Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cancer Res Commun ; 3(5): 884-895, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37377898

RESUMEN

Purpose: Patients with metastatic uveal melanoma have limited therapeutic options and high mortality rate so new treatment options are needed. Patients and Methods: We previously reported that patients treated with the PD-1 inhibitor pembrolizumab and the histone deacetylase inhibitor entinostat in the PEMDAC trial, experienced clinical benefits if their tumor originated from iris or was wildtype for BAP1 tumor suppressor gene. Here we present the 2-year follow-up of the patients in the PEMDAC trial and identify additional factors that correlate with response or survival. Results: Durable responses were observed in 4 patients, with additional 8 patients exhibiting a stable disease. The median overall survival was 13.7 months. Grade 3 adverse events were reported in 62% of the patients, but they were all manageable. No fatal toxicity was observed. Activity of thymidine kinase 1 in plasma was higher in patients with stable disease or who progressed on treatment, compared with those with partial response. Chemokines and cytokines were analyzed in plasma. Three chemokines were significantly different when comparing patients with and without response. One of the factors, CCL21, was higher in the plasma of responding patients before treatment initiation but decreased in the same patients upon treatment. In tumors, CCL21 was expressed in areas resembling tertiary lymphoid structures (TLS). High plasma levels of CCL21 and presence of TLS-like regions in the tumor correlated with longer survival. Conclusions: This study provides insight into durable responses in the PEMDAC trial, and describes dynamic changes of chemokines and cytokines in the blood of these patients. Significance: The most significant finding from the 2-year follow-up study of the PEMDAC trial was that high CCL21 levels in blood was associated with response and survival. CCL21 was also expressed in TLS-like regions and presence of these regions was associated with longer survival. These analyses of soluble and tumor markers can inform on predictive biomarkers needing validation and become hypothesis generating for experimental research.


Asunto(s)
Quimiocina CCL21 , Melanoma , Humanos , Quimiocina CCL21/genética , Quimiocinas/sangre , Epigénesis Genética , Estudios de Seguimiento , Inmunoterapia , Melanoma/tratamiento farmacológico
2.
Tidsskr Nor Laegeforen ; 142(15)2022 10 25.
Artículo en Inglés, Noruego | MEDLINE | ID: mdl-36286558
3.
Melanoma Res ; 32(4): 241-248, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34753889

RESUMEN

Targeted therapy and immunotherapy have revolutionized the treatment of metastatic skin melanoma but around half of all patients develop resistance early or late during treatment. The situation is even worse for patients with metastatic uveal melanoma (UM). Here we hypothesized that the immunotherapy of therapy-resistant skin melanoma or UM can be enhanced by epigenetic inhibitors. Cultured B16F10 cells and human UM cells were treated with the histone deacetylase inhibitor (HDACi) entinostat or BETi JQ1. Entinostat-induced HLA expression and PD-L1, but JQ1 did not. A syngeneic mouse model carrying B16-F10 melanoma cells was treated with PD-1 and CTLA4 inhibitors, which was curative. Co-treatment with the bioavailable BETi iBET726 impaired the immunotherapy effect. Monotherapy of a B16-F10 mouse model with anti-PD-1 resulted in a moderate therapeutic effect that could be enhanced by entinostat. Mice carrying PD-L1 knockout B16-F10 cells were also sensitive to entinostat. This suggests HDAC inhibition and immunotherapy could work in concert. Indeed, co-cultures of UM with HLA-matched melanoma-specific tumor-infiltrating lymphocytes (TILs) resulted in higher TIL-mediated melanoma killing when entinostat was added. Further exploration of combined immunotherapy and epigenetic therapy in metastatic melanoma resistant to PD-1 inhibition is warranted.


Asunto(s)
Melanoma Experimental , Neoplasias Cutáneas , Animales , Antígeno B7-H1 , Epigénesis Genética , Melanoma , Melanoma Experimental/tratamiento farmacológico , Ratones , Melanoma Cutáneo Maligno
4.
Tidsskr Nor Laegeforen ; 1412021 09 28.
Artículo en Noruego | MEDLINE | ID: mdl-34596998
5.
Nat Commun ; 12(1): 5155, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34453044

RESUMEN

Preclinical studies have suggested that epigenetic therapy could enhance immunogenicity of cancer cells. We report the results of the PEMDAC phase 2 clinical trial (n = 29; NCT02697630) where the HDAC inhibitor entinostat was combined with the PD-1 inhibitor pembrolizumab in patients with metastatic uveal melanoma (UM). The primary endpoint was objective response rate (ORR), and was met with an ORR of 14%. The clinical benefit rate at 18 weeks was 28%, median progression free survival was 2.1 months and the median overall survival was 13.4 months. Toxicities were manageable, and there were no treatment-related deaths. Objective responses and/or prolonged survival were seen in patients with BAP1 wildtype tumors, and in one patient with an iris melanoma that exhibited a UV signature. Longer survival also correlated with low baseline ctDNA levels or LDH. In conclusion, HDAC inhibition and anti-PD1 immunotherapy results in durable responses in a subset of patients with metastatic UM.Trial registration ClinicalTrials.gov registration number: NCT02697630 (registered 3 March 2016). EudraCT registration number: 2016-002114-50.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Benzamidas/administración & dosificación , Melanoma/tratamiento farmacológico , Piridinas/administración & dosificación , Neoplasias de la Úvea/tratamiento farmacológico , Humanos , Melanoma/patología , Supervivencia sin Progresión , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias de la Úvea/patología
6.
J Immunother Cancer ; 9(7)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34215689

RESUMEN

We report a case of rapid eradication of melanoma brain metastases and simultaneous near-fatal encephalomyelitis following double immune checkpoint blockade. Brain damage marker S-100B and C reactive protein increased before symptoms or signs of encephalomyelitis and peaked when the patient fell into a coma. At that point, additional brain damage markers and peripheral T cell phenotype was analyzed. The analyses were repeated four times during the patient's recovery. Axonal damage marker neurofilament light polypeptide (NFL) and astrocytic damage marker glial fibrillar acidic protein (GFAP) were very high in blood and cerebrospinal fluid and gradually normalized after immunosuppression and intensive care. The costimulatory receptor inducible T cell costimulatory receptor (ICOS) was expressed on a high proportion of CD4+ and CD8+T cells as encephalomyelitis symptoms peaked and then gradually decreased in parallel with clinical improvement. Both single and double immune checkpoint inhibitor-treated melanoma patients with other serious immune-related adverse events (irAE) (n=9) also expressed ICOS on a significantly higher proportion of CD4+ and CD8+T cells compared with controls without irAE (n=12). In conclusion, our results suggest a potential role for ICOS on CD4+ and CD8+T cells in mediating encephalomyelitis and other serious irAE. In addition, brain damage markers in blood could facilitate early diagnosis of encephalitis.


Asunto(s)
Biomarcadores/metabolismo , Daño Encefálico Crónico/inducido químicamente , Daño Encefálico Crónico/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Encefalomielitis/inducido químicamente , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Anciano , Daño Encefálico Crónico/patología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Masculino
8.
Nat Commun ; 11(1): 1894, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32313009

RESUMEN

Metastatic uveal melanoma is less well understood than its primary counterpart, has a distinct biology compared to skin melanoma, and lacks effective treatments. Here we genomically profile metastatic tumors and infiltrating lymphocytes. BAP1 alterations are overrepresented and found in 29/32 of cases. Reintroducing a functional BAP1 allele into a deficient patient-derived cell line, reveals a broad shift towards a transcriptomic subtype previously associated with better prognosis of the primary disease. One outlier tumor has a high mutational burden associated with UV-damage. CDKN2A deletions also occur, which are rarely present in primaries. A focused knockdown screen is used to investigate overexpressed genes associated withcopy number gains. Tumor-infiltrating lymphocytes are in several cases found tumor-reactive, but expression of the immune checkpoint receptors TIM-3, TIGIT and LAG3 is also abundant. This study represents the largest whole-genome analysis of uveal melanoma to date, and presents an updated view of the metastatic disease.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Neoplasias Primarias Secundarias/genética , Neoplasias de la Úvea/genética , Animales , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Dosificación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Linfocitos , Linfocitos Infiltrantes de Tumor/patología , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Ratones , Mutación , Neoplasias Primarias Secundarias/inmunología , Neoplasias Primarias Secundarias/metabolismo , Neoplasias Primarias Secundarias/patología , Pronóstico , Análisis de Secuencia de ADN , Transcriptoma , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias de la Úvea/inmunología , Neoplasias de la Úvea/metabolismo , Neoplasias de la Úvea/patología
10.
Acta Oncol ; 58(7): 962-966, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31151362

RESUMEN

Introduction: Phase III studies of PD-1 inhibitors have demonstrated remarkable improvements in the survival of patients with metastatic melanoma (MM). If these results are generalizable to an unselected patient population treated in clinical routine is unknown. This study aimed to investigate and describe clinical efficacy and safety of PD-1 inhibitors in patients with MM treated in routine clinical practice. Material and methods: A retrospective descriptive study of patients with metastatic or inoperable cutaneous melanoma treated with PD-1 inhibitors at a single institution (Department of Oncology, Sahlgrenska University Hospital) from 1 September 2015 to 31 August 2017. Data were obtained from medical records. Results: A total of 116 patients were included in the analyses. The overall survival (OS) at 12-month follow-up was 70.2% and the median OS was 27.9 months. Patients with BRAF mutated tumors had increased OS, whereas ECOG PS ≥2, LDH > ULN and presence or history of brain metastases (stage M1d) were associated with impaired survival. Immune-related AEs of any grade occurred in 64 (55.2%) patients and 15 (12.9%) patients experienced immune-related AEs of grades 3 and 4. Notably, rheumatic adverse events occurred at a higher rate (15.5%) than previously reported. The occurrence of immune-related AEs was associated with a benefit in OS, while the severity of immune-related AEs did not affect survival, nor did the use of systemic corticosteroids. Conclusions: The efficacy and safety of PD1 inhibitors in routine clinical practice appear comparable to that described in clinical trials.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Melanoma/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Enfermedades Reumáticas/epidemiología , Neoplasias Cutáneas/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Inmunológicos/efectos adversos , Ensayos Clínicos Fase III como Asunto , Femenino , Estudios de Seguimiento , Humanos , Estimación de Kaplan-Meier , Masculino , Melanoma/mortalidad , Melanoma/secundario , Persona de Mediana Edad , Mutación , Receptor de Muerte Celular Programada 1/inmunología , Proteínas Proto-Oncogénicas B-raf/genética , Estudios Retrospectivos , Enfermedades Reumáticas/inducido químicamente , Enfermedades Reumáticas/inmunología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Suecia/epidemiología
11.
BMC Cancer ; 19(1): 415, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31046743

RESUMEN

BACKGROUND: While recent years have seen a revolution in the treatment of metastatic cutaneous melanoma, no treatment has yet been able to demonstrate any prolonged survival in metastatic uveal melanoma. Thus, metastatic uveal melanoma remains a disease with an urgent unmet medical need. Reports of treatment with immune checkpoint inhibitors have thus far been disappointing. Based on animal experiments, it is reasonable to hypothesize that the effect of immunotherapy may be augmented by epigenetic therapy. Proposed mechanisms include enhanced expression of HLA class I and cancer antigens on cancer cells, as well as suppression of myeloid suppressor cells. METHODS: The PEMDAC study is a multicenter, open label phase II study assessing the efficacy of concomitant use of the PD1 inhibitor pembrolizumab and the class I HDAC inhibitor entinostat in adult patients with metastatic uveal melanoma. Primary endpoint is objective response rate. Eligible patients have histologically confirmed metastatic uveal melanoma, ECOG performance status 0-1, measurable disease as per RECIST 1.1 and may have received any number of prior therapies, with the exception of anticancer immunotherapy. Twenty nine patients will be enrolled. Patients receive pembrolizumab 200 mg intravenously every third week in combination with entinostat 5 mg orally once weekly. Treatment will continue until progression of disease or intolerable toxicity or for a maximum of 24 months. DISCUSSION: The PEMDAC study is the first trial to assess whether the addition of an HDAC inhibitor to anti-PD1 therapy can yield objective anti-tumoral responses in metastatic UM. TRIAL REGISTRATION: ClinicalTrials.gov registration number: NCT02697630 . (Registered 3 March 2016). EudraCT registration number: 2016-002114-50.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Benzamidas/administración & dosificación , Melanoma/tratamiento farmacológico , Piridinas/administración & dosificación , Neoplasias de la Úvea/tratamiento farmacológico , Administración Intravenosa , Administración Oral , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas/uso terapéutico , Esquema de Medicación , Femenino , Humanos , Masculino , Estudios Prospectivos , Piridinas/uso terapéutico , Proyectos de Investigación , Resultado del Tratamiento
12.
Cancer Res ; 79(5): 899-904, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30622115

RESUMEN

Chimeric antigen receptors (CAR) can transmit signals akin to those from activated T-cell receptors when bound to a cell surface target. CAR-expressing T cells against CD19 can cause curative effects in leukemia and lymphoma and is approved for clinical use. However, no CAR-T therapy is currently approved for use in solid tumors. We hypothesize that the resistance of solid tumors to CAR-T can be overcome by similar means as those used to reactivate tumor-infiltrating T lymphocytes (TIL), for example, by cytokines or immune checkpoint blockade. Here we demonstrate that CAR-T cells directed against HER2 can kill uveal and cutaneous melanoma cells in vitro and in vivo. Curative effects in vivo were only observed in xenografts grown in a NOD/SCID IL2 receptor gamma (NOG) knockout mouse strain transgenic for human IL2. The effect was target-specific, as CRISPR/Cas9-mediated disruption of HER2 in the melanoma cells abrogated the killing effect of the CAR-T cells. The CAR-T cells were also able to kill melanoma cells from patients resistant to adoptive T-cell transfer (ACT) of autologous TILs. Thus, CAR-T therapy represents an option for patients that do not respond to immunotherapy with ACT of TIL or immune checkpoint blockade. In addition, our data highlight the use of IL2 transgenic NOG mice as models to prove efficacy of CAR-T-cell products, possibly even in a personalized manner. SIGNIFICANCE: These findings demonstrate that a novel humanized mouse model can help clinical translation of CAR-T cells against uveal and cutaneous melanoma that do not respond to TIL therapy or immune checkpoint blockade.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma/terapia , Receptor ErbB-2/inmunología , Linfocitos T/trasplante , Neoplasias de la Úvea/terapia , Animales , Línea Celular Tumoral , Humanos , Subunidad gamma Común de Receptores de Interleucina/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/trasplante , Melanoma/enzimología , Melanoma/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Receptor ErbB-2/metabolismo , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/terapia , Linfocitos T/inmunología , Neoplasias de la Úvea/enzimología , Neoplasias de la Úvea/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Melanoma Cutáneo Maligno
13.
Cell Death Dis ; 9(8): 810, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30042422

RESUMEN

Karonudib (TH1579) is a novel compound that exerts anti-tumor activities and has recently entered phase I clinical testing. The aim of this study was to conduct a pre-clinical trial in patient-derived xenografts to identify the possible biomarkers of response or resistance that could guide inclusion of patients suffering from metastatic melanoma in phase II clinical trials. Patient-derived xenografts from 31 melanoma patients with metastatic disease were treated with karonudib or a vehicle for 18 days. Treatment responses were followed by measuring tumor sizes, and the models were categorized in the response groups. Tumors were harvested and processed for RNA sequencing and protein analysis. To investigate the effect of karonudib on T-cell-mediated anti-tumor activities, tumor-infiltrating T cells were injected in mice carrying autologous tumors and the mice treated with karonudib. We show that karonudib has heterogeneous anti-tumor effect on metastatic melanoma. Thus, based on the treatment responses, we could divide the 31 patient-derived xenografts in three treatment groups: progression group (32%), suppression group (42%), and regression group (26%). Furthermore, we show that karonudib has anti-tumor effect, irrespective of major melanoma driver mutations. Also, we identify high expression of ABCB1, which codes for p-gp pumps as a resistance biomarker. Finally, we show that karonudib treatment does not hamper T-cell-mediated anti-tumor responses. These findings can be used to guide future use of karonudib in clinical use with a potential approach as precision medicine.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores Enzimáticos/uso terapéutico , Melanoma/tratamiento farmacológico , Pirimidinas/uso terapéutico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Humanos , Melanoma/patología , Ratones , Pirimidinas/farmacología , Trasplante Heterólogo , Células Tumorales Cultivadas , Regulación hacia Arriba
15.
Artículo en Inglés | MEDLINE | ID: mdl-32913988

RESUMEN

PURPOSE: Cancer of unknown primary is a group of metastatic tumors in which the standard diagnostic workup fails to identify the site of origin of the tumor. The potential impact of precision oncology on this group of patients is large, because actionable driver mutations and a correct diagnosis could provide treatment options otherwise not available for patients with these fatal cancers. This study investigated if comprehensive genomic analyses could provide information on the origin of the tumor. PATIENTS AND METHODS: Here we describe a patient whose tumor was misdiagnosed at least three times. Next-generation sequencing, a patient-derived xenograft mouse model, and bioinformatics were used to identify an actionable mutation, predict resistance development to the targeted therapy, and correctly diagnose the origin of the tumor. Transcriptomic classification was benchmarked using The Cancer Genome Atlas (TCGA). RESULTS: Despite the lack of a known primary tumor site and the absence of diagnostic immunohistochemical markers, the origin of the patient's tumor was established using the novel bioinformatic workflow. This included a mutational signature analysis of the sequenced metastases and comparison of their transcriptomic profiles to a pan-cancer panel of tumors from TCGA. We further discuss the strengths and limitations of the latter approaches in the context of three potentially incorrectly diagnosed TCGA lung tumors. CONCLUSION: Comprehensive genomic analyses can provide information on the origin of tumors in patients with cancer of unknown primary.

16.
Nat Commun ; 8(1): 707, 2017 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-28955032

RESUMEN

Immune checkpoint inhibitors and adoptive cell transfer (ACT) of autologous tumor-infiltrating T cells have shown durable responses in patients with melanoma. To study ACT and immunotherapies in a humanized model, we have developed PDXv2.0 - a melanoma PDX model where tumor cells and tumor-infiltrating T cells from the same patient are transplanted sequentially in non-obese diabetic/severe combined immune-deficient/common gamma chain (NOG/NSG) knockout mouse. Key to T-cell survival/effect in this model is the continuous presence of interleukin-2 (IL-2). Tumors that grow in PDXv2.0 are eradicated if the autologous tumor cells and T cells come from a patient that exhibited an objective response to ACT in the clinic. However, T cells from patients that are non-responders to ACT cannot kill tumor cells in PDXv2.0. Taken together, PDXv2.0 provides the potential framework to further model genetically diverse human cancers for assessing the efficacy of immunotherapies as well as combination therapies.Combining different types of immune therapies might benefit certain patients. Here, the authors develop an autologous immune-humanized melanoma mouse model that allows the preclinical assessment of cancer cell-T cell interactions from each individual patient and the benefits of immunotherapies combinations.


Asunto(s)
Interleucina-2/genética , Linfocitos Infiltrantes de Tumor/trasplante , Melanoma/terapia , Neoplasias Cutáneas/terapia , Linfocitos T/trasplante , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Humanos , Inmunoterapia Adoptiva , Subunidad gamma Común de Receptores de Interleucina/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Melanoma/inmunología , Melanoma/patología , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Linfocitos T/inmunología , Linfocitos T/patología
17.
Oncotarget ; 5(20): 9609-18, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25228592

RESUMEN

The development of novel therapies against melanoma would benefit from individualized tumor models to ensure the rapid and accurate identification of biomarkers of therapy response. Previous studies have suggested that patient-derived xenografts (PDXes) could be useful. However, the utility of PDXes in guiding real-time treatment decisions has only been reported in anecdotal forms. Here tumor biopsies from patients with stage III and IV metastatic malignant melanoma were transplanted into immunocompromised mice to generate PDXes. 23/26 melanoma biopsies generated serially transplantable PDX models, and their histology, mutation status and expression profile resembled their corresponding patient biopsy. The potential treatment for one patient was revealed by an in vitro drug screen and treating PDXes with the MEK inhibitor trametinib. In another patient, the BRAF mutation predicted the response of both the patient and its corresponding PDXes to MAPK-targeted therapy. Importantly, in this unselected group of patients, the time from biopsy for generation of PDXes until death was significantly longer than the time required to reach the treatment phase of the PDXes. Thus, it could be clinically meaningful to use this type of platform for melanoma patients as a pre-selection tool in clinical trials.


Asunto(s)
Melanoma/tratamiento farmacológico , Melanoma/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Adulto , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Metástasis Linfática , Masculino , Melanoma/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Medicina de Precisión , Neoplasias Cutáneas/genética
18.
Biochimie ; 94(1): 2-10, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21839138

RESUMEN

Molecular shape and its impact on bilayer curvature stress are powerful concepts for describing the effects of lipids and fatty acids on fundamental membrane properties, such as passive permeability and derived properties like drug transport across liposomal membranes. We illustrate these relationships by studying the effects of fatty acids and lysolipids on the permeation of a potent anti-cancer drug, doxorubicin, across the bilayer of a liposome in which the drug is encapsulated. Using a simple fluorescence assay, we have systematically studied the passive permeation of doxorubicin across liposomal membranes in different lipid phases: the solid-ordered phase (DPPC bilayers), the liquid-disordered phase (POPC lipid bilayers), and the liquid-ordered phase induced by high levels of cholesterol (DOPC + cholesterol lipid bilayers). The effect of different free fatty acids (FA) and lysolipids (LL), separately and in combination, on permeability was assessed to elucidate the possible mechanism of phospholipase A(2)-triggered release in cancer tissue of liposomal doxorubicin formulations. In all cases, FAs applied separately lead to significant enhancement of permeability, most pronounced in liquid-disordered bilayers and less pronounced in solid and solid-ordered bilayers. LLs applied separately had only a marginal effect on permeability. FA and LL applied in combination lead to a synergistic enhancement of permeability in solid bilayers, whereas in liquid-disordered bilayers, the combined effect suppressed the otherwise strong permeability enhancement due to the FAs.


Asunto(s)
Ácidos Grasos no Esterificados/química , Lípidos/química , Membranas Artificiales , Profármacos/farmacología , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Lípidos/farmacología , Liposomas , Profármacos/farmacocinética
19.
Anticancer Drugs ; 21(7): 674-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20559126

RESUMEN

Several fatty acids and lysolipids have been shown earlier to increase the permeability of membranes of artificial liposomes, thereby increasing the release of drugs such as doxorubicin (Dox) contained within them. Free fatty acids can also inhibit cancer cell growth in vitro, and it has been suggested that this inhibition results from increased membrane permeability. Clearly, therefore, increased membrane permeability could be used in the design of liposomes for targeted drug delivery. For example, as free fatty acids and lysolipids are released upon phospholipase degradation of the liposome, the liposome could deliver membrane permeability enhancers in addition to the drug to increase the targeted anticancer effect. In this study, we examined the effect on Dox uptake in the breast cancer cell lines MDA-MB-231, MCF7, and MCF7-MDR when incubated with a large panel of different free fatty acids and lysolipids. Dox uptake was quantified by flow cytometry and fluorescence microscopy. We observed no increased Dox uptake in any of the breast cancer cell lines, suggesting that growth inhibitory effects observed earlier subsequent to the addition of free fatty acids to cancer cells are not caused by increased cell membrane permeability.


Asunto(s)
Antibióticos Antineoplásicos/metabolismo , Neoplasias de la Mama/metabolismo , Doxorrubicina/metabolismo , Ácidos Grasos no Esterificados/farmacología , Lípidos/farmacología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...