Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Photochem Photobiol B ; 255: 112910, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663337

RESUMO

The prognosis for patients with advanced-stage pancreatic ductal adenocarcinoma (PDAC) remains dismal. It is generally accepted that combination cancer therapies offer the most promise, such as Folforinox, despite their associated high toxicity. This study addresses the issue of chemoresistance by introducing a complementary dual priming approach to attenuate the DNA repair mechanism and to improve the efficacy of a type 1 topoisomerase (Top1) inhibitor. The result is a regimen that integrates drug-repurposing and nanotechnology using 3 clinically relevant FDA-approved agents (1) Top1 inhibitor (irinotecan) at subcytotoxic doses (2) benzoporphyrin derivative (BPD) as a photoactive molecule for photodynamic priming (PDP) to improve the delivery of irinotecan within the cancer cell and (3) minocycline priming (MNP) to modulate DNA repair enzyme Tdp1 (tyrosyl-DNA phosphodiesterase) activity. We demonstrate in heterotypic 3D cancer models that incorporate cancer cells and pancreatic cancer-associated fibroblasts that simultaneous targeting of Tdp1 and Top1 were significantly more effective by employing MNP and photoactivatable multi-inhibitor liposomes encapsulating BPD and irinotecan compared to monotherapies or a cocktail of dual or triple-agents. These data are encouraging and warrant further work in appropriate animal models to evolve improved therapeutic regimens.


Assuntos
Carcinoma Ductal Pancreático , Irinotecano , Minociclina , Neoplasias Pancreáticas , Fotoquimioterapia , Humanos , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/metabolismo , Linhagem Celular Tumoral , Minociclina/farmacologia , Minociclina/uso terapêutico , Irinotecano/farmacologia , Irinotecano/uso terapêutico , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia , Diester Fosfórico Hidrolases/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Inibidores da Topoisomerase I/farmacologia , Inibidores da Topoisomerase I/uso terapêutico , Inibidores da Topoisomerase I/química , Lipossomos/química
2.
J Photochem Photobiol B ; 250: 112811, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38000171

RESUMO

Desmoplasia in pancreatic ductal adenocarcinoma (PDAC) is characterized by elevated levels of tumor collagen. Desmoplasia restricts drug delivery in PDAC, contributes to treatment resistance, and is associated with poor survival outcomes. We have previously shown that photodynamic therapy (PDT)-based treatment remediates desmoplasia in orthotopic PDAC tumors by reducing second harmonic generation signals from collagen by >90% and by reducing collagen alignment by >103-fold [19]. Remediating desmoplasia correlated with improved survival outcomes in mice. To understand this phenomenon at a fundamental level, it is important to dissect the impact of therapy on collagen subtypes. In this study, we demonstrate that immunofluorescence profiling of collagen subtypes I, II, III and IV in PDAC tumors 72 h following multiple treatment regimens is predictive of long-term outcomes. Treatment regimens include nanoliposomal irinotecan chemotherapy (nal-IRI; akin to ONIVYDE™), a combination of nal-IRI chemotherapy with PDT encapsulated in a single photoactivable multi-inhibitor liposome (PMIL) and an EGFR-targeted PMIL construct (TPMIL). Results show that the relative tumor content of collagen I, II and III was inversely correlated with overall survival (P ≤ 0.0013, P ≤ 0.0001, P ≤ 0.0011, respectively), while, surprisingly, the relative tumor content of collagen IV was directly correlated with overall survival (P ≤ 0.0001). Similar relationships were observed between the relative tumor content of collagen subtypes and the residual tumor volume at day 88 following treatment. Considering that the relationship between collagen subtypes and treatment outcomes is observed across multiple treatment regimens, immunofluorescence profiling at 72 h following treatment appears to be predictive of tumor growth inhibition and survival in PDAC. Early immunofluorescence collagen subtype profiling may therefore aid in treatment personalization and may inform the dosimetry and scheduling of combination regimens for PDAC, such as chemotherapy and emerging PDT-based combinations, to maximize patient survival benefit.


Assuntos
Neoplasias Pancreáticas , Humanos , Animais , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Resultado do Tratamento , Colágeno , Colágeno Tipo I , Imunofluorescência , Lipossomos
3.
Adv Sci (Weinh) ; 9(24): e2104594, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35748165

RESUMO

Desmoplasia is characteristic of pancreatic ductal adenocarcinoma (PDAC), which exhibits 5-year survival rates of 3%. Desmoplasia presents physical and biochemical barriers that contribute to treatment resistance, yet depleting the stroma alone is unsuccessful and even detrimental to patient outcomes. This study is the first demonstration of targeted photoactivable multi-inhibitor liposomes (TPMILs) that induce both photodynamic and chemotherapeutic tumor insult, while simultaneously remediating desmoplasia in orthotopic PDAC. TPMILs targeted with cetuximab (anti-EGFR mAb) contain lipidated benzoporphyrin derivative (BPD-PC) photosensitizer and irinotecan. The desmoplastic tumors comprise human PDAC cells and patient-derived cancer-associated fibroblasts. Upon photoactivation, the TPMILs induce 90% tumor growth inhibition at only 8.1% of the patient equivalent dose of nanoliposomal irinotecan (nal-IRI). Without EGFR targeting, PMIL photoactivation is ineffective. TPMIL photoactivation is also sixfold more effective at inhibiting tumor growth than a cocktail of Visudyne-photodynamic therapy (PDT) and nal-IRI, and also doubles survival and extends progression-free survival by greater than fivefold. Second harmonic generation imaging reveals that TPMIL photoactivation reduces collagen density by >90% and increases collagen nonalignment by >103 -fold. Collagen nonalignment correlates with a reduction in tumor burden and survival. This single-construct phototoxic, chemotherapeutic, and desmoplasia-remediating regimen offers unprecedented opportunities to substantially extend survival in patients with otherwise dismal prognoses.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/tratamento farmacológico , Receptores ErbB/uso terapêutico , Humanos , Irinotecano/farmacologia , Irinotecano/uso terapêutico , Lipossomos/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias Pancreáticas
4.
Nano Res ; 14(5): 1344-1354, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33717420

RESUMO

The significance and ability for receptor targeted nanoliposomes (tNLs) to bind to their molecular targets in solid tumors in vivo has been questioned, particularly as the efficiency of their tumor accumulation and selectivity is not always predictive of their efficacy or molecular specificity. This study presents, for the first time, in situ NIR molecular imaging-based quantitation of the in vivo specificity of tNLs for their target receptors, as opposed to tumor selectivity, which includes influences of enhanced tumor permeability and retention. Results show that neither tumor delivery nor selectivity (tumor-to-normal ratio) of cetuximab and IRDye conjugated tNLs correlate with EGFR expression in U251, U87 and 9L tumors, and in fact underrepresent their imaging-derived molecular specificity by up to 94.2%. Conversely, their in vivo specificity, which we quantify as the concentration of tNL-reported tumor EGFR provided by NIR molecular imaging, correlates positively with EGFR expression levels in vitro and ex vivo (Pearson's r= 0.92 and 0.96, respectively). This study provides a unique opportunity to address the problematic disconnect between tNL synthesis and in vivo specificity. The findings encourage their continued adoption as platforms for precision medicine, and facilitates intelligent synthesis and patient customization in order to improve safety profiles and therapeutic outcomes.

5.
Cancers (Basel) ; 14(1)2021 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-35008362

RESUMO

The newly developed multimodal imaging system combining raster-scan optoacoustic (OA) microscopy and fluorescence (FL) wide-field imaging was used for characterizing the tumor vascular structure with 38/50 µm axial/transverse resolution and assessment of photosensitizer fluorescence kinetics during treatment with novel theranostic agents. A multifunctional photoactivatable multi-inhibitor liposomal (PMILs) nano platform was engineered here, containing a clinically approved photosensitizer, Benzoporphyrin derivative (BPD) in the bilayer, and topoisomerase I inhibitor, Irinotecan (IRI) in its inner core, for a synergetic therapeutic impact. The optimized PMIL was anionic, with the hydrodynamic diameter of 131.6 ± 2.1 nm and polydispersity index (PDI) of 0.05 ± 0.01, and the zeta potential between -14.9 ± 1.04 to -16.9 ± 0.92 mV. In the in vivo studies on BALB/c mice with CT26 tumors were performed to evaluate PMILs' therapeutic efficacy. PMILs demonstrated the best inhibitory effect of 97% on tumor growth compared to the treatment with BPD-PC containing liposomes (PALs), 81%, or IRI containing liposomes (L-[IRI]) alone, 50%. This confirms the release of IRI within the tumor cells upon PMILs triggering by NIR light, which is additionally illustrated by FL monitoring demonstrating enhancement of drug accumulation in tumor initiated by PDT in 24 h after the treatment. OA monitoring revealed the largest alterations of the tumor vascular structure in the PMILs treated mice as compared to BPD-PC or IRI treated mice. The results were further corroborated with histological data that also showed a 5-fold higher percentage of hemorrhages in PMIL treated mice compared to the control groups. Overall, these results suggest that multifunctional PMILs simultaneously delivering PDT and chemotherapy agents along with OA and FL multi-modal imaging offers an efficient and personalized image-guided platform to improve cancer treatment outcomes.

6.
Nanophotonics ; 10(12): 3199-3214, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37485044

RESUMO

Photodynamic priming (PDP), a collateral effect of photodynamic therapy, can transiently alter the tumor microenvironment (TME) beyond the cytotoxic zone. Studies have demonstrated that PDP increases tumor permeability and modulates immune-stimulatory effects by inducing immunogenic cell death, via the release of damage-associated molecular patterns and tumor-associated antigens. Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest of cancers with a stubborn immunosuppressive TME and a dense stroma, representing a challenge for current molecular targeted therapies often involving macromolecules. We, therefore, tested the hypothesis that PDP's TME modulation will enable targeted therapy and result in immune stimulation. Using triple-receptor-targeted photoimmuno-nanoconjugate (TR-PINs)-mediated PDP, targeting epidermal growth factor receptor, transferrin receptor, and human epidermal growth factor receptor 2 we show light dose-dependent TR-PINs mediated cytotoxicity inhuman PDA Ccells (MIAPaCa-2),co-cultured with human pancreatic cancer-associated fibroblasts (PCAFs) in spheroids. Furthermore, TR-PINs induced the expression of heat shock proteins (Hsp60, Hsp70), Calreticulin, and high mobility group box 1 in a light dose and time-dependent manner.TR-PINs-mediated T cell activation was observed in co-cultures of immune cells with the MIA PaCa-2-PCAF spheroids. Both CD4+ T and CD8+ T cells showed light dose and time-dependant antitumor reactivity by upregulating degranulation marker CD107a and interferon-gamma post-PDP. Substantial tumor cell death in immune cell-spheroid co-cultures by day 3 shows the augmentation by antitumor T cell activation and their ability to recognize tumors for a light dose-dependent kill. These data confirm enhanced destruction of heterogeneous pancreatic spheroids mediated by PDP-induced phototoxicity, TME modulation and increased immunogenicity with targeted nanoconstructs.

7.
J Clin Med ; 9(8)2020 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-32726945

RESUMO

Receptor heterogeneity in cancer is a major limitation of molecular targeting for cancer therapeutics. Single-receptor-targeted treatment exerts selection pressures that result in treatment escape for low-receptor-expressing tumor subpopulations. To overcome this potential for heterogeneity-driven resistance to molecular targeted photodynamic therapy (PDT), we present for the first time a triple-receptor-targeted photoimmuno-nanoconjugate (TR-PIN) platform. TR-PIN functionalization with cetuximab, holo-transferrin, and trastuzumab conferred specificity for epidermal growth factor receptor (EGFR), transferrin receptor (TfR), and human epidermal growth factor receptor 2 (HER-2), respectively. The TR-PINs exhibited up to a 24-fold improvement in cancer cell binding compared with EGFR-specific cetuximab-targeted PINs (Cet-PINs) in low-EGFR-expressing cell lines. Photodestruction using TR-PINs was significantly higher than the monotargeted Cet-PINs in heterocellular 3D in vitro models of heterogeneous pancreatic ductal adenocarcinoma (PDAC; MIA PaCa-2 cells) and heterogeneous head and neck squamous cell carcinoma (HNSCC, SCC9 cells) containing low-EGFR-expressing T47D (high TfR) or SKOV-3 (high HER-2) cells. Through their capacity for multiple tumor target recognition, TR-PINs can serve as a unique and amenable platform for the effective photodynamic eradication of diverse tumor subpopulations in heterogeneous cancers to mitigate escape for more complete and durable treatment responses.

8.
Cancers (Basel) ; 12(6)2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32485915

RESUMO

Extensive desmoplasia is a hallmark of pancreatic ductal adenocarcinoma (PDAC), which frequently associates with treatment resistance. Recent findings indicate that a combination of photodynamic therapy and the multi-kinase inhibitor cabozantinib achieved local tumor control and a significant decrease in tumor metastases in preclinical PDAC models, but the underlying therapeutic mechanisms remain unclear. This study elucidates the molecular basis of this multi-agent regimen, focusing on the role of MET signaling. Since MET activation stems from its interaction with hepatocyte growth factor (HGF), which is typically secreted by fibroblasts, we developed heterotypic PDAC microtumor models that recapitulate these interactions. In these models, MET signaling can be constitutively activated through paracrine and autocrine mechanisms. Photodynamic therapy caused significant elevations in HGF secretion by fibroblasts, suggesting it plays a complex role in the modulation of the paracrine HGF-MET signaling cascade in desmoplastic tumors. Blocking MET phosphorylation with adjuvant cabozantinib caused a significant improvement in photodynamic therapy efficacy, most notably by elevating spheroid necrosis at low radiant exposures. These findings highlight that adjuvant photodynamic therapy can augment chemotherapy efficacies, and potentially achieve improved management of desmoplastic PDAC in a more tolerable manner.

9.
J Porphyr Phthalocyanines ; 24(11n12): 1320-1360, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-37425217

RESUMO

Photodynamic therapy is a photochemistry-based approach, approved for the treatment of several malignant and non-malignant pathologies. It relies on the use of a non-toxic, light activatable chemical, photosensitizer, which preferentially accumulates in tissues/cells and, upon irradiation with the appropriate wavelength of light, confers cytotoxicity by generation of reactive molecular species. The preferential accumulation however is not universal and, depending on the anatomical site, the ratio of tumor to normal tissue may be reversed in favor of normal tissue. Under such circumstances, control of the volume of light illumination provides a second handle of selectivity. Singlet oxygen is the putative favorite reactive molecular species although other entities such as nitric oxide have been credibly implicated. Typically, most photosensitizers in current clinical use have a finite quantum yield of fluorescence which is exploited for surgery guidance and can also be incorporated for monitoring and treatment design. In addition, the photodynamic process alters the cellular, stromal, and/or vascular microenvironment transiently in a process termed photodynamic priming, making it more receptive to subsequent additional therapies including chemo- and immunotherapy. Thus, photodynamic priming may be considered as an enabling technology for the more commonly used frontline treatments. Recently, there has been an increase in the exploitation of the theranostic potential of photodynamic therapy in different preclinical and clinical settings with the use of new photosensitizer formulations and combinatorial therapeutic options. The emergence of nanomedicine has further added to the repertoire of photodynamic therapy's potential and the convergence and co-evolution of these two exciting tools is expected to push the barriers of smart therapies, where such optical approaches might have a special niche. This review provides a perspective on current status of photodynamic therapy in anti-cancer and anti-microbial therapies and it suggests how evolving technologies combined with photochemically-initiated molecular processes may be exploited to become co-conspirators in optimization of treatment outcomes. We also project, at least for the short term, the direction that this modality may be taking in the near future.

10.
Nano Lett ; 19(11): 7573-7587, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31518145

RESUMO

Despite untiring efforts to develop therapies for pancreatic ductal adenocarcinoma (PDAC), survival statistics remain dismal, necessitating distinct approaches. Photodynamic priming (PDP), which improves drug delivery and combination regimens, as well as tumor photodestruction are key attributes of photodynamic therapy (PDT), making it a distinctive clinical option for PDAC. Localized, high-payload nanomedicine-assisted delivery of photosensitizers (PSs), with molecular specificity and controlled photoactivation, thus becomes critical in order to reduce collateral toxicity during more expansive photodynamic activation procedures with curative intent. As such, targeted photoactivable lipid-based nanomedicines are an ideal candidate but have failed to provide greater than two-fold cancer cell selectivity, if at all, due to their extensive multivariant physical, optical, and chemical complexity. Here, we report (1) a systematic multivariant tuning approach to engineer (Cet, anti-EGFR mAb) photoimmunonanoconjugates (PINs), and (2) stroma-rich heterotypic PDAC in vitro and in vivo models incorporating patient-derived pancreatic cancer-associated fibroblasts (PCAFs) that recapitulate the desmoplasia observed in the clinic. These offer a comprehensive, disease-specific framework for the development of Cet-PINs. Specificity-tuning of the PINs, in terms of PS lipid anchoring, electrostatic modulation, Cet orientation, and Cet surface densities, achieved ∼16-fold binding specificities and rapid penetration of the heterotypic organoids within 1 h, thereby providing a ∼16-fold enhancement in molecular targeted NIR photodestruction. As a demonstration of their inherent amenability for multifunctionality, encapsulation of high payloads of gemcitabine hydrochloride, 5-fluorouracil, and oxaliplatin within the Cet-PINs further improved their antitumor efficacy in the heterotypic organoids. In heterotypic desmoplastic tumors, the Cet-PINs efficiently penetrated up to 470 µm away from blood vessels, and photodynamic activation resulted in substantial tumor necrosis, which was not elicited in T47D tumors (low EGFR) or when using untargeted constructs in both tumor types. Photodynamic activation of the Cet-PINs in the heterotypic desmoplastic tumors resulted in collagen photomodulation, with a 1.5-fold reduction in collagen density, suggesting that PDP may also hold potential for conquering desmoplasia. The in vivo safety profile of photodynamic activation of the Cet-PINs was also substantially improved, as compared to the untargeted constructs. While treatment using the Cet-PINs did not cause any detriment to the mice's health or to healthy proximal tissue, photodynamic activation of untargeted constructs induced severe acute cachexia and weight loss in all treated mice, with substantial peripheral skin necrosis, muscle necrosis, and bowel perforation. This study is the first report demonstrating the true value of molecular targeting for NIR-activable PINs. These constructs integrate high payload delivery, efficient photodestruction, molecular precision, and collagen photomodulation in desmoplastic PDAC tumors in a single treatment using a single construct. Such combined PIN platforms and heterocellular models open up an array of further multiplexed combination therapies to synergistically control desmoplastic tumor progression and extend PDAC patient survival.


Assuntos
Carcinoma Ductal Pancreático/tratamento farmacológico , Imunoconjugados/uso terapêutico , Nanoconjugados/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/patologia , Carcinoma Ductal Pancreático/patologia , Sistemas de Liberação de Medicamentos/métodos , Receptores ErbB/antagonistas & inibidores , Humanos , Imunoconjugados/administração & dosagem , Camundongos , Nanoconjugados/administração & dosagem , Nanomedicina/métodos , Organoides/efeitos dos fármacos , Organoides/patologia , Neoplasias Pancreáticas/patologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/administração & dosagem
11.
Photochem Photobiol ; 95(1): 364-377, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30125366

RESUMO

With the rapidly emerging designs and applications of light-activated, photodynamic therapy (PDT)-based nanoconstructs, photonanomedicines (PNMs), an unmet need exists to establish whether conventional methods of photochemical and photophysical characterization of photosensitizers are relevant for evaluating new PNMs in order to intelligently guide their design. As a model system, we build on the clinical formulation of benzoporphyrin derivative (BPD), Visudyne® , by developing a panel of nanolipid formulations entrapping new lipidated chemical variants of BPD with differing chemical, photochemical and photophysical properties. These are 16:0 and 20:0 lysophosphocholine-BPD (16:0/20:0 BPD-PC), DSPE-PEG-BPD and BPD-cholesterol. We show that Visudyne® was the most phototoxic formulation to OVCAR-5 cells, and the least effective was liposomal DSPE-PEG-BPD. However, these differences did not match their optical, photophysical and photochemical properties, as the static BPD quenching was highest in Visudyne, which also exhibited the lowest generation of singlet oxygen. Furthermore, we establish that OVCAR-5 cell phototoxicity also does not correlate with rates of photosensitizer photobleaching and fluorescence quantum yields in any nanolipid formulations. These findings warrant critical future studies into subcellular targets and molecular mechanisms of phototoxicity of photodynamic nanoconstructs, as more reliable prognostic surrogates for predicting efficacy to appropriately and intelligently guide their design.


Assuntos
Lipídeos/química , Nanopartículas/química , Neoplasias Ovarianas/tratamento farmacológico , Processos Fotoquímicos , Porfirinas/química , Porfirinas/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Ovarianas/patologia
12.
Photochem Photobiol ; 95(1): 419-429, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30499113

RESUMO

A major objective in developing new treatment approaches for lethal tumors is to reduce toxicity to normal tissues while maintaining therapeutic efficacy. Photodynamic therapy (PDT) provides a mechanistically distinct approach to treat tumors without the systemic toxicity of chemotherapy drugs. PDT involves the light-based activation of a small molecule, a photosensitizer (PS), to generate reactive molecular species (RMS) that are toxic to target tissue. Depending on the PS localization, various cellular and subcellular components can be targeted, causing selective photodamage. It has been shown that targeted lysosomal photodamage followed by, or simultaneous with, mitochondrial photodamage using two different PS results in a considerable enhancement in PDT efficacy. Here, two liposomal formulations of benzoporphyrin derivative (BPD): (1) Visudyne (clinically approved) and (2) an in-house formulation entrapping a lipid conjugate of BPD are used in combination with direct PS localization to mitochondria, endoplasmic reticulum and lysosomes, enabling simultaneous photodamage to all three organelles using a single wavelength of light. Building on findings by our group, and others, this study demonstrates, for the first time in a 3D model for ovarian cancer, that BPD-mediated photodestruction of lysosomes and mitochondria/ER significantly enhances PDT efficacy at lower light doses than treatment with either PS formulation alone.


Assuntos
Lipídeos/química , Lipossomos , Modelos Biológicos , Neoplasias Ovarianas/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/uso terapêutico , Verteporfina/uso terapêutico , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Quimioterapia Combinada , Feminino , Humanos , Fármacos Fotossensibilizantes/administração & dosagem , Porfirinas/administração & dosagem , Verteporfina/administração & dosagem
13.
Lasers Surg Med ; 50(5): 499-505, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29527710

RESUMO

OBJECTIVE: A relatively low level of lysosomal photodamage has been shown capable of promoting the efficacy of photodamage simultaneously or subsequently directed to mitochondrial/ER sites. The procedure has hitherto involved the use of two photosensitizing agents that require irradiation at two different wavelengths and different formulation techniques. This, together with different pharmacokinetic profiles of the photosensitizers, adds a layer of complexity to a protocol that we have sought to circumvent. In this study, liposomal formulations were used to direct photodamage created by benzoporphyrin derivative (BPD, Verteporfin) to lysosomes, mitochondria and the ER. This resulted in the development of an optimal targeting profile using a single agent and a single wavelength of activating irradiation. MATERIALS/METHODS: These studies were carried out in monolayer cultures of OVCAR5 tumor cells. BPD localization was modified by lipid anchoring and formulation in liposomes, and was assessed by fluorescence microscopy. Irradiation was carried out at 690 ± 10 nm with photodamage assessed also using fluorescent probes and microscopy. RESULTS: BPD normally localizes in a wide variety of sub-cellular loci that include both mitochondria and the ER, but lysosomes are spared from photodamage. Using a liposomal formulation containing BPD anchored to a lipid resulted in the targeting of lysosomes. A mixture of liposomes containing "free" and "anchored" BPD was shown to significantly promote photokilling. Eliminating cholesterol from the formulation of the anchored product enhanced lysosomal photodamage; prior studies had revealed that excess cholesterol can have a cytoprotective effect when lysosomes are the PDT target. DISCUSSION: The ability of a liposomal formulation to change localization patterns permits directing photodynamic therapy toward specific sub-cellular loci, thereby promoting photokilling. Incorporating chemotherapeutic agents into such formulations could represent a logical next step in assessing the ability of directed photodamage to enhance tumor eradication. Lasers Surg. Med. 50:499-505, 2018. © 2018 Wiley Periodicals, Inc.


Assuntos
Lipossomos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/administração & dosagem , Verteporfina/administração & dosagem , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Retículo Endoplasmático/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos
14.
J Biomed Opt ; 22(12): 1-6, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28853247

RESUMO

The emergence of fluorescently labeled therapeutic antibodies has given rise to molecular probes for image-guided surgery. However, the extraneous interstitial presence of an unbound and nonspecifically accumulated probe gives rise to false-positive detection of tumor tissue and margins. Thus, the concept of tumor-cell activation of smart probes provides a potentially superior mechanism of delineating tumor margins as well as small tumor deposits. The combination of molecular targeting with intracellular activation circumvents the presence of extracellular, nonspecific signals of targeted probe accumulation. Here, we present a demonstration of the clinical antibodies cetuximab (cet, anti-EGFR mAb) and trastuzumab (trast, anti-HER-2 mAb) conjugated to Alexa Fluor molecules and IRDye QC-1 quencher optimized at the ratio of 1∶2∶6 to provide the greatest degree of proteolytic fluorescence activation, synonymous with intracellular lysosomal degradation. The cet-AF-Q-C1 conjugate (1∶2∶6) provides up to 9.8-fold proteolytic fluorescence activation. By preparing a spectrally distinct, irrelevant sham IgG-AF-QC-1 conjugate, a dual-activatable probe approach is shown to enhance the specificity of imaging within an orthotopic AsPC-1 pancreatic cancer xenograft model. The dual-activatable approach warrants expedited clinical translation to improve the specificity of image-guided surgery by spectrally decomposing specific from nonspecific probe accumulation, binding, and internalization.


Assuntos
Cetuximab , Corantes Fluorescentes , Sondas Moleculares , Cirurgia Assistida por Computador , Trastuzumab , Animais , Feminino , Imunoglobulina G
15.
Int J Pharm ; 513(1-2): 554-563, 2016 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-27651326

RESUMO

In this study, Paclitaxel (PTX) containing, bovine serum albumin (BSA) nanoparticles were fabricated via a simple approach. Folic acid (FA) was conjugated to chitosan (CS)/carboxymethyl cellulose (CMC) through an esterification reaction to produce BSA-CS-FA or BSA-CMC-FA conjugates. NiFe2O4 noncore (NFs) and PTX were loaded through a heat treatment and by a diffusion process. NFs-BSA-CS and NFs-BSA-CMC-FA with size of about 80nm, showed superior transversal R2 relaxation rate of 349 (mM)-1s-1 along with folate receptor-targeted and magnetically directed functions. NFs-BSA-CS-FA or NFs-BSA-CS-FA were found stable and biocompatible. Application of an external magnetic field effectively enhanced the PTX release from PTX-NFs-BSA-CS-FA or PTX-NFs-BSA-CS-FA and hence tumor inhibition rate. This study validate that NFs-BSA-CS-FA or NFs-BSA-CMC-FA and PTX-NFs-BSA-CS-FA or PTX-NFs-BSA-CS-FA are suitable systems for tumor diagnosis and therapy.


Assuntos
Antineoplásicos Fitogênicos , Sistemas de Liberação de Medicamentos , Nanocompostos , Paclitaxel , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Quitosana/administração & dosagem , Quitosana/análogos & derivados , Quitosana/química , Quitosana/uso terapêutico , Liberação Controlada de Fármacos , Eritrócitos/efeitos dos fármacos , Compostos Férricos/administração & dosagem , Compostos Férricos/química , Compostos Férricos/uso terapêutico , Ácido Fólico/administração & dosagem , Ácido Fólico/química , Ácido Fólico/uso terapêutico , Hemólise , Humanos , Células MCF-7 , Fenômenos Magnéticos , Nanocompostos/administração & dosagem , Nanocompostos/química , Nanocompostos/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Níquel/administração & dosagem , Níquel/química , Níquel/uso terapêutico , Paclitaxel/administração & dosagem , Paclitaxel/química , Paclitaxel/uso terapêutico , Soroalbumina Bovina/administração & dosagem , Soroalbumina Bovina/química , Soroalbumina Bovina/uso terapêutico
16.
Int J Nanomedicine ; 11: 3833-48, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27570452

RESUMO

Superparamagnetic iron oxide nanoparticles (SPIONs) have the potential to be used as multimodal imaging and cancer therapy agents due to their excellent magnetism and ability to generate reactive oxygen species when exposed to light. We report the synthesis of highly biocompatible SPIONs through a facile green approach using fruit peel extracts as the biogenic reductant. This green synthesis protocol involves the stabilization of SPIONs through coordination of different phytochemicals. The SPIONs were functionalized with polyethylene glycol (PEG)-6000 and succinic acid and were extensively characterized by X-ray diffraction analysis, field emission scanning electron microscopy, energy-dispersive X-ray spectroscopy, atomic force microscopy, Rutherford backscattering spectrometry, diffused reflectance spectroscopy, fluorescence emission, Fourier-transform infrared spectroscopy, ultraviolet-visible spectroscopy, and magnetization analysis. The developed SPIONs were found to be stable, almost spherical with a size range of 17-25 nm. They exhibited excellent water dispersibility, colloidal stability, and relatively high R 2 relaxivity (225 mM(-1) s(-1)). Cell viability assay data revealed that PEGylation or carboxylation appears to significantly shield the surface of the particles but does not lead to improved cytocompatibility. A highly significant increase of reactive oxygen species in light-exposed samples was found to play an important role in the photokilling of human cervical epithelial malignant carcinoma (HeLa) cells. The bio-SPIONs developed are highly favorable for various biomedical applications without risking interference from potentially toxic reagents.


Assuntos
Dextranos/química , Frutas/química , Química Verde/métodos , Espectroscopia de Ressonância Magnética , Nanopartículas de Magnetita/química , Micro-Ondas , Fotoquimioterapia , Extratos Vegetais/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Dextranos/síntese química , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Células HeLa , Humanos , Nanopartículas de Magnetita/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Análise Espectral , Difração de Raios X
17.
Int J Nanomedicine ; 11: 3159-66, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27471383

RESUMO

We report "smart" nickel oxide nanoparticles (NOPs) as multimodal cancer therapy agent. Water-dispersible and light-sensitive NiO core was synthesized with folic acid (FA) connected bovine serum albumin (BSA) shell on entrapped doxorubicin (DOX). The entrapped drug from NOP-DOX@BSA-FA was released in a sustained way (64 hours, pH=5.5, dark conditions) while a robust release was found under red light exposure (in 1/2 hour under λmax=655 nm, 50 mW/cm(2), at pH=5.5). The cell viability, thiobarbituric acid reactive substances and diphenylisobenzofuran assays conducted under light and dark conditions revealed a high photodynamic therapy potential of our construct. Furthermore, we found that the combined effect of DOX and NOPs from NOP-DOX@BSA-FA resulted in cell death approximately eightfold high compared to free DOX. We propose that NOP-DOX@BSA-FA is a potential photodynamic therapy agent and a collective drug delivery system for the systemic administration of cancer chemotherapeutics resulting in combination therapy.


Assuntos
Antineoplásicos/uso terapêutico , Nanopartículas/química , Neoplasias/tratamento farmacológico , Níquel/química , Fotoquimioterapia/métodos , Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Ácido Fólico/farmacologia , Ácido Fólico/uso terapêutico , Células HeLa , Humanos , Microscopia de Fluorescência , Nanopartículas/ultraestrutura , Nanosferas/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Soroalbumina Bovina/química
18.
Acta Radiol Short Rep ; 2(7): 2047981613512484, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349716

RESUMO

BACKGROUND: Diffusion-weighted imaging (DWI) along with the calculation of apparent diffusion coefficient (ADC), is a novel, non-invasive, and reliable technique of choice for accurate assessment and for the treatment planning of different types of brain tumors. It is more advantageous in the distinction and differentiation of benign from malignant meningiomas on the basis of ADC values. PURPOSE: To investigate the utility of DW magnetic resonance imaging (MRI), and to compare the apparent diffusion coefficient (ADC) obtained at two b-values for an authentic and preoperative characterization of meningiomas. MATERIAL AND METHODS: Twenty-six patients with clinically diagnosed or histologically verified meningioma (18 benign and 8 malignant) underwent imaging including DWI at 1.5 T. DW images were obtained at b = 1000 s/mm(2) and b = 2000 s/mm(2), ADC maps were generated at both the b-values. Signal intensities (SIs) and ADCs for solid tumorous tissues, contralateral normal tissues, and peritumoral edema were calculated and normalized ADC (NADC) ratio were determined for tumorous tissues. SI scores, ADC maps, and ADC values were analyzed visually and quantitatively, and were compared at both the b-values. RESULTS: DW images at b = 2000 s/mm(2) were more conspicuity (either hyperintense or hypointense) with improved contrast. The mean ADC of malignant meningiomas (0.64 ± 0.05 and 0.42 ± 0.03) was significantly lower (P < 0.05) as compared with benign meningiomas (1.04 ± 0.12 and 0.80 ± 0.07) at both the b-values. Mean NADC ratio in the malignant type was also significantly lower (P < 0.05) than the benign type at both the b-values. Mean ADC values for peritumoral edema do not differ between benign and malignant meningiomas. CONCLUSION: 1.5-T DWI using high b-values improved our ability to differentiate benign from malignant meningiomas. DWI may play an important role in the preoperative radiological evaluation and the recognition of these types for proper surgical treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA