Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 484
1.
Int J Nanomedicine ; 19: 3737-3751, 2024.
Article En | MEDLINE | ID: mdl-38699684

Background: Chemo-photodynamic combination therapy has demonstrated significant potential in the treatment of cancer. Triptolide (TPL), a naturally derived anticancer agent, when combined with the photosensitizer Chlorin e6 (Ce6), has shown to provide enhanced anti-tumor benefits. However, the development of stimuli-responsive nanovehicles for the co-delivery of TPL and Ce6 could further enhance the efficacy of this combination therapy. Methods: In this study, we synthesized a pH/ROS dual-responsive mPEG-TK-PBAE copolymer, which contains a pH-sensitive PBAE moiety and a ROS-sensitive thioketal (TK) linkage. Through a self-assembly process, TPL and Ce6 were successfully co-loaded into mPEG-TK-PBAE nanoparticles, hereafter referred to as TPL/Ce6 NPs. We evaluated the pH- and ROS-sensitive drug release and particle size changes. Furthermore, we investigated both the in vitro suppression of cellular proliferation and induction of apoptosis in HepG2 cells, as well as the in vivo anti-tumor efficacy of TPL/Ce6 NPs in H22 xenograft nude mice. Results: The mPEG-TK-PBAE copolymer was synthesized through a one-pot Michael-addition reaction and successfully co-encapsulated both TPL and Ce6 by self-assembly. Upon exposure to acid pH values and high ROS levels, the payloads in TPL/Ce6 NPs were rapidly released. Notably, the abundant ROS generated by the released Ce6 under laser irradiation further accelerated the degradation of the nanosystem, thereby amplifying the tumor microenvironment-responsive drug release and enhancing anticancer efficacy. Consequently, TPL/Ce6 NPs significantly increased PDT-induced oxidative stress and augmented TPL-induced apoptosis in HepG2 cells, leading to synergistic anticancer effects in vitro. Moreover, administering TPL/Ce6 NPs (containing 0.3 mg/kg of TPL and 4 mg/kg of Ce6) seven times, accompanied by 650 nm laser irradiation, efficiently inhibited tumor growth in H22 tumor-bearing mice, while exhibiting lower systemic toxicity. Conclusion: Overall, we have developed a tumor microenvironment-responsive nanosystem for the co-delivery of TPL and Ce6, demonstrating amplified synergistic effects of chemo-photodynamic therapy (chemo-PDT) for hepatocellular carcinoma (HCC) treatment.


Apoptosis , Chlorophyllides , Diterpenes , Liver Neoplasms , Mice, Nude , Phenanthrenes , Photochemotherapy , Photosensitizing Agents , Porphyrins , Reactive Oxygen Species , Animals , Humans , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Hep G2 Cells , Liver Neoplasms/drug therapy , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/administration & dosage , Porphyrins/pharmacokinetics , Diterpenes/chemistry , Diterpenes/pharmacology , Diterpenes/pharmacokinetics , Diterpenes/administration & dosage , Hydrogen-Ion Concentration , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Apoptosis/drug effects , Mice , Carcinoma, Hepatocellular/drug therapy , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Epoxy Compounds/administration & dosage , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Drug Liberation , Cell Proliferation/drug effects , Polyethylene Glycols/chemistry , Combined Modality Therapy
2.
ACS Macro Lett ; 13(5): 599-606, 2024 May 21.
Article En | MEDLINE | ID: mdl-38683197

The high glutathione (GSH) level of the tumor microenvironment severely affects the efficacy of photodynamic therapy (PDT). The current GSH depletion strategies have difficulty meeting the dual needs of security and efficiency. In this study, we report a photosensitizer Chlorin e6 (Ce6) and hypoxia-activated prodrug tirapazamine (TPZ) coloaded cross-linked multifunctional polymersome (TPZ/Ce6@SSPS) with GSH-triggered continuous GSH depletion for enhanced photodynamic therapy and hypoxia-activated chemotherapy. At tumor sites, the disulfide bonds of TPZ/Ce6@SSPS react with GSH to realize decross-linking for on-demand drug release. Meanwhile, the generated highly reactive quinone methide (QM) can further deplete GSH. This continuous GSH depletion will amplify tumor oxidative stress, enhancing the PDT effect of Ce6. Aggravated tumor hypoxia induced by PDT activates the prodrug TPZ, resulting in an enhanced combination of PDT and hypoxia-activated chemotherapy. Both in vitro and in vivo results demonstrate the efficient GSH depletion and potent antitumor activities by TPZ/Ce6@SSPS. This work provides a strategy for the design of a continuous GSH depletion platform, which holds great promise for enhanced combination tumor therapy.


Chlorophyllides , Glutathione , Photochemotherapy , Photosensitizing Agents , Prodrugs , Tirapazamine , Glutathione/metabolism , Photochemotherapy/methods , Tirapazamine/pharmacology , Animals , Mice , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Prodrugs/pharmacology , Porphyrins/pharmacology , Porphyrins/administration & dosage , Porphyrins/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Tumor Microenvironment/drug effects
3.
Photodiagnosis Photodyn Ther ; 47: 104096, 2024 Jun.
Article En | MEDLINE | ID: mdl-38643893

BACKGROUND: Port wine stains (PWS) are vascular malformations, and photodynamic therapy (PDT) is a promising treatment. Emerging drug delivery methods employ nanoparticles (NPs) to enhance drug permeability and retention in diseased blood vessels and improve drug bioavailability. (-) -epigallocatechin-3-gallate glycine (EGCG) has anti-angiogenetic effects and boosts photodynamic therapy. Chlorin e6 (Ce6) is capable of efficiently producing singlet oxygen, rendering it a very promising photosensitizer for utilization in nanomedicine. MATERIAL AND METHODS: EGCG-Ce6-NPs were synthesized and characterized using various techniques. The photodynamic effects of EGCG-Ce6-NPs on endothelial cells were evaluated. The compatibility and toxicity of the nanoparticle was tested using the CCK-8 assay. The intracellular uptake of the nanoparticle was observed using an inverted fluorescence microscope, and the intracellular fluorescence intensity was detected using flow cytometry. The ROS generation and apoptosis induced by EGCG-Ce6-NPs was observed using confocal laser scanning microscopy and flow cytometry respectively. RESULTS: EGCG-Ce6-NPs exhibited stability, spherical shape of uniform size while reducing the particle diameter, low polydisperse profile and retaining the ability to effectively generate singlet oxygen. These characteristics suggest promising potential for enhancing drug permeability and retention. Additionally, EGCG-Ce6-NPs demonstrated good compatibility with endothelial cells and enhanced intracellular uptake of Ce6. Furthermore, EGCG-Ce6-NPs increased activation efficiency, induced significant toxicity, more reactive oxygen species, and a higher rate of late apoptosis after laser irradiation. CONCLUSION: This in vitro study showed the potentials EGCG-Ce6-NPs for the destruction of endothelial cells in vasculature.


Catechin , Chlorophyllides , Nanoparticles , Photochemotherapy , Photosensitizing Agents , Porphyrins , Photosensitizing Agents/pharmacology , Photosensitizing Agents/pharmacokinetics , Photochemotherapy/methods , Nanoparticles/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/pharmacokinetics , Catechin/chemistry , Humans , Porphyrins/pharmacology , Porphyrins/pharmacokinetics , Endothelial Cells/drug effects , Polyphenols/pharmacology , Apoptosis/drug effects , Singlet Oxygen/metabolism , Cell Survival/drug effects
4.
Acta Biomater ; 179: 272-283, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38460931

Anticancer drugs used for systemic chemotherapy often exhibit off-target toxicity and uncontrolled drug release due to their lack of targeting. To improve the bioavailability of drugs and reduce side effects, we have developed a mixed micelle of nanomedicine composed of two prodrugs with surface modified monoclonal antibody for cancer therapy. In this system, Nimotuzumab was used as targeting ligands of the mixed micelles (named as DCMMs) that is composed of polymer-doxorubicin prodrug (abbreviated as PEG-b-P(GMA-ss-DOX)) and maleimide polyethylene glycol-chlorin e6 (abbreviated as Mal-PEG-Ce6). The mixed micelles modified with Nimotuzumab (named as NTZ-DCMMs) bind to overexpressed EGFR receptors on Hepatoma-22 (H22) cells. Disulfide bonds in PEG-b-P(GMA-ss-DOX) are disrupted in tumor microenvironment, inducing the reduction-responsive release of DOX and leading to tumor cell apoptosis. Simultaneously, Chlorin e6 (Ce6) produced plenty of singlet oxygen (1O2) under laser irradiation to kill tumor cells. In vivo biological distribution and antineoplastic effect experiments demonstrate that NTZ-DCMMs enhanced drug enrichment at tumor sites through targeting function of antibody, dramatically suppressing tumor growth and mitigating cardiotoxicity of drugs. All results prove that NTZ-DCMMs have the ability to actively target H22 cells and quickly respond to tumor microenvironment, which is expected to become an intelligent and multifunctional drug delivery carrier for efficient chemotherapy and photodynamic therapy of hepatoma. STATEMENT OF SIGNIFICANCE: Anticancer drugs used for systemic chemotherapy often exhibit off-target toxicity due to their lack of targeting. Therefore, it's necessary to develop effective, targeted, and collaborative treatment strategies. We construct a mixed micelle of nanomedicine based on two polymer prodrugs and modified with monoclonal antibody on surface for cancer therapy. Under the tumor cell microenvironment, the disulfide bonds of polymer-ss-DOX were broken, effectively triggering DOX release. The photosensitizer Ce6 could generate a large amount of ROS under light, which synergistically promotes tumor cell apoptosis. By coupling antibodies to the hydrophilic segments of polymer micelles, drugs can be specifically delivered. Compared with monotherapy, the combination of chemotherapy and photodynamic therapy can significantly enhance the therapeutic effect of liver cancer.


Chlorophyllides , Doxorubicin , Micelles , Nanomedicine , Photochemotherapy , Porphyrins , Prodrugs , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/chemistry , Animals , Photochemotherapy/methods , Cell Line, Tumor , Nanomedicine/methods , Porphyrins/chemistry , Porphyrins/pharmacology , Porphyrins/pharmacokinetics , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Mice , Polymers/chemistry , Polymers/pharmacology , Mice, Inbred BALB C , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Apoptosis/drug effects
5.
Bioconjug Chem ; 32(6): 1067-1077, 2021 06 16.
Article En | MEDLINE | ID: mdl-34033716

Passing through the blood-brain barrier (BBB) to treat neurological conditions is one of the main hurdles in modern medicine. Many drugs with promising in vitro profiles become ineffective in vivo due to BBB restrictive permeability. In particular, this includes drugs such as antiviral porphyrins, with the ability to fight brain-resident viruses causing diseases such as HIV-associated neurocognitive disorders (HAND). In the last two decades, BBB shuttles, particularly peptide-based ones, have shown promise in carrying various payloads across the BBB. Thus, peptide-drug conjugates (PDCs) formed by covalent attachment of a BBB peptide shuttle and an antiviral drug may become key therapeutic tools in treating neurological disorders of viral origin. In this study, we have used various approaches (guanidinium, phosphonium, and carbodiimide-based couplings) for on-resin synthesis of new peptide-porphyrin conjugates (PPCs) with BBB-crossing and potential antiviral activity. After careful fine-tuning of the synthetic chemistry, DIC/oxyma has emerged as a preferred method, by which 14 different PPCs have been made and satisfactorily characterized. The PPCs are prepared by coupling a porphyrin carboxyl group to an amino group (either N-terminal or a Lys side chain) of the peptide shuttle and show effective in vitro BBB translocation ability, low cytotoxicity toward mouse brain endothelial cells, and low hemolytic activity. Three of the PPCs, MP-P5, P4-MP, and P4-L-MP, effectively inhibiting HIV infectivity in vitro, stand out as most promising. Their efficacy against other brain-targeting viruses (Dengue, Zika, and SARS-CoV-2) is currently under evaluation, with preliminary results confirming that PPCs are a promising strategy to treat viral brain infections.


Anti-HIV Agents/pharmacokinetics , Blood-Brain Barrier/metabolism , Peptides/pharmacokinetics , Porphyrins/pharmacokinetics , Animals , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Biological Transport , Cell Line , Drug Discovery , HEK293 Cells , HIV/drug effects , HIV Infections/drug therapy , Humans , Mice , Peptides/chemistry , Peptides/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology
6.
Int J Nanomedicine ; 16: 3241-3254, 2021.
Article En | MEDLINE | ID: mdl-34007176

PURPOSE: Immune checkpoint inhibitors (ICIs) and sonodynamic therapy (SDT) are types of immunotherapy. In order to combine soluble programmed cell death protein 1 (sPD-1)-mediated immune checkpoint therapy and chlorin e6 (Ce6)-assisted SDT, nanobubbles (NBs) were generated to simultaneously load sPD-1 and Ce6. MATERIALS AND METHODS: The sPD-1/Ce6-NBs, which were prepared by thin-film hydration and mechanical oscillation, had a stable physical condition, and delivered sPD-1 and Ce6 in a targeted manner. NBs could strengthen tumor suppression by increasing tumor-targeting accumulation of Ce6 and sPD-1, and by inducing ultrasound-targeted NB destruction. A mouse H22 cell hepatoma xenograft model was used to evaluate the synergetic immunotherapeutic effect and mechanism of sPD-1/Ce6-NBs. RESULTS: By observing the tumor inhibition rate, tissue and cell apoptosis, apoptosis-related genes and protein expression, the best immunotherapeutic effect was exhibited by the sPD-1/Ce6-NBs group. The immunotherapeutic mechanism initially demonstrated that when tumor cells were transfected by sPD-1 delivered by NBs, which downregulated the expression of programmed death-ligand 1 (PD-L1) in tumor cells, and blocked the PD-1/PD-L1 signaling pathway, which improved T-cell-mediated tumor inhibition. Furthermore, ICIs combined with SDT induced immunogenic cell death by translocating calreticulin to the cell surface and then synergistically enhancing antitumor immune responses. CONCLUSION: In conclusion, sPD-1/Ce6-NBs were successfully designed. Ultrasound-mediated sPD-1/Ce6-NBs are potentially effective delivery systems for combination immunotherapy of hepatocellular carcinoma.


Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Nanoparticles/chemistry , Porphyrins/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Chlorophyllides , Humans , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Porphyrins/pharmacokinetics , Porphyrins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Burden
7.
Eur J Pharm Biopharm ; 162: 50-58, 2021 May.
Article En | MEDLINE | ID: mdl-33691169

The topical photodynamic therapy (PDT) is mainly used in the treatment of dermato-oncological diseases. The distribution and functionality of the photosensitizer Tetrahydroporphyrin-Tetratosylat (THPTS) was investigated using microscopic and spectroscopic methods after topical application to excised porcine skin followed by irradiation. The distribution of THPTS was determined by two-photon tomography combined with fluorescence lifetime imaging (TPT/FLIM) and confocal Raman microspectroscopy (CRM). The radicals were quantified and characterized by electron paramagnetic resonance (EPR) spectroscopy. Results show a penetration depth of THPTS into the skin down to around 12 ± 5 µm. A penetration of THPTS through the stratum corneum was not clearly observable after 1 h penetration time, but cannot be excluded. The irradiation within the phototherapeutic window (spectral range of visible and near infrared light in the range ≈ 650-850 nm) is needed to activate THPTS. An incubation time of 10 min showed the highest radical production. A longer incubation time affected the functionality of THPTS, whereby significant less radicals were detectable. During PDT mainly reactive oxygen species (ROS) and lipid oxygen species (LOS) are produced. Overall, the irradiation dose per se influences the radical types formed in skin. While ROS are always prominent at low doses, LOS increase at high doses, independent of previous skin treatment and the irradiation wavelength used.


Photosensitizing Agents/pharmacokinetics , Porphyrins/pharmacokinetics , Reactive Oxygen Species/metabolism , Skin/metabolism , Administration, Cutaneous , Animals , Infrared Rays , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Reactive Oxygen Species/analysis , Spatio-Temporal Analysis , Swine , Time Factors , Tissue Distribution/radiation effects
8.
ACS Appl Mater Interfaces ; 13(12): 14004-14014, 2021 Mar 31.
Article En | MEDLINE | ID: mdl-33728894

Developing novel activatable photosensitizers with excellent plasma membrane targeting ability is urgently needed for smart photodynamic therapy (PDT). Herein, a tumor acidity-activatable photosensitizer combined with a two-step bioorthogonal pretargeting strategy to anchor photosensitizers on the plasma membrane for effective PDT is developed. Briefly, artificial receptors are first anchored on the cell plasma membrane using cell-labeling agents (Az-NPs) via the enhanced permeability and retention effect to achieve the tumor cell labeling. Then, pH-sensitive nanoparticles (S-NPs) modified with dibenzocyclooctyne (DBCO) and chlorin e6 (Ce6) accumulate in tumor tissue and disassemble upon protonation of their tertiary amines in response to the acidic tumor environment, exposing the contained DBCO and Ce6. The selective, highly specific click reactions between DBCO and azide groups enable Ce6 to be anchored on the tumor cell surface. Upon laser irradiation, the cell membrane is severely damaged by the cytotoxic reactive oxygen species, resulting in remarkable cellular apoptosis. Taken together, the membrane-localized PDT by our bioorthogonal pretargeting strategy to anchor activatable photosensitizers on the plasma membrane provides a simple but effective method for enhancing the therapeutic efficacy of photosensitizers in anticancer therapy.


Cell Membrane/metabolism , Cyclooctanes/administration & dosage , Drug Delivery Systems , Neoplasms/drug therapy , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Animals , Cell Line, Tumor , Chlorophyllides , Cyclooctanes/pharmacokinetics , Cyclooctanes/therapeutic use , Humans , Mice , Nanoparticles/administration & dosage , Nanoparticles/therapeutic use , Neoplasms/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Receptors, Artificial/metabolism
9.
J Microencapsul ; 38(2): 81-88, 2021 Mar.
Article En | MEDLINE | ID: mdl-32964772

AIMS: To construct a self-assembly supramolecular drug delivery system (DDS) to co-deliver chlorin e6 (Ce6) and tripeptide tyroseroleutide (YSL) and evaluate the anti-tumour effects. METHODS: A supramolecular DDS was constructed via self-assembly of Ce6 and YSL based on π-π stacking and hydrogen-bond interaction. The size, morphology, stability, in vitro drug release, cellular uptake, cytotoxicity, pharmacokinetics analysis and pharmacodynamics analysis were respectively studied. RESULTS: Ce6-YSL nanoparticles with a uniform size of 75 ± 3.5 nm (PDI = 0.128) and monodispersed spherical morphology were constructed. The nanoparticles exhibited good stability with zeta potential -21.2 ± 1.73 mV. Under the weak acidic conditions, the accumulative drug release was 82.8% (w/w) (pH = 6.0) and 91.5% (w/w) (pH = 5.0), respectively, indicating that nanoparticles performed smart responsive properties and achieved controlled release characteristics in acidic tumour microenvironment. In addition, nanoparticles could easily enter the tumour cells and induce ROS production and inhibit cell proliferation in SMMCC-7721 cells with IC50 value 3.4 ± 0.023 µg/mL under laser irradiation. Furthermore, the nanoparticles could retain a much higher blood concentration in vivo and displayed excellent antitumor effect in tumour-bearing mice, showing no influence on body weight. CONCLUSIONS: This self-assembly supramolecular DDS can be used for combination of photodynamic therapy and chemotherapy in future research.


Antineoplastic Agents/administration & dosage , Nanoparticles/administration & dosage , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chlorophyllides , Drug Delivery Systems , Drug Liberation , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/analysis , Nanoparticles/therapeutic use , Oligopeptides/pharmacokinetics , Oligopeptides/therapeutic use , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Rats, Sprague-Dawley
10.
J Mater Chem B ; 8(40): 9251-9257, 2020 10 21.
Article En | MEDLINE | ID: mdl-32929430

The major limitations of photodynamic therapy (PDT) are the poor tissue penetration of excitation light and the neutralization of reactive oxygen species (ROS) generated by overexpressed glutathione (GSH) in cancer cells. Despite tremendous efforts to design nanoplatforms, PDT still suffers from unsatisfactory effects. Furthermore, the residual of nanomaterials in the body has restricted their clinical application. To address these issues, Janus nanocomposites containing an Yb/Er codoped NaYF4 upconverting nanocrystal head and a disulfide-bridged mesoporous organosilicon body (UCN/MON) with loaded chlorin e6 (Ce6) were designed. On one hand, the upconverting nanocrystal head can convert near-infrared (NIR) light into visible light to activate Ce6 to release ROS. On the other hand, the silica body can be degraded though a redox reaction with GSH, to not only improve the tumor selectivity of the photosensitizer by redox- and pH-triggered Ce6 release, but also diminish the concentration of GSH in cancer cells to reduce the depletion of ROS. Thereby, an enhanced PDT triggered by NIR irradiation was achieved. Furthermore, UCN/MONs showed a higher clearance rate after therapeutic actions than nonbiodegradable UCN/MSNs due to their biocompatibility. Taken together, this work revealed the potential of UCN/MONs for highly efficient and NIR-induced PDT, highlighting the prospects of UCN/MONs in the clinic.


Antineoplastic Agents/therapeutic use , Glutathione/metabolism , Nanocomposites/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Porphyrins/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Chlorophyllides , Erbium/chemistry , Erbium/radiation effects , Erbium/therapeutic use , Female , Fluorides/chemistry , Fluorides/pharmacokinetics , Fluorides/radiation effects , Fluorides/therapeutic use , Humans , Infrared Rays , Mice, Inbred BALB C , Nanocomposites/chemistry , Nanocomposites/radiation effects , Nanoparticles/chemistry , Nanoparticles/radiation effects , Nanoparticles/therapeutic use , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/radiation effects , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Porphyrins/radiation effects , Silicon Dioxide/chemistry , Silicon Dioxide/metabolism , Silicon Dioxide/pharmacokinetics , Singlet Oxygen/metabolism , Ytterbium/chemistry , Ytterbium/radiation effects , Ytterbium/therapeutic use , Yttrium/chemistry , Yttrium/pharmacokinetics , Yttrium/radiation effects , Yttrium/therapeutic use
11.
Eur J Med Chem ; 207: 112715, 2020 Dec 01.
Article En | MEDLINE | ID: mdl-32846322

This study aimed to improve the biological effectiveness and pharmacokinetic properties of chlorin e6, a second-generation photosensitizer (PS), for tumor photodynamic therapy (PDT). Herein, the novel 31-hexyloxy chlorin e6-based 152- or 131-amino acid derivatives 3a, 3b, 3c and 8 were synthesized and their photophysical properties and in vitro bioactivities such as phototoxicity against A549, HeLa and melanoma B16-F10 cells, reactive oxygen species (ROS) production and subcellular localization were evaluated. In addition, preferred target compounds were also investigated for their in vivo pharmacokinetic in SD rats and in vivo antitumor efficacies in C57BL/6 mice bearing melanoma B16-F10 cells. Apparently, simultaneous introduction of amino acid residue and n-hexyloxy chain in chlorin e6 made a significant improvement in photophysical properties, ROS production, in vitro and in vivo PDT efficacy. Encouragingly, all target compounds showed higher in vitro phototoxicity than Talaporfin, and that 3c (152-Lys) exhibited strongest phototoxicity and highest dark toxicity/phototoxicity ratio, followed by 8 (131-Asp), 3a (152-Asp) and 3b (152-Glu). Moreover, in vivo PDT antitumor efficacy of 3a, 3c and 8 was all better than that of Talaporfin, and that both 3c and 8 had stronger PDT antitumor efficiency than 3a. The overall results suggested that these novel 31-hexyloxy chlorin e6-based 152- or 131-amino acid derivatives, especially 3c and 8, might be potential antitumor candidate drugs for clinical treatment of melanoma by PDT.


Amino Acids/chemistry , Amino Acids/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , A549 Cells , Amino Acids/pharmacokinetics , Amino Acids/therapeutic use , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyllides , Drug Design , HeLa Cells , Humans , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacokinetics , Photosensitizing Agents/therapeutic use , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Rats, Sprague-Dawley
12.
Photodiagnosis Photodyn Ther ; 30: 101789, 2020 Jun.
Article En | MEDLINE | ID: mdl-32361041

Photodynamic therapy is a clinically approved procedure for the treatment of neoplastic and other non-malignant diseases. Meso-tetrakis(N-methyl-4-pyridyl)porphyrin (TMPyP) is a photosensitizing agent which has been used in many applications. However, the use of TMPyP topically is limited due to its hydrophilicity. To overcome this problem, TMPyP was loaded in ethosomes. Three ethosomal formulae (A), (B) and (C) were prepared and characterized. Preparation (A) was chosen to be used in the in vitro and in vivo study, having the greatest encapsulation efficiency, the smallest size and the highest cumulative release percentage. The results of in vitro permeation study revealed that the ethosomal TMPyP was superior to the drug in the free form with permeation flux (3.92 µg cm-2 h-1). In the in vivo animal study done on Swiss albino mice, after 19 days of Ehrlich tumor implantation, the group treated with the ethosomal preparation showed significantly smaller tumor size (143.28 ±â€¯13.2 mm3) compared to the group treated with the free TMPyP (219 ±â€¯11.9 mm3). It showed also significant longer survival time (21 days) compared to that treated with the free drug (18.2 ±â€¯1.2 days). Based on the obtained results, transdermal delivery of TMPyP was potentiated by incorporating it in ethosomes.


Drug Delivery Systems/methods , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Animals , Drug Liberation , Drug Stability , Male , Mice , Microscopy, Electron, Transmission , Particle Size , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Porphyrins/administration & dosage , Porphyrins/pharmacokinetics , Skin Absorption , Survival Analysis
13.
Mol Pharm ; 17(7): 2532-2545, 2020 07 06.
Article En | MEDLINE | ID: mdl-32407125

Recent cancer immunotherapy has attracted much attention due to high specificity and recurrence prevention of tumor. Nevertheless, its therapeutic effects are still challenging in solid cancer. To establish superior antitumor immunity, chlorin e6 (Ce6)-loaded pH sensitive carbon dots were investigated (Ce6@IDCDs). At tumoral pH 6.5, Ce6 was released four times compared with the release at physiological pH 7.4 due to an imbalance between hydrophilic and hydrophobic forces via protonation of imidazole groups in Ce6@IDCDs. This result led to the superior singlet oxygen generating activity of Ce6@IDCDs without Ce6 quenching. The maturation effects of dendritic cells after co-incubation with supernatant media obtained from Ce6@IDCDs with laser-treated cells at pH 6.5 were much higher than at physiological pH. Furthermore, Ce6@IDCDs following a laser at pH 6.5 significantly promoted calreticulin exposure and high-mobility group box 1 release, as major immunogenic cell death markers. In bilateral CT-26-bearing mice model, the Ce6@IDCDs elicited significant antitumoral effects at laser treated-primary tumor regions via therapeutic reactive oxygen species. Furthermore, Ce6@IDCDs upon laser irradiation induced a large amount of activated CD8+ T cells, natural killer cells, and mature dendritic cells recruitment into tumoral tissue and hampered tumor growth even at untreated sites approximately four-fold compared with those of others. Overall, this pH-sensitive immunoinducer can accomplish primary and distant tumor ablation via photomediated cancer immunotherapy.


Carbon/chemistry , Dendritic Cells/drug effects , Immunotherapy/methods , Nanoparticles/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Porphyrins/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Animals , Calreticulin/metabolism , Cell Death/drug effects , Cell Death/immunology , Cell Death/radiation effects , Cell Line, Tumor , Chlorophyllides , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemistry , Lasers , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Nanoparticles/radiation effects , Nanoparticles/ultrastructure , Neoplasms/immunology , Neoplasms/radiotherapy , Particle Size , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/radiation effects , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/therapeutic use , Spectroscopy, Fourier Transform Infrared , Tumor Microenvironment , Xenograft Model Antitumor Assays
14.
Nanoscale ; 12(16): 8890-8897, 2020 Apr 30.
Article En | MEDLINE | ID: mdl-32266902

Selectively attenuating the protection offered by heat shock protein 90 (HSP90), which is indispensable for the stabilization of the essential regulators of cell survival and works as a cell guardian under oxidative stress conditions, is a potential approach to improve the efficiency of cancer therapy. Here, we designed a biodegradable nanoplatform (APCN/BP-FA) based on a Zr(iv)-based porphyrinic porous coordination network (PCN) and black phosphorus (BP) sheets for efficient photodynamic therapy (PDT) by enhancing the accumulation of the nanoplatforms in the tumor area and attenuating the protection of cancer cells. Owing to the favorable degradability of BP, the nanosystem exhibited accelerated the release of the HSP90 inhibitor tanespimycin (17-AAG) and an apparent promotion in the reactive oxygen species (ROS) yield of PCN as well as expedited the degradation of the PCN-laden BP nanoplatforms. Both in vitro and in vivo results revealed that the elevated amounts of ROS and reduced cytoprotection in tumor cells were caused by the nanoplatforms. This strategy may provide a promising method for attenuating cytoprotection to aid efficient photodynamic therapy.


Metal-Organic Frameworks/chemistry , Neoplasms/drug therapy , Phosphorus/chemistry , Photochemotherapy/methods , Animals , Benzoquinones/chemistry , Benzoquinones/therapeutic use , Cell Line, Tumor , Drug Delivery Systems , Folic Acid/chemistry , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/chemistry , Lactams, Macrocyclic/therapeutic use , Metal-Organic Frameworks/pharmacokinetics , Metal-Organic Frameworks/therapeutic use , Mice , Nanostructures/chemistry , Nanostructures/therapeutic use , Neoplasms/metabolism , Phosphorus/pharmacokinetics , Phosphorus/therapeutic use , Porosity , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays , Zirconium/chemistry , Zirconium/pharmacokinetics , Zirconium/therapeutic use
15.
Nat Commun ; 11(1): 1724, 2020 04 07.
Article En | MEDLINE | ID: mdl-32265490

Vesicular photothermal therapy agents (PTAs) are highly desirable in photothermal therapy (PTT) for their excellent light-harvesting ability and versatile hollow compartments. However, up to now, the reported vesicular PTAs are generally self-assembled from small molecules like liposomes, and polymer vesicles have seldom been used as PTAs due to the unsatisfactory photothermal conversion efficiency resulting from the irregular packing of chromophores in the vesicle membranes. Here we report a nano-sized polymer vesicle from hyperbranched polyporphyrins with favorable photothermal stability and extraordinarily high photothermal efficiency (44.1%), showing great potential in imaging-guided PTT for tumors through in vitro and in vivo experiments. These excellent properties are attributed to the in situ supramolecular polymerization of porphyrin units inside the vesicle membrane into well-organized 1D monofilaments driven by π-π stacking. We believe the supramolecular polymerization-enhanced self-assembly process reported here will shed a new light on the design of supramolecular materials with new structures and functions.


Cell Survival/drug effects , Hyperthermia, Induced/methods , Nanoparticles/chemistry , Phototherapy/methods , Polymers/chemistry , Porphyrins/chemistry , Animals , Circular Dichroism , Female , Humans , Hydrophobic and Hydrophilic Interactions , MCF-7 Cells , Membranes, Artificial , Mice , Mice, Nude , Microscopy, Electron, Scanning , Molecular Dynamics Simulation , NIH 3T3 Cells , Nanoparticles/therapeutic use , Nanoparticles/ultrastructure , Polymerization , Polymers/chemical synthesis , Polymers/pharmacokinetics , Polymers/therapeutic use , Porphyrins/chemical synthesis , Porphyrins/pharmacokinetics , Porphyrins/therapeutic use , Rats , Spectrometry, Fluorescence , Temperature , Transplantation, Heterologous
16.
Int J Pharm ; 582: 119321, 2020 May 30.
Article En | MEDLINE | ID: mdl-32289483

Combining functional proteins with small molecular drugs into one entity may endow distinct synergistic advantages. However, on account of completely different physicochemical properties of such payloads, co-delivery through systemic administration for therapeutic purpose is challenging. Herein, we designed the protein-drug conjugate HSAP-DC-CAT (human serum albumin/Pt (IV)-dibenzocyclooctyne/chlorin e6-catalase) by modification of CAT and cisplatin pro-drug loaded HSA with pH-sensitive azide linker 3-(azidomethyl)-4-methyl-2,5-furandione (AzMMMan) followed by click chemistry assembly with DC. The dynamic covalent bonds between linker and proteins, on the one hand, can bridge proteins and small molecular drugs in the intermediate state for systemic delivery in the harsh in vivo environment; on the other hand, it can trigger traceless cleavage and release of drugs and proteins with full bioactivity in acidic microenvironment of tumor. The multifunctional HSAP-DC-CAT provides efficient cytosolic transduction in vitro, excellent blood half-lives after systemic administration, and significant antitumor outcome via integrated cisplatin-based chemotherapy and Ce6-based photodynamic therapy enhanced by catalase-induced manipulation of tumor hypoxia microenvironment. This study describes a universal formulation strategy for protein and small molecular drug by a bifunctional linker through amide reaction and click chemistry, with traceless in vivo release of therapeutic units.


Antioxidants/pharmacology , Breast Neoplasms/drug therapy , Catalase/pharmacology , Cisplatin/pharmacology , Photochemotherapy , Porphyrins/pharmacology , Prodrugs/pharmacology , Radiation-Sensitizing Agents/pharmacology , Serum Albumin, Human/chemistry , Animals , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catalase/chemistry , Catalase/pharmacokinetics , Cell Line, Tumor , Chlorophyllides , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Click Chemistry , Delayed-Action Preparations , Drug Carriers , Drug Compounding , Female , Hydrogen-Ion Concentration , Mice, Nude , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacokinetics , Tumor Hypoxia , Tumor Microenvironment
17.
Sci Adv ; 6(10): eaaz0575, 2020 03.
Article En | MEDLINE | ID: mdl-32181355

Although photodynamic therapy (PDT) has been clinically applied tumor hypoxia still greatly restricts the performance of this oxygen-dependent oncological treatment. The delivery of oxygen donors to tumor may produce excessive reactive oxygen species (ROS) and damage the peripheral tissues. Herein, we developed a strategy to solve the hypoxia issue by enhancing the lethality of ROS. Before PDT, the ROS-defensing system of the cancer cells was obstructed by an inhibitor to MTH1, which is a key for the remediation of ROS-caused DNA damage. As a result, both nuclei and mitochondrial DNA damages were increased, remarkably promoting cellular apoptosis. The therapeutic results demonstrated that the performance of PDT can be improved by the MTH1 inhibitor, leading to efficient cancer cell killing effect in the hypoxic tumor. This strategy makes better use of the limited oxygen, holding the promise to achieve satisfactory therapeutic effect by PDT without generating redundant cytotoxic ROS.


Antineoplastic Agents/pharmacology , DNA Repair Enzymes/genetics , DNA, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Melanoma, Experimental/drug therapy , Phosphoric Monoester Hydrolases/genetics , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Chlorophyllides , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/metabolism , DNA, Neoplasm/antagonists & inhibitors , DNA, Neoplasm/metabolism , Drug Carriers/administration & dosage , Drug Carriers/chemical synthesis , Drug Compounding/methods , Enzyme Inhibitors/chemistry , Female , Gene Expression , HCT116 Cells , HeLa Cells , Humans , Light , MCF-7 Cells , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacokinetics , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Pyrimidines/chemistry , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Nano Lett ; 20(3): 2062-2071, 2020 03 11.
Article En | MEDLINE | ID: mdl-32096643

Tumor hypoxia is the Achilles heel of oxygen-dependent photodynamic therapy (PDT), and tremendous challenges are confronted to reverse the tumor hypoxia. In this work, an oxidative phosphorylation inhibitor of atovaquone (ATO) and a photosensitizer of chlorine e6 (Ce6)-based self-delivery nanomedicine (designated as ACSN) were prepared via π-π stacking and hydrophobic interaction for O2-economized PDT against hypoxic tumors. Specifically, carrier-free ACSN exhibited an extremely high drug loading rate and avoided the excipient-induced systemic toxicity. Moreover, ACSN not only dramatically improved the solubility and stability of ATO and Ce6 but also enhanced the cellular internalization and intratumoral permeability. Abundant investigations confirmed that ACSN effectively suppressed the oxygen consumption to reverse the tumor hypoxia by inhibiting mitochondrial respiration. Benefiting from the synergistic mechanism, an enhanced PDT effect of ACSN was observed on the inhibition of tumor growth. This self-delivery system for oxygen-economized PDT might be a potential appealing clinical strategy for tumor eradication.


Mammary Neoplasms, Experimental , Nanomedicine , Nanoparticles , Photochemotherapy , Porphyrins , Animals , Cell Hypoxia/drug effects , Cell Line, Tumor , Chlorophyllides , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Mitochondria/pathology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Porphyrins/pharmacology
19.
Biomater Sci ; 8(4): 1106-1116, 2020 Feb 21.
Article En | MEDLINE | ID: mdl-31994549

In this study, we sought to design a bionanomaterial that could exert anticancer effects against primary tumors and protect against rechallenged tumors via photodynamic immunotherapy. As a biomaterial, we used an amphiphilic phenylalanine derivative of poly-gamma glutamic acid, which forms nanoparticles by self-assembly. For anticancer effects, we co-entrapped hydrophobic chlorin e6 and monophosphoryl lipid A in the core of the plain amphiphilic phenylalanine nanoparticles (AN), to generate M/C/AN. For comparison, we used plain AN and chlorin e6-loaded AN (C/AN). In vitro studies showed that B16F10 cancer cells treated with C/AN or M/C/AN generated reactive oxygen species and exhibited an enhanced surface display of calreticulin upon exposure to 660 nm light irradiation. C/AN and M/C/AN exerted similar photodynamic anticancer effects; however, M/C/AN, but not C/AN, induced in vitro dendritic cell maturation. Our biodistribution study revealed that C/AN and M/C/AN showed higher accumulation at the tumor tissues compared to that seen in the free chlorin e6-treated group. In B16F10 tumor-bearing mice, the intravenous injection of C/AN or M/C/AN showed similar photodynamic anticancer effects against primary tumors. However, the growth of rechallenged tumors was more significantly inhibited in the M/C/AN group compared to the C/AN group. At day 40 after inoculation of the primary tumor, M/C/AN-treated mice showed 100% survival, whereas the other groups showed 0% survival. In the tumor microenvironment, higher infiltration of CD8+ T cells was observed in the M/C/AN group compared to the other groups. Our results suggest that AN co-loaded with a photosensitizer and an immune stimulant may hold great potential for use in photodynamic immunotherapy to inhibit both primary and metastatic tumors.


Biomimetics/methods , Lipid A/analogs & derivatives , Melanoma, Experimental/drug therapy , Porphyrins/administration & dosage , Administration, Intravenous , Animals , Capsules , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyllides , Immunotherapy , Lipid A/administration & dosage , Lipid A/chemistry , Lipid A/pharmacokinetics , Lipid A/pharmacology , Melanoma, Experimental/immunology , Mice , Nanoparticles , Photochemotherapy , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/chemistry , Polymers/chemistry , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Porphyrins/pharmacology , Tissue Distribution , Tumor Microenvironment/drug effects
20.
Curr Drug Discov Technol ; 17(5): 661-669, 2020.
Article En | MEDLINE | ID: mdl-30973109

BACKGROUND: Detailed photochemical and photocytotoxicity studies of two new porphyrins: 5,10,15,20-meso-tetrakis-(4-acetoxy-3-methoxyphenyl) porphyrin (P2.1) and 5-(4-hydroxy-3- methoxyphenyl)-10,15,20-tris-(4-acetoxy-3-methoxyphenyl)porphyrin (P2.2) are reported, as potential candidates for theranostics. For powdered samples of P2.1 and P2.2 adsorbed onto a powdered biocompatible substrate, polyethylene glycol (PEG), a concentration study was performed, correlating the fluorescence emission intensity with sample absorption to determine the useful concentration range for photodynamic therapy of cancer (PDT) in which aggregation does not occur. Cytotoxicity studies were performed in dark and illuminated conditions. METHODS: The laser induced luminescence set-up is home-made, a N2 laser is used as the excitation source and a time gated charged-coupled device (ICCD) as the detector. Fluorescence lifetime determinations were made using pulsed light sources from the excitation LEDs and measures of the fluorescence intensities at different time delays after the excitation pulse. The singlet oxygen formation quantum yields ΦΔ measurements were obtained by comparing the total area of the emission spectra for the reference compound and also for the samples under study in the same solvent and with the same optical density at the excitation wavelength (405 nm). An integrating sphere for relative and absolute measurements was used in this work as an alternative methodology to obtain the values for the fluorescence emission quantum yields (ΦF) of the adsorbed porphyrin under study. The cytotoxicity evaluation was made in the dark and under irradiation, using four different human tumor cell lines and one non-tumor primary cell culture. RESULTS: In order to establish the useful range of concentrations of the sensitizer for PDT, and due to the use of powdered samples, a special methodology was needed: the variations of the fluorescence lifetimes and fluorescence quantum yields were evaluated as a function of the concentration of the dye, measured by (1-R)*fdye. Both ΦF and τF are constant in the range from 0.002 to about 0.050 µmol g-1, and only after that a concentration quenching effect becomes visible, decreasing both ΦF and τF. This methodology is based in the correlations established between the Remission Function values and ΦF and τF obtained for increasing values of the sensitizer concentrations. CONCLUSIONS: The study of the aggregation effects of P2.1 and P2.2 porphyrins into a PEG matrix allowed us to determine the usable concentration range for photodynamic therapy use, where the aggregation of porphyrins decreases, therefore reducing the PDT action. The use of an integrating sphere for relative and absolute measurements of fluorescence quantum yields and also the lifetime studies as a function of the dye loading confirms the useful range for the use of P2.1 and P2.2 in PEG as powdered samples. The determination of the GI50, the porphyrin concentration which inhibits 50% of the cell growth, evidences that P2.2, the A3B porphyrin overtakes P2.1 (the A4 porphyrin) in terms of PDT efficiency and both porphyrins are much better PDT agents than the unsubstituted porphyrin, TPP. These data clearly show that porphyrins P2.2 and P2.1 exhibit an excellent behaviour in terms of its photocytotoxicity. These results encourage us to pursuit in the study of this family of porphyrins in which a balance of hydrophobic versus hydrophilic substituents in the phenyl group was achieved.


Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Porphyrins/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Carriers/chemistry , Drug Evaluation, Preclinical , Humans , Lasers, Gas , Nanoparticles/chemistry , Neoplasms/pathology , Photochemotherapy/instrumentation , Photosensitizing Agents/pharmacokinetics , Polyethylene Glycols/chemistry , Porphyrins/pharmacokinetics , Theranostic Nanomedicine/instrumentation , Theranostic Nanomedicine/methods , Tissue Distribution/radiation effects
...