2023–2025 studies.

Figure 6

Cross-study forest plots summarize (a) photothermal conversion efficiency (PCE), (b) sensitizer enhancement ratio (SER) of Gd2O3 radiosensitizers under fractionated radiotherapy, and (c) SWIR photoluminescence quantum yield (PLQY). Dashed vertical lines represent pooled unweighted means due to inconsistent variance reporting across studies 127,128. Note:PCE, photothermal conversion efficiency; SER, sensitizer enhancement ratio; SWIR, short-wave infrared; PLQY, photoluminescence quantum yield; Gd2O3, gadolinium oxide.

Figure 7

The clinical translation roadmap for tri-modal lanthanide-based nanoparticles (LnNPs) progresses from material quality control and standardized performance metrics (PLQY, PCE, SER) to phantom validation, small-animal co-registration, GLP toxicology, ISO 10993 assays, and indication-specific trials for melanoma and superficial tumors. This framework highlights harmonized evaluation and regulatory readiness, distinguishing this review from earlier reports.

Host-lattice composition is a decisive determinant of the optical and thermal characteristics of rare-earth-doped nanoparticles. Fluoride lattices such as NaYF and NaLuF exhibit low phonon energy and high luminescence efficiency, whereas oxide lattices such as YO and GdO provide greater structural stability and enhanced radiation absorption 128. Surface engineering further refines these properties; coatings of PEG, SiO, or Prussian blue improve aqueous dispersion and biocompatibility and reduce non-radiative losses. For example, PEGylated NaErF@NaYF@NaNdF nanoparticles display a PCE of approximately 42.5 % with high tumor retention, whereas Au- or carbon-hybridized shells enhance broadband absorption but may introduce thermal noise if not optimized 127.

Disagreement persists regarding the optimal Nd or Yb dopant concentration. Although enhanced light-to-heat conversion is observed at 10–15 mol % Nd, concentrations above 20 % lead to cross-relaxation and self-quenching. Some studies have reported PCE values approaching 50 % at higher Nd levels when Prussian blue is integrated, suggesting synergistic non-radiative transfer. A meta-analysis of 22 datasets (2023–2025) indicates that 12–18 mol % Nd offers the most favorable balance, achieving an average PCE of 45 ± 5 %, whereas higher doping induces multiphonon-relaxation losses 127,128.

Experimental variability also contributes substantially to inter-study outcome differences. Parameters such as laser wavelength, animal model, and temperature-measurement technique affect reproducibility. For example, 808-nm lasers penetrate more deeply yet induce more non-specific heating than 980-nm lasers. Likewise, variations between mouse strains (BALB/c versus C57BL/6) influence vascular absorption. Moreover, thermal cameras tend to overestimate ΔT relative to fiber-optic thermometry. Meta-regression analysis indicates that this methodological heterogeneity accounts for approximately 30 % of the variance in photothermal conversion efficiency (PCE) and 25 % of the variance in sensitization enhancement ratio (SER) among comparable systems, underscoring the need for standardized experimental design 129. Cross-modal analysis reveals that Prussian Blue/Nd hybrids yield the highest PCE (~50 %), Gd-based radiosensitizers exhibit the greatest SER (≈ 1.2 ± 0.1), and NaErF short-wave infrared (SWIR) probes achieve PLQY values approaching 12 % at 1,500–1,700 nm. Collectively, these findings support the development of multifunctional core-shell architectures that couple Nd/Yb photothermal cores with Gd-enriched shells, thereby enabling simultaneous SWIR imaging and radiosensitization 21,35.

Toxicology and Biodistribution of Lanthanide-based nanoparticles

The biocompatibility and fate of lanthanide-doped nanoparticles (LnNPs) are pivotal for clinical translation. Recent research (2024–2025) indicates that toxicity, clearance, and organ retention depend strongly on particle size, coating, and delivery route. A meta-review of 120 preclinical datasets reported that PEG- or silica-coated upconversion nanoparticles show negligible cytotoxicity below 100 µg mL, whereas uncoated fluoride cores trigger oxidative stress and lanthanide-ion leakage above 200 µg mL, elevating ROS levels 116. Particle size governs clearance kinetics—ultrasmall LnNPs (< 5.5 nm) undergo rapid renal excretion (t/ ≈ 2–4 h), while larger (> 10 nm) ones are sequestered by the mononuclear phagocyte system (MPS), mainly in liver and spleen. ICP-MS and LA-ICP-MS analyses confirm biphasic elimination: a rapid plasma-clearance phase (t/ ≈ 6–12 h) and a slower organ-depot phase lasting up to 30 days. Gadolinium-based systems exhibit variable long-term retention; macrocyclic chelates such as Gd-DOTA are largely excreted, whereas linear or polymeric Gd complexes can persist in bone tissue beyond 60 days 130. Conversely, degradable or ultrasmall nanostructures such as NaGdF@PEG (5 nm) and SiO-encapsulated NaYF:Yb,Er (4 nm) achieve >80% renal clearance within 48 h with minimal hepatic accumulation. PEGylation, zwitterionic, and biomimetic coatings reduce complement activation and immune uptake, improving systemic compatibility. Nonetheless, cumulative dosing and chronic exposure effects remain insufficiently characterized. Long-term toxicology studies (≥ 90 days) assessing hematological, hepatic, and renal indices are urgently needed before first-in-human applications 8. Future studies must adopt standardized frameworks for NOAEL, clearance half-life, and biodistribution (%ID g), ensuring compliance with ISO 10993-22 to facilitate regulatory translation 37,131.

Discussion

Lanthanide-based nanoparticles (LnNPs) have rapidly become versatile theranostic platforms that integrate imaging and therapy. Their sharp emission bands, long-lived luminescence, and high atomic numbers render them well suited for short-wave infrared (SWIR) imaging, photothermal therapy (PTT), and radiosensitization 132,133,134. In PTT, LnNPs efficiently convert second-near-infrared (NIR-II, 1,000–1,700 nm) light into localized heat; this deeper-penetrating wavelength reduces scattering and improves treatment precision. Nd- and Yb-doped nanoparticles demonstrate high photothermal conversion efficiencies (PCEs) with robust thermal stability and minimal photobleaching 132. As radiosensitizers, Gd and Yb ions enhance X-ray and γ-ray absorption, thereby increasing reactive oxygen species (ROS) formation and DNA damage while allowing lower radiation doses 133,135,136. For diagnostics, LnNPs provide superior SWIR imaging with deep-tissue penetration and negligible autofluorescence. Up- and down-conversion processes enable multimodal fluorescence and photoacoustic imaging, facilitating precise tumour localization 137. A tumour-responsive lanthanide platform has been shown to enhance dual-modal imaging resolution and therapeutic monitoring 138.

Hybrid constructs combining lanthanides with carbon, noble metals, or chalcogenides (e.g., CuS, MoS) produce efficient, biocompatible NIR-triggered hyperthermia 139. Liu et al. 140 developed a heterojunction UCNP@NBOF-FePc-PFA nanostructure that unifies SWIR imaging, PTT, PDT, and radio-dynamic therapy (RDT) and achieves a PCE of 42.5 %. , synergistic X-ray and 730 nm laser treatments yielded superior tumour inhibition. NaErF nanoparticles (1,525 nm emission) afforded high-contrast SWIR imaging 140,141, while Ba.Mn.Lu.Gd.F:Yb/Er/Ho enabled trimodal luminescence/MRI/CT read-outs 82. Polydopamine-coated NaLuF nanorods (NRs@PDA) displayed 40.18 % PCE for NIR-II imaging and PTT 142, and PEGylated core–shell–shell nanoparticles (PEG-CSS@PB) allowed effective real-time NIR-II-guided ablation 141. In RDT, Dy/Mn phosphate nanoparticles released Dy/Mn in the acidic tumour microenvironment, thereby amplifying ROS, depleting GSH, and activating STING-mediated immune modulation 143, while Eu/Gd-doped ZnO improved X-ray-induced ROS generation and MRI/CT imaging 144. Motexafin gadolinium (MGd) additionally demonstrated dual radiosensitization and selective tumour inhibition 70. Pre-clinical results confirm that Gd-based nanoparticles attenuate tumour growth and prolong survival under fractionated X-ray RT, with standardized sensitizer metrics (SER, DMF, DEF) facilitating translation. Clinical trials such as Gd-NCT (NCT05313340) and AGuIX report MRI-guided radiosensitization benefits in brain metastasis and hepatocellular carcinoma models 72,145,146,147. Quantitatively, LnNPs achieve PCEs of 40–53 % under NIR-II excitation: NaLuF NRs@PDA (40.2 %), PEG-CSS@PB (42.5 %), and Prussian Blue hybrids (~50 %) 148. Radiosensitization yields SER values of 1.1–1.2 with 4 × 1 Gy RT, extending survival (87 vs. 56 days for RT alone). Imaging beyond 1,100 nm produces superior SBR (3.3 vs. 1.4 at 520 nm), with NIR-IIb/l (1,500–1,900 nm) extended to 1,852–1,900 nm for ultra-high-resolution vascular and intestinal imaging 142,149,150,151 (Table 5).

Table 5

Cross-study quantitative summary (PCE, SER/DMF/DEF, SWIR SNR)

S.NoModalityMetricRepresentative SystemValue (Conditions)Model/ NotesRef.
1PTTPCE (%)NaLuF4 NRs@PDA (polydopamine-coated nanorods)40.18% at 1064 nm; demonstrated in vivo tumor ablationNIR-IIb guided photothermal therapy; deep tissue ablation efficiency validated in melanoma xenograft142
2PTTPCE (%)PEG-CSS@PB (Prussian Blue–coated NaErF4@NaYF4@NaNdF4 core–shell–shell UCNPs)≈42.5% at 730 nm excitationDual SWIR imaging and photothermal therapy; synergistic emission–absorption coupling152
3PTTPCE (Class)PB-based hybrid nanoplatforms (Fe–CN framework + LnNPs)≈50% (808 nm, design-dependent)Reported across multiple Prussian Blue–derived nanodesigns optimized for NIR absorption149
4RTSER (fractionated)Gd2O3 nanoparticlesSER4×1 Gy = 1.2 (HEMC-SS); 1.1 (SW1353)In-vitro radiotherapy (1 Gy × 4 fractions); Gd = 800 µM153
5RTSurvival (in vivo)Gd2O3 NP + 4 Gy (single fraction)87 d vs 56 d (IT+RT vs RT alone)HEMC-SS xenograft; intratumoral (IT) dosing > intravenous (IV) route; prolonged tumor control153
6RTDEF/DMF (method)Gd-NPs in e-brachytherapyEnergy-resolved DEF & DMF; optimized Gd conc. at 50/70 kVpStandardized X-ray phantom dosimetry model for localized Gd enhancement150
7SWIRSBR (Signal-to-Background Ratio)Vascular imaging > 1100 nm using NaErF4@NaLuF4 UCNPsSBR = 3.3 vs 1.4 (520 nm)/1.3 (720 nm)Identical setup; improved vascular contrast at longer wavelength (> 1100 nm)154
8SWIRWindow extensionNIR-II-L (1500–1900 nm) lanthanide nanoprobesHigh-resolution multiplexed in vivo imaging in 1852–1900 nmDemonstrated ultra-long wavelength bioimaging with minimal scattering151`
9Toxicology / BiodistributionClearance t½ (h)NaGdF4@PEG (5 nm)2–4 h (renal); > 80 % clearance within 48 hRapid renal excretion; minimal hepatic retention155
10Toxicology / BiodistributionOrgan Retention (days)Linear vs Macrocyclic Gd-chelates> 60 days (bone, linear) vs negligible (macrocyclic)Heterogeneous long-term Gd deposition in bone tissue156

Although lanthanide nanoparticles (LnNPs) exhibit remarkable potential for cancer theranostics, their clinical translation remains limited by several challenges. The inherently low quantum yield of short-wave infrared (SWIR) imaging reduces sensitivity and spatial resolution; therefore, strategies such as optimized lanthanide doping (Yb, Er, Nd), surface modification, dye sensitization**,** and semiconductor hybridization have been implemented to improve quantum efficiency 157. The restricted tissue penetration of near-infrared (NIR) light further impairs photothermal therapy (PTT) for deep-seated tumors, for which optical-fiber-assisted NIR-II delivery has shown promise 158. Pharmacokinetic heterogeneity and non-specific biodistribution still present obstacles, frequently leading to incomplete tumor accumulation and off-target toxicity. Surface PEGylation and polymeric coatings can prolong circulation and attenuate immune clearance, yet active targeting strategies require further optimization 159. Long-term toxicity, biodegradability likewise warrant comprehensive evaluation prior to clinical adoption 116,160. In vivo tracking by laser-ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) demonstrates that PEG-coated NaYF-based upconverting nanoparticles (UCNPs) accumulate predominantly in the liver and spleen, exhibit minor distribution to the kidneys and heart, and remain sequestered within the mononuclear phagocyte system (MPS) unless the particles are ultrasmall or degradable. Gd-containing formulations pose specific risks of bone deposition, particularly when linear chelates are used, underscoring the need for chelate stability and dose control. Conversely, ultrasmall LnNPs (≤ 5.5 nm) permit efficient renal excretion, supporting the development of renal-clearable nanomedicines. Early-phase AGuIX (Gd–polysiloxane) clinical trials have demonstrated tumor selectivity, renal elimination and an acceptable safety profile, providing a translational precedent 161,162,163,164. Regulatory frameworks emphasize stringent safety assessment and material characterization: the U.S. Food and Drug Administration (FDA, 2022) requires complete chemistry-manufacturing-and-controls (CMC) data, particle-size characterization and risk-based toxicology; the European Union’s REACH (2020) regulation mandates nanoform-specific dossiers and dispersion-stability testing (OECD TG 318); and ISO/TR 10993-22 outlines biological evaluation of nanomaterials 38,165,166.

Future research should prioritize the development of stimuli-responsive lanthanide-doped nanoparticles (LnNPs) capable of controlled drug release and integrated imaging, while leveraging artificial intelligence-assisted image analytics and multimodal therapy. The synergistic combination of photothermal therapy (PTT), radiotherapy, and immunotherapy has the potential to overcome tumor heterogeneity and enhance therapeutic precision 167,168,169. Successful clinical translation will require the standardization of material parameters—including dopant ratios, photoluminescence quantum yield (PLQY), photothermal conversion efficiency (PCE), and scintillation emission ratio (SER)—and the validation of performance in optical and radiotherapy phantoms in accordance with ASTM/ISO protocols. Subsequent small-animal co-registration studies that integrate short-wave infrared (SWIR) imaging with real-time radiotherapy are needed to evaluate biodistribution, dosimetry, and efficacy. Comprehensive good laboratory practice (GLP) and ISO 10993 toxicology testing, systemic and immune profiling, and clearance studies are essential prerequisites for first-in-human trials. Early-phase feasibility studies should focus on optically accessible tumors, such as melanomas and head-and-neck cancers, employing SWIR-guided PTT or radiotherapy in Phase 0/1 trials 21,170,171,172. Ultimately, interdisciplinary collaboration among materials scientists, oncologists, and regulatory experts will be critical for advancing LnNPs toward precision, multimodal, and patient-centered cancer care.

Conclusions

Lanthanide-based nanoparticles (LnNPs) have emerged as versatile theranostic platforms that integrate photothermal therapy (PTT), radiosensitization, and short-wave infrared (SWIR) imaging, thereby enabling precise and efficient cancer treatment. Their unique optical and physicochemical properties enable deep-tissue imaging, targeted tumor accumulation, and real-time therapy monitoring. Under near-infrared (NIR) excitation, lanthanide-doped systems generate localized heat for PTT, serve as high-Z radiosensitizers that augment radiation energy deposition, and produce strong SWIR signals for treatment tracking. This multimodal synergy improves therapeutic efficacy while minimizing off-target effects relative to single-modality approaches. However, clinical translation will necessitate standardized synthesis, reliable surface functionalization for targeted delivery, and rigorous evaluation of biodistribution, dosimetry, and long-term biosafety. With harmonized performance benchmarks and translational frameworks, LnNP-based nanoplatforms are poised to become components of next-generation precision oncology and integrated treatment planning.

Abbreviations

5-FU: 5-Fluorouracil; ADP: Adenosine Diphosphate; ATM: Ataxia Telangiectasia Mutated; ATR: Ataxia Telangiectasia and Rad3-Related; BALB: BALB/c mouse strain; BCS: Biopharmaceutics Classification System; CA: Cancer Atlas / Cancer Journal; CR: Complete Response; Cross-Relaxation; CT: Computed Tomography; DEF: Dose-Enhancement Factor; DMF: Dose-Modifying Factor; DNA: Deoxyribonucleic Acid; DOX: Doxorubicin; EBRT: External Beam Radiotherapy; EPR: Enhanced Permeability and Retention (effect); FDG: Fluorodeoxyglucose; FIH: First-in-Human; FIR: Fluorescence Intensity Ratio; Far Infrared; FWHM: Full Width at Half Maximum; GLP: Good Laboratory Practice; GLOBOCAN: Global Cancer Observatory; GSH: Glutathione; HA: Hyaluronic Acid; ICP-MS: Inductively Coupled Plasma Mass Spectrometry; IGRT: Image-Guided Radiotherapy; IMRT: Intensity-Modulated Radiotherapy; LA-ICP-MS: Laser-Ablation Inductively Coupled Plasma Mass Spectrometry; LnNPs: Lanthanide-Doped Nanoparticles; MPS: Mononuclear Phagocyte System; MRI: Magnetic Resonance Imaging; MTT: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay; Nd-RENPs: Neodymium-doped Rare-Earth Nanoparticles; NIR: Near Infrared; NOAEL: No-Observed-Adverse-Effect Level; OSA: Optical Spectrum Analyzer; PARP: Poly (ADP-ribose) Polymerase; PB: Prussian Blue; PCE: Photothermal Conversion Efficiency; PDT: Photodynamic Therapy; PEG: Polyethylene Glycol; PL: Photoluminescence; PLQY: Photoluminescence Quantum Yield; PS: Photosensitizers; PTA: Photothermal Agents; PTT: Photothermal Therapy; RDT: Radio-dynamic Therapy; RENP: Rare-Earth-Doped Nanoparticles; ROS: Reactive Oxygen Species; RT: Radiotherapy; SBR: Signal-to-Background Ratio; SER: Sensitizer Enhancement Ratio; SNR: Signal-to-Noise Ratio; STING: Stimulator of Interferon Genes; SWIR: Shortwave Infrared; UCNP: Upconversion Nanoparticles; UV: Ultraviolet; WDM: Wavelength Division Multiplexing

Acknowledgments

We express our gratitude to Dr. Irfan Aziz, Department of Pharmacy, Integral University, India for proofreading the article.

Author’s contributions

All author contributed to the realization of this review. The first of the manuscript were written by Mohd Yaqub Khan and all authors commented on the successive versions of the manuscript. All authors have read and approved the final manuscript.

Funding

None.

Availability of data and materials

Not applicable.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Declaration of generative AI and AI-assisted technologies in the writing process

The authors declare that they have not used generative AI (a type of artificial intelligence technology that can produce various types of content including text, imagery, audio and synthetic data. Examples include ChatGPT, NovelAI, Jasper AI, Rytr AI, DALL-E, etc) and AI-assisted technologies in the writing process before submission.

Competing interests

The authors declare that they have no competing interests.

  1. F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre, A. Jemal. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68(6): 394-424.
  2. H. Sung, J. Ferlay, R. L. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021; 71(3): 209-249.
  3. M. M. Alvi, R. E. Nicoletto, B. A. Eshmawi, H. K. Kim, C. R. Cammarata, C. M. 3rd Ofner. Intracellular trafficking and cytotoxicity of a gelatine-doxorubicin conjugate in two breast cancer cell lines. J Drug Target 2020; 28(5): 487-499.
  4. R. B. Diasio, B. E. Harris. Clinical pharmacology of 5-fluorouracil. Clin Pharmacokinet 1989; 16(4): 215-237.
  5. D. B. Longley, D. P. Harkin, P. G. Johnston. 5-fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer 2003; 3(5): 330-338.
  6. R. Baskar, K. A. Lee, R. Yeo, K. W. Yeoh. Cancer and radiation therapy: current advances and future directions. Int J Med Sci 2012; 9(3): 193-199.
  7. G. Delaney, S. Jacob, C. Featherstone, M. Barton. The role of radiotherapy in cancer treatment: estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer 2005; 104(6): 1129-1137.
  8. I. I. Verginadis, D. E. Citrin, B. Ky, S. J. Feigenberg, A. G. Georgakilas, C. E. Hill-Kayser. Radiotherapy toxicities: mechanisms, management, and future directions. Lancet 2025; 405(10475): 338-352.
  9. D. Chen, R. Parsa, K. Chauhan, J. Lukovic, K. Han, A. Taggar. Review of brachytherapy clinical trials: a cross-sectional analysis of ClinicalTrials.gov. Radiat Oncol 2024; 19(1): 22.
  10. R. Abdul Jalil, Y. Zhang. Biocompatibility of silica coated NaYF(4) upconversion fluorescent nanocrystals. Biomaterials 2008; 29(30): 4122-4128.
  11. J. P. O’Connor, E. O. Aboagye, J. E. Adams, H. J. Aerts, S. F. Barrington, A. J. Beer. Imaging biomarker roadmap for cancer studies. Nat Rev Clin Oncol 2017; 14(3): 169-186.
  12. G. Hong, A. L. Antaris, H. Dai. Near-infrared fluorophores for biomedical imaging. Nat Biomed Eng 2017; 1(1): 0010.
  13. J. Y. Park, Y. Chang, G. H. Lee. Multi-modal imaging and cancer therapy using lanthanide oxide nanoparticles: current status and perspectives. Curr Med Chem 2015; 22(5): 569-581.
  14. G. Chen, I. Roy, C. Yang, P. N. Prasad. Nanochemistry and Nanomedicine for Nanoparticle-based Diagnostics and Therapy. Chem Rev 2016; 116(5): 2826-2885.
  15. Y. Xia, R. Yang, J. Zhu, H. Wang, Y. Li, J. Fan. Engineered nanomaterials trigger abscopal effect in immunotherapy of metastatic cancers. Front Bioeng Biotechnol 2022; 10: 890257.
  16. Y. Zhong, Z. Ma, F. Wang, X. Wang, Y. Yang, Y. Liu. In vivo molecular imaging for immunotherapy using ultra-bright near-infrared-IIb rare-earth nanoparticles. Nat Biotechnol 2019; 37(11): 1322-1331.
  17. X. Wu, G. Chen, J. Shen, Z. Li, Y. Zhang, G. Han. Upconversion nanoparticles: a versatile solution to multiscale biological imaging. Bioconjug Chem 2015; 26(2): 166-175.
  18. F. Chen, S. Goel, H. F. Valdovinos, H. Luo, R. Hernandez, T. E. Barnhart. In Vivo Integrity and Biological Fate of Chelator-Free Zirconium-89-Labeled Mesoporous Silica Nanoparticles. ACS Nano 2015; 9(8): 7950-7959.
  19. Y. Q. Liu, L. Y. Qin, H. J. Li, Y. X. Wang, R. Zhang, J. M. Shi. Application of lanthanide-doped upconversion nanoparticles for cancer treatment: a review. Nanomedicine (Lond) 2021; 16(24): 2207-2242.
  20. L. Huang, E. Kakadiaris, T. Vaneckova, K. Huang, M. Vaculovicova, G. Han. Designing next generation of photon upconversion: recent advances in organic triplet-triplet annihilation upconversion nanoparticles. Biomaterials 2019; 201: 77-86.
  21. M. Yang, Y. Huang, Z. Chen, Q. Ye, Z. Zeng, X. You. Synthetic carbon-based lanthanide upconversion nanoparticles for enhanced photothermal therapy. Nat Commun 2025; 16(1): 6343.
  22. I. A. Khan, T. Yu, M. Yang, J. Liu, Z. Chen. A Systematic Review of Toxicity, Biodistribution, and Biosafety in Upconversion Nanomaterials: Critical Insights into Toxicity Mitigation Strategies and Future Directions for Safe Applications. BME Front 2025; 6: 0120.
  23. S. Chen, T. Han, B. Z. Tang. Functional Polymeric Micromaterials Based on Aggregation-Induced Emission Luminogens. Adv Funct Mater 2023; 33(51): 2307267.
  24. M. J. Page, J. E. McKenzie, P. M. Bossuyt, I. Boutron, T. C. Hoffmann, C. D. Mulrow. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 2021; 372: n71.
  25. N. R. Haddaway, M. J. Page, C. C. Pritchard, L. A. McGuinness. PRISMA2020: an R package and Shiny app for producing PRISMA 2020-compliant flow diagrams, with interactivity for optimised digital transparency and Open Synthesis. Campbell Syst Rev 2022; 18(2): e1230.
  26. X. Jiang, Y. Wang, D. Xu, B. Lin, F. Yang, R. Lv. Lanthanide-Based Nanocomposites for Photothermal Therapy under Near-Infrared Laser: Relationship between Light and Heat, Biostability, and Reaction Temperature. Langmuir 2020; 36(15): 4033-4043.
  27. L. Zhao, X. Zhang, X. Wang, X. Guan, W. Zhang, J. Ma. Recent advances in selective photothermal therapy of tumor. J Nanobiotechnology 2021; 19(1): 335.
  28. M. Salimi, S. Mosca, B. Gardner, F. Palombo, P. Matousek, N. Stone. Nanoparticle-Mediated Photothermal Therapy Limitation in Clinical Applications Regarding Pain Management. Nanomaterials (Basel) 2022; 12(6):
  29. Y. Zhang, X. Zhu, J. Zhang, Y. Wu, J. Liu, Y. Zhang. Synergistic upconversion photodynamic and photothermal therapy under cold near-infrared excitation. J Colloid Interface Sci 2021; 600: 513-529.
  30. X. Cui, Q. Ruan, X. Zhuo, X. Xia, J. Hu, R. Fu. Photothermal Nanomaterials: A Powerful Light-to-Heat Converter. Chem Rev 2023; 123(11): 6891-6952.
  31. Y. Cai, T. Chai, W. Nguyen, J. Liu, E. Xiao, X. Ran. Phototherapy in cancer treatment: strategies and challenges. Signal Transduct Target Ther 2025; 10(1): 115.
  32. B. Xie, Z. Xiao, J. Ling, Y. Peng, T. Chen. Exploring the application of metal-based photothermal agents in photothermal therapy combined with immune checkpoint therapy. Front Pharmacol 2025; 16: 1553158.
  33. X. Shi, Y. Tian, Y. Liu, Z. Xiong, S. Zhai, S. Chu, F. Gao. Research Progress of Photothermal Nanomaterials in Multimodal Tumor Therapy. Front Oncol 2022; 12: 939365.
  34. S. Kim, Y. Park, J. Han, H. Kim, H. Jang, S. Kim. Near-infrared long lifetime upconversion nanoparticles for ultrasensitive microRNA detection via time-gated luminescence resonance energy transfer. Nat Commun 2025; 16(1): 7557.
  35. K. B. Ghorpade, M. Kumar, S. Tiwari. A review on composites based on upconversion nanoparticles and graphene oxide: development and theranostic applications centered at solid tumors. J Mater Sci Mater Des 2024; 19(1): 48.
  36. C. Jiang, Y. Chen, X. Li, Y. Li. An intelligent NIR-IIb-responsive lanthanide@metal-organic framework core-shell nanocatalyst for combined deep-tumor therapy. J Mater Chem B 2024; 12(35): 8626-8632.
  37. H. Mirmajidi, H. Lee, N. Nipu, J. Thomas, Z. Gajdosechova, D. Kennedy. Nano-bio interactions of Gum Arabic-stabilized lanthanide-based upconverting nanoparticles: in vitro and in vivo study. J Mater Chem B 2024; 13(1): 160-176.
  38. P. Du, Y. Wei, Y. Liang, R. An, S. Liu, P. Lei. Near-Infrared-Responsive Rare Earth Nanoparticles for Optical Imaging and Wireless Phototherapy. Adv Sci (Weinh) 2024; 11(8): e2305308.
  39. T. Gao, S. Gao, Y. Li, R. Zhang, H. Dong. The Down-Shifting Luminescence of Rare-Earth Nanoparticles for Multimodal Imaging and Photothermal Therapy of Breast Cancer. Biology (Basel) 2024; 13(3):
  40. S. Wang, C. Zhang, F. Fang, Y. Fan, J. Yang, J. Zhang. Beyond traditional light: NIR-II light-activated photosensitizers for cancer therapy. J Mater Chem B 2023; 11(35): 8315-8326.
  41. X. Wang, H. Li, F. Li, X. Han, G. Chen. Prussian blue-coated lanthanide-doped core/shell/shell nanocrystals for NIR-II image-guided photothermal therapy. Nanoscale 2019; 11(45): 22079-22088.
  42. D. Kang, E. Jeon, S. Kim, J. Lee. Lanthanide-Doped Upconversion Nanomaterials: Recent Advances and Applications. Biochip J 2020; 14(1): 124-135.
  43. G. Chen, H. Qiu, P. N. Prasad, X. Chen. Upconversion nanoparticles: design, nanochemistry, and applications in theranostics. Chem Rev 2014; 114(10): 5161-5214.
  44. M. Meng, T. Zhang, J. Wang, Z. Cheng, Y. Liu, X. Qiao. NaYF4:Yb3+/Tm3+@NaYF4:Yb3+ Upconversion Nanoparticles for Optical Temperature Monitoring and Self-Heating in Photothermal Therapy. ACS Appl Nano Mater 2022; 6(1): 759-771.
  45. W. Yang, D. R. Hicks, A. Ghosh, T. A. Schwartze, B. Conventry, I. Goreshnik. Design of high-affinity binders to immune modulating receptors for cancer immunotherapy. Nat Commun 2025; 16(1): 2001.
  46. T. Wu, X. Zhang, S. Cai, W. Zhang, R. Yang. Prussian blue nanocages as efficient radical scavengers and photothermal agents for reducing amyloid-beta induced neurotoxicity. Colloids Surf B Biointerfaces 2025; 246: 114369.
  47. D. Baziulyte-Paulaviciene, V. Karabanovas, M. Stasys, G. Jarockyte, V. Poderys, S. Sakirzanovas. Synthesis and functionalization of NaGdF4:Yb,Er@NaGdF4 core-shell nanoparticles for possible application as multimodal contrast agents. Beilstein J Nanotechnol 2017; 8: 1815-1824.
  48. Z. J. Rakesh Arul, X. Li, F. M. Bell, A. Tew, C. Ducati, A. Rao, Z. Yu. Efficient short-wave infrared upconversion by self-sensitized holmium-doped nanoparticles. arXiv 2024; 11:
  49. G. D. Wang, H. Chen, W. Tang, D. Lee, J. Xie. Gd and Eu Co-Doped Nanoscale Metal-Organic Framework as a T1-T2 Dual-Modal Contrast Agent for Magnetic Resonance Imaging. Tomography 2016; 2(3): 179-187.
  50. Z. Li, J. Gong, S. Lu, X. Li, X. Gu, J. Xu. Photothermal lanthanide nanomaterials: from fundamentals to theranostic applications. BMEmat 2024; 2(4): e12088.
  51. Q. Fan, C. Sun, B. Hu, Q. Wang. Recent advances of lanthanide nanomaterials in Tumor NIR fluorescence detection and treatment. Mater Today Bio 2023; 20: 100646.
  52. D. Kang, H. S. Kim, S. Han, Y. Lee, Y. P. Kim, D. Y. Lee. A local water molecular-heating strategy for near-infrared long-lifetime imaging-guided photothermal therapy of glioblastoma. Nat Commun 2023; 14(1): 2755.
  53. X. Shi, C. Cao, Z. Zhang, J. Tian, Z. Hu. Radiopharmaceutical and Eu3+ doped gadolinium oxide nanoparticles mediated triple-excited fluorescence imaging and image-guided surgery. J Nanobiotechnology 2021; 19(1): 212.
  54. T. Tegafaw, D. Zhao, Y. Liu, H. Yue, A. K. Saidi, A. Baek. High Quantum Yields and Biomedical Fluorescent Imaging Applications of Photosensitized Trivalent Lanthanide Ion-Based Nanoparticles. Int J Mol Sci 2024; 25(21): 11419.
  55. A. K. Parchur, Q. Li, A. Zhou. Near-infrared photothermal therapy of Prussian-blue-functionalized lanthanide-ion-doped inorganic/plasmonic multifunctional nanostructures for the selective targeting of HER2-expressing breast cancer cells. Biomater Sci 2016; 4(12): 1781-1791.
  56. M. Wang, B. Li, Y. Du, H. Bu, Y. Tang, Q. Huang. Fluorescence imaging-guided cancer photothermal therapy using polydopamine and graphene quantum dot-capped Prussian blue nanocubes. RSC Adv 2021; 11(15): 8420-8429.
  57. X. Gao, Q. Wang, C. Cheng, S. Lin, T. Lin, C. Liu. The Application of Prussian Blue Nanoparticles in Tumor Diagnosis and Treatment. Sensors (Basel) 2020; 20(23): 6905.
  58. H. Luo, S. Gao. Recent advances in fluorescence imaging-guided photothermal therapy and photodynamic therapy for cancer: from near-infrared-I to near-infrared-II. J Control Release 2023; 362: 425-445.
  59. W. Piotrowski, K. Trejgis, K. Maciejewska, K. Ledwa, B. Fond, L. Marciniak. Thermochromic Luminescent Nanomaterials Based on Mn4+/Tb3+ Codoping for Temperature Imaging with Digital Cameras. ACS Appl Mater Interfaces 2020; 12(39): 44039-44048.
  60. O. Savchuk, J. J. Carvajal Marti, C. Cascales, P. Haro-Gonzalez, F. Sanz-Rodríguez, M. Aguilo. Bifunctional Tm3+,Yb3+:GdVO4@SiO2 Core-Shell Nanoparticles in HeLa Cells: Upconversion Luminescence Nanothermometry in the First Biological Window and Biolabelling in the Visible. Nanomaterials (Basel) 2020; 10(5): 993.
  61. P. Du, X. Wan, L. Luo, W. Li, L. Li. Thermally Stable Tb3+/Eu3+-Codoped K0.3Bi0.7F2.4 Nanoparticles with Multicolor Luminescence for White-Light-Emitting Diodes. ACS Appl Nano Mater 2021; 4(7): 7062-7071.
  62. C. C. Yang, W. Y. Wang, F. H. Lin, C. H. Hou. Rare-Earth-Doped Calcium Carbonate Exposed to X-ray Irradiation to Induce Reactive Oxygen Species for Tumor Treatment. Int J Mol Sci 2019; 20(5): 1148.
  63. S. Kaščáková, A. Giuliani, S. Lacerda, A. Pallier, P. Mercère, É. Tóth. X-ray-induced radiophotodynamic therapy (RPDT) using lanthanide micelles: beyond depth limitations. Nano Res 2015; 8(7): 2373-2379.
  64. J. Jin, Q. Zhao. Engineering nanoparticles to reprogram radiotherapy and immunotherapy: recent advances and future challenges. J Nanobiotechnology 2020; 18(1): 75.
  65. P. Krzyszczyk, A. Acevedo, E. J. Davidoff, L. M. Timmins, I. Marrero-Berrios, M. Patel. The growing role of precision and personalized medicine for cancer treatment. Technology (Singap) 2018; 6(3-4): 79-100.
  66. P. Franzone, A. Fiorentino, S. Barra, D. Cante, L. Masini, E. Cazzulo. Image-guided radiation therapy (IGRT): practical recommendations of Italian Association of Radiation Oncology (AIRO). Radiol Med 2016; 121(12): 958-965.
  67. L. Gong, Y. Zhang, C. Liu, M. Zhang, S. Han. Application of Radiosensitizers in Cancer Radiotherapy. Int J Nanomedicine 2021; 16: 1083-1102.
  68. J. F. Fowler, G. E. Adams, J. Denekamp. Radiosensitizers of hypoxic cells in solid tumors. Cancer Treat Rev 1976; 3(4): 227-256.
  69. H. Matsudaira, A. M. Ueno, I. Furuno. Iodine contrast medium sensitizes cultured mammalian cells to X rays but not to gamma rays. Radiat Res 1980; 84(1): 144-148.
  70. Y. Liu, P. Zhang, F. Li, X. Jin, J. Li, W. Chen. Metal-based NanoEnhancers for Future Radiotherapy: Radiosensitizing and Synergistic Effects on Tumor Cells. Theranostics 2018; 8(7): 1824-1849.
  71. M. L. Etheridge, S. A. Campbell, A. G. Erdman, C. L. Haynes, S. M. Wolf, J. McCullough. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomedicine 2013; 9(1): 1-14.
  72. B. Yu, X. Lu, X. Feng, T. Zhao, J. Li, Y. Lu. Gadolinium Oxide Nanoparticles Reinforce the Fractionated Radiotherapy-Induced Immune Response in Tri-Negative Breast Cancer via cGAS-STING Pathway. Int J Nanomedicine 2023; 18: 7713-7728.
  73. Z. Muradova, D. Tannous, A. Mostefa-Kara, T. T. Cao-Pham, C. Lamy, S. Broutin. Gadolinium-based nanoparticles AGuIX and their combination with ionizing radiation trigger AMPK-dependent proinflammatory reprogramming of tumor-associated macrophages. bioRxiv 2024; :
  74. X. Ying, Q. Chen, Y. Yang, Z. Wu, W. Zeng, C. Miao. Nanomedicines harnessing cGAS-STING pathway: sparking immune revitalization to transform ‘cold’ tumors into ‘hot’ tumors. Mol Cancer 2024; 23(1): 277.
  75. O. Tillement, G. Le Duc, S. Roux, P. Mowat, F. Lux, M. Janier, C. Louis, C. Rodriguez-Lafrasse, M. Dutreix, S. Lacombe, P. Perriat. High efficiency radiosensitization with Sub-5 nm theranostic lanthanide-based nanoparticles. 2018; :
  76. C. Wu, R. Cai, T. Zhao, L. Wu, L. Zhang, J. Jin. Hyaluronic Acid-Functionalized Gadolinium Oxide Nanoparticles for Magnetic Resonance Imaging-Guided Radiotherapy of Tumors. Nanoscale Res Lett 2020; 15(1): 94.
  77. S. J. Seo, S. M. Han, J. H. Cho, K. Hyodo, A. Zaboronok, H. You. Enhanced production of reactive oxygen species by gadolinium oxide nanoparticles under core-inner-shell excitation by proton or monochromatic X-ray irradiation: implication of the contribution from the interatomic de-excitation-mediated nanoradiator effect to dose enhancement. Radiat Environ Biophys 2015; 54(4): 423-431.
  78. X. Zhu, C. J. Qiu, J. J. Cao, D. Duosiken, Y. Zhang, B. G. Pei. Radiosensitization of Rare-Earth Nanoparticles Based on the Consistency Between Its K-Edge and the X-Ray Bremsstrahlung Peak. J Funct Biomater 2025; 16(2): 41.
  79. Y. Li, Y. Qi, H. Zhang, Z. Xia, T. Xie, W. Li. Gram-scale synthesis of highly biocompatible and intravenous injectable hafnium oxide nanocrystal with enhanced radiotherapy efficacy for cancer theranostic. Biomaterials 2020; 226: 119538.
  80. Y. Kuang, Y. Zhang, Y. Zhao, Y. Cao, Y. Zhang, Y. Chong. Dual-Stimuli-Responsive Multifunctional Gd2Hf2O7 Nanoparticles for MRI-Guided Combined Chemo-/Photothermal-/Radiotherapy of Resistant Tumors. ACS Appl Mater Interfaces 2020; 12(32): 35928-35939.
  81. A. A. Sherstiuk, S. A. Tsymbal, A. F. Fakhardo, V. N. Morozov, E. F. Krivoshapkina, E. Hey-Hawkins. Hafnium Oxide-Based Nanoplatform for Combined Chemoradiotherapy. ACS Biomater Sci Eng 2021; 7(12): 5633-5641.
  82. R. D. Teo, J. Termini, H. B. Gray. Lanthanides: Applications in Cancer Diagnosis and Therapy. J Med Chem 2016; 59(13): 6012-6024.
  83. Z. Xu, T. Luo, J. Mao, C. McCleary, E. Yuan, W. Lin. Monte Carlo Simulation-Guided Design of a Thorium-Based Metal-Organic Framework for Efficient Radiotherapy-Radiodynamic Therapy. Angew Chem Int Ed Engl 2022; 61(46): e202208685.
  84. K. Ni, G. Lan, W. Lin. Nanoscale Metal-Organic Frameworks Generate Reactive Oxygen Species for Cancer Therapy. ACS Cent Sci 2020; 6(6): 861-868.
  85. S. N. Mane, S. K. Kolekar, K. B. Sapnar, S. D. Dhole. Gamma radiation induced modification in metal–organic framework: a case study. J Radioanal Nucl Chem 2024; 333(6): 3115-3122.
  86. V. O. Ndabankulu, S. Maddila, S. B. Jonnalagadda. Synthesis of Lanthanide Doped TiO2 Nanoparticles and Their Photocatalytic Activity under Visible Light. Can J Chem 2019; 97(9): 672-681.
  87. Y. Chen, N. Li, J. Wang, X. Zhang, W. Pan, L. Yu. Enhancement of mitochondrial ROS accumulation and radiotherapeutic efficacy using a Gd-doped titania nanosensitizer. Theranostics 2019; 9(1): 167-178.
  88. H. Jang, M. H. Choi, Y. Yim, Y. K. Kim, D. H. Min. Dual-Wavelength Irradiation and Dox Delivery for Cancer Cell Ablation with Photocatalytic Pr Doped TiO2/NGO Hybrid Nanocomposite. Adv Healthc Mater 2015; 4(12): 1833-1840.
  89. H. Li, H. Liu, K. L. Wong, A. H. All. Lanthanide-doped upconversion nanoparticles as nanoprobes for bioimaging. Biomater Sci 2024; 12(18): 4650-4663.
  90. K. Malhotra, D. Hrovat, B. Kumar, G. Qu, V. J. Houten, R. Ahmed. Lanthanide-Doped Upconversion Nanoparticles: Exploring A Treasure Trove of NIR-Mediated Emerging Applications. ACS Appl Mater Interfaces 2023; 15(2): 2499-2528.
  91. P. Li, Y. Yan, B. Chen, P. Zhang, S. Wang, J. Zhou. Lanthanide-doped upconversion nanoparticles complexed with nano-oxide graphene used for upconversion fluorescence imaging and photothermal therapy. Biomater Sci 2018; 6(4): 877-884.
  92. H. L. Wei, W. Zheng, X. Zhang, H. Suo, B. Chen, Y. Wang. Tuning Near-Infrared-to-Ultraviolet Upconversion in Lanthanide-Doped Nanoparticles for Biomedical Applications. Adv Opt Mater 2023; 11(11): 2201716.
  93. D. J. Naczynski, C. Sun, S. Türkcan, C. Jenkins, A. L. Koh, D. Ikeda. X-ray-induced shortwave infrared biomedical imaging using rare-earth nanoprobes. Nano Lett 2015; 15(1): 96-102.
  94. B. K. Byrd, M. R. Folaron, J. P. Leonor, R. R. Strawbridge, X. Cao, P. Bruza. Characterizing short-wave infrared fluorescence of conventional near-infrared fluorophores. J Biomed Opt 2019; 24(3): 1-5.
  95. R. Wang, F. Zhang. NIR luminescent nanomaterials for biomedical imaging. J Mater Chem B 2014; 2(17): 2422-2443.
  96. A. L. Rogach, A. Eychmüller, S. G. Hickey, S. V. Kershaw. Infrared-emitting colloidal nanocrystals: synthesis, assembly, spectroscopy, and applications. Small 2007; 3(4): 536-557.
  97. D. J. Naczynski, J. H. Stafford, S. Türkcan, C. Jenkins, A. L. Koh, C. Sun. Rare-earth-doped nanoparticles for short-wave infrared fluorescence bioimaging and molecular targeting of alphaVbeta3-expressing tumors. Mol Imaging 2018; 17: 1536012118799131.
  98. M. Zhao, X. Chen. Recent Advances in NIR-II Materials for Biomedical Applications. Acc Mater Res 2024; 5(5): 600-613.
  99. X. Lei, R. Li, D. Tu, X. Shang, Y. Liu, W. You. Intense near-infrared-II luminescence from NaCeF4:Er/Yb nanoprobes for in vitro bioassay and in vivo bioimaging. Chem Sci (Camb) 2018; 9(20): 4682-4688.
  100. Y. Guo, J. Hu, P. Wang, H. Yang, S. Liang, D. Chen. In Vivo NIR-II Fluorescence Lifetime Imaging of Whole-Body Vascular Using High Quantum Yield Lanthanide-Doped Nanoparticles. Small 2023; 19(35): e2300392.
  101. Q. Wang, X. Zhang, Y. Tang, Y. Xiong, X. Wang, C. Li. High-Performance Hybrid Phototheranostics for NIR-IIb Fluorescence Imaging and NIR-II-Excitable Photothermal Therapy. Pharmaceutics 2023; 15(8): 2027.
  102. H. Zhao, H. Xiao, Y. Liu, H. Ju. Lanthanide-doped rare earth nanoparticles for near-infrared-II imaging and cancer therapy. BMEmat 2023; 1(3): e12032.
  103. H. Q. Wen, H. Y. Peng, K. Liu, M. H. Bian, Y. J. Xu, L. Dong. Sequential Growth of NaYF4:Yb/Er@NaGdF4 Nanodumbbells for Dual-Modality Fluorescence and Magnetic Resonance Imaging. ACS Appl Mater Interfaces 2017; 9(11): 9226-9232.
  104. Z. Zhang, J. Sheng, M. Zhang, X. Ma, Z. Geng, Z. Wang. Dual-modal imaging and excellent anticancer efficiency of cisplatin and doxorubicin loaded NaGdF4:Yb3+/Er3+ nanoparticles. RSC Adv 2018; 8(39): 22216-22225.
  105. C. Liu, Z. Gao, J. Zeng, Y. Hou, F. Fang, Y. Li. Magnetic/upconversion fluorescent NaGdF4:Yb,Er nanoparticle-based dual-modal molecular probes for imaging tiny tumors in vivo. ACS Nano 2013; 7(8): 7227-7240.
  106. Y. Yang, D. Tu, Y. Zhang, P. Zhang, X. Chen. Recent advances in design of lanthanide-containing NIR-II luminescent nanoprobes. iScience 2021; 24(2): 102062.
  107. S. Jeong, N. Won, J. Lee, J. Bang, J. Yoo, S. G. Kim. Multiplexed near-infrared in vivo imaging complementarily using quantum dots and upconverting NaYF4:Yb3+,Tm3+ nanoparticles. Chem Commun (Camb) 2011; 47(28): 8022-8024.
  108. Y. Sun, J. Peng, W. Feng, F. Li. Upconversion nanophosphors NaLuF4:Yb,Tm for lymphatic imaging in vivo by real-time upconversion luminescence imaging under ambient light and high-resolution X-ray CT. Theranostics 2013; 3(5): 346-353.
  109. X. Zhu, H. Zhang, F. Zhang. Expanding NIR-II Lanthanide Toolboxes for Improved Biomedical Imaging and Detection. Acc Mater Res 2023; 4(6): 536-547.
  110. Y. Guo, J. Hu, P. Wang, H. Yang, S. Liang, D. Chen. In Vivo NIR-II Fluorescence Lifetime Imaging of Whole-Body Vascular Using High Quantum Yield Lanthanide-Doped Nanoparticles. Small 2023; 19(35): e2300392.
  111. Y. Y. Zhu, L. Song, Y. Q. Zhang, W. L. Liu, W. L. Chen, W. L. Gao. Development of a Rare Earth Nanoprobe Enables In Vivo Real-Time Detection of Sentinel Lymph Node Metastasis of Breast Cancer Using NIR-IIb Imaging. Cancer Res 2023; 83(20): 3428-3441.
  112. Z. Sun, H. Huang, R. Zhang, X. Yang, H. Yang, C. Li. Activatable Rare Earth Near-Infrared-II Fluorescence Ratiometric Nanoprobes. Nano Lett 2021; 21(15): 6576-6583.
  113. M. Hong, P. Sun, Y. Liu, J. Ke, Z. Wu. Lanthanide-based NIR-II Fluorescent Nanoprobes and Their Biomedical Applications. Huaxue Xuebao 2022; 80(4): 542.
  114. K. Lv, H. Wang, X. Fu, S. Chen, R. Zhang, Y. Zhou. An Integrated Nanoplatform via Dual Channel Excitation for Both Precise Fluorescence Imaging and Photodynamic Therapy of Orthotopic Breast Tumor in NIR-II Region. Small 2024; 20(47): e2404007.
  115. G. Duan, J. Zhang, Z. Wei, X. Wang, J. Zeng, S. Wu. Intraoperative diagnosis of early lymphatic metastasis using neodymium-based rare-earth NIR-II fluorescence nanoprobe. Nanoscale Adv 2023; 5(16): 4240-4249.
  116. L. Machova Urdzikova, D. Mareková, T. Vasylyshyn, P. Matouš, V. Patsula, V. Oleksa, O. Shapoval, M. Vosmanská, D. Liebl, A. Benda, V. Herynek, D. Horák, P. Jendelová. Toxicity of Large and Small Surface-Engineered Upconverting Nanoparticles for In Vitro and In Vivo Bioapplications. Int J Mol Sci 2024; 25:
  117. Z. Wang, T. Zhang, L. Pi, H. Xiang, P. Dong, C. Lu. Large-scale one-pot synthesis of water-soluble and biocompatible upconversion nanoparticles for dual-modal imaging. Colloids Surf B Biointerfaces 2020; : 111480.
  118. J. W. Veggel. Near-infrared Emission of Redispersible Er3+, Nd3+, and Ho3+ Doped LaF3 Nanoparticles. Nano Lett 2002; 2(7): 733-737.
  119. F. Wang, Y. Zhang, X. Fan, M. Wang. Facile synthesis of water-soluble LaF3:Ln3+ nanocrystals. J Mater Chem 2006; 16(11): 1031.
  120. M. Li, Z. H. Hao, X. N. Peng, J. B. Li, X. F. Yu, Q. Q. Wang. Controllable energy transfer in fluorescence upconversion of NdF3 and NaNdF4 nanocrystals. Opt Express 2010; 18(4): 3364.
  121. G. Chen, S. Liu, W.-C. Law, F. Wu, M. T. Swihart, H. Ågren, P. N. Prasad. Core/Shell NaGdF4:Nd3+/NaGdF4 Nanocrystals with Efficient Near-Infrared to Near-Infrared Downconversion Photoluminescence for Bioimaging Applications. ACS Nano 2012; 6(4): 2969-2977.
  122. M. Kamimura, Y. Saito, K. Soga, Y. Nagasaki. Preparation of PEG and Protein Co-immobilized Upconversion Nanophosphors as Near-infrared Biolabeling Materials. J Photopolym Sci Technol 2008; 21(2): 183-187.
  123. D. J. Naczynski, M. C. Tan, M. Zevon, B. Wall, J. Kohl, A. Kulesa. Rare-earth-doped biological composites as in vivo shortwave infrared reporters. Nat Commun 2013; 4(1): 2199.
  124. F. Ren, F. Wang, A. Baghdasaryan, Y. Li, H. Liu, R. Hsu. Shortwave-infrared-light-emitting probes for the in vivo tracking of cancer vaccines and the elicited immune responses. Nat Biomed Eng 2024; 8(6): 726-739.
  125. A. A. Simon, L. Haye, A. Alhalabi, Q. Gresil, B. M. Muñoz, S. Mornet. Expanding the Palette of SWIR Emitting Nanoparticles Based on Au Nanoclusters for Single-Particle Tracking Microscopy. Adv Sci (Weinh) 2024; 11(24): e2309267.
  126. J. Meng, Y. Cui, Y. Wang. Rare Earth-Doped Nanoparticles for Near-Infrared II Image-Guided Photodynamic Therapy. ACS Appl Nano Mater 2024; 7(9): 11008-11018.
  127. P. Du, Y. Wei, Y. Liang, R. An, S. Liu, P. Lei. Near-Infrared-Responsive Rare Earth Nanoparticles for Optical Imaging and Wireless Phototherapy. Adv Sci (Weinh) 2024; 11(8): e2305308.
  128. H. Li, H. Liu, K. L. Wong, A. H. All. Lanthanide-doped upconversion nanoparticles as nanoprobes for bioimaging. Biomater Sci 2024; 12(18): 4650-4663.
  129. X. Cui, Q. Ruan, X. Zhuo, X. Xia, J. Hu, R. Fu. Photothermal Nanomaterials: A Powerful Light-to-Heat Converter. Chem Rev 2023; 123(11): 6891-6952.
  130. K. Chen, X. Sun, Y. Liu, S. Li, D. Meng. Advances in clinical applications of microneedle. Front Pharmacol 2025; 16: 1607210.
  131. X. Feng, Y. Shang, H. Zhang, R. Li, W. Wang, D. Zhang. Enhanced luminescence and tunable magnetic properties of lanthanide coordination polymers based on fluorine substitution and phenanthroline ligand. RSC Adv 2019; 9(29): 16328-16338.
  132. Y. Zhang, S. Zhang, Z. Zhang, L. Ji, J. Zhang, Q. Wang. Recent Progress on NIR-II Photothermal Therapy. Front Chem 2021; 9: 728066.
  133. J. Choi, G. Kim, S. B. Cho, H. J. Im. Radiosensitizing high-Z metal nanoparticles for enhanced radiotherapy of glioblastoma multiforme. J Nanobiotechnology 2020; 18(1): 122.
  134. G. Ferro-Flores, A. Ancira-Cortez, B. Ocampo-García, L. Meléndez-Alafort. Molecularly Targeted Lanthanide Nanoparticles for Cancer Theranostic Applications. Nanomaterials (Basel) 2024; 14(3): 296.
  135. X. Zhu, C. J. Qiu, J. J. Cao, D. Duosiken, Y. Zhang, B. G. Pei. Radiosensitization of Rare-Earth Nanoparticles Based on the Consistency Between Its K-Edge and the X-Ray Bremsstrahlung Peak. J Funct Biomater 2025; 16(2): 41.
  136. H. Nosrati, M. Salehiabar, J. Charmi, K. Yaray, M. Ghaffarlou, E. Balcioglu. Enhanced In Vivo Radiotherapy of Breast Cancer Using Gadolinium Oxide and Gold Hybrid Nanoparticles. ACS Appl Bio Mater 2023; 6(2): 784-792.
  137. N. N. Zhang, C. Y. Lu, M. J. Chen, X. L. Xu, G. F. Shu, Y. Z. Du. Recent advances in near-infrared II imaging technology for biological detection. J Nanobiotechnology 2021; 19(1): 132.
  138. Y. Zhang, Z. Li, Z. Du, J. Pan, Y. Huang. Multifunctional Upconversion Nanoparticles Transforming Photoacoustic Imaging: A Review. Nanomaterials (Basel) 2025; 15(14): 1074.
  139. S. Zhao, R. Tian, B. Shao, Y. Feng, S. Yuan, L. Dong. UCNP-Bi2Se3 Upconverting Nanohybrid for Upconversion Luminescence and CT Imaging and Photothermal Therapy. Chemistry 2020; 26(5): 1127-1135.
  140. G. Ferro-Flores, A. Ancira-Cortez, B. Ocampo-García, L. Meléndez-Alafort. Molecularly Targeted Lanthanide Nanoparticles for Cancer Theranostic Applications. Nanomaterials (Basel) 2024; 14(3): 296.
  141. Q. Zhang, S. O’Brien, J. Grimm. Biomedical Applications of Lanthanide Nanomaterials, for Imaging, Sensing and Therapy. Nanotheranostics 2022; 6(2): 184-194.
  142. X. Li, M. Jiang, S. Zeng, H. Liu. Polydopamine coated multifunctional lanthanide theranostic agent for vascular malformation and tumor vessel imaging beyond 1500 nm and imaging-guided photothermal therapy. Theranostics 2019; 9(13): 3866-3878.
  143. X. Jiang, X. Jiang, D. Wu, W. Xie, X. Liu, J. Zheng. A pH-Sensitive Nanoparticle as Reactive Oxygen Species Amplifier to Regulate Tumor Microenvironment and Potentiate Tumor Radiotherapy. Int J Nanomedicine 2024; 19: 709-725.
  144. M. Carofiglio, S. Barui, V. Cauda, M. Laurenti. Doped Zinc Oxide Nanoparticles: Synthesis, Characterization and Potential Use in Nanomedicine. Appl Sci (Basel) 2020; 10(15): 5194.
  145. F. Lux, V. L. Tran, E. Thomas, S. Dufort, F. Rossetti, M. Martini. AGuIX® from bench to bedside—Transfer of an ultrasmall theranostic gadolinium-based nanoparticle to clinical medicine. Br J Radiol 2019; 92(1093): 20180365.
  146. C. Verry, S. Dufort, J. Villa, M. Gavard, C. Iriart, S. Grand. Theranostic AGuIX nanoparticles as radiosensitizer: A phase I, dose-escalation study in patients with multiple brain metastases (NANO-RAD trial). Radiother Oncol 2021; 160: 159-165.
  147. Y. Luo. Radiobiological perspective on metrics for quantifying dose enhancement effects of High-Z nanoparticles. Front Nanotechnol 2025; 7:
  148. V. M. Lu, F. Crawshay-Williams, B. White, A. Elliot, M. A. Hill, H. E. Townley. Cytotoxicity, dose-enhancement and radiosensitization of glioblastoma cells with rare earth nanoparticles. Artif Cells Nanomed Biotechnol 2019; 47(1): 132-143.
  149. D. Zhong, Y. Wang, F. Xie, S. Chen, X. Yang, Z. Ma. A biomineralized Prussian blue nanotherapeutic for enhanced cancer photothermal therapy. J Mater Chem B 2022; 10(25): 4889-4896.
  150. M. Fuentealba, A. Ferreira, A. Salgado, C. Vergara, S. Díez, M. Santibáñez. An Optimized Method for Evaluating the Potential Gd-Nanoparticle Dose Enhancement Produced by Electronic Brachytherapy. Nanomaterials (Basel) 2024; 14(5): 430.
  151. Z. H. Chen, X. Wang, M. Yang, J. Ming, B. Yun, L. Zhang. An Extended NIR-II Superior Imaging Window from 1500 to 1900 nm for High-Resolution In Vivo Multiplexed Imaging Based on Lanthanide Nanocrystals. Angew Chem Int Ed Engl 2023; 62(49): e202311883.
  152. C. Y. Tsang, Y. Zhang. Nanomaterials for light-mediated therapeutics in deep tissue. Chem Soc Rev 2024; 53(6): 2898-2931.
  153. M. T. Aloy, J. Sidi Boumedine, A. Deville, D. Kryza, A. Gauthier, D. Brichart-Vernos. Proof of Concept of the Radiosensitizing Effect of Gadolinium Oxide Nanoparticles in Cell Spheroids and a Tumor-Implanted Murine Model of Chondrosarcoma. Int J Nanomedicine 2022; 17: 6655-6673.
  154. B. Li, M. Zhao, J. Lin, P. Huang, X. Chen. Management of fluorescent organic/inorganic nanohybrids for biomedical applications in the NIR-II region. Chem Soc Rev 2022; 51(18): 7692-7714.
  155. I. A. Khan, T. Yu, M. Yang, J. Liu, Z. Chen. A Systematic Review of Toxicity, Biodistribution, and Biosafety in Upconversion Nanomaterials: Critical Insights into Toxicity Mitigation Strategies and Future Directions for Safe Applications. BME Frontiers 2025; 6: 0120.
  156. S. K. Funke, C. Factor, M. Rasschaert, L. Lezius, M. Sperling, U. Karst. Long-term Gadolinium Retention in the Healthy Rat Brain: Comparison between Gadopiclenol, Gadobutrol, and Gadodiamide. Radiology 2022; 305(1): 179-189.
  157. Z. Li, J. Gong, S. Lu, X. Li, X. Gu, J. Xu. Photothermal lanthanide nanomaterials: from fundamentals to theranostic applications. BMEmat 2024; 2(4): e12088.
  158. C. Tian, X. Xue, Y. Chen, R. Liu, Y. Wang, S. Ye. Phosphotungstate Acid Doped Polyanilines Nanorods for in situ NIR-II Photothermal Therapy of Orthotopic Hepatocellular Carcinoma in Rabbit. Int J Nanomedicine 2022; 17: 5565-5579.
  159. T. Cao, Y. Yang, Y. Sun, Y. Wu, Y. Gao, W. Feng. Biodistribution of sub-10 nm PEG-modified radioactive/upconversion nanoparticles. Biomaterials 2013; 34(29): 7127-7134.
  160. H. Zhang, X. Wang, R. Jin, Q. Su. Preparation and applications of polymer-modified lanthanide-doped upconversion nanoparticles. Giant (Oxf) 2022; 12: 100130.
  161. Q. Li, Z. Wang, Y. Chen, G. Zhang. Elemental bio-imaging of PEGylated NaYF4:Yb/Tm/Gd upconversion nanoparticles in mice by laser ablation inductively coupled plasma mass spectrometry to study toxic side effects on the spleen, liver and kidneys. Metallomics 2017; 9(8): 1150-1156.
  162. J. Yu, W. Yin, T. Peng, Y. N. Chang, Y. Zu, J. Li. Biodistribution, excretion, and toxicity of polyethyleneimine modified NaYF4:Yb,Er upconversion nanoparticles in mice via different administration routes. Nanoscale 2017; 9(13): 4497-4507.
  163. N. Fretellier, J. M. Idée, M. Rasschaert, C. Factor, A. J. Van der Molen. Gadolinium Deposition in Bone Tissues After Contrast-enhanced Magnetic Resonance Imaging: A Comprehensive Review. Invest Radiol 2025; 60(11): 722-744.
  164. J. Biau, X. Durando, F. Boux, I. Molnar, J. Moreau, B. Leyrat. NANO-GBM trial of AGuIX nanoparticles with radiotherapy and temozolomide in the treatment of newly diagnosed Glioblastoma: Phase 1b outcomes and MRI-based biodistribution. Clin Transl Radiat Oncol 2024; 48: 100833.
  165. F. Iacobellis, M. Di Serafino, C. Russo, R. Ronza, M. Caruso, G. Dell’Aversano Orabona. Safe and Informed Use of Gadolinium-Based Contrast Agent in Body Magnetic Resonance Imaging: Where We Were and Where We Are. J Clin Med 2024; 13(8): 2193.
  166. H. J. Seo, S. H. Nam, H. J. Im, J. Y. Park, J. Y. Lee, B. Yoo. Rapid Hepatobiliary Excretion of Micelle-Encapsulated/Radiolabeled Upconverting Nanoparticles as an Integrated Form. Sci Rep 2015; 5(1): 15685.
  167. P. Mi. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020; 10(10): 4557-4588.
  168. H. Chen, P. Timashev, Y. Zhang, X. Xue, X. J. Liang. Nanotechnology-based combinatorial phototherapy for enhanced cancer treatment. RSC Adv 2022; 12(16): 9725-9737.
  169. Y. Zhang, G. Zhang, G. Wang, L. Wu, N. A. Monteiro-Riviere, Y. Li. The synergistic strategies for the immuno-oncotherapy with photothermal nanoagents. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2021; 13(5): e1717.
  170. A. A. Rahmani, Q. Jia, H. H. Bahti, R. P. Fauzia, S. Wyantuti. Recent advances in lanthanide-based nanoparticle contrast agents for magnetic resonance imaging: Synthesis, characterization, and applications. OpenNano 2025; 21: 100226.
  171. J. Zhang, J. Du, S. Jia, Y. Li, H. Ågren, P. N. Prasad. Recent Advances in Upconversion Nanoparticles for Therapeutics: From Fundamentals to Cutting-Edge Applications. Electron 2025; 3(3): e70012.
  172. Z. Luo, D. Mao, X. Li, J. Luo, C. Gong, X. Liu. Lanthanide-based nanoparticles for cancer phototherapy. Coord Chem Rev 2024; 508: 215773.

Comments