Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Control Release ; 354: 19-33, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36503069

RESUMEN

Triggered drug delivery strategies have been shown to enhance drug accumulation at target diseased sites in comparison to administration of free drug. In particular, many studies have demonstrated improved targetability of chemotherapeutics when delivered via thermosensitive liposomes. However, most studies continue to focus on encapsulating doxorubicin while many other drugs would benefit from this targeted and localized delivery approach. The proposed study explores the therapeutic potential of a thermosensitive liposome formulation of the commonly used chemotherapy drug vinorelbine in combination with mild hyperthermia (39-43 °C) in a murine model of rhabdomyosarcoma. Rhabdomyosarcoma, the most common soft tissue sarcoma in children, is largely treated using conventional chemotherapy which is associated with significant adverse long-term sequelae. In this study, mild hyperthermia was pursued as a non-invasive, non-toxic means to improve the efficacy and safety profiles of vinorelbine. Thorough assessment of the pharmacokinetics, biodistribution, efficacy and toxicity of vinorelbine administered in the thermosensitive liposome formulation was compared to administration in a traditional, non-thermosensitive liposome formulation. This study shows the potential of an advanced formulation technology in combination with mild hyperthermia as a means to target an untargeted therapeutic agent and result in a significant improvement in its therapeutic index.


Asunto(s)
Hipertermia Inducida , Rabdomiosarcoma , Niño , Ratones , Humanos , Animales , Liposomas , Vinorelbina , Distribución Tisular , Sistemas de Liberación de Medicamentos , Doxorrubicina , Línea Celular Tumoral
2.
Drug Deliv Transl Res ; 13(4): 1059-1073, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36577832

RESUMEN

Chemotherapy plays an important role in debulking tumors in advance of surgery and/or radiotherapy, tackling residual disease, and treating metastatic disease. In recent years many promising advanced drug delivery strategies have emerged that offer more targeted delivery approaches to chemotherapy treatment. For example, thermosensitive liposome-mediated drug delivery in combination with localized mild hyperthermia can increase local drug concentrations resulting in a reduction in systemic toxicity and an improvement in local disease control. However, the majority of solid tumor-associated deaths are due to metastatic spread. A therapeutic approach focused on a localized target area harbors the risk of overlooking and undertreating potential metastatic spread. Previous studies reported systemic, albeit limited, anti-tumor effects following treatment with thermosensitive liposomal chemotherapy and localized mild hyperthermia. This work explores the systemic treatment capabilities of a thermosensitive liposome formulation of the vinca alkaloid vinorelbine in combination with mild hyperthermia in an immunocompetent murine model of rhabdomyosarcoma. This treatment approach was found to be highly effective at heated, primary tumor sites. However, it demonstrated limited anti-tumor effects in secondary, distant tumors. As a result, the addition of immune checkpoint inhibition therapy was pursued to further enhance the systemic anti-tumor effect of this treatment approach. Once combined with immune checkpoint inhibition therapy, a significant improvement in systemic treatment capability was achieved. We believe this is one of the first studies to demonstrate that a triple combination of thermosensitive liposomes, localized mild hyperthermia, and immune checkpoint inhibition therapy can enhance the systemic treatment capabilities of thermosensitive liposomes.


Asunto(s)
Antineoplásicos , Hipertermia Inducida , Neoplasias , Ratones , Animales , Liposomas , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Hipertermia Inducida/métodos , Sistemas de Liberación de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Inmunoterapia , Doxorrubicina
3.
Nutr Clin Pract ; 37(4): 935-944, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35072294

RESUMEN

BACKGROUND: Clinicians may be reluctant to feed patients on high-flow nasal cannula (HFNC) therapy, despite studies suggesting it is beneficial and safe. We describe the implementation of a feeding protocol for patients with bronchiolitis on HFNC and determine its effect on nutrition goals. METHODS: Prospective bedside data on enteral volume, feed interruptions, and aspiration events were collected on patients with bronchiolitis who were <24 months of age, treated with HFNC, and fed per a developed protocol. Exclusion criteria included history of prematurity <32 weeks, congenital heart disease, or positive-pressure ventilation before feeding. Length of intensive care unit and hospital stay was compared with both a concurrent cohort (CC) of patients not fed per the protocol and a retrospective cohort (RC) admitted prior to protocol creation. RESULTS: Seventy-eight patients met the criteria for the prospective study arm: 24 patients were included in the CC, and 74 were included in the RC. Seventy-one percent of prospective patients received enteral nutrition (EN) on HFNC day 1 vs 42% of the CC. In the prospective cohort, feed interruption occurred in 23% of patients and was associated with higher flow rates; however, no aspiration events occurred. Patients fed per protocol were fed 8-10 h sooner and discharged 1 day earlier than those in the RC. CONCLUSION: The use of a feeding protocol for patients with bronchiolitis on HFNC was safe and associated with shorter time to initiate EN and shorter length of hospital stay.


Asunto(s)
Bronquiolitis , Cánula , Bronquiolitis/terapia , Humanos , Lactante , Terapia por Inhalación de Oxígeno/métodos , Estudios Prospectivos , Estudios Retrospectivos
4.
Nanomedicine ; 40: 102484, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34748961

RESUMEN

"A single disappointing study does not mean an end to the future of ThermoDox®", writes Michael Tardugno (CEO of Celsion Corporation), after announcing the termination of Celsion's second Phase III clinical trial. The OPTIMA trial, as it was known, evaluated their thermosensitive liposome (TSL) formulation of doxorubicin (ThermoDox®) in combination with radiofrequency ablation for the treatment of hepatocellular carcinoma (HCC). The purpose of this perspective is to review the case of ThermoDox and to address questions related to its clinical translation. Specifically, what has prevented the clinical translation of this once highly regarded breakthrough technology? Is this the end of TSLs? What can we learn from the challenges faced in the clinical development of this multi-modal therapy? As formulation scientists working in the field, we continue to believe that heat-triggered drug delivery platforms have tremendous potential as chemotherapy. Herein, we highlight potential limitations in the design of many of the Thermodox clinical trials, and we propose that despite these setbacks, TSLs have the potential to become an effective component of cancer therapy.


Asunto(s)
Carcinoma Hepatocelular , Hipertermia Inducida , Neoplasias Hepáticas , Carcinoma Hepatocelular/tratamiento farmacológico , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Sistemas de Liberación de Medicamentos , Calor , Humanos , Liposomas , Neoplasias Hepáticas/tratamiento farmacológico
6.
Adv Drug Deliv Rev ; 163-164: 98-124, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32681862

RESUMEN

Hyperthermia has demonstrated clinical success in improving the efficacy of both chemo- and radio-therapy in solid tumors. Pre-clinical and clinical research studies have demonstrated that targeted hyperthermia can increase tumor blood flow and increase the perfused fraction of the tumor in a temperature and time dependent manner. Changes in tumor blood circulation can produce significant physiological changes including enhanced vascular permeability, increased oxygenation, decreased interstitial fluid pressure, and reestablishment of normal physiological pH conditions. These alterations in tumor physiology can positively impact both small molecule and nanomedicine chemotherapy accumulation and distribution within the tumor, as well as the fraction of the tumor susceptible to radiation therapy. Hyperthermia can trigger drug release from thermosensitive formulations and further improve the accumulation, distribution, and efficacy of chemotherapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Hipertermia Inducida/métodos , Hipertermia/fisiopatología , Neoplasias/terapia , Radioterapia/métodos , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Permeabilidad Capilar/fisiología , Terapia Combinada , Sistemas de Liberación de Medicamentos/métodos , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Neoplasias/irrigación sanguínea , Neoplasias/fisiopatología , Oxígeno/sangre , Factores de Tiempo , Microambiente Tumoral/fisiología
7.
J Control Release ; 282: 35-45, 2018 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-29673642

RESUMEN

Triple negative breast cancer is an aggressive disease that accounts for at least 15% of breast cancer diagnoses, and a disproportionately high percentage of breast cancer related morbidity. Intensive research efforts are focused on the development of more efficacious treatments for this disease, for which therapeutic options remain limited. The high incidence of mutations in key DNA repair pathways in triple negative breast cancer results in increased sensitivity to DNA damaging agents, such as platinum-based chemotherapies. Hyperthermia has been successfully used in breast cancer treatment to sensitize tumors to radiation therapy and chemotherapy. It has also been used as a mechanism to trigger drug release from thermosensitive liposomes. In this study, mild hyperthermia is used to trigger release of cisplatin from thermosensitive liposomes in the vasculature of human triple negative breast cancer tumors implanted orthotopically in mice. This heat-triggered liposomal formulation of cisplatin resulted in significantly delayed tumor growth and improved overall survival compared to treatment with either non-thermosensitive liposomes containing cisplatin or free cisplatin, as was observed in two independent tumor models (i.e. MDA-MB-231 and MDA-MB-436). The in vitro sensitivity of the cell lines to cisplatin and hyperthermia alone and in combination was characterized extensively using enzymatic assays, clonogenic assays, and spheroid growth assays. Evaluation of correlations between the in vitro and in vivo results served to identify the in vitro approach that is most predictive of the effects of hyperthermia in vivo. Relative expression of several heat shock proteins and the DNA damage repair protein BRCA1 were assayed at baseline and in response to hyperthermia both in vitro and in vivo. Interestingly, delivery of cisplatin in thermosensitive liposomes in combination with hyperthermia resulted in the most significant tumor growth delay, relative to free cisplatin, in the less cisplatin-sensitive cell line (i.e. MDA-MB-231). This work demonstrates that thermosensitive cisplatin liposomes used in combination with hyperthermia offer a novel method for effective treatment of triple negative breast cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Preparaciones de Acción Retardada/química , Sistemas de Liberación de Medicamentos/métodos , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Mama/irrigación sanguínea , Mama/efectos de los fármacos , Mama/patología , Línea Celular Tumoral , Cisplatino/uso terapéutico , Femenino , Humanos , Hipertermia Inducida/métodos , Liposomas/química , Ratones SCID , Neoplasias de la Mama Triple Negativas/patología
8.
J Control Release ; 249: 63-73, 2017 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-28122204

RESUMEN

Thermosensitive liposomes have been put forward as a strategy to improve upon the limited tumor drug availability associated with conventional non-thermosensitive liposomes. ThermoDox® is the first and only thermosensitive liposome formulation to reach clinical development. The initial Phase III clinical trial on ThermoDox® (i.e. HEAT trial) evaluating the combination of ThermoDox® and radiofrequency ablation (RFA) in comparison to RFA alone for treatment of inoperable hepatocellular carcinoma (HCC) failed to reach its primary endpoint in progression-free survival (PFS). Nevertheless, a subgroup analysis demonstrated a marked improvement in PFS and a significant improvement in overall survival (OS) for patients who underwent RFA treatment for at least 45min. Potential reasons for failure of the HEAT trial have been summarized in this review and include issues with clinical trial design, lack of supporting preclinical data, and improvements in the control arm (i.e. RFA alone). In recent years, there have been many developments and improvements in heating infrastructure, thermometry and treatment planning of hyperthermia and ablation treatments. Still, there are many barriers to the clinical implementation and accessibility of heat treatment. This review provides an in-depth analysis of the current status, as well as potential challenges faced by continued clinical translation of thermosensitive liposomes.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/terapia , Terapia Combinada/métodos , Preparaciones de Acción Retardada/uso terapéutico , Liposomas/uso terapéutico , Neoplasias Hepáticas/terapia , Animales , Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/cirugía , Ablación por Catéter/métodos , Ensayos Clínicos como Asunto , Preparaciones de Acción Retardada/administración & dosificación , Calor , Humanos , Hipertermia Inducida/métodos , Liposomas/administración & dosificación , Hígado/patología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía
9.
J Drug Target ; 24(9): 865-877, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27310112

RESUMEN

Treatment efficacy of a heat-activated thermosensitive liposome formulation of cisplatin (CDDP), known as HTLC, was determined in xenograft models of non-small-cell lung carcinoma. The short-term impact of local hyperthermia (HT) on tumor morphology, microvessel density and local inflammatory response was also evaluated. The HTLC formulation in combination with local HT resulted in a significant advantage in therapeutic effect in comparison with free drug and a non-thermosensitive liposome formulation of CDDP (i.e. LipoplatinTM) when administered at their maximum tolerated doses. Local HT-induced widespread cell necrosis and a significant reduction in microvessel density in the necrotic regions of tumors. CD11b-expressing innate leukocytes were demonstrated to infiltrate and reside preferentially at the necrotic rim of tumors, likely as a means to phagocytose-damaged tissue. Colocalization of CD11b with a marker of DNA damage (i.e. γH2AX) revealed a small portion of CD11b-expressing leukocytes that were possibly undergoing apoptosis as a result of HT-induced damage and/or the short lifespan of leukocytes. Overall, HT-induced tissue damage (i.e. at 24-h post-treatment) alone did not result in significant improvements in treatment effect, rather, the enhancement in tumor drug availability was correlated with improved therapeutic outcomes.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Cisplatino/administración & dosificación , Liposomas/administración & dosificación , Neoplasias Pulmonares/terapia , Neoplasias Experimentales/terapia , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cisplatino/química , Cisplatino/uso terapéutico , Femenino , Xenoinjertos , Calor , Humanos , Hipertermia Inducida , Liposomas/química , Ratones , Ratones SCID , Distribución Aleatoria
10.
Mol Pharm ; 12(11): 3973-85, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26394060

RESUMEN

Current chemotherapy strategies for second-line treatment of relapsed ovarian cancer are unable to effectively treat residual disease post-cytoreduction. The findings presented herein suggest that tissue penetration of drug is not only an issue for large, unresectable tumors, but also for invisible, microscopic lesions. The present study sought to investigate the potential of a block copolymer micelle (BCM) formulation, which may reduce toxicities of doxorubicin (DOX) in a similar way to pegylated liposomal doxorubicin (PLD, Doxil/Caelyx), while enhancing penetration into tumor tissue and improving intratumoral availability of drug. To achieve this goal, 50 nm-sized BCMs capable of high DOX encapsulation (BCM-DOX) at drug levels ranging from 2 to 7.6 mg/mL were formulated using an ultrafiltration technique. BCM-DOX was evaluated in 2D and 3D cell culture of the human ovarian cancer cell lines HEYA8, OV-90, and SKOV3. Additionally, the current study examines the impact of mild hyperthermia (MHT) on the cytotoxicity of DOX. The BCM-DOX formulation fulfilled the goal of controlling drug release while providing up to 9-fold greater cell monolayer cytotoxicity in comparison to PLD. In 3D cell culture, using multicellular tumor spheroids (MCTS) as a model of residual disease postsurgery, BCM-DOX achieved the benefits of an extended release formulation of DOX and resulted in improvements in drug accumulation over PLD, while yielding drug levels approaching that achievable by exposure to DOX alone. In comparison to PLD, this translated into superior MCTS growth inhibition in the short term and comparable inhibition in the long term. Overall, although MHT appeared to enhance drug accumulation in HEYA8 MCTS treated with BCM-DOX and DOX alone in the short term, improved growth inhibition of MCTS by MHT was not observed after 48 h of drug treatment. Evaluation of BCM-DOX in comparison to PLD as well as the effects of MHT is warranted in vivo.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/análogos & derivados , Sistemas de Liberación de Medicamentos , Hipertermia Inducida , Neoplasia Residual/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Polímeros/química , Apoptosis/efectos de los fármacos , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Femenino , Humanos , Micelas , Neoplasia Residual/patología , Neoplasias Ováricas/patología , Polietilenglicoles/administración & dosificación , Esferoides Celulares , Células Tumorales Cultivadas
11.
J Control Release ; 219: 313-330, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26409122

RESUMEN

The majority of animal models of cancer have performed poorly in terms of predicting clinical performance of new therapeutics, which are most often first evaluated in patients with advanced, metastatic disease. The development and use of metastatic models of cancer may enhance clinical translatability of preclinical studies focused on the development of nanotechnology-based drug delivery systems and macromolecular therapeutics, potentially accelerating their clinical implementation. It is recognized that the development and use of such models are not without challenge. Preclinical imaging tools offer a solution by allowing temporal and spatial characterization of metastatic lesions. This paper provides a review of imaging methods applicable for evaluation of novel therapeutics in clinically relevant models of advanced cancer. An overview of currently utilized models of oncology in small animals is followed by image-based development and characterization of visceral metastatic cancer models. Examples of imaging tools employed for metastatic lesion detection, evaluation of anti-tumor and anti-metastatic potential and biodistribution of novel therapies, as well as the co-development and/or use of imageable surrogates of response, are also discussed. While the focus is on development of macromolecular and nanotechnology-based therapeutics, examples with small molecules are included in some cases to illustrate concepts and approaches that can be applied in the assessment of nanotechnologies or macromolecules.


Asunto(s)
Diagnóstico por Imagen , Modelos Animales de Enfermedad , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Evaluación Preclínica de Medicamentos , Humanos , Nanotecnología , Metástasis de la Neoplasia/diagnóstico , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Distribución Tisular
12.
J Control Release ; 178: 69-78, 2014 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-24440663

RESUMEN

Cisplatin (CDDP) has been identified as the primary chemotherapeutic agent for the treatment of cervical cancer, but dose limiting toxicity is a key issue associated with its clinical application. A suite of liposome formulations of CDDP has been developed in efforts to reduce systemic toxicity, but their therapeutic advantage over the free drug has been modest due to insufficient drug release at the tumor site. This report describes the development of a novel heat-activated thermosensitive liposome formulation containing CDDP (HTLC) designed to release approximately 90% of the loaded drug in less than 5min under mild heating conditions (42°C). Physico-chemical characteristics of HTLC were assessed in terms of gel to liquid crystalline phase transition temperature (Tm), drug loading efficiency, particle size, and stability. The pharmacokinetic profile and biodistribution of HTLC in non-tumor-bearing mice were evaluated over a 24h period. A sophisticated spatio-temporal elucidation of HTLC release in tumor-bearing mice was achieved by way of real-time monitoring using a magnetic resonance (MR) imaging protocol, wherein a custom-built laser-based conformal heat source was applied at the tumor volume to trigger the release of HTLC co-encapsulated with the MR contrast agent gadoteridol (Gd-HP-DO3A). MR thermometry (MRT) demonstrated that a relatively uniform temperature distribution was achieved in the tumor volume using the external laser-based heating setup. In mice bearing subcutaneously-implanted ME-180 cervical tumors, the combination of HTLC and heat resulted in a 2-fold increase in tumor drug levels at 1h post-administration compared to HTLC without heating. Furthermore, the overall tumor accumulation levels for the HTLC groups (with and without heat) at 1h post-injection were significantly higher than the corresponding free CDDP group. This translated into a significant improvement in therapeutic efficacy evaluated as tumor growth delay (p<0.05) for the heated HTLC treatment group compared to the unheated HTLC, heated or unheated free CDDP, and saline groups. Overall, findings from this study demonstrate that a heat-activated, triggered release formulation of CDDP results in a significant enhancement in the therapeutic index of this drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Hipertermia Inducida , Neoplasias del Cuello Uterino/terapia , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Cisplatino/farmacocinética , Terapia Combinada , Femenino , Calor , Humanos , Rayos Láser , Liposomas , Ratones , Ratones SCID , Distribución Tisular , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación , Neoplasias del Cuello Uterino/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA