Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; 11(17): e2302872, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38445882

RESUMO

Glioblastoma (GBM) is hard to treat due to cellular invasion into functioning brain tissues, limited drug delivery, and evolved treatment resistance. Recurrence is nearly universal even after surgery, chemotherapy, and radiation. Photodynamic therapy (PDT) involves photosensitizer administration followed by light activation to generate reactive oxygen species at tumor sites, thereby killing cells or inducing biological changes. PDT can ablate unresectable GBM and sensitize tumors to chemotherapy. Verteporfin (VP) is a promising photosensitizer that relies on liposomal carriers for clinical use. While lipids increase VP's solubility, they also reduce intracellular photosensitizer accumulation. Here, a pure-drug nanoformulation of VP, termed "NanoVP", eliminating the need for lipids, excipients, or stabilizers is reported. NanoVP has a tunable size (65-150 nm) and 1500-fold higher photosensitizer loading capacity than liposomal VP. NanoVP shows a 2-fold increase in photosensitizer uptake and superior PDT efficacy in GBM cells compared to liposomal VP. In mouse models, NanoVP-PDT improved tumor control and extended animal survival, outperforming liposomal VP and 5-aminolevulinic acid (5-ALA). Moreover, low-dose NanoVP-PDT can safely open the blood-brain barrier, increasing drug accumulation in rat brains by 5.5-fold compared to 5-ALA. NanoVP is a new photosensitizer formulation that has the potential to facilitate PDT for the treatment of GBM.


Assuntos
Neoplasias Encefálicas , Sistemas de Liberação de Medicamentos , Fotoquimioterapia , Fármacos Fotossensibilizantes , Verteporfina , Animais , Fotoquimioterapia/métodos , Verteporfina/farmacologia , Verteporfina/uso terapêutico , Camundongos , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Glioblastoma/tratamento farmacológico , Nanopartículas/química , Modelos Animais de Doenças , Humanos , Ratos , Lipossomos , Linhagem Celular Tumoral , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos
2.
Sci Adv ; 9(36): eadi3441, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37672582

RESUMO

Fluorescence-guided intervention can bolster standard therapies by detecting and treating microscopic tumors before lethal recurrence. Tremendous progress in photoimmunotherapy and nanotechnology has been made to treat metastasis. However, many are lost in translation due to heterogeneous treatment effects. Here, we integrate three technological advances in targeted photo-activable multi-agent liposome (TPMAL), fluorescence-guided intervention, and laser endoscopy (ML7710) to improve photoimmunotherapy. TPMAL consists of a nanoliposome chemotherapy labeled with fluorophores for tracking and photosensitizer immunoconjugates for photoimmunotherapy. ML7710 is connected to Modulight Cloud to capture and analyze multispectral emission from TPMAL for fluorescence-guided drug delivery (FGDD) and fluorescence-guided light dosimetry (FGLD) in peritoneal carcinomatosis mouse models. FGDD revealed that TPMAL enhances drug delivery to metastases by 14-fold. ML7710 captured interpatient variability in TPMAL uptake and prompted FGLD in >50% of animals. By combining TPMAL, ML7710, and fluorescence-guided intervention, variation in treatment response was substantially reduced and tumor control improved without side effects.


Assuntos
Neoplasias Peritoneais , Animais , Camundongos , Neoplasias Peritoneais/terapia , Imunoterapia , Fototerapia , Nanotecnologia , Sistemas de Liberação de Medicamentos , Lipossomos
3.
Metabolites ; 12(11)2022 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-36422237

RESUMO

Breast cancer is the most diagnosed cancer type in women, with it being the second most deadly cancer in terms of total yearly mortality. Due to the prevalence of this disease, better methods are needed for both detection and treatment. Reduced nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) are autofluorescent biomarkers that lend insight into cell and tissue metabolism. As such, we developed an endoscopic device to measure these metabolites in tissue to differentiate between malignant tumors and normal tissue. We performed initial validations in liquid phantoms as well as compared to a previously validated redox imaging system. We also imaged ex vivo tissue samples after modulation with carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP) and a combination of rotenone and antimycin A. We then imaged the rim and the core of MDA-MB-231 breast cancer tumors, with our results showing that the core of a cancerous lesion has a significantly higher optical redox ratio ([FAD]/([FAD] + [NADH])) than the rim, which agrees with previously published results. The mouse muscle tissues exhibited a significantly lower FAD, higher NADH, and lower redox ratio compared to the tumor core or rim. We also used the endoscope to measure NADH and FAD after photodynamic therapy treatment, a light-activated treatment methodology. Our results found that the NADH signal increases in the malignancy rim and core, while the core of cancers demonstrated a significant increase in the FAD signal.

4.
J Biomed Opt ; 25(9)2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32981239

RESUMO

SIGNIFICANCE: Previous studies have been performed to image photosensitizers in certain organs and tumors using fluorescence laminar optical tomography. Currently, no work has yet been published to quantitatively compare the signal compensation of fluorescence laminar optical tomography with two-dimensional (2-D) imaging in tissues. AIM: The purpose of this study is to quantify the benefit that fluorescence laminar optical tomography holds over 2-D imaging. We compared fluorescence laminar optical tomography with maximum intensity projection imaging to simulate 2-D imaging, as this would be the most similar and stringent comparison. APPROACH: A capillary filled with a photosensitizer was placed in a phantom and ex vivo rodent brains, with fluorescence laminar optical tomography and maximum intensity projection images obtained. The signal loss in the Z direction was quantified and compared to see which methodology could compensate better for signal loss caused by tissue attenuation. RESULTS: The results demonstrated that we can reconstruct a capillary filled with benzoporphyrin derivative photosensitizers faithfully in phantoms and in ex vivo rodent brain tissues using fluorescence laminar optical tomography. We further demonstrated that we can better compensate for signal loss when compared with maximum intensity projection imaging. CONCLUSIONS: Using fluorescence laminar optical tomography (FLOT), one can compensate for signal loss in deeper parts of tissue when imaging in ex vivo rodent brain tissue compared with maximum intensity projection imaging.


Assuntos
Fármacos Fotossensibilizantes , Tomografia Óptica , Animais , Encéfalo/diagnóstico por imagem , Imagens de Fantasmas , Fármacos Fotossensibilizantes/farmacologia , Roedores
5.
Photochem Photobiol ; 96(2): 301-309, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31441057

RESUMO

Fluorescence-guided surgery (FGS) is routinely utilized in clinical centers around the world, whereas the combination of FGS and photodynamic therapy (PDT) has yet to reach clinical implementation and remains an active area of translational investigations. Two significant challenges to the clinical translation of PDT for brain cancer are as follows: (1) Limited light penetration depth in brain tissues and (2) Poor selectivity and delivery of the appropriate photosensitizers. To address these shortcomings, we developed nanoliposomal protoporphyrin IX (Nal-PpIX) and nanoliposomal benzoporphyrin derivative (Nal-BPD) and then evaluated their photodynamic effects as a function of depth in tissue and light fluence using rat brains. Although red light penetration depth (defined as the depth at which the incident optical energy drops to 1/e, ~37%) is typically a few millimeters in tissues, we demonstrated that the remaining optical energy could induce PDT effects up to 2 cm within brain tissues. Photobleaching and singlet oxygen yield studies between Nal-BPD and Nal-PpIX suggest that deep-tissue PDT (>1 cm) is more effective when using Nal-BPD. These findings indicate that Nal-BPD-PDT is more likely to generate cytotoxic effects deep within the brain and allow for the treatment of brain invading tumor cells centimeters away from the main, resectable tumor mass.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacocinética , Animais , Neoplasias Encefálicas/metabolismo , Fármacos Fotossensibilizantes/uso terapêutico , Ratos , Espectrometria de Fluorescência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA