Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 1.
Published in final edited form as: Nat Rev Bioeng. 2024 Jun 19;2(9):752–769. doi: 10.1038/s44222-024-00196-z

Engineering photodynamics for treatment, priming and imaging

Girgis Obaid 1, Jonathan P Celli 2, Mans Broekgaarden 3, Anne-Laure Bulin 3, Petteri Uusimaa 4, Brian Pogue 5, Tayyaba Hasan 6, Huang-Chiao Huang 7,
PMCID: PMC11801064  NIHMSID: NIHMS2011117  PMID: 39927170

Abstract

Photodynamic therapy (PDT) is a photochemistry-based treatment approach that relies on the activation of photosensitizers by light to locally generate reactive oxygen species that induce cellular cytotoxicity, in particular for the treatment of tumours. The cytotoxic effects of PDT are depth-limited owing to light penetration limits in tissue. However, photodynamic priming (PDP), which inherently occurs during PDT, can prime the tissue microenvironment to adjuvant therapies beyond the direct PDT ablative zone. In this Review, we discuss the underlying mechanisms of PDT and PDP, and their application to the treatment of cancer, outlining how PDP can permeabilize the tumour vasculature, overcome biological barriers, modulate multidrug resistance, enhance immune responses, increase tumour permeability and enable the photochemical release of drugs. We further examine the molecular engineering of photosensitizers to improve their pharmacodynamic and pharmacokinetic properties, increase their molecular specificity and allow image guidance of PDT, and investigate engineered cellular models for the design and optimization of PDT and PDP. Finally, we discuss alternative activation sources, including ultrasound, X-rays and self-illuminating compounds, and outline key barriers to the clinical translation of PDT and PDP.

Introduction

Photodynamic therapy (PDT) is a photochemistry-based treatment modality that involves the light activation of photosensitizers to generate reactive oxygen species (ROS), such as singlet oxygen (1O2), and free radicals1. ROS can be cytotoxic, deleterious or biomodulatory to biological targets, depending on the light dose and photosensitizer concentration2, and they can thus be used for the treatment of diseases. PDT was first approved by the US Food and Drug Administration (FDA) for bladder cancer, followed by indications in ophthalmology, dermatology and oncology3. Although PDT has been clinically proven as a minimally invasive treatment for certain local tumours (such as, prostate cancer, brain cancer and bladder cancer) (Fig. 1a) with high spatiotemporal control and minimal-to-no side effects, its widespread clinical implementation for cancers and metastatic diseases remains limited. This limitation is likely due to various factors, including perceptions from studies using first-generation photosensitizers with limited tumour specificity, training barriers to bring optical technologies (for example, lasers) into mainstream oncology, insufficient knowledge (and misconceptions) about the biomodulatory effects of PDT beyond the tissue ablation zone, the need for preclinical models to optimize PDT dosimetry and study its biological responses, and the complicated regulatory pathway for what is considered a combination product (drug and device). Bioengineering approaches can address some of these concerns to help integrate PDT into clinical workflows. Here, the science and clinical application of using photodynamic activation to achieve intended biological responses in target cells and tissues is referred to broadly as ‘photodynamics’.

Fig. 1 |. Photodynamic therapy.

Fig. 1 |

a, Major advances in photodynamic therapy (PDT) and related engineering approaches since the first application of PDT in patients in 1976220. The following references are cited in the timeline: refs. 17,53,6163,65,66,76,164,220222. b, Examples of PDT agents include monoclonal antibody–photosensitizer (mAb–PS) conjugates (photoimmunconjugates)17, F(ab’)2–PS conjugates66, nanobody–PS conjugates76, virus-like particle drug conjugates88, porphysomes63, photoactivable multi-inhibitor nanoliposomes61, photoimmunconjugate nanoliposomes62,86 and photoimmunconjugate nanoparticles87. c, Endoscopic ultrasound (EUS)-guided Visudyne-PDT procedure in a patient with locally advanced pancreatic cancer55. The patient first receives an intravenous administration of Visudyne (liposomal verteporfin commercialized by Novartis AG; 0.4 mg per kg) at 60–90 min before the EUS procedure. Under EUS guidance, a diffuser is then placed into the tumour, delivering light at 50 J cm−1 for 333 s. Computed tomography images are obtained after PDT to identify tumour necrosis55 and photodynamic priming (PDP) effects54. d, General procedure in clinical PDT, including formulation selection and administration routes of photosensitizers, illumination procedures, technologies involved in photoactivation and image-guidance in treatment planning, monitoring, and evaluation. AK, actinic keratosis; ALA, 5-aminolevulinic acid; AMD, age-related macular degeneration; F(ab’), fragment antigen-binding region; HpD, haematoporphyrin derivative; LED, light-emitting diode; PCI, photochemical internalization; PIT, photoimmunotherapy.

Multifunctionality can be imparted on photosensitizers, for example, by tuning and regulating their pharmacokinetic and pharmacodynamic behaviour, adding molecular specificity for tumour tissue, improving their transport across cellular barriers, and enabling stimuli-responsive controlled release with image guidance. In 2000, Visudyne (a liposomal formulation of verteporfin, Novartis AG), along with diode lasers at 689 nm (Coherent and Zeiss lasers), was approved by the FDA as the first liposomal photosensitizer delivery system for macular degeneration4. In 2020, the first antibody–photosensitizer conjugate, cetuximab saratolacan sodium5, with a near-infrared laser system (BioBlade, Rakuten Medical Inc.), was approved by the Ministry of Health, Labour and Welfare in Japan for photoimmunotherapy of head and neck cancer (Fig. 1a). Iterations and amalgamations of nanoscale systems and molecule-targeted nanoconstructs (Fig. 1b) may further improve the clinical translation of PDT (Fig. 1c,d).

Cancer therapy should consider resistance mechanisms, physical tumour barriers, microenvironmental factors and immunologic heterogeneity to improve treatment responses. Photodynamic priming (PDP) is a collateral phenomenon of PDT beyond the direct PDT ablative zone caused by the exponential decrease of light intensity on its way through tissues (although the incident optical energy drops to ~37% within a few millimetres, diffused red light penetrates up to several centimetres). PDP can thus transiently alter the tumour microenvironment, including modulating vascular permeability, cellular processes and subcellular compartments of tumour and non-tumour cells, as well as altering extracellular matrix components and the local tumour immune landscape (Fig. 2). Therefore, PDP is a powerful tool to control vascular permeability6, target drug transporter proteins7, modulate evolutionary processes in disease progression6, remodel the tumour stroma8,9 and potentially modulate the immune landscape10. PDP may thus enable other treatment modalities (such as, chemotherapy, radiation and immunotherapy) and potentially reverse chemoresistance. To identify and optimize photodynamic modalities in preclinical settings, high-throughput heterocellular models have been developed that recapitulate key physiological and architectural cues of disease (Fig. 3a); for example, 3D and organoid cultures enable high-throughput dosimetry analyses, mechanistic studies and evaluation of treatment response (Fig. 3b).

Fig. 2 |. Manipulation of tissue architecture and microenvironment by photodynamics.

Fig. 2 |

Light irradiation of solid tumours results in a gradient of fluences that decrease exponentially along the tumour cross-section. A zone of acute tumour tissue death arises in the areas of high light fluences. Beyond that zone of photodynamic therapy (PDT)-induced tissue death, a finite photodynamic priming (PDP) volume exists, which is characterized by transient priming events, such as loosening of stroma and permeabilization of vasculature, as well as mechanistic sensitization to adjuvant therapies and immunological modulation. The combined effects of PDT and PDP lead to local tumour destruction and allow the control of locoregional and distant metastases through the abscopal effect. DAMPs, damage-associated molecular patterns; ICI, immune checkpoint inhibitor; RTKi, receptor tyrosine kinase inhibitor.

Fig. 3 |. Models and analysis of photodynamic therapy.

Fig. 3 |

a, Tissue-engineered 3D cellular models can be used to optimize photodynamic therapy in vitro, including monotypic and heterotypic spheroid cultures, architecturally complex tumouroids grown on 3D-printed matrix substrates, tumouroids containing immunological subtypes (T cells), and organoids derived from patient tumours. b, Biomaterials can be utilized to develop 3D cellular models. Spheroids can be cultured in ultra-low-attachment microwell plates. Substrates such as Matrigel, collagen, alginate and 3D-printed hydrogels can be used to develop matrix-embedded models, including architecturally complex tumouroids and immunologically active tumouroids. c, High-throughput image analysis workflows can be applied to evaluate and optimize photodynamic therapy dosimetry and combinations, including LIVE/DEAD imaging, comprehensive high-throughput image analyses for architecturally complex tumouroids, volumetric imaging of matrix-embedded tumouroids, assessment of photosensitizer microdistribution and imaging of optical redox ratios. Part c adapted with permission from ref. 223, International Society for Optics and Photonics; adapted from ref. 101, Springer Nature Limited; adapted with permission from ref. 224, American Chemical Society; and adapted with permission from ref. 108, Elsevier.

Effective PDT also requires the coordination of photosensitizers, light sources and oxygen at the target tissue. The choice of photonics device depends on the disease location, treatment schedule, type of photosensitizer and many other variables. To improve the consistency and safety of PDT, implantable photonic devices are being developed to treat large and disseminated malignancies, whereas image-guided strategies can inform the timing and dosimetry of PDT. However, the limited penetration depth of visible light in tissue confines PDT to superficial tumours. Deep-seated lesions thus have to be treated by multiple interstitial illuminators and intraoperative illumination. Alternatively, materials can be applied that can be excited by other type of energy sources, such as, ionizing radiation, ultrasound, chemiluminescence or bioluminescence, and Cerenkov radiation. If energy can be delivered in a minimally invasive manner to activate low-cost, off-label, clinically approved photosensitizers using patient-customized applicators and affordable, transportable light delivery systems, PDT also becomes well-suited as a low-cost treatment modality in resource-limited settings (Box 1).

Box 1 |. Photodynamic therapy in resource-limited settings.

By 2030, 75% of global cancer deaths are expected to occur in low-income and middle-income countries225. Therefore, user-friendly technologies for cancer treatment are required that are portable, allow battery-powered operation, ensure cost-effectiveness and enable telemedicine integration. Photodynamic therapy (PDT) is well-suited to meeting these requirements, through the use of low-cost light sources and handheld fluorescence imaging tools integrated with technologies such as smartphones and artificial intelligence, and well-tolerated topical or oral photosensitizer formulations.

For example, the PDT Brazil program introduced PDT for nonmelanoma skin cancer treatment in over 100 clinics across Brazil, Bolivia, Chile, Ecuador, El Salvador, Colombia, Cuba, Mexico, Peru and Venezuela226,227. Through industry partnership, an affordable, simple-to-use light-emitting diode (LED)-based irradiation device (LINCE device, MM Optics LTDA) was disseminated to participating clinics. This portable device includes handheld probes for treatment (630-nm LED array) and fluorescence-based monitoring of protoporphyrin IX accumulation and photobleaching. Using this approach, an 80% cure rate of lesions could be achieved for patients treated since 2012227.

Over 135,000 new cases of oral cancer occur annually in India228. A low-cost, portable, battery-operated system was developed for image-guided PDT of oral cancer229232. This platform is based on a cost-effective fibre-coupled LED for PDT, coupled with smartphone-based image guidance using an LED array over the camera sensor229 or an attached intraoral probe230. Ergonomic light delivery is achieved using 3D-printed intraoral applicators, which also standardize beam size and dosimetry231. Using this PDT device, complete and durable tumour response was reported after a single PDT session in 22 out of 30 patients (NCT03638622), with no serious adverse events, scarring, fibrosis or loss of function232.

Understanding photosensitizer and photonic device design considerations is key to the optimization and clinical translation of PDT for advanced cancers (Box 2). In this Review, we discuss photodynamics, in particular PDP, and the molecular engineering of photosensitizers, with an emphasis on design considerations that cover compartmentalization, physical entrapment and chemical linkage, photophysical and photochemical alteration, degradation, and controlled drug release. We further highlight PDP as a powerful clinical tool for tumour modulation at the molecular, cellular, tissue microenvironmental and immunological level. We also examine engineering strategies for photonic devices and alternative light sources, as well as image-guided and radiomics methods for the personalization of PDT, and provide a roadmap for their integration into clinical protocols. Finally, we investigate materials chemistry, image analysis and smartphone interface approaches, which are particularly relevant in resource-limited settings.

Box 2 |. Regulatory and translational considerations.

Photodynamic therapy (PDT) is typically regulated as a combination product that includes a pharmaceutical and a medical device used for light activation. In the USA, this regulatory pathway involves pre-market approval and is executed by the US Food and Drug Administration (FDA). The dosimetry for PDT and photodynamic priming (PDP) is complex and translationally challenging, involving the photosensitizer agent, potential excipients (for example, nanoscale carriers) and light irradiation parameters. Although photosensitizers are generally considered non-cytotoxic and safe in the absence of light, explicit toxicological evaluation of photosensitizer doses and the associated concentration of excipients are required. With respect to light dosimetry, clinical dosimetry values must explicitly specify the wavelength of the light source, the total light dose in J cm−2 and the irradiance in mW cm−2. Therefore, preclinical research should identify and report these parameters.

Importantly, intraluminal, interstitial and/or endovascular light irradiation need to be safe. In particular, multimodal and multiwavelength light sources can provide real-time feedback on treatment efficiency (for example, drug localization and implicit dosimetry). The diverse therapeutic and biomodulatory effects of PDT and PDP may be achieved using alternative activation mechanisms, such as ultrasound, radiotherapy and radioluminescence. However, the mechanisms of sonodynamic therapy, radiodynamic therapy and X-ray-activated PDT or PDP remain unclear and require further elucidation.

Three-dimensional models can enable translationally relevant evaluation of PDT, PDP and combination therapies. For example, desmoplastic models can be applied to investigate PDP, and (vascularized) flow models can be used to study resistance, drug delivery and enhanced permeability and retention.

Patients with locally advanced or recurrent diseases may be selected for PDT safety and feasibility studies3. However, such clinical studies are not designed to investigate the full potential of PDT and its long-term effects, in particular with regard to PDP. Preclinical studies using large animal models may also have an important role in evaluating the safety, efficacy and long-term effects of PDT-based therapies, and can be applied to train surgical personnel in the application of PDT and PDP.

Photodynamic priming

Tumour ablation is the first and highest priority for PDT. However, as light traverses through the tissue, the irradiances and fluences of light drop below the threshold required to ablate tumour tissue. In areas that extend beyond the direct ablative zone of PDT, sub-cytotoxic PDT regimens inadvertently exhibit a range of tumour-modulating phenomena, which are referred to as PDP6. PDP effects prime the tumour microenvironment (vasculature, stroma, immune landscape and oxygenation) in a way that may synergize with adjuvant modalities, including immune checkpoint therapy, sensitization to radiotherapy, modulation of tumour tissue and stroma for improved drug delivery, and photochemical internalization for endosomal escape. Such subablative priming mechanistically sensitizes tumour tissue that survives beyond the PDT ablative zone (Fig. 2).

PDT can serve as an adjuvant to a number of modalities, including chemotherapy, receptor tyrosine kinase inhibitors, biologics, immune checkpoint therapies and radiation therapy. For example, PDT can be applied as an adjuvant for chemotherapeutics, combining the tumour-ablative effects of PDT with the systemic cytotoxic and antiproliferative effects of chemotherapy11. Such PDT–chemo combinations are likely to benefit from PDP, which can increase the chemo-sensitivity of tissue that survives beyond the direct ablative zone of PDT. In addition, PDP can be applied to maintain the integrity and perfusion of tumour vasculature to enhance subsequent systematically administered therapies. However, the effects of PDP on tumour tissue that had received subablative regimens of PDT need to be thoroughly investigated to design the sequence of light activation and administration of concomitant secondary therapies, such as chemotherapy.

Upon light activation, the photosensitizer generates ROS that can oxidize or destroy biomolecules, such as anti-apoptotic proteins (B-cell lymphoma 2 (BCL-2) and B-cell lymphoma-extra large (BCL-xL))12,13 and drug efflux transporters7,14 responsible for the development of multidrug resistance. PDP can also increase vascular permeability and alter the tumour extracellular matrix, improving drug transport to the tumour site and sensitizing cancer cells to chemotherapeutic agents. Moreover, oxidative stress and biomolecule oxidation triggered by PDP can lead to immunogenic cell death responses, promoting the release of tumour-associated antigens and anti-tumour immune responses.

Priming the tumour vasculature

Vascular responses to PDT (such as, permeability and occlusion) are elicited by short drug–light intervals (that is, the time interval between photosensitizer administration and subsequent light activation), which ensure that photosensitizers are in circulation during activation. Vascular responses are also dictated by the PDT dose, with low doses typically inducing vascular permeability, and high doses typically inducing vascular occlusion and ablation. For example, PDT using a 15-min drug–light interval can impede blood flow 6 h after treatment, leading to a 36% reduction in tumour regrowth rates in the RIF-1 mouse fibrosarcoma model15. By contrast, a drug–light interval of 3 h does not result in a therapeutic effect. Similarly, vascular perfusion, tumour vessel density and growth of KLN205 mouse lung tumours and LM8 mouse osteosarcoma tumours can be reduced by applying PDT in a 15-min drug–light interval, whereas treatment with a 3-h drug–light interval is less efficacious16. Vascular responses are also specific to the photosensitizer. Using a photoimmunoconjugate, a drug–light interval of 192 h delays tumour growth and increases survival in an M-1 myosarcoma mouse model with respect to animals undergoing light activation immediately after photoimmunoconjugate administration17.

Vascular effects of PDT have first been described using fluorescent spheres with diameters ≥100 nm, which show increased tumour extravasation in mouse models of colon cancer following 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-a PDT. This phenomenon has been utilized to improve the efficacy of liposomal doxorubicin18. In general, liposomes can be applied for PDT-controlled chemotherapy; for example, chemotherapeutics, such as doxorubicin and irinotecan, can be delivered by porphyrin–phospholipid liposomes to increase their local concentration in the tumour and improve their efficacy following light activation in preclinical animal models1921. This ‘chemophototherapy’ approach initially reduces blood flow in the tumour; this effect is followed by a transient increase in blood flow and blood flow stasis19,20,22. The acute drop in blood flow may be associated with immediate vascular permeability, followed by narrowing of the blood vessels and an increase in flow during thrombosis as well as subsequent vascular occlusion. The efficacy of chemophototherapy is influenced by the drug–light interval; shorter drug–light intervals of 0.5–1 h are typically more efficacious than longer drug–light intervals20,21 owing to the vascular presence of porphyrin–phospholipid liposomes early after administration. However, these effects may differ across cancer models20, and the native vascularization and vessel diameters may play a role in vascular permeabilization by PDT and PDP23. To fully take advantage of vascular permeabilization by porphyrin–phospholipid liposomes, a short drug–light interval can initially be applied to induce vascular permeability, followed by a longer drug–light interval to give porphyrin–phospholipid liposomes sufficient time to accumulate in tumours. This two-step treatment improves tumour growth inhibition and survival in a pancreatic cancer xenograft mouse model24.

Arginylglycylasparatic acid (RGD)–ferritin conjugates loaded with photosensitizers target the vasculature, improving vascular permeability and albumin extravasation, thereby augmenting the efficacy of liposomal doxorubicin in a mouse colon cancer model18. The vasculature can also be targeted with cationic liposomes25,26; for example, cationic liposomes delivering a chemotherapeutic may increase vascular permeability owing to their specific adhesion to neoangiogenic vessels, resulting in necrosis of the microvascular endothelium27. Combination of low-dose PDT (20 J cm−2) and doxorubicin using cationic doxorubicin-loaded porphyrin–phospholipid liposomes achieves synergistic inhibition of tumour growth in a MIA PaCa-2 mouse model22. Interestingly, the delivery of high doxorubicin payloads to tumour vascular endothelial cells does not reduce tumour growth, whereas treatment with cationic porphyrin–phospholipid liposomes devoid of doxorubicin is effective in reducing tumour growth in the MIA PaCa-2 mouse model, suggesting that vascular permeability is not the sole contributor to the enhanced doxorubicin efficacies observed after PDT. Therefore, non-ablative PDP modulation of tumour tissue, reduced stromal cell densities and extracellular matrix remodelling may also impact the efficacy of chemotherapy.

Overcoming drug delivery barriers

Low-dose PDT can improve the delivery and distribution of liposomal chemotherapies in colorectal tumours18 and lung tumours28 in rodents through vascular permeabilization. PDP modulation of vasculature is also relevant in combination therapies29,30. Here, the recruitment of leukocytes is essential to the endothelial permeabilizing effects of photochemistry31. PDP can also shift human brain microvascular endothelial cell junctions (vascular endothelial-cadherin and zonula occludens-1) from a continuous phenotype to a more punctate and perpendicular phenotype in vitro, resulting in increased endothelial permeability to nanoscale liposomes and macromolecules32. In addition to vascular modulation, PDP can decrease tumour interstitial fluid pressure in vivo33, soften the extracellular matrix and increase nanoparticle penetration in 3D co-culture models34, overcome tumour desmoplasia by modulating tumour collagen content and the extracellular matrix in vivo8, enable chemotherapy dose-reduction in mouse models35, and enhance the cytotoxicity of radiation therapy in 3D tumour nodules36. Improved permeability of the cancer microenvironment likely stems from direct oxidation of extracellular matrix proteins, such as collagen and fibronectin37. PDP can also reduce tumour collagen deposition and alignment in a pancreatic ductal adenocarcinoma mouse model9. Tumour collagen content and extracellular matrix density can further be reduced by PDP in mouse prostate tumour models, improving nanoparticle penetration8.

Modulation of multidrug resistance

Efforts to overcome cancer multidrug resistance using small-molecule inhibitors of drug efflux transporters have largely failed in the clinic owing to systemic toxicities. PDP can also inhibit ATP-binding cassette drug efflux transporters (such as, ATP-binding cassette sub-family G member 2 (ABCG2) and ATP-binding cassette sub-family B member 1 (ABCB1)); for example, using a verteporfin photosensitizer and a 690-nm laser, PDP can reduce ABCG2 expression in pancreatic cancer cells and increase irinotecan accumulation in tumours more than tenfold in a MIA PaCa-2 human pancreatic carcinoma orthotopic xenograft mouse model14. Verteporfin can attach to and induce photodamage in ABCG2 and ABCB138; for example, low light doses (690 nm, 0.05 J cm−2) and a nanomolar concentration of verteporfin is sufficient to inhibit the ATPase activity and induce protein crosslinking of ABCB1 and ABCG27.

Increasing tumour permeability without promoting migration

Increasing tumour permeability to chemotherapeutics by depleting or enzymatically degrading cancer stroma has not improved survival in patients with pancreatic cancer39,40. PDP can increase pancreatic tumour permeability to drugs, and surviving cancer cells may be less capable of escaping the primary tumour and form distant metastases. Of note, invasive pancreatic cancer cells with the potential for epithelial–mesenchymal transition exhibit higher sensitivity to PDT than cells with a distinct epithelial phenotype in the same 3D in vitro culture41. For example, porfimer sodium-PDP can suppress migration and inhibit the invasive process of glioblastoma cells in vitro42. Similarly, PDP using 5-aminolevulinic acid suppresses migration and invasion of head and neck cancer cells in vitro, along with inhibition of the focal adhesion kinase-Src-kinase-extracellular-signal-regulated kinase (FAK-Src-ERK) signalling pathway43. Furthermore, proteomic analyses of PDT-treated cancer cells revealed that integrins are a major target of photooxidation events44,45, which can prevent cellular interactions with the extracellular matrix and potentially explain the reduced migration potential of cancer cells following PDT4650. Epidermal growth factor receptor (EGFR)-targeted PDP combined with prostaglandin EP4 receptor inhibition attenuates cancer-promoting cell signalling and behaviours linked to metastasis in ovarian cancer cells in vitro51. PDP can also reduce the tumoural expression of CD44 and CXCR4 stemness markers in a pancreatic cancer mouse model, overcoming the development of resistance after multicycle chemotherapy6. Moreover, combining PDP with vitamin D receptor activation in fibroblasts suppresses pro-tumorigenic CXCL12/CXCR7 crosstalk and further improves chemotherapy efficacy in vivo35. Interestingly, low doses of PDT, which are sub-lethal to pancreatic tumour nodules in 3D homoculture, become lethal if the same cells are co-cultured with fibroblasts, suggesting that crosstalk with stromal fibroblasts may enhance tumour response to PDT52.

Therefore, not only can PDP increase the permeability of the tumour microenvironment, but it can also prevent cancer cells from interacting with the extracellular matrix, resulting in reduced migration, epithelial–mesenchymal transition and stemness37. Investigating the effect of PDP on the development of metastatic disease is thus crucial to provide information on the safety of PDP.

Photochemical release of endolysosomally sequestered agents

Beyond the initial ablative PDT zone, surviving cancer cells may experience PDP through damage of intracellular organelles, including the membrane of endolysosomes. Most water-soluble agents, proteins and nanoparticles, including therapeutic and immunologic agents, are internalized by cancer cells through endocytosis and are frequently sequestered in the endolysosomes. Photochemical internalization relies on a photodynamic trigger to enable the endolysosomal escape of cancer therapeutic agents53, including chemotherapeutics, such as bleomycin (NCT00993512) and gemcitabine (NCT04099888), and antigens for vaccine delivery (NCT02947854), by confining photosensitizers to the endolysosomal membrane. Typically, lysosome-specific amphiphilic photosensitizers, such as fimaporfin, are used for photochemical internalization; however, hydrophobic photosensitizers may also show lysosomal localization. Therefore, photochemical internalization may be a common mechanism by which cancer cells are sensitized to exogenous adjuvant agents. Photochemical internalization is thus a type of PDP enabling the priming of tumour cells that are located beyond the direct ablative zone to respond to agents sequestered and inactivated in the endolysosomes.

Clinical implementation

PDT is clinically used for the treatment of prostate cancer, brain cancer, bladder cancer and non-cancer applications, such as actinic keratosis and macular degeneration3. In addition, PDP effects on tumour tissue beyond the PDT ablative zones can be utilized to augment the effects of standard treatment modalities. Computed tomography (CT) is routinely used to detect PDT-induced necrosis in cancer tissues, and CT texture analysis can be applied to identify PDP effects54. For example, early-phase clinical studies showed that interstitial endoscopic ultrasound-guided PDT using Visudyne55 or porfimer sodium56 can induce necrosis up to 15.7 ± 5.5 mm in diameter and 18 ± 22% necrosis, respectively, in unresectable pancreatic tumours. Beyond this zone of necrosis, a range of PDP effects may be induced.

To take advantage of the effects of PDP in clinical practice, PDP may be leveraged to eradicate residual disease and reduce recurrence by enhancing anti-tumour immunity. The enhancement of the immune response following PDT or PDP can go beyond the local cytotoxic zone, leading to sustained immune-mediated surveillance and suppression of neoplastic cell growth in other parts of the body2,57,58. For example, PDP using a triple-receptor-targeted formulation can enhance immune responses by upregulating the membrane exposure and through release of damage-associated molecular patterns, inducing T cell activation in a 3D tumour model10. The mechanisms of PDP-induced anti-tumour immune enhancement remain to be fully investigated; however, PDP may be a promising addition to immunotherapies.

PDP can also be applied to improve the delivery and selectivity of chemotherapy to tumours. For example, low-dose PDT can increase the enhanced permeability and retention effect in breast tumour59 and pancreatic cancer6 mouse models, thereby increasing the efficacies of adjuvant chemotherapy. However, the impact of PDP depends on the light dose60 and drug–light interval20. Nanoscale liposomes allow the co-delivery of both photosensitizers and adjuvant therapeutics within a single formulation9,61,62, and they can be further engineered to release their content only in light-exposed areas, thereby reducing off-target drug toxicity63,64. Therefore, cellular priming, stroma remodelling and immunomodulatory effects of PDP may be utilized to improve cancer treatment.

Molecular engineering of photosensitizers

Pharmacokinetic and pharmacodynamic behaviour

The pharmacokinetic and pharmacodynamic profiles of photosensitizers can be improved by increasing the water solubility of hydrophobic and amphiphilic photosensitizers or by reducing the clearance rate of hydrophilic photosensitizers (Table 1). Most photosensitizers are hydrophobic or amphiphilic to enable plasma membrane penetrance and intracellular photodamage, and thus require solubilization. These photosensitizers are typically delivered using a nanoscale construct (also called a photonanomedicine); alternatively, photosensitizing nanoparticles can be applied65. For example, Visudyne is a liposomal formulation of verteporfin4. In addition, macromolecules can be conjugated to photosensitizers to impart molecular specificity17,66.

Table 1 |.

Examples of photosensitizer carriers and enhancers

Chemistry Degradation mechanisms Release Application Advantages Disadvantages Refs.
Photoimmunoconjugates and targeted conjugates
Photoimmuno-conjugates with stochastically conjugated photosensitizers Intracellular reduction, disassembly and proteolysis Photosensitizers are released after proteolysis Approved: photoimmunotherapy of head and neck cancer (Ministry of Health, Labour and Welfare in Japan)
Clinical trials: phase III clinical trial of photoimmunotherapy of head and neck cancer (NCT03769506)
Pre-clinical: photoimmunotherapy of solid and disseminated tumours (for example, head and neck cancer, ovarian cancer, oral pre-cancers, myosarcoma and mesothelioma)
Specificity improves efficacy and makes high, curative doses safe
Long circulation times
Receptor heterogeneity can result in incomplete responses
Inhomogeneous tumour distribution owing to binding site barrier effect
Photosensitizer-to-antibody ratio limited by reactivity in antigen-binding regions
17,75,195,196
Photoimmuno-conjugates with site-specifically conjugated photosensitizers Intracellular reduction, disassembly and proteolysis Photosensitizers are released after proteolysis and/or as a result of the reduction of thioether linkers Pre-clinical:
Photoimmunotherapy of solid and disseminated tumours
If a polyvalent carrier is used, the photosensitizer-to-antibody ratio can exceed the stochastic labelling method
No impedance of antigen binding
If no polyvalent carrier is used, the photosensitizer-to-antibody ratio is limited by the number of site-specific modification sites 197,198
Nanobodies Intracellular reduction, disassembly and proteolysis Photosensitizers are released after proteolysis Pre-clinical: Nanobody-targeted photodynamic therapy of resistant tumours, triggering immune responses Efficient tumour penetration and homogenous distribution
Rapid clearance from healthy tissue minimizes drug-to-light interval
Rapid clearance may limit bulk accumulation in solid tumours 199,200
Lipid and polymer nanoformulations
Liposomes Light-triggered degradation, disassembly in endolysosomes or tumour parenchyma Passive photosensitizer transfer to lipophilic molecules in serum or to target cell membrane Approved: PDT for age-related macular degeneration (Visudyne; FDA)
Clinical trials: PDT and non-PDT (dark effects) using Visudyne for solid tumours, including glioblastoma (NCT04590664, phase I/II), pancreatic cancer (NCT03033225, phase II) and prostate cancer (NCT03033225, phase I/II)
Pre-clinical: PDT-based combinations (for example, chemotherapy, immunotherapy, targeted inhibitors and biologics)
Improves solubility, photoactivity, PK/PD and tumour selectivity of hydrophobic, amphiphilic and hydrophilic photosensitizers
Light-triggered drug release
Enables multiagent co-delivery and potentiates efficacy
Premature photosensitizer leakage and off-target accumulation
Can exhibit instability in serum
Short circulation half-lives without polyethylene glycol coating
61,67,201,202
Porphysomes Light-triggered degradation, disassembly in endolysosomes or tumour parenchyma Encapsulated agents (if included) are released from the carrier upon light activation Pre-clinical: photothermal therapy, PDT, light-triggered combination therapies, photoacoustic and multimodal imaging No photosensitizer leakage owing to efficient self-assembly
Simplified system, in which the carrier also serves as the photosensitizer
Improved photosensitizer pharmacokinetics with no requirement for a nanoscale vehicle
Light-triggered drug release
Multiagent co-delivery and potentiated efficacy
PDT efficacy is contingent on partial or full dissociation of the construct. Otherwise photothermal effects dominate 63,203205
Liposomes with lipid-anchored photosensitizers Light-triggered degradation, disassembly in endolysosomes or tumour parenchyma Encapsulated agents are released from the carrier upon light activation Pre-clinical: PDT, light-triggered combination therapies and lysosome-targeted photodamage PDT efficacy not contingent on carrier dissociation
No photosensitizer leakage owing to stable lipid anchoring into liposomes
Improved PK/PD of photosensitizer and encapsulated agents
Light-triggered drug release of large and small therapeutic agents
Photodamage is restricted to lysosomes, so treatment is less cytotoxic than mitochondrial photodamage 9,19,67,82,202,206,207
Micelles Hydrolysis Encapsulated agents are released from the carrier upon light activation Pre-clinical: PDT-based combinations and image-guided PDT Small size favours tumour penetration
High loading capacity for hydrophobic photosensitizers compared to liposomes
Short circulation half-lives
Low serum stability
Loading restricted to hydrophobic photosensitizers
208212
AIE photosensitizer formulations (Contingent on formulation) Formulations release only the encapsulated agent while maintaining the photosensitizer in an aggregated, active state Pre-clinical: PDT-based combinations Formulations maintain the aggregated state of AIE photosensitizers, minimize molecular rotations and maintain their phototoxicity
Degree of AIE photosensitizer loading provides control over the degree of phototoxicity
Destabilization may lead to de-aggregation and inactivation of AIE photosensitizers 96,97,213,214
Nanoscale metal–organic frameworks
Hafnium – photosensitizer frameworks Low-pH-induced disassembly Secondary drug release triggered by photoactivation and/or low pH in tumour microenvironment and endolysosomes Pre-clinical: PDT-based combinations Improved photosensitizer PK/PD with no requirement for a nanoscale vehicle Photochemistry limited by static quenching of tightly packed photosensitizers
Low stability
Short circulation times
73,215
Iron – photosensitizer frameworks Low-pH-induced disassembly Secondary drug release triggered by photoactivation and/or low pH in tumour microenvironment and endolysosomes Pre-clinical: PDT-based combinations (for example, chemotherapy and immunotherapy) Hypoxia activity
Improved photosensitizer PK/PD with no requirement for a nanoscale vehicle
Photochemistry limited by static quenching of tightly packed photosensitizers
Low stability
Short circulation times
216,217
Copper – photosensitizer frameworks Low-pH-induced disassembly Secondary drug release triggered by photoactivation, redox chemistry and/or low pH in tumour microenvironment and endolysosomes Pre-clinical: PDT-based combinations (for example, chemotherapy and immunotherapy) Enhanced PDT efficacy through glutathione depletion
Improved photosensitizer PK/PD with no requirement for a nanoscale vehicle
Photochemistry limited by static quenching of tightly packed photosensitizers
Low stability
Short circulation times
218,219

AIE, aggregation-induced emission; FDA, US Food and Drug Administration; PDT, photodynamic therapy; PK/PD, pharmacokinetic/pharmacodynamic.

Hydrophobic photosensitizers typically partition within lipophilic regions of a lipid-based delivery vehicle; therefore, the pharmacokinetic and pharmacodynamic profiles of the vehicle should be considered, for example, for porphyrin–phospholipid liposomes delivering doxorubicin20. Prior to photoactivation, these doxorubicin-loaded porphyrin–phospholipid liposomes exhibit almost identical pharmacokinetic and pharmacodynamic profiles to the FDA-approved doxorubicin liposomal formulation19. Importantly, the hydrophobic porphyrin–phospholipid photosensitizer remains stably entrapped during circulation. Pharmacokinetic and pharmacodynamic modelling also revealed that phototriggered release of doxorubicin in the vasculature is responsible for the marked improvement in anti-tumour efficacy20. Polyethylene glycol coating of liposomes can further prevent leakage of unmodified benzoporphyrin derivative (BPD) photosensitizer molecules into serum; however, lipid anchoring of BPD to liposomes is needed to prevent non-specific switching and transfer of the photosensitizer to cancer cell membranes67. Interestingly, lipid anchoring of BPD to liposomes re-routes the photosensitizer to lysosomes from the mitochondria and endoplasmic reticulum68. Combined with liposomes containing native BPD, multiorganelle-targeted PDT potentiates apoptosis in 3D cancer models67.

Self-assembled nanovesicles comprised of porphyrin–phospholipid liposomes63 are potent photothermal and photoacoustic contrast agents in their intact quenched state; however, their disassembly can be triggered by cancer receptor targeting, which causes de-quenching of the porphyrin–phospholipids, thereby enabling PDT and fluorescence imaging69. In addition, various metals can be chelated within porphysomes to allow imaging, such as magnetic resonance imaging (MRI)70 and positron-emission tomography imaging71. Similarly, photosensitive metal–organic frameworks can be applied that combine PDT, chemotherapy and immune checkpoint blockade72. Unifying the pharmacokinetic and pharmacodynamic profiles of multiple treatment regimens would allow further control of combination therapies and simplify treatment protocols9,20,61,62,73.

The membrane configuration of lipid-anchored photosensitizers (membrane protruding versus membrane inserting) also has a key role in these photosensitizers’ pharmacokinetics, pharmacodynamics, photochemistry and therapeutic efficacy; for example, a membrane-protruding lipid conjugate of the photosensitizer IRDye700DX reduces tumour delivery of liposomes 7.2-fold as compared with a membrane-inserting lipid conjugate of the photosensitizer verteporfin74. In addition, the phototherapeutic efficacy of the membrane-protruding configuration is decreased tenfold, despite notably higher ROS production. Therefore, the surface configuration of photosensitizers tethered to liposomes is a crucial and often overlooked parameter in modulating the pharmacokinetics and pharmacodynamics of photosensitizers.

Molecular specificity

Molecular specificity of photosensitizers can be achieved by tethering photosensitizers to macromolecular targeting moieties that exhibit affinities towards tumour-associated receptors and biomolecules (Table 1). Molecular specificity provides tumour-targeted photodamage, thereby minimizing healthy tissue toxicity and allowing the safe use of escalated doses of PDT for a complete curative regimen5. Antibodies and engineered antibody formats are often applied as macromolecular photosensitizer conjugates, an approach referred to as antibody-targeted PDT or photoimmunotherapy17. For example, photoimmunotherapy using cetuximab saratolacan sodium5 has been approved for head and neck cancer in Japan, and it is currently in phase III clinical trials in the US (NCT03769506).

Targeting moieties, such as antibodies, antibody fragments and nanobodies, can be tethered either directly to photosensitizers through covalent conjugation7578 or to photosensitizer-carrying macromolecular structures, including dendrons79, liposomes8082 and polymeric nanoparticles83, amongst others84,85. Tethering antibody–photosensitizer conjugates to liposomes carrying chemotherapeutics62,86 or to polymeric nanoparticles87 further improves tumour-selective photosensitizer uptake and anti-tumour responses in vivo. In addition, a virus-like particle–phthalocyanine photosensitizer conjugate (AU-011) can target cell-surface heparan sulfate proteoglycans for PDT of uveal melanoma (NCT03052127)88. Molecular specificity, and subsequent cancer cell internalization, of targeted photosensitizer constructs may also be important for improving anti-tumour efficacy with respect to bulk delivery to the tumour interstitium, even if bulk delivery is selective89.

Stimuli responsiveness and image guidance

Stimuli-responsive platforms enable spatiotemporal control over treatment induction, for example, by controlling the release of drugs90, such as chemotherapeutics19, small-molecule inhibitors61 and immune checkpoint inhibitors91, by light (Table 1). Such phototriggered treatment induction typically improves anti-tumour responses compared with cocktails of PDT and secondary or tertiary therapeutics9,61. Photochemical internalization also utilizes photodynamics to spatiotemporally control the intracellular release of chemotherapeutics from endolysosomes by light. For example, this approach can be applied for the endosomal escape of lipid nanoparticles carrying single-guide RNA for CRISPR–Cas9 gene editing92, RNAi to inhibit angiogenesis93, and small interfering RNA (siRNA), such as in the clinically approved siRNA-carrying lipid nanoparticle patisiran94. Endosomal escape of these therapeutic payloads enables them to reach their intracellular targets, which can in turn complement the therapeutic efficacy of PDT that is applied for endosomal escape.

Instead of using macromolecules to solubilize hydrophobic and amphiphilic photosensitizers, aggregation-induced emission (AIE) photosensitizers can be applied; in this case, phototoxicity is contingent on aggregation, which restricts intramolecular motions95. A number of macromolecular structures can preserve the stacking and aggregation of AIE photosensitizers, thereby maximizing phototoxicity. For example, nanoscale lipid and polymer carriers improve the efficacy of AIE photosensitizers96. In addition, site-directed aggregation and phototoxicity can be engineered in mitochondria-directed AIE photosensitizers conjugated with pyridinium moieties97. Once the conjugates localize and aggregate within the mitochondria, they become phototoxic in vitro and in vivo. Similarly, AIE photosensitizers directed to intracellular lipid droplets, the plasma membrane or the endoplasmic reticulum can exhibit site-specific aggregation and activation of photochemical properties98.

Models for optimizing photodynamic therapy

Engineered models are key to preclinically evaluate PDT approaches. For example, 2D cell culture models are valuable for studying treatment effects; however, they lack translational power. By contrast, in vivo models have higher translational value, partly recapitulating the complexity of human cancer tissues within a living organism. However, to minimize the use of animals in research and better recapitulate human conditions, compared with 2D culture models99, 3D human cell culture models can be engineered that mimic the complexity of cancer tissues and offer the investigational accessibility and throughput of in vitro models100102 (Fig. 3). Three-dimensional culture models can be non-adherent spheroids, composed of cancer cells and supplemented with stromal cells; matrix-adherent or matrix-embedded microtumours, composed of established cancer and stroma cell lines; or matrix-adherent and matrix-embedded cancer organoids, which are obtained from dissociated surgically resected tumour tissues.

Imaging-based assays

The heterogeneity of models and the absence of functional assays are major challenges in the utilization of 3D cancer models for evaluating treatment effects103,104. Predominantly measured by probing cellular metabolic activity, treatment effects can also materialize by changes in tumour size, tumour growth rates, viability and redox state, among other factors. Imaging-based assays, which are capable of multiparametric analysis of hundreds to thousands of cancer organoids, have been developed to embrace the heterogeneity of these models as a strength, rather than as a weakness. For example, quantitatively analysing images obtained by darkfield microscopy enables the identification of distinct alterations in the size distribution of ovarian cancer organoids following carboplatin chemotherapy and PDT105. This image analysis procedure can also be combined with a LIVE/DEAD cell staining procedure to extract cancer organoid volumes and viability indices for each individual organoid imaged by low-magnification microscopy100. This protocol can be adapted to asymmetrical cancer organoid cultures, providing the readouts in area rather than in volume101.

Quantitative imaging approaches can thus be implemented to identify specific treatment responses in 3D cell cultures. For example, high-throughput image analysis revealed that chemotherapeutics reduce the viability of architecturally complex heterotypic organoids, that PDT decreases the relative live areas or volumes of microtumours by causing peripheral necrosis, and that radiotherapy inhibits cancer organoid growth without affecting viability101. Image analysis can also inform on the uptake and localization of photosensitizers106,107 as well as on perturbations in redox state using NAD(P)H and FAD autofluorescence108111. Moreover, imaging approaches can be adapted for any biomarker expression analysis by immunofluorescence.

Spheroid and microtumour models

Spheroid cancer models are cell aggregates cultured on U-bottom multiplate wells with a non-adherent coating. Spheroids can be composed of cancer cell lines and may be supplemented with endothelial cells, fibroblasts or immune cells to more closely mimic the cancer microenvironment. As spheroid models are inexpensive and homogeneous, they constitute the most widely used 3D models in PDT research. For example, spheroids composed of both cancer and stroma cells have been used to demonstrate that antibody-functionalized liposomes can facilitate selective PDT of cancer cells82, that PDT–radiotherapy combination therapy results in smaller and more necrotic pancreatic cancer spheroids than each individual therapy36, and that such spheroids are useful to evaluate the uptake and penetration depth of various nanocarriers for photosensitizers112. However, not all cell types can grow in non-adherent environments. In addition, the formation of non-spherical concave structures rather than true spheroids may not represent cancer tissues as accurately as intended.

Alternatively, cancer cells can be grown in extracellular matrix scaffolds to form spherical microtumours at the gel–medium interface, thereby simulating the mechanobiological environment of a tumour. For example, ovarian microtumours have been used to demonstrate synergistic, yet sequence-dependent, enhancement of carboplatin chemotherapy by PDT113. Similarly, short-term and long-term effects of oxaliplatin–PDT combinations have been studied in pancreatic microtumours114. More complexity can be added by introducing multiple cell types; for example, in pancreatic microtumours grown on extracellular matrix hydrogels, the addition of cancer-associated fibroblasts substantially alters the morphology and redox state of microtumours as well as their susceptibility to oxaliplatin and PDT108. However, a limitation of both spheroid and hydrogel-based models is the lack of fluid flow, which can be introduced through microfluidics. Under fluid flow, human ovarian cancer microtumours undergo epithelial-to-mesenchymal transition115, which induces carboplatin resistance but improves the cellular delivery of photosensitizers formulations and increases photoimmunotherapy efficacy116,117.

Patient-derived organoids

Cells can be extracted from patient biopsies and grown into cancer organoids to identify the most effective treatment regimens for a specific patient118. This requires the dissociation of tissues into single-cell suspensions, which can then be grown on or in extracellular matrix scaffolds118. For example, patient-derived head and neck cancer organoids were utilized to examine the selectivity of immunotargeted PDT by comparing phototoxicity in the cancer organoids with phototoxicity in healthy organoids developed from nearby normal tissue tissue from the same patients119. Similarly, patient-derived cholangiocarcinoma organoids demonstrated higher protoporphyrin IX biosynthesis following 5-aminolevulinic acid treatment than non-cancerous intrahepatic and extrahepatic bile duct organoids derived from the same patients120. Therefore, establishing selectivity of photosensitization approaches in patient-derived organoids may inform the suitability of patients to undergo targeted PDT or fluorescence-guided surgery.

Taken together, evidence from studies indicates that 3D cancer models recapitulate PDT dose responses better than 2D cell cultures and that the responses of the former are similar to the in vivo responses. Importantly, rather than information-poor biochemical assays, imaging-based assays can be applied to 3D culture models to track tumour growth and treatment-induced shrinkage, which is similar to how ultrasound, CT and positron-emission tomography are used to evaluate treatment responses in patients. Further utilization of 3D cancer models will greatly benefit from rapid optical imaging systems, optical clearing methods121 combined with adaptive optics122 or light-sheet microscopy123, and artificial intelligence-based image analysis124.

Photonic devices and image-guided strategies

Light sources

The light threshold to induce PDT phototoxicity in tissues and cells is in the range of 109–1010 photons absorbed per cell125127. Although the delivered radiant energy can be as low as 1–10 J cm−2 in vitro, depending on the photosensitizer and its concentration, in vivo, macroscopic PDT typically requires more than 50 J cm−2 for bulk tissue damage128,129. PDP is elicited at fluences that are lower than these values6,54,82. Large class IV laser systems were originally used as light sources for experimental PDT, whereas most high-power needs are now supplied by diode laser modules or light-emitting diode (LED) lamps130. In particular, if focal light delivery to isolated locations is required, multichannel diode lasers can be delivered by fibres, for example, for intraocular, interstitial, intraoperative intraluminal and diffuse illumination131133. These fibres are terminated by specialized diffusers or applicators that are specific to the tissue or organ134136. Interstitial and superficial light delivery enable bulk tumour irradiation, in particular for inner organ cancer therapy. Clinical light delivery systems have also been developed for different cancers (such as, peritoneal metastasis and brain cancer) and for online treatment monitoring and analytics62,137139. In ophthalmology, light delivery systems are also cloud-connected and tablet-controlled to improve usability and clinical adoption140. External dermatological PDT is typically performed with LED-based lamps or LED-based blankets that cover the tissue141144. Deep-tissue PDT can be achieved with LED implants145147 or by chemiluminescence, bioluminescence, X-ray, ultrasound or Cerenkov radiation.

Image guidance

Image guidance in PDT has mainly focused on light delivery verification and localization to confirm that a sufficient amount of light has been delivered. However, geometric guidance can also be achieved by CT148, ultrasound149, MRI150,151 and surface and cavity capture152154. Moreover, photosensitizers can be indirectly guided through quantifying fluorescence pre-treatment62 or photobleaching to assess absorbance during treatment62,155. In addition, 1O2 guidance can be applied to quantify photochemical delivery during treatment156,157. Although these image guidance approaches require additional instrumentation or procedures, they can ensure that there is no under-treatment or over-treatment. Clinical light delivery systems typically integrate treatment monitoring functions based on tissue absorption or fluorescence to inform dosimetry and report on efficacy62,137139. In particular, over-treatment can have devastating effects in specific anatomical locations, such as oesophageal structures158,159 and the bladder. Under-treatment may often occur in PDT, requiring repeated and customized treatments for full efficacy62. Of note, under-treatment essentially is the core concept of PDP, which subtly causes alterations in permeability and tissue response.

Surrogate and minimal approaches to dosimetry

Experimental PDT typically takes an extreme dosimetry approach; here, a number of parameters (such as, photosensitizer delivery, photobleaching, tissue optical properties and 1O2 levels) are measured to understand the response and determine dominant factors in treatment control. However, clinical PDT aims for delivery with low dosimetry to avoid over-treatment. Clinical light delivery systems are typically minimally invasive single-probe systems that provide treatment monitoring for usability, repeatability and efficacy. Surrogate endpoints, as typically used in pre-CT and post-CT scans with contrast or pre-skin and post-skin measurements of photosensitizers, can be useful to quantify the delivery and efficacy of PDT and PDP54. In addition, laser, optical imaging and photonanomedicines may improve treatment planning, dosimetry and treatment response monitoring.

Alternative activation sources

Ultrasound

Ultrasound as a diagnostic tool benefits from deep tissue penetration (for example, low frequency ≈1 MHz pressure pulses can reach up to a depth of 10 cm) to probe deep-seated tumours. In particular, sonodynamic therapy relies on low-frequency ultrasound to activate an acoustically sensitive agent, that is, a sonosensitizer or photosensitizer. However, the mechanisms underlying sonodynamic therapy remain to be fully unravelled. In sonodynamic therapy, ultrasound itself can induce a therapeutic effect. Although typically performed with minimally hyperthermic ultrasound doses, sonomechanical tissue damage can occur160. However, sonodynamic therapy primarily relies on mechanisms that generate ROS,- in particular, 1O2. Two mechanisms have been proposed to explain the origin of ROS production in sonodynamic therapy: sonoluminescence and pyrolysis. Sonoluminescence is an emission of photons generated during cavitation, peaking around 400–450 nm, a range that overlaps well with the absorption peak of commonly used photosensitizers. Sonoluminescence may excite photosensitizers to induce PDT in tumour regions that may be typically inaccessible to direct light activation. However, although an overlap between the sonoluminescence spectrum and the absorption of porphyrin sonosensitizers has been reported, photoinactive compounds can also produce 1O2 upon ultrasound excitation161. By contrast, pyrolysis may be triggered by inertial cavitation, which causes high local temperatures and pressures. Although pyrolysis is independent of the presence of a sonosensitizer, the proximity of a drug may amplify the efficacy of pyrolysis observed with ultrasound alone162,163. Therefore, sonodynamic therapy may be able to induce a therapeutic effect even in tumours localized at a depth of 10 cm within tissue; however, the underlying mechanisms remain to be fully investigated.

X-rays

X-rays are commonly used in radiology and radiotherapy for diagnosis and treatment, respectively. X-rays readily penetrate soft tissue and may thus provide a promising tool to activate PDT regardless of tumour depth164166. In particular, radiodynamic therapy relies on the direct X-ray excitation of a photosensitizer, which in this context is referred to as radiosensitizer. Alternatively, indirect excitation of photosensitizers can be achieved using nanotransducers that locally down-convert high-energy X-rays into light to excite nearby photosensitizers in an approach referred to as XPDT.

Radiodynamic therapy.

Several photosensitizers can be excited by X-rays to produce ROS, including 1O2167. X-rays may excite photosensitizers through direct electronic excitation or through indirect excitation by what is speculated to be Cerenkov radiation. Cerenkov radiation causes optical emission if charged particles, such as electrons or positrons, travel faster than the phase velocity of light in a given dielectric medium. The spectrum of Cerenkov radiation is mainly in the ultraviolet–visible range, which overlaps with the strong absorption (Soret) bands of commonly used photosensitizers. During X-ray radiotherapy, secondary electrons are created by the interaction of ionizing photons with tissue. These electrons can generate Cerenkov emission if their energy is higher than approximately 250 keV168,169. Cerenkov radiation can be measured in tissues that are exposed to a 6–18 MeV photon or electron irradiation, causing the excitation of protoporphyrin IX embedded in biological phantom structures168. For example, 5-aminolevulinic acid, the precursor of endogenous protoporphyrin IX, shows radiosensitizing properties upon 15 MV photon irradiation in a subcutaneous model of lung cancer (KP1 cells implanted in C57BL/6 mice)170. However, although protoporphyrin IX may produce ROS, including 1O2, upon X-ray irradiation below 250 keV167,171173, such energies are insufficient to induce Cerenkov radiation, emphasizing the fact that Cerenkov radiation cannot be the only mechanism at play here. 5-Aminolevulinic acid has also been shown to possess radiosensitizing properties in colorectal cancer models in vitro and in vivo, using an X-ray generator operating at 150 kVp, which is also insufficient to trigger Cerenkov emission174. However, other photosensitizers, such as a haematoporphyrin derivative, have been reported to be directly excited by X-rays; it nevertheless remains unclear what this depends on, except for the concentration of the photosensitizer167.

Therefore, the role of Cerenkov radiation in the radiodynamic process and the radiosensitizing properties of specific photosensitizers remain to be fully understood. Because of the origin of the Cerenkov radiation and the associated energy threshold, the role of X-ray energy in radiotherapy should be reported and investigated. Nevertheless, radiodynamic therapy has clinical potential, as illustrated by a clinical trial for first-time relapse of malignant glioma (NCT05590689). This phase I/II clinical trial was designed to investigate the tolerance of repeated administration of 5-aminolevulinic acid, usually applied as a single dose, in combination with radiotherapy. In addition, the efficacy of additional 5-aminolevulinic acid administration is being investigated.

Nanoscintillators.

XPDT relies on nanotransducers called nanoscintillators. These nanoparticles locally down-convert X-rays into radioluminescence, which can excite nearby photosensitizers164,175. Nanoscintillators are usually made of insulating materials doped with rare-earth elements. Various nanoscintillators have been preclinically investigated165,166 at doses of radiation (≈5 Gy) that are lower than common radiotherapy doses (up to 80 Gy), applying computer simulations to estimate the maximal radioluminescence-induced PDT effect176,177. Interestingly, the radioluminescence emitted by nanoscintillators upon radiotherapy is orders of magnitude lower than the light dose required to induce such an effect. Therefore, the efficacy of XPDT cannot be solely explained by radioluminescence, and other parameters are likely to be at play. In particular, the existence of non-resonant Förster energy transfer between a nanoscintillator and photosensitizer may enhance treatment efficacy178. In addition, radiotherapy and PDT have different cellular targets, activating different pathways, and may thus synergize to improve outcomes36. Another (synergistic) contribution comes from the heavy elements present in nanoscintillators, which can locally enhance the radiation dose owing to the energy deposited by Auger and photoelectrons emitted during photoelectric interactions between X-rays and nanoscintillators179. Moreover, Cerenkov emission may be considered if radiotherapy is delivered by high-energy X-rays or particle beams.

The design of XPDT protocols should consider the energy of X-rays. Unlike Cerenkov radiation, which is maximized at high-energy X-rays, the absorption cross-section of nanoscintillators is maximized for X-ray energies below 250 keV, for which the photoelectric effect dominates176,177. Preclinical studies are typically performed using small animal irradiators, which operate in the keV energy range; by contrast, clinical irradiators operate in the MeV range. If the efficacy drops at high energy, strategies such as brachytherapy may have to be considered in the clinic. Furthermore, the bioavailability and toxicity of nanotransducers containing heavy elements need to be considered. Encouragingly, Gd-based and Hf-based nanoparticles were shown to be safe in in phase I clinical trials180,181.

Self-illuminating compounds

Bioluminescence originates from the oxidation of luciferins (for example, D-luciferin and coelenterazine)182 catalysed by luciferase enzymes. Bioluminescence may be able to locally excite photosensitizers to induce PDT, regardless of tumour location. However, the intensity of bioluminescence is orders of magnitude lower than the intensity of the light applied to activate PDT183. Therefore, bioluminescence-induced PDT likely originates from a combination of radiative and non-radiative energy transfer. A bioluminescence energy of around 100 μJ may induce a photochemical effect superior to a laser delivering an energy of ~100 mJ183. Bioluminescence-induced PDT has been shown in vitro in human glioblastoma cells (U87-MG)184, mouse Lewis lung cancer (LCC) cells185, mouse melanoma cells (B16F10)183 and mouse colon cancer cells (CT26)183, as well as in vivo for LCC, B16F10 and CT26 tumours183. However, cancer cell-specific expression of luciferase will need to be demonstrated for clinical translation, as shown for intratumourally administered D-luciferase conjugated to QD-655 quantum dots183. Intratumoural photosensitizer delivery by minimally invasive techniques may increase PDT efficacy186, in particular if combined with implantable biomaterials and self-illuminating light sources.

Radionuclides can also excite photosensitizers regardless of the tumour depth and without the need for an external stimulus169,185,187,188. Although not yet fully elucidated189,190, the synergy of radionuclides injected together with a photosensitizer may initiate from the activation of the photosensitizer by Cerenkov radiation, created by the electron or positron emitted during radioactive decays. Although this hypothesis is challenged by the dim intensity of Cerenkov radiation, the activity of radionuclides may correlate with the activation of meso-tetrakis(4-carboxyphenyl)porphyrin191, mesotetraphenylporphin192 and TiO2 nanoparticles185,187, acting as photosensitizers. Here, radioactive elements were applied that are commonly used as radiotracers in nuclear imaging.

In addition to investigating the mechanisms of action leading to photosensitizer activation (Cerenkov radiation or other mechanisms), the role of the specific emitters (electron, positron, gamma), the energies of emitted particles, and the half-life of the radioactive elements remain to be fully investigated. For example, 68Ga was found to be more efficient than 18F at producing ROS after activating TiO2 nanoparticles both in vitro and in vivo in mouse breast cancer cells187. In addition, it has been theoretically shown that the lifetime of the emitter is a critical parameter to consider when selecting the radionuclide: the longer the lifetime, the more efficient the Cerenkov emitter is for PDT169. Regardless of the origin of the effect, radionuclides have been repeatedly shown to activate photosensitizers without an external stimulus.

Outlook

PDT and, in particular, photodynamics-based precision therapy will benefit from the development of new sensitizing agents, a thorough mechanistic understanding of photodynamic processes, and the integration and emergence of new nanotechnology and imaging tools, as well as microtumour models. Importantly, the low and nonoverlapping toxicity of PDT (when combined with chemotherapeutics) and the absence of adverse effects, such as fibrosis, make this approach promising for the treatment of diseases. However, many nanotechnology and image-guided strategies for PDT and PDP have not yet been implemented in the clinic, and may require further characterization and optimization in microtumours and in animal models.

In addition, controlled and robust clinical trials will be required to move more PDT approaches to the clinic. Typically, PDT is tested as a monotherapy if other approaches (such as chemotherapy and radiation) have failed or are not suitable, and it often results in tumour necrosis, reduction in tumour volume, and increased survival55,153,193. However, these promising results are often only observed in a small sample size (fewer than 100 participants). Larger controlled clinical trials of PDT should thus be guided and informed by an organized framework, equivalent to the Radiation Therapy Oncology Group, to clarify its clinical implementation, establish guidelines for dosimetry and treatment design, and allow for appropriate comparisons with other therapies.

Although the regulatory process of photodynamic approaches is complex, as it involves the regulation of both a photosensitizer drug and a laser device, several PDT approaches have already been approved for the clinic3. For example, photodynamic treatment became the first line of therapy for age-related macular degeneration, a leading cause of blindness in the elderly. Importantly, industry involvement is key to commercialization (Box 2).

PDT is particularly suitable for the treatment of deep-seated tumours, and it could also be applied to extend the treatment potential of other therapies. For example, immunotherapy is not applicable to all cancer types or all patients, owing to dose-limiting immune-related toxicities and the inability of immune cells to reach tumours, particularly stroma-rich tumours, such as in pancreatic cancer. PDT can transiently permeabilize tumours (stroma, cells and vasculature) to increase T cell trafficking to tumours and blood. In addition, optical systems and devices as well as artificial intelligence approaches in imaging may be utilized to identify immune cells and microenvironmental changes. The ability of PDT to modulate tumour and vascular permeability could further control patient-specific dosing and timing of checkpoint inhibitors and cell therapies, improving their therapeutic index. Effective dosimetry by incorporating radiomics features of tumours and imaging would enable precise patient-specific administration of PDT therapies. Therefore, PDT should not only be considered a local therapy but as part of an arsenal of treatment combinations.

Moreover, photodynamically active nanomaterials should be explored that can switch photodynamic processes on and off to offer quantitative control of photodynamic effects. In addition to delivering multiple therapeutic agents, whose delivery and efficacy are augmented by photodynamic action, control of delivery by nanomaterials will allow more precise dosimetry and spatial control to limit undesired tissue damage. Such an approach would remove uncertainties related to the different pharmacokinetics of drugs when used in combination. Combining nanomaterials with on-line hyperspectral imaging opens up possibilities of precision medicine, for example, by applying different levels of light to different parts of the body or tissue. Finally, PDT may also be applied beyond cancer; for example, for age-related macular degeneration (Visudyne)4, and infectious diseases, such as SARS-CoV-2194.

Key points.

  • Photodynamic therapy (PDT), including photodynamic priming, allows the localized destruction of diseased cells and tissues by light, benefitting from minimal side effects, minimally invasive administration and the potential for combination therapies.

  • Nanomaterials can be designed to enable controlled delivery of combination therapies, improve the pharmacokinetics of photosensitizers and enable optical imaging-guided PDT.

  • PDT can be applied for tumour ablation, modulation of the tumour environment and boosting of the immune responses against local and metastatic disease.

  • Robust clinical trials, industry involvement and collaboration between different research fields (such as, physics, mathematics, chemistry and engineering) are essential for the clinical translation of PDT-based therapies.

Acknowledgements

G.O. acknowledges support from the UT Dallas Jonsson School Research Initiative and the US National Institutes of Health (NIH) award (R00CA215301). J.P.C. acknowledges support from the US NIH (U54CA156734, UH3CA1889901 and U01CA279862). The European Union is acknowledged for the support of M.B. (ERC STG, RADIOCONTROL, 101078392) and A.-L.B. (ERC STG, RADIANCE, 101116304). Views and opinions expressed are however those of the authors only and do not necessarily reflect those of the European Union or the European Research Council Executive Agency. Neither the European Union nor the granting authority can be held responsible for them. T.H. acknowledges support from the US NIH (P01 CA084203, R01CA231606, R01CA266855, UH3CA189901, R44DE026083) and the US Air Force Office of Scientific Research (FA9550-20-1-0063). H.H. acknowledges support from the US NIH Trailblazer Award (NIBIB R21EB028508), US National Science Foundation (CBET-2030253, CBET-2037815) and the US NIH (R01CA260340, R01CA256710).

Footnotes

Competing interests

P.U. is a co-founder of and has equity in Modulight Corporation, Finland. H.H. has research agreements with Modulight Corporation, Finland. The remaining authors declare no competing interests.

References

  • 1.Dolmans DE, Fukumura D & Jain RK Photodynamic therapy for cancer. Nat. Rev. Cancer 3, 380–387 (2003). [DOI] [PubMed] [Google Scholar]
  • 2.Castano AP, Mroz P & Hamblin MR Photodynamic therapy and anti-tumour immunity. Nat. Rev. Cancer 6, 535–545 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Li X, Lovell JF, Yoon J & Chen X Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol 17, 657–674 (2020). [DOI] [PubMed] [Google Scholar]
  • 4.Verteporfin Roundtable Participants Guidelines for using verteporfin (Visudyne) in photodynamic therapy for choroidal neovascularization due to age-related macular degeneration and other causes: update. Retina 25, 119–134 (2005). [DOI] [PubMed] [Google Scholar]
  • 5.Mitsunaga M et al. Cancer cell-selective in vivo near infrared photoimmunotherapy targeting specific membrane molecules. Nat. Med 17, 1685–1691 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]; This article reports the conjugation of the phthalocyanine dye IR700 to monoclonal antibodies for photoimmunotherapy of epidermal growth factor receptor-expressing epidermoid carcinoma in vivo.
  • 6.Huang HC et al. Photodynamic priming mitigates chemotherapeutic selection pressures and improves drug delivery. Cancer Res 78, 558–571 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Liang B, Lusvarghi S, Ambudkar S & Huang H-C Mechanistic insights into photodynamic regulation of adenosine 5′-triphosphate-binding cassette drug transporters. ACS Pharmacol. Transl. Sci 4, 1578–1587 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Overchuk M et al. Subtherapeutic photodynamic treatment facilitates tumor nanomedicine delivery and overcomes desmoplasia. Nano Lett 21, 344–352 (2021). [DOI] [PubMed] [Google Scholar]; This article demonstrates photodynamic priming to alleviate tumour desmoplasia and enhance nanomedicine accumulation in prostate tumour-bearing mice.
  • 9.Obaid G et al. Remediating desmoplasia with EGFR-Targeted photoactivable multi-inhibitor liposomes doubles overall survival in pancreatic cancer. Adv. Sci 9, e2104594 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.De Silva P et al. Photodynamic priming with triple-receptor targeted nanoconjugates that trigger T cell-mediated immune responses in a 3D in vitro heterocellular model of pancreatic cancer. Nanophotonics 10, 3199–3214 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Luo D, Carter KA, Miranda D & Lovell JF Chemophototherapy: an emerging treatment option for solid tumors. Adv. Sci 4, 1600106 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kim HR, Luo Y, Li G & Kessel D Enhanced apoptotic response to photodynamic therapy after bcl-2 transfection. Cancer Res 59, 3429–3432 (1999). [PMC free article] [PubMed] [Google Scholar]
  • 13.Xue LY, Chiu SM, Fiebig A, Andrews DW & Oleinick NL Photodamage to multiple Bcl-xL isoforms by photodynamic therapy with the phthalocyanine photosensitizer Pc 4. Oncogene 22, 9197–9204 (2003). [DOI] [PubMed] [Google Scholar]
  • 14.Huang HC et al. Photodynamic therapy synergizes with irinotecan to overcome compensatory mechanisms and improve treatment outcomes in pancreatic cancer. Cancer Res 76, 1066–1077 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen B et al. Blood flow dynamics after photodynamic therapy with verteporfin in the RIF-1 tumor. Radiat. Res 160, 452–459 (2003). [DOI] [PubMed] [Google Scholar]
  • 16.Osaki T, Takagi S, Hoshino Y, Okumura M & Fujinaga T Antitumor effects and blood flow dynamics after photodynamic therapy using benzoporphyrin derivative monoacid ring A in KLN205 and LM8 mouse tumor models. Cancer Lett 248, 47–57 (2007). [DOI] [PubMed] [Google Scholar]
  • 17.Mew D, Wat CK, Towers GH & Levy JG Photoimmunotherapy: treatment of animal tumors with tumor-specific monoclonal antibody-hematoporphyrin conjugates. J. Immunol 130, 1473–1477 (1983). [PubMed] [Google Scholar]
  • 18.Snyder JW, Greco WR, Bellnier DA, Vaughan L & Henderson BW Photodynamic therapy: a means to enhanced drug delivery to tumors. Cancer Res 63, 8126–8131 (2003). [PubMed] [Google Scholar]
  • 19.Luo D et al. Doxorubicin encapsulated in stealth liposomes conferred with light-triggered drug release. Biomaterials 75, 193–202 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Luo D et al. Pharmacokinetics and pharmacodynamics of liposomal chemophototherapy with short drug-light intervals. J. Control. Release 297, 39–47 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Luo D, Carter KA, Geng J, He X & Lovell JF Short drug–light intervals improve liposomal chemophototherapy in mice bearing MIA PaCa-2 xenografts. Mol. Pharm 15, 3682–3689 (2018). [DOI] [PubMed] [Google Scholar]
  • 22.Luo D et al. Vessel-targeted chemophototherapy with cationic porphyrin-phospholipid liposomes. Mol. Cancer Ther 16, 2452–2461 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Araki T et al. Augmented EPR effect by photo-triggered tumor vascular treatment improved therapeutic efficacy of liposomal paclitaxel in mice bearing tumors with low permeable vasculature. J. Control. Release 200, 106–114 (2015). [DOI] [PubMed] [Google Scholar]
  • 24.Ghosh S & Lovell JF Two laser treatments can improve tumor ablation efficiency of chemophototherapy. Pharmaceutics 13, 2183 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Abu Lila AS, Ishida T & Kiwada H Targeting anticancer drugs to tumor vasculature using cationic liposomes. Pharm. Res 27, 1171–1183 (2010). [DOI] [PubMed] [Google Scholar]
  • 26.Campbell RB, Ying B, Kuesters GM & Hemphill R Fighting cancer: from the bench to bedside using second generation cationic liposomal therapeutics. J. Pharm. Sci 98, 411–429 (2009). [DOI] [PubMed] [Google Scholar]
  • 27.Strieth S et al. Paclitaxel encapsulated in cationic liposomes increases tumor microvessel leakiness and improves therapeutic efficacy in combination with Cisplatin. Clin. Cancer Res 14, 4603–4611 (2008). [DOI] [PubMed] [Google Scholar]
  • 28.Wang Y et al. Photodynamic induced uptake of liposomal doxorubicin to rat lung tumors parallels tumor vascular density. Lasers Surg. Med 44, 318–324 (2012). [DOI] [PubMed] [Google Scholar]
  • 29.Debefve E, Pegaz B, Ballini JP, Konan YN & van den Bergh H Combination therapy using aspirin-enhanced photodynamic selective drug delivery. Vasc. Pharmacol 46, 171–180 (2007). [DOI] [PubMed] [Google Scholar]
  • 30.Debefve E, Pegaz B, Ballini J-P & van den Bergh H Combination therapy using verteporfin and ranibizumab; optimizing the timing in the CAM Model. Photochem. Photobiol 85, 1400–1408 (2009). [DOI] [PubMed] [Google Scholar]
  • 31.Debefve E et al. Leukocyte–endothelial cell interaction is necessary for photodynamic therapy induced vascular permeabilization. Lasers Surg. Med 43, 696–704 (2011). [DOI] [PubMed] [Google Scholar]
  • 32.Inglut CT et al. Photodynamic priming modulates endothelial cell-cell junction phenotype for light-activated remote control of drug delivery. IEEE J. Sel. Top. Quantum Electron 27, 7200311 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Perentes JY et al. Low-dose vascular photodynamic therapy decreases tumor interstitial fluid pressure, which promotes liposomal doxorubicin distribution in a murine sarcoma metastasis model. Transl. Oncol 7, 393–399 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Karimnia V et al. Photodynamic stromal depletion enhances therapeutic nanoparticle delivery in 3D pancreatic ductal adenocarcinoma tumor models. Photochem. Photobiol 99, 120–131 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Anbil S et al. Vitamin D receptor activation and photodynamic priming enables durable low-dose chemotherapy. Mol. Cancer Ther 19, 1308–1319 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bulin A-L, Broekgaarden M, Simeone D & Hasan T Low dose photodynamic therapy harmonizes with radiation therapy to induce beneficial effects on pancreatic heterocellular spheroids. Oncotarget 10, 2625–2643 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Carigga Gutierrez NM et al. Increasing cancer permeability by photodynamic priming: from microenvironment to mechanotransduction signaling. Cancer Metastasis Rev 41, 899–934 (2022). [DOI] [PubMed] [Google Scholar]
  • 38.Baglo Y et al. Porphyrin-lipid assemblies and nanovesicles overcome ABC transporter-mediated photodynamic therapy resistance in cancer cells. Cancer Lett 457, 110–118 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Catenacci DV et al. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J. Clin. Oncol 33, 4284–4292 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Van Cutsem E et al. Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J. Clin. Oncol 38, 3185–3194 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cramer GM, Jones DP, El-Hamidi H & Celli JP ECM composition and rheology regulate growth, motility, and response to photodynamic therapy in 3D models of pancreatic ductal adenocarcinoma. Mol. Cancer Res 15, 15–25 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jiang F et al. Photodynamic therapy with photofrin reduces invasiveness of malignant human glioma cells. Lasers Med. Sci 17, 280–288 (2002). [DOI] [PubMed] [Google Scholar]
  • 43.Yang TH, Chen CT, Wang CP & Lou PJ Photodynamic therapy suppresses the migration and invasion of head and neck cancer cells in vitro. Oral Oncol 43, 358–365 (2007). [DOI] [PubMed] [Google Scholar]
  • 44.Tsaytler PA, O’Flaherty MC, Sakharov DV, Krijgsveld J & Egmond MR Immediate protein targets of photodynamic treatment in carcinoma cells. J. Proteome Res 7, 3868–3878 (2008). [DOI] [PubMed] [Google Scholar]
  • 45.Hsieh YJ 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 7, 1370 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mao W et al. A combined modality of carboplatin and photodynamic therapy suppresses epithelial-mesenchymal transition and matrix metalloproteinase-2 (MMP-2)/MMP-9 expression in HEp-2 human laryngeal cancer cells via ROS-mediated inhibition of MEK/ERK signalling pathway. Lasers Med. Sci 31, 1697–1705 (2016). [DOI] [PubMed] [Google Scholar]
  • 47.Ma H et al. Photodynamic effect of chlorin e6 on cytoskeleton protein of human colon cancer SW480 cells. Photodiagnosis Photodyn. Ther 33, 102201 (2021). [DOI] [PubMed] [Google Scholar]
  • 48.Liu LY, Man XX, Yao HX & Tan YY Effects of pheophorbide a-mediated photodynamic therapy on proliferation and metastasis of human prostate cancer cells. Eur. Rev. Med. Pharmacol. Sci 21, 5571–5579 (2017). [DOI] [PubMed] [Google Scholar]
  • 49.Calvo G et al. Reversal of the migratory and invasive phenotype of Ras-transfected mammary cells by photodynamic therapy treatment. J. Cell. Biochem 118, 464–477 (2017). [DOI] [PubMed] [Google Scholar]
  • 50.Ma C et al. Nanoparticle delivery of Wnt-1 siRNA enhances photodynamic therapy by inhibiting epithelial-mesenchymal transition for oral cancer. Biomater. Sci 5, 494–501 (2017). [DOI] [PubMed] [Google Scholar]
  • 51.Sorrin AJ et al. Photodynamic priming improves the anti-migratory activity of prostaglandin e receptor 4 antagonist in cancer cells in vitro. Cancers 13, 5259 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Karimnia V, Rizvi I, Slack FJ & Celli JP Photodestruction of stromal fibroblasts enhances tumor response to PDT in 3D pancreatic cancer coculture models. Photochem. Photobiol 97, 416–426 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Berg K et al. Photochemical internalization: a novel technology for delivery of macromolecules into cytosol. Cancer Res 59, 1180–1183 (1999). [PubMed] [Google Scholar]; This article reports the photochemical release of macromolecules from endosomes and lysosomes into the cytosol of cells.
  • 54.Vincent P et al. CT radiomic features of photodynamic priming in clinical pancreatic ductal adenocarcinoma treatment. Phys. Med. Biol 66, 175006 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hanada Y et al. EUS-guided verteporfin photodynamic therapy for pancreatic cancer. Gastrointest. Endosc 94, 179–186 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.DeWitt JM et al. Phase 1 study of EUS-guided photodynamic therapy for locally advanced pancreatic cancer. Gastrointest. Endosc 89, 390–398 (2019). [DOI] [PubMed] [Google Scholar]
  • 57.Falk-Mahapatra R & Gollnick SO Photodynamic therapy and immunity: an update. Photochem. Photobiol 96, 550–559 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Cramer GM, Moon EK, Cengel KA & Busch TM Photodynamic therapy and immune checkpoint blockade. Photochem. Photobiol 96, 954–961 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zhen Z et al. Tumor vasculature targeted photodynamic therapy for enhanced delivery of nanoparticles. ACS Nano 8, 6004–6013 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wang Y et al. Fluence plays a critical role on the subsequent distribution of chemotherapy and tumor growth delay in murine mesothelioma xenografts pre-treated by photodynamic therapy. Lasers Surg. Med 47, 323–330 (2015). [DOI] [PubMed] [Google Scholar]
  • 61.Spring BQ et al. A photoactivable multi-inhibitor nanoliposome for tumour control and simultaneous inhibition of treatment escape pathways. Nat. Nanotechnol 11, 378–387 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]; This article reports a liposome that is doped with photosensitizers and encapsulates a nanoparticle containing cabozantinib for the treatment of pancreatic tumours in mice.
  • 62.Liang BJ et al. Fluorescence-guided photoimmunotherapy using targeted nanotechnology and ML7710 to manage peritoneal carcinomatosis. Sci. Adv 9, eadi3441 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lovell JF et al. Porphysome nanovesicles generated by porphyrin bilayers for use as multimodal biophotonic contrast agents. Nat. Mater 10, 324–332 (2011). [DOI] [PubMed] [Google Scholar]; This article reports porphyrin–phospholipid conjugates that can be self-assembled into porphysome nanovesicles for imaging and phototherapy of KB tumour-bearing mice.
  • 64.Carter KA et al. Porphyrin–phospholipid liposomes permeabilized by near-infrared light. Nat. Commun 5, 3546 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Jori G et al. Preferential delivery of liposome-incorporated porphyrins to neoplastic cells in tumour-bearing rats. Br. J. Cancer 48, 307–309 (1983). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Duska LR, Hamblin MR, Bamberg MP & Hasan T Biodistribution of charged F(ab’)2 photoimmunoconjugates in a xenograft model of ovarian cancer. Br. J. Cancer 75, 837–844 (1997). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rizvi I et al. A combination of Visudyne and a lipid-anchored liposomal formulation of benzoporphyrin derivative enhances photodynamic therapy efficacy in a 3D model for ovarian cancer. Photochem. Photobiol 95, 419–429 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Inglut CT et al. Systematic evaluation of light-activatable biohybrids for anti-glioma photodynamic therapy. J. Clin. Med 8, 1269 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Jin CS, Cui L, Wang F, Chen J & Zheng G Targeting-triggered porphysome nanostructure disruption for activatable photodynamic therapy. Adv. Healthc. Mater 3, 1240–1249 (2014). [DOI] [PubMed] [Google Scholar]
  • 70.Muhanna N et al. Phototheranostic porphyrin nanoparticles enable visualization and targeted treatment of head and neck cancer in clinically relevant models. Theranostics 5, 1428–1443 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liu TW, MacDonald TD, Shi J, Wilson BC & Zheng G Intrinsically copper-64-labeled organic nanoparticles as radiotracers. Angew. Chem. Int. Ed 51, 13128–13131 (2012). [DOI] [PubMed] [Google Scholar]
  • 72.He C et al. Core-shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. Nat. Commun 7, 12499 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]; This article reports a core-shell nanoparticle encapsulating oxaliplatin and a photosensitizer pyropheophorbide–lipid conjugate, which can be intravenously administered and combined with immunotherapy to treat colorectal tumours in mice.
  • 73.He C, Liu D & Lin W Self-assembled core-shell nanoparticles for combined chemotherapy and photodynamic therapy of resistant head and neck cancers. ACS Nano 9, 991–1003 (2015). [DOI] [PubMed] [Google Scholar]
  • 74.Guirguis M et al. Membrane composition is a functional determinant of NIR-activable liposomes in orthotopic head and neck cancer. Nanophotonics 10, 3169–3185 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Spring BQ et al. Selective treatment and monitoring of disseminated cancer micrometastases in vivo using dual-function, activatable immunoconjugates. Proc. Natl Acad. Sci. USA 111, E933–E942 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Heukers R, van Bergen en Henegouwen PM & Oliveira S Nanobody–photosensitizer conjugates for targeted photodynamic therapy. Nanomedicine 10, 1441–1451 (2014). [DOI] [PubMed] [Google Scholar]
  • 77.Jin J et al. Evaluating near-infrared photoimmunotherapy for targeting fibroblast activation protein-α expressing cells in vitro and in vivo. Cancer Sci 114, 236–246 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sadraeian M et al. Photoimmunotherapy using cationic and anionic photosensitizer-antibody conjugates against HIV Env-expressing cells. Int. J. Mol. Sci 21, 9151 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bryden F et al. Assembly of high-potency photosensitizer–antibody conjugates through application of dendron multiplier technology. Bioconjugate Chem 29, 176–181 (2018). [DOI] [PubMed] [Google Scholar]
  • 80.Broekgaarden M et al. Site-specific conjugation of single domain antibodies to liposomes enhances photosensitizer uptake and photodynamic therapy efficacy. Nanoscale 8, 6490–6494 (2016). [DOI] [PubMed] [Google Scholar]
  • 81.Mesquita BS et al. The impact of nanobody density on the targeting efficiency of PEGylated liposomes. Int. J. Mol. Sci 23, 14974 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Obaid G et al. Impacting pancreatic cancer therapy in heterotypic in vitro organoids and in vivo tumors with specificity-tuned, NIR-activable photoimmunonanoconjugates: towards conquering desmoplasia? Nano Lett 19, 7573–7587 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhang Z, Lu Z, Yuan Q, Zhang C & Tang Y ROS-responsive and active targeted drug delivery based on conjugated polymer nanoparticles for synergistic chemo-/photodynamic therapy. J. Mater. Chem. B 9, 2240–2248 (2021). [DOI] [PubMed] [Google Scholar]
  • 84.Bryden F et al. Regioselective and stoichiometrically controlled conjugation of photodynamic sensitizers to a HER2 targeting antibody fragment. Bioconjugate Chem 25, 611–617 (2014). [DOI] [PubMed] [Google Scholar]
  • 85.Staneloudi C et al. Development and characterization of novel photosensitizer : scFv conjugates for use in photodynamic therapy of cancer. Immunology 120, 512–517 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Liang BJ et al. Breaking the selectivity-uptake trade-off of photoimmunoconjugates with nanoliposomal irinotecan for synergistic multi-tier cancer targeting. J. Nanobiotechnol 18, 1 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Huang HC, Pigula M, Fang Y & Hasan T Immobilization of photo-immunoconjugates on nanoparticles leads to enhanced light-activated biological effects. Small 14, e1800236 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kines RC et al. An infrared dye-conjugated virus-like particle for the treatment of primary uveal melanoma. Mol. Cancer Ther 17, 565–574 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bhandari C et al. What NIR photodynamic activation offers molecular targeted nanomedicines: perspectives into the conundrum of tumor specificity and selectivity. Nano Today 36, 101052 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lucky SS, Soo KC & Zhang Y Nanoparticles in photodynamic therapy. Chem. Rev 115, 1990–2042 (2015). [DOI] [PubMed] [Google Scholar]
  • 91.Lu K et al. Chlorin-based nanoscale metal–organic framework systemically rejects colorectal cancers via synergistic photodynamic therapy and checkpoint blockade immunotherapy. J. Am. Chem. Soc 138, 12502–12510 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Deng S et al. Codelivery of CRISPR-Cas9 and chlorin e6 for spatially controlled tumor-specific gene editing with synergistic drug effects. Sci. Adv 6, eabb4005 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]; This article reports a nanoparticle containing the photosensitizer chlorin e6 and Cas9/single-guide RNA for photodynamic therapy and Nrf2 gene editing of nasopharyngeal carcinoma in vivo.
  • 93.Yue D et al. An alternating irradiation strategy-driven combination therapy of PDT and RNAi for highly efficient inhibition of tumor growth and metastasis. Adv. Healthc. Mater 10, e2001850 (2021). [DOI] [PubMed] [Google Scholar]
  • 94.Mo Y et al. Light-activated siRNA endosomal release (LASER) by porphyrin lipid nanoparticles. ACS Nano 17, 4688–4703 (2023). [DOI] [PubMed] [Google Scholar]
  • 95.Dai J et al. Aggregation-induced emission photosensitizers: from molecular design to photodynamic therapy. J. Med. Chem 63, 1996–2012 (2020). [DOI] [PubMed] [Google Scholar]
  • 96.Yang Y et al. Photodynamic therapy with liposomes encapsulating photosensitizers with aggregation-induced emission. Nano Lett 19, 1821–1826 (2019). [DOI] [PubMed] [Google Scholar]
  • 97.Wang D et al. Highly efficient photosensitizers with far-red/near-infrared aggregation-induced emission for in vitro and in vivo cancer theranostics. Adv. Mater 30, e1802105 (2018). [DOI] [PubMed] [Google Scholar]; This article reports aggregation-induced emission luminogens (AIEgen) that can be intratumourally administered for fluorescence imaging and photodynamic therapy of A431 tumours in mice.
  • 98.Wu Y et al. Endoplasmic reticulum-targeted aggregation-induced emission luminogen for synergetic tumor ablation with glibenclamide. ACS Appl. Mater. Interfaces 15, 50821–50835 (2023). [DOI] [PubMed] [Google Scholar]
  • 99.Grimm D EPA plan to end animal testing splits scientists. Science 365, 1231 (2019). [DOI] [PubMed] [Google Scholar]
  • 100.Celli JP et al. An imaging-based platform for high-content, quantitative evaluation of therapeutic response in 3D tumour models. Sci. Rep 4, 3751 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bulin AL, Broekgaarden M & Hasan T Comprehensive high-throughput image analysis for therapeutic efficacy of architecturally complex heterotypic organoids. Sci. Rep 7, 16645 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Phan N et al. A simple high-throughput approach identifies actionable drug sensitivities in patient-derived tumor organoids. Commun. Biol 2, 78 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zhao Z et al. Organoids. Nat. Rev. Methods Primers 2, 94 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hofer M & Lutolf MP Engineering organoids. Nat. Rev. Mater 6, 402–420 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Celli JP, Rizvi I, Evans CL, Abu-Yousif AO & Hasan T Quantitative imaging reveals heterogeneous growth dynamics and treatment-dependent residual tumor distributions in a three-dimensional ovarian cancer model. J. Biomed. Opt 15, 051603 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Glidden MD et al. Image-based quantification of benzoporphyrin derivative uptake, localization, and photobleaching in 3D tumor models, for optimization of PDT parameters. Theranostics 2, 827–839 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Le Clainche T, Carigga Gutierrez NM, Pujol-Solé N, Coll J-L & Broekgaarden M Optimizing the pharmacological and optical dosimetry for photodynamic therapy with methylene blue and nanoliposomal benzoporphyrin on pancreatic cancer spheroids. Onco 2, 19–33 (2022). [Google Scholar]
  • 108.Broekgaarden M et al. Modulation of redox metabolism negates cancer-associated fibroblasts-induced treatment resistance in a heterotypic 3D culture platform of pancreatic cancer. Biomaterials 222, 119421 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Favreau PF et al. Label-free redox imaging of patient-derived organoids using selective plane illumination microscopy. Biomed. Opt. Express 11, 2591–2606 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gil DA, Deming D & Skala MC Patient-derived cancer organoid tracking with wide-field one-photon redox imaging to assess treatment response. J. Biomed. Opt 26, 036005 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Gaitan B et al. Development of an endoscopic auto-fluorescent sensing device to aid in the detection of breast cancer and inform photodynamic therapy. Metabolites 12, 1097 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Yakavets I et al. Effect of stroma on the behavior of temoporfin-loaded lipid nanovesicles inside the stroma-rich head and neck carcinoma spheroids. J. Nanobiotechnol 19, 3 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Rizvi I et al. Synergistic enhancement of carboplatin efficacy with photodynamic therapy in a three-dimensional model for micrometastatic ovarian cancer. Cancer Res 70, 9319–9328 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Broekgaarden M et al. Neoadjuvant photodynamic therapy augments immediate and prolonged oxaliplatin efficacy in metastatic pancreatic cancer organoids. Oncotarget 9, 13009–13022 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Rizvi I et al. Flow induces epithelial-mesenchymal transition, cellular heterogeneity and biomarker modulation in 3D ovarian cancer nodules. Proc. Natl Acad. Sci. USA 110, E1974–E1983 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Sorrin AJ et al. Transient fluid flow improves photoimmunoconjugate delivery and photoimmunotherapy efficacy. iScience 26, 107221 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Nath S et al. Flow-induced shear stress confers resistance to carboplatin in an adherent three-dimensional model for ovarian cancer: a role for EGFR-targeted photoimmunotherapy informed by physical stress. J. Clin. Med 9, 924 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Drost J & Clevers H Organoids in cancer research. Nat. Rev. Cancer 18, 407–418 (2018). [DOI] [PubMed] [Google Scholar]
  • 119.Driehuis E et al. Patient-derived head and neck cancer organoids recapitulate EGFR expression levels of respective tissues and are responsive to EGFR-targeted photodynamic therapy. J. Clin. Med 8, 1880 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Fujiwara H et al. 5-Aminolevulinic acid-mediated photodynamic activity in patient-derived cholangiocarcinoma organoids. Surg. Oncol 35, 484–490 (2020). [DOI] [PubMed] [Google Scholar]
  • 121.Dekkers JF et al. High-resolution 3D imaging of fixed and cleared organoids. Nat. Protoc 14, 1756–1771 (2019). [DOI] [PubMed] [Google Scholar]
  • 122.Kner P, Sedat JW, Agard DA & Kam Z High-resolution wide-field microscopy with adaptive optics for spherical aberration correction and motionless focusing. J. Microsc 237, 136–147 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Alladin A et al. Tracking cells in epithelial acini by light sheet microscopy reveals proximity effects in breast cancer initiation. eLife 9, e54066 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.de Medeiros G et al. Multiscale light-sheet organoid imaging framework. Nat. Commun 13, 4864 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhu TC, Kim MM, Liang X, Finlay JC & Busch TM In-vivo singlet oxygen threshold doses for PDT. Photonics Lasers Med 4, 59–71 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Farrell TJ, Wilson BC, Patterson MS & Olivo MC Comparison of the in vivo photodynamic threshold dose for photofrin, mono- and tetrasulfonated aluminum phthalocyanine using a rat liver model. Photochem. Photobiol 68, 394–399 (1998). [PubMed] [Google Scholar]
  • 127.Patterson MS, Wilson BC & Graff R In vivo tests of the concept of photodynamic threshold dose in normal rat liver photosensitized by aluminum chlorosulphonated phthalocyanine. Photochem. Photobiol 51, 343–349 (1990). [DOI] [PubMed] [Google Scholar]
  • 128.Standish BA et al. Interstitial Doppler optical coherence tomography monitors microvascular changes during photodynamic therapy in a Dunning prostate model under varying treatment conditions. J. Biomed. Opt 12, 034022 (2007). [DOI] [PubMed] [Google Scholar]
  • 129.Johansson A et al. In vivo measurement of parameters of dosimetric importance during interstitial photodynamic therapy of thick skin tumors. J. Biomed. Opt 11, 34029 (2006). [DOI] [PubMed] [Google Scholar]
  • 130.Xiang M et al. A review of light sources and enhanced targeting for photodynamic therapy. Curr. Med. Chem 28, 6437–6457 (2021). [DOI] [PubMed] [Google Scholar]
  • 131.Wang KK & Zhu TC Reconstruction of in-vivo optical properties for human prostate using interstitial diffuse optical tomography. Opt. Express 17, 11665–11672 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Beck TJ et al. Interstitial photodynamic therapy of nonresectable malignant glioma recurrences using 5-aminolevulinic acid induced protoporphyrin IX. Lasers Surg. Med 39, 386–393 (2007). [DOI] [PubMed] [Google Scholar]
  • 133.Finlay JC et al. Interstitial fluorescence spectroscopy in the human prostate during motexafin lutetium-mediated photodynamic therapy. Photochem. Photobiol 82, 1270–1278 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Yassine AA, Lilge L & Betz V Optimizing interstitial photodynamic therapy with custom cylindrical diffusers. J. Biophotonics 12, e201800153 (2019). [DOI] [PubMed] [Google Scholar]
  • 135.Lilge L, Vesselov L & Whittington W Thin cylindrical diffusers in multimode Ge-doped silica fibers. Lasers Surg. Med 36, 245–251 (2005). [DOI] [PubMed] [Google Scholar]
  • 136.Murrer LH, Marijnissen JP & Star WM Improvements in the design of linear diffusers for photodynamic therapy. Phys. Med. Biol 42, 1461–1464 (1997). [DOI] [PubMed] [Google Scholar]
  • 137.Schipmann S et al. Combination of ALA-induced fluorescence-guided resection and intraoperative open photodynamic therapy for recurrent glioblastoma: case series on a promising dual strategy for local tumor control. J. Neurosurg 134, 426–436 (2020). [DOI] [PubMed] [Google Scholar]
  • 138.Ylöniemi Z et al. Cloud-based medical laser platform for phototherapy and treatment monitoring for glioblastoma. In Proc. SPIE 11629, Optical Techniques in Neurosurgery, Neurophotonics, and Optogenetics 116290F (SPIE, 2021). [Google Scholar]
  • 139.Kaivosoja V et al. Configurable cloud based medical laser platform with real-time treatment monitoring. In Proc. SPIE 11628, Mechanisms and Techniques in Photodynamic Therapy and Photobiomodulation 116280Q (SPIE, 2021). [Google Scholar]
  • 140.Vesala L et al. Novel ophthalmic PDT laser platform to target oncology and various other retinal indications. In Proc. SPIE 12359, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XXXI 1235905 (SPIE, 2023). [Google Scholar]
  • 141.Cochrane C, Mordon SR, Lesage JC & Koncar V New design of textile light diffusers for photodynamic therapy. Mater. Sci. Eng C 33, 1170–1175 (2013). [DOI] [PubMed] [Google Scholar]
  • 142.Mordon S et al. Light emitting fabric technologies for photodynamic therapy. Photodiagnosis Photodyn. Ther 12, 1–8 (2015). [DOI] [PubMed] [Google Scholar]
  • 143.Dubois M et al. Real-life evaluation of the treatment of actinic keratoses by textile photodynamic therapy (FLUXMEDICARE® device). Photodiagnosis Photodyn. Ther 34, 102213 (2021). [DOI] [PubMed] [Google Scholar]
  • 144.Lecomte F et al. Photodynamic therapy using a new painless light-emitting fabrics device in the treatment of extramammary Paget disease of the vulva (the PAGETEX Study): protocol for an interventional efficacy and safety trial. JMIR Res. Protoc 8, e15026 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bansal A, Yang F, Xi T, Zhang Y & Ho JS In vivo wireless photonic photodynamic therapy. Proc. Natl Acad. Sci. USA 115, 1469–1474 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Kim WS et al. AI-enabled, implantable, multichannel wireless telemetry for photodynamic therapy. Nat. Commun 13, 2178 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Choi J et al. Implantable micro-scale LED device guided photodynamic therapy to potentiate antitumor immunity with mild visible light. Biomater. Res 26, 56 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Jermyn M et al. CT contrast predicts pancreatic cancer treatment response to verteporfin-based photodynamic therapy. Phys. Med. Biol 59, 1911–1921 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Rohrbach DJ et al. Preoperative mapping of nonmelanoma skin cancer using spatial frequency domain and ultrasound imaging. Acad. Radiol 21, 263–270 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Quach S et al. Interstitial photodynamic therapy for newly diagnosed glioblastoma. J. Neurooncol 162, 217–223 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Davidson SRH et al. Treatment planning and dose analysis for interstitial photodynamic therapy of prostate cancer. Phys. Med. Biol 54, 2293–2313 (2009). [DOI] [PubMed] [Google Scholar]
  • 152.Zhu TC et al. Light fluence dosimetry in lung-simulating cavities. In Proc. SPIE 10476, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XXVII 104760F (SPIE, 2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Friedberg JS et al. Phase II trial of pleural photodynamic therapy and surgery for patients with non-small-cell lung cancer with pleural spread. J. Clin. Oncol 22, 2192–2201 (2004). [DOI] [PubMed] [Google Scholar]
  • 154.Zhu TC et al. Real-time PDT dose dosimetry for pleural photodynamic therapy. In Proc. SPIE 11940, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic and Photobiomodulation Therapy XXX 1194002 (SPIE, 2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Tyrrell J, Campbell S & Curnow A Protoporphyrin IX photobleaching during the light irradiation phase of standard dermatological methyl-aminolevulinate photodynamic therapy. Photodiagnosis Photodyn. Ther 7, 232–238 (2010). [DOI] [PubMed] [Google Scholar]
  • 156.Kim MM et al. A comparison of singlet oxygen explicit dosimetry (SOED) and singlet oxygen luminescence dosimetry (SOLD) for Photofrin-mediated photodynamic therapy. Cancers 8, 109 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Zhao Y et al. High optical-throughput spectroscopic singlet oxygen and photosensitizer luminescence dosimeter for monitoring of photodynamic therapy. J. Biophotonics 14, e202100088 (2021). [DOI] [PubMed] [Google Scholar]
  • 158.Panjehpour M, Overholt BF, Phan MN & Haydek JM Optimization of light dosimetry for photodynamic therapy of Barrett’s esophagus: efficacy vs. incidence of stricture after treatment. Gastrointest. Endosc 61, 13–18 (2005). [DOI] [PubMed] [Google Scholar]
  • 159.Pereira SP et al. Photodynamic therapy of malignant biliary strictures using meso-tetrahydroxyphenylchlorin. Eur. J. Gastroenterol. Hepatol 19, 479–485 (2007). [DOI] [PubMed] [Google Scholar]
  • 160.Choi V, Rajora MA & Zheng G Activating drugs with sound: mechanisms behind sonodynamic therapy and the role of nanomedicine. Bioconjugate Chem 31, 967–989 (2020). [DOI] [PubMed] [Google Scholar]
  • 161.Canavese G et al. Nanoparticle-assisted ultrasound: a special focus on sonodynamic therapy against cancer. Chem. Eng. J 340, 155–172 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Rosenthal I, Sostaric JZ & Riesz P Sonodynamic therapy–a review of the synergistic effects of drugs and ultrasound. Ultrason. Sonochem 11, 349–363 (2004). [DOI] [PubMed] [Google Scholar]
  • 163.Um W et al. Recent advances in nanomaterial-based augmented sonodynamic therapy of cancer. Chem. Commun 57, 2854–2866 (2021). [DOI] [PubMed] [Google Scholar]
  • 164.Chen W & Zhang J Using nanoparticles to enable simultaneous radiation and photodynamic therapies for cancer treatment. J. Nanosci. Nanotechnol 6, 1159–1166 (2006). [DOI] [PubMed] [Google Scholar]
  • 165.Sun W, Zhou Z, Pratx G, Chen X & Chen H Nanoscintillator-mediated X-ray induced photodynamic therapy for deep-seated tumors: from concept to biomedical applications. Theranostics 10, 1296–1318 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Souris JS et al. X-ray activated nanoplatforms for deep tissue photodynamic therapy. Nanomaterials 13, 673 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Larue L et al. Using X-rays in photodynamic therapy: an overview. Photochem. Photobiol. Sci 17, 1612–1650 (2018). [DOI] [PubMed] [Google Scholar]
  • 168.Axelsson J, Davis SC, Gladstone DJ & Pogue BW Cerenkov emission induced by external beam radiation stimulates molecular fluorescence. Med. Phys 38, 4127–4132 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Daouk J et al. Can Cerenkov light really induce an effective photodynamic therapy? Radiation 1, 5–17 (2021). [Google Scholar]
  • 170.Yang DM, Cvetkovic D, Chen L & Ma CC Therapeutic effects of in-vivo radiodynamic therapy (RDT) for lung cancer treatment: a combination of 15MV photons and 5-aminolevulinic acid (5-ALA). Biomed. Phys. Eng. Express 8, 065031 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Berg K, Luksiene Z, Moan J & Ma L Combined treatment of ionizing radiation and photosensitization by 5-aminolevulinic acid-induced protoporphyrin IX. Radiat. Res 142, 340–346 (1995). [PubMed] [Google Scholar]
  • 172.Takahashi J & Masaki M Characterization of reactive oxygen species generated by protoporphyrin IX under X-ray irradiation. Radiat. Phys. Chem 78, 889–898 (2009). [Google Scholar]
  • 173.Takahashi J et al. 5-Aminolevulinic acid enhances cancer radiotherapy in a mouse tumor model. Springerplus 2, 602 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Yamamoto J et al. Radiosensitizing effect of 5-aminolevulinic acid-induced protoporphyrin IX in glioma cells in vitro. Oncol. Rep 27, 1748–1752 (2012). [DOI] [PubMed] [Google Scholar]
  • 175.Chen H et al. Nanoscintillator-mediated X-ray inducible photodynamic therapy for in vivo cancer treatment. Nano Lett 15, 2249–2256 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]; This article reports a nanoscintillator with photosensitizer-loaded mesoporous silica coating that converts X-rays to photons for anti-tumour photodynamic therapy in vivo.
  • 176.Morgan NY, Kramer-Marek G, Smith PD, Camphausen K & Capala J Nanoscintillator conjugates as photodynamic therapy-based radiosensitizers: calculation of required physical parameters. Radiat. Res 171, 236–244 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Bulin AL et al. Modelling energy deposition in nanoscintillators to predict the efficiency of the X-ray-induced photodynamic effect. Nanoscale 7, 5744–5751 (2015). [DOI] [PubMed] [Google Scholar]
  • 178.Bulin A-L et al. X-ray-induced singlet oxygen activation with nanoscintillator-coupled porphyrins. J. Phys. Chem C 117, 21583–21589 (2013). [Google Scholar]
  • 179.Bulin AL et al. Radiation dose-enhancement is a potent radiotherapeutic effect of rare-earth composite nanoscintillators in preclinical models of glioblastoma. Adv. Sci 7, 2001675 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Lux F et al. AGuIX from bench to bedside-transfer of an ultrasmall theranostic gadolinium-based nanoparticle to clinical medicine. Br. J. Radiol 92, 20180365 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Bonvalot S et al. NBTXR3, a first-in-class radioenhancer hafnium oxide nanoparticle, plus radiotherapy versus radiotherapy alone in patients with locally advanced soft-tissue sarcoma (Act.In.Sarc): a multicentre, phase 2–3, randomised, controlled trial. Lancet Oncol 20, 1148–1159 (2019). [DOI] [PubMed] [Google Scholar]
  • 182.Syed AJ & Anderson JC Applications of bioluminescence in biotechnology and beyond. Chem. Soc. Rev 50, 5668–5705 (2021). [DOI] [PubMed] [Google Scholar]
  • 183.Kim YR et al. Bioluminescence-activated deep-tissue photodynamic therapy of cancer. Theranostics 5, 805–817 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Ng J et al. Bioluminescence-activated photodynamic therapy for luciferase transfected, grade 4 astrocytoma cells in vitro. Photodiagnosis Photodyn. Ther 38, 102856 (2022). [DOI] [PubMed] [Google Scholar]
  • 185.Kotagiri N, Sudlow GP, Akers WJ & Achilefu S Breaking the depth dependency of phototherapy with Cerenkov radiation and low-radiance-responsive nanophotosensitizers. Nat. Nanotechnol 10, 370–379 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]; This article reports intratumourally administered nanophotosensitizers that are activated by Cerenkov radiation from radionuclides to treat tumours in vivo.
  • 186.Ma CH, Yang J, Mueller JL & Huang HC Intratumoral photosensitizer delivery and photodynamic therapy. Nano Life 11, 2130003 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Duan D et al. Activating TiO2 nanoparticles: gallium-68 serves as a high-yield photon emitter for Cerenkov-induced photodynamic therapy. ACS Appl. Mater. Interfaces 10, 5278–5286 (2018). [DOI] [PubMed] [Google Scholar]
  • 188.Ni D et al. Magnetic targeting of nanotheranostics enhances Cerenkov radiation-induced photodynamic therapy. J. Am. Chem. Soc 140, 14971–14979 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Pratx G & Kapp DS Is Cherenkov luminescence bright enough for photodynamic therapy? Nat. Nanotechnol 13, 354 (2018). [DOI] [PubMed] [Google Scholar]
  • 190.Kotagiri N, Laforest R & Achilefu S Reply to ‘Is Cherenkov luminescence bright enough for photodynamic therapy?’. Nat. Nanotechnol 13, 354–355 (2018). [DOI] [PubMed] [Google Scholar]
  • 191.Ni K, Luo T, Nash GT & Lin W Nanoscale metal–organic frameworks for cancer immunotherapy. Acc. Chem. Res 53, 1739–1748 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Hartl BA, Hirschberg H, Marcu L & Cherry SR Activating photodynamic therapy in vitro with Cerenkov radiation generated from yttrium-90. J. Environ. Pathol. Toxicol. Oncol 35, 185–192 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Chhatre S, Murgu S, Vachani A & Jayadevappa R Photodynamic therapy for stage I and II non-small cell lung cancer: a SEER-Medicare analysis 2000–2016. Medicine 101, e29053 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Rusk R & Hodge J Impact of nasal photodisinfection on SARS-CoV-2 infection in an industrial workplace. Public Health Pract 6, 100393 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Cognetti DM et al. Phase 1/2a, open-label, multicenter study of RM-1929 photoimmunotherapy in patients with locoregional, recurrent head and neck squamous cell carcinoma. Head Neck 43, 3875–3887 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Nishinaga Y et al. Targeted phototherapy for malignant pleural mesothelioma: near-infrared photoimmunotherapy targeting podoplanin. Cells 9, 1019 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Sadiki A et al. Site-specific bioconjugation and convergent click chemistry enhances antibody–chromophore conjugate binding efficiency. Photochem. Photobiol 96, 596–603 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Alonso CM et al. Site-specific and stoichiometric conjugation of cationic porphyrins to antiangiogenic monoclonal antibodies. Bioconjugate Chem 21, 302–313 (2010). [DOI] [PubMed] [Google Scholar]
  • 199.van Driel PBAA et al. EGFR targeted nanobody-photosensitizer conjugates for photodynamic therapy in a pre-clinical model of head and neck cancer. J. Control. Release 229, 93–105 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Deken MM et al. Nanobody-targeted photodynamic therapy induces significant tumor regression of trastuzumab-resistant HER2-positive breast cancer, after a single treatment session. J. Control. Release 323, 269–281 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Bovis MJ et al. Improved in vivo delivery of m-THPC via pegylated liposomes for use in photodynamic therapy. J. Control. Release 157, 196–205 (2012). [DOI] [PubMed] [Google Scholar]
  • 202.Obaid G, Jin W, Bano S, Kessel D & Hasan T Nanolipid formulations of benzoporphyrin derivative: exploring the dependence of nanoconstruct photophysics and photochemistry on their therapeutic index in ovarian cancer cells. Photochem. Photobiol 95, 364–377 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.MacDonald TD, Liu TW & Zheng G An MRI-sensitive, non-photobleachable porphysome photothermal agent. Angew. Chem. Int. Ed 53, 6956–6959 (2014). [DOI] [PubMed] [Google Scholar]
  • 204.Guidolin K, Ding L, Chen J & Zheng G Development of a clinically viable strategy for nanoparticle-based photodynamic therapy of colorectal cancer. Adv. Ther 6, 2200342 (2023). [Google Scholar]
  • 205.Kinoshita T et al. A novel laser fiberscope for simultaneous imaging and phototherapy of peripheral lung cancer. Chest 156, 571–578 (2019). [DOI] [PubMed] [Google Scholar]
  • 206.Carter KA et al. Sphingomyelin liposomes containing porphyrin-phospholipid for irinotecan chemophototherapy. Theranostics 6, 2329–2336 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Carter KA et al. Metal chelation modulates phototherapeutic properties of mitoxantrone-loaded porphyrin–phospholipid liposomes. Mol. Pharm 13, 420–427 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Cole HD et al. Establishing a robust and reliable response from a potent osmium-based photosensitizer via lipid nanoformulation. Photochem. Photobiol 99, 751–760 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Shah D et al. Enabling in vivo optical imaging of an osmium photosensitizer by micellar formulation. Pharmaceutics 14, 2426 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Nishiyama N, Morimoto Y, Jang WD & Kataoka K Design and development of dendrimer photosensitizer-incorporated polymeric micelles for enhanced photodynamic therapy. Adv. Drug Deliv. Rev 61, 327–338 (2009). [DOI] [PubMed] [Google Scholar]
  • 211.Xu J, Zeng F, Wu H, Hu C & Wu S Enhanced photodynamic efficiency achieved via a dual-targeted strategy based on photosensitizer/micelle structure. Biomacromolecules 15, 4249–4259 (2014). [DOI] [PubMed] [Google Scholar]
  • 212.Liu X et al. Photosensitizer cross-linked nano-micelle platform for multimodal imaging guided synergistic photothermal/photodynamic therapy. Nanoscale 8, 15323–15339 (2016). [DOI] [PubMed] [Google Scholar]
  • 213.Cai X et al. Multifunctional liposome: a bright AIEgen–lipid conjugate with strong photosensitization. Angew. Chem. Int. Ed 57, 16396–16400 (2018). [DOI] [PubMed] [Google Scholar]
  • 214.Mao D et al. Chemiluminescence-guided cancer therapy using a chemiexcited photosensitizer. Chem 3, 991–1007 (2017). [Google Scholar]
  • 215.Sun X et al. A DNA functionalized metal–organic framework combined with magnesium peroxide nanoparticles: targeted and enhanced photodynamic therapy. Mater. Chem. Front 6, 956–965 (2022). [Google Scholar]
  • 216.Lan G et al. Nanoscale metal–organic framework overcomes hypoxia for photodynamic therapy primed cancer immunotherapy. J. Am. Chem. Soc 140, 5670–5673 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Mo Z et al. MOF(Fe)-derived composites as a unique nanoplatform for chemophotodynamic tumor therapy. J. Mater. Chem. B 10, 8760–8770 (2022). [DOI] [PubMed] [Google Scholar]
  • 218.Zhang W et al. Enhanced photodynamic therapy by reduced levels of intracellular glutathione obtained by employing a nano-MOF with CuII as the active center. Angew. Chem. Int. Ed 57, 4891–4896 (2018). [DOI] [PubMed] [Google Scholar]
  • 219.Wang Y et al. Cancer-cell-activated photodynamic therapy assisted by Cu(II)-based metal–organic framework. ACS Nano 13, 6879–6890 (2019). [DOI] [PubMed] [Google Scholar]
  • 220.Kelly JF & Snell ME Hematoporphyrin derivative: a possible aid in the diagnosis and therapy of carcinoma of the bladder. J. Urol 115, 150–151 (1976). [DOI] [PubMed] [Google Scholar]
  • 221.Schmidt S, Wagner U, Oehr P & Krebs D Clinical use of photodynamic therapy in gynecologic tumor patients — antibody-targeted photodynamic laser therapy as a new oncologic treatment procedure. Zentralbl. Gynakol 114, 307–311 (1992). [PubMed] [Google Scholar]
  • 222.Ran C, Zhang Z, Hooker J & Moore A In vivo photoactivation without “light”: use of Cherenkov radiation to overcome the penetration limit of light. Mol. Imaging Biol 14, 156–162 (2012). [DOI] [PubMed] [Google Scholar]
  • 223.Celli J, Rizvi I, Evans C, Abu-Yousif A & Hasan T Three-dimensional ovarian cancer models: imaging and therapeutic combinations. In Proc. SPIE 7551, Optical Methods for Tumor Treatment and Detection: Mechanisms and Techniques in Photodynamic Therapy XIX 75510I (SPIE, 2010). [Google Scholar]
  • 224.Saad MA et al. Photoimmunotherapy retains its anti-tumor efficacy with increasing stromal content in heterotypic pancreatic cancer spheroids. Mol. Pharm 19, 2549–2563 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Shah SC, Kayamba V, Peek RM & Heimburger D Cancer control in low- and middle-income countries: is it time to consider screening? JCO Glob. Oncol 5, 1–8 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Buzza HH et al. Photodynamic therapy: progress toward a scientific and clinical network in Latin America. Photodiagnosis Photodyn. Ther 13, 261–266 (2016). [DOI] [PubMed] [Google Scholar]
  • 227.Buzzá HH et al. Overall results for a national program of photodynamic therapy for basal cell carcinoma: a multicenter clinical study to bring new techniques to social health care. Cancer Control 26, 1073274819856885 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Sung H et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin 71, 209–249 (2021). [DOI] [PubMed] [Google Scholar]
  • 229.Khan S et al. Clinical evaluation of smartphone-based fluorescence imaging for guidance and monitoring of ALA-PDT treatment of early oral cancer. J. Biomed. Opt 25, 1–10 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Khan S et al. Clinical assessment of a low-cost, hand-held, smartphone-attached intraoral imaging probe for 5-aminolevulinic acid photodynamic therapy monitoring and guidance. J. Biomed. Opt 28, 082809 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Mallidi S et al. Platform for ergonomic intraoral photodynamic therapy using low-cost, modular 3D-printed components: design, comfort and clinical evaluation. Sci. Rep 9, 15830 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Siddiqui SA et al. Clinical evaluation of a mobile, low-cost system for fluorescence guided photodynamic therapy of early oral cancer in India. Photodiagnosis Photodyn. Ther 38, 102843 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES