Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Vet Comp Oncol ; 21(4): 605-615, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37653682

RESUMEN

Hyperthermia is a form of a cancer treatment which is frequently applied in combination with radiotherapy (RT) to improve therapy responses and radiosensitivity. The mode of action of hyperthermia is multifactorial; the one hand by altering the amount of the blood circulation in the treated tissue, on the other hand by modulating molecular pathways involved in cell survival processes and immunogenic interactions. One of the most dominant proteins induced by hyperthermia is the major stress-inducible heat shock protein 70 (Hsp70). Hsp70 can be found in the blood either as a free-protein (free HSP70) derived from necrotic cells, or lipid-bound (liposomal Hsp70) when it is actively released in extracellular vesicles (EVs) by living cells. The aim of the study was to evaluate the levels of free and liposomal Hsp70 before and after treatment with RT alone or hyperthermia combined with radiotherapy (HTRT) in dogs and cats to evaluate therapy responses. Peripheral blood was collected from feline and canine patients before and at 2, 4, 6 and 24 h after treatment with RT or HTRT. Hsp70 enzyme-linked immunosorbent assays (ELISAs) were performed to determine the free and liposomal Hsp70 concentrations in the serum. The levels were analysed after the first fraction of radiation to study immediate effects and after all applied fractions to study cumulative effects. The levels of free and liposomal Hsp70 levels in the circulation were not affected by the first singular treatment and cumulative effects of RT in cats however, after finalizing all treatment cycles with HTRT free and liposomal Hsp70 levels significantly increased. In dogs, HTRT, but not treatment with RT alone, significantly affected liposomal Hsp70 levels during the first fraction. Free Hsp70 levels were significantly increased after RT, but not HTRT, during the first fraction in dogs. In dogs, on the other hand, RT alone resulted in a significant increase in liposomal Hsp70, but HTRT did not significantly affect the liposomal Hsp70 when cumulative effects were analysed. Free Hsp70 was significantly induced in dogs after both, RT and HTRT when cumulative effects were analysed. RT and HTRT treatments differentially affect the levels of free and liposomal Hsp70 in dogs and cats. Both forms of Hsp70 could potentially be further investigated as potential liquid biopsy markers to study responses to RT and HTRT treatment in companion animals.


Asunto(s)
Enfermedades de los Gatos , Enfermedades de los Perros , Hipertermia Inducida , Neoplasias , Humanos , Gatos , Animales , Perros , Proteínas HSP70 de Choque Térmico/metabolismo , Enfermedades de los Gatos/radioterapia , Hipertermia Inducida/veterinaria , Hipertermia Inducida/métodos , Enfermedades de los Perros/radioterapia , Neoplasias/radioterapia , Neoplasias/veterinaria
2.
Clin Transl Radiat Oncol ; 27: 109-113, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33598571

RESUMEN

INTRODUCTION: Driven by the current unsatisfactory outcomes for patients with locally advanced pancreatic cancer (LAPC), a biologically intensified clinical protocol was developed to explore the feasibility and efficacy of FOLFORINOX chemotherapy followed by deep hyperthermia concomitant with chemoradiation and subsequent FOLFORINOX chemotherapy in patients with LAPC. METHODS: Nine patients with LAPC were treated according to the HEATPAC Phase II trial protocol which consists of 4 cycles of FOLFORINOX chemotherapy followed by gemcitabine-based chemoradiation to 56 Gy combined with weekly deep hyperthermia and then a further 8 cycles of FOLFORINOX chemotherapy. RESULTS: One grade three related toxicity was reported and two tumours became resectable. The median overall survival was 24 months and 1 year overall survival was 100%. CONCLUSIONS: Intensification of chemoradiation with deep hyperthermia was feasible in nine consecutive patients with LAPC.

3.
Vet Comp Oncol ; 16(2): 202-213, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28892246

RESUMEN

Hyperthermia (HT) as an adjuvant to radiation therapy (RT) is a multimodality treatment method to enhance therapeutic efficacy in different tumours. High demands are placed on the hardware and treatment planning software to guarantee adequately planned and applied HT treatments. The aim of this prospective study was to determine the effectiveness and safety of the novel HT system in tumour-bearing dogs and cats in terms of local response and toxicity as well as to compare planned with actual achieved data during heating. A novel applicator with a flexible number of elements and integrated closed-loop temperature feedback control system, and a tool for patient-specific treatment planning were used in a combined thermoradiotherapy protocol. Good agreement between predictions from planning and clinical outcome was found in 7 of 8 cases. Effective HT treatments were planned and verified with the novel system and provided improved quality of life in all but 1 patient. This individualized treatment planning and controlled heat exposure allows adaptive, flexible and safe HT treatments in palliatively treated animal patients.


Asunto(s)
Enfermedades de los Gatos/terapia , Enfermedades de los Perros/terapia , Hipertermia Inducida/veterinaria , Neoplasias/veterinaria , Animales , Enfermedades de los Gatos/radioterapia , Gatos , Terapia Combinada/métodos , Terapia Combinada/veterinaria , Enfermedades de los Perros/radioterapia , Perros , Diseño de Equipo , Hipertermia Inducida/efectos adversos , Hipertermia Inducida/métodos , Neoplasias/radioterapia , Neoplasias/terapia , Proyectos Piloto , Estudios Prospectivos , Radioterapia Adyuvante/métodos , Radioterapia Adyuvante/veterinaria , Facultades de Medicina Veterinaria , Suiza , Resultado del Tratamiento
4.
Cancer Treat Rev ; 50: 217-227, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27756009

RESUMEN

Effective multimodal cancer management requires the optimal integration of diagnostic and therapeutic modalities. Radiation therapy, chemotherapy and immunotherapy, alone or in combination, are integral parts of various cancer treatment protocols. Hyperthermia at 39-45°C is a potent radiosensitiser and has been shown to improve therapeutic outcomes in various tumours through its synergy with chemotherapy. Gene silencing approaches, using small interfering RNAs and microRNAs, are also being explored in clinical trials in oncology. The rapid developments in multifunctional nanoparticles provide ample opportunities to integrate both diagnostic and therapeutic modalities into a single effective cancer "theranostic" vector. Nanoparticles could extravasate passively into the tumour tissues in preference to the adjacent normal tissues by capitalizing on the enhanced permeability and retention effect. Tumour targeting might be further augmented by conjugating tumour-specific peptides and antibodies onto the surface of these nanoparticles or by activation through electromagnetic radiations, laser or ultrasound. Magnetic nanoparticles can induce hyperthermia in the presence of an alternating magnetic field, thereby multifunctionally with tumour-specific payloads empowering tumour specific radiotheranostics (for both imaging and radiotherapy), chemotherapy drug delivery, immunotherapy and gene silencing therapy. Such a (nano)bullet could realise the "magic bullet" conceived by Paul Ehrlich more than a century ago. This article discusses the various aspects of this "magic (nano)bullet" and the challenges that need to be addressed to usher in this new paradigm in modern cancer diagnostics and therapeutics.


Asunto(s)
Antineoplásicos/uso terapéutico , Compuestos Férricos/uso terapéutico , Hipertermia Inducida/métodos , Nanopartículas de Magnetita/uso terapéutico , Neoplasias/terapia , Tratamiento con ARN de Interferencia/métodos , Terapia Combinada , Sistemas de Liberación de Medicamentos , Humanos , Campos Magnéticos , Imanes , Nanopartículas/uso terapéutico , Neoplasias/diagnóstico por imagen , Nanomedicina Teranóstica
5.
Cancer Treat Rev ; 41(9): 742-53, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26051911

RESUMEN

Hyperthermia, one of the oldest forms of cancer treatment involves selective heating of tumor tissues to temperatures ranging between 39 and 45°C. Recent developments based on the thermoradiobiological rationale of hyperthermia indicate it to be a potent radio- and chemosensitizer. This has been further corroborated through positive clinical outcomes in various tumor sites using thermoradiotherapy or thermoradiochemotherapy approaches. Moreover, being devoid of any additional significant toxicity, hyperthermia has been safely used with low or moderate doses of reirradiation for retreatment of previously treated and recurrent tumors, resulting in significant tumor regression. Recent in vitro and in vivo studies also indicate a unique immunomodulating prospect of hyperthermia, especially when combined with radiotherapy. In addition, the technological advances over the last decade both in hardware and software have led to potent and even safer loco-regional hyperthermia treatment delivery, thermal treatment planning, thermal dose monitoring through noninvasive thermometry and online adaptive temperature modulation. The review summarizes the outcomes from various clinical studies (both randomized and nonrandomized) where hyperthermia is used as a thermal sensitizer of radiotherapy and-/or chemotherapy in various solid tumors and presents an overview of the progresses in loco-regional hyperthermia. These recent developments, supported by positive clinical outcomes should merit hyperthermia to be incorporated in the therapeutic armamentarium as a safe and an effective addendum to the existing oncological treatment modalities.


Asunto(s)
Hipertermia Inducida/métodos , Neoplasias/terapia , Ensayos Clínicos como Asunto , Terapia Combinada , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia
6.
Ann Oncol ; 20(9): 1522-1528, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19465425

RESUMEN

BACKGROUND: This multicenter phase II study investigated the efficacy and feasibility of preoperative induction chemotherapy followed by chemoradiation and surgery in patients with esophageal carcinoma. PATIENTS AND METHODS: Patients with locally advanced resectable squamous cell carcinoma or adenocarcinoma of the esophagus received induction chemotherapy with cisplatin 75 mg/m(2) and docetaxel (Taxotere) 75 mg/m(2) on days 1 and 22, followed by radiotherapy of 45 Gy (25 x 1.8 Gy) and concurrent chemotherapy comprising cisplatin 25 mg/m(2) and docetaxel 20 mg/m(2) weekly for 5 weeks, followed by surgery. RESULTS: Sixty-six patients were enrolled at eleven centers and 57 underwent surgery. R0 resection was achieved in 52 patients. Fifteen patients showed complete, 16 patients nearly complete and 26 patients poor pathological remission. Median overall survival was 36.5 months and median event-free survival was 22.8 months. Squamous cell carcinoma and good pathologically documented response were associated with longer survival. Eighty-two percent of all included patients completed neoadjuvant therapy and survived for 30 days after surgery. Dysphagia and mucositis grade 3/4 were infrequent (<9%) during chemoradiation. Five patients (9%) died due to surgical complications. CONCLUSIONS: This neoadjuvant, taxane-containing regimen was efficacious and feasible in patients with locally advanced esophageal cancer in a multicenter, community-based setting and represents a suitable backbone for further investigation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma/terapia , Neoplasias Esofágicas/terapia , Terapia Neoadyuvante , Adulto , Anciano , Carcinoma/mortalidad , Cisplatino/administración & dosificación , Cisplatino/efectos adversos , Terapia Combinada , Procedimientos Quirúrgicos del Sistema Digestivo , Docetaxel , Neoplasias Esofágicas/mortalidad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Radioterapia , Taxoides/administración & dosificación , Taxoides/efectos adversos , Resultado del Tratamiento
7.
Ann Oncol ; 18(7): 1196-202, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17429100

RESUMEN

BACKGROUND: The aim of this multicenter trial was to prospectively evaluate neo-adjuvant chemotherapy followed by extrapleural pneumonectomy (EPP) and radiotherapy, including quality of life as outcome. PATIENTS AND METHODS: Eligible patients had malignant pleural mesothelioma of all histological types, World Health Organization performance status of zero to two and clinical stage T1-T3, N0-2, M0 disease considered completely resectable. Neo-adjuvant chemotherapy consisted of three cycles of cisplatin and gemcitabine followed by EPP. Postoperative radiotherapy was considered for all patients. RESULTS: In all, 58 of 61 patients completed three cycles of neo-adjuvant chemotherapy. Forty-five patients (74%) underwent EPP and in 37 patients (61%) the resection was complete. Postoperative radiotherapy was initiated in 36 patients. The median survival of all patients was 19.8 months [95% confidence interval (CI) 14.6-24.5]. For the 45 patients undergoing EPP, the median survival was 23 months (95% CI 16.6-32.9). Psychological distress showed minor variations over time with distress above the cut-off score indicating no morbidity with 82% (N = 36) at baseline and 76% (N = 26) at 3 months after surgery (P = 0.5). CONCLUSIONS: The observed rate of operability is promising. A median survival of 23 months for patients undergoing EPP compares favourably with the survival reported from single center studies of upfront surgery. This approach was not associated with an increase in psychological distress.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Mesotelioma/terapia , Terapia Neoadyuvante , Neoplasias Pleurales/terapia , Neumonectomía , Adulto , Anciano , Cisplatino/administración & dosificación , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Mesotelioma/mortalidad , Mesotelioma/psicología , Persona de Mediana Edad , Neoplasias Pleurales/mortalidad , Neoplasias Pleurales/psicología , Calidad de Vida , Radioterapia , Gemcitabina
8.
J Thorac Cardiovasc Surg ; 131(3): 697-703, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16515926

RESUMEN

OBJECTIVE: We sought to investigate whether intrapleural topical application of cisplatin with a surgical carrier has a prolonged local tissue level in comparison with cisplatin solution while reducing systemic toxicity. METHODS: Forty immune-competent Fischer rats were inoculated with 10(6) mesothelioma cells. Ten days later, left pneumonectomy with tumor debulking was performed. Twenty animals underwent local application of cisplatin solution (100 mg/m2), whereas the same quantity of cisplatin was topically applied as a gel with the Vivostat (Vivolution) system in 20 other animals. In each group 5 subgroups of 4 animals were defined according to the harvesting time of blood and tissue samples (2, 4, 24, and 72 hours and 1 week) after local therapy. Platinum concentrations in serum and tissue and systemic toxicity were analyzed. RESULTS: Platinum concentrations in tissue were significantly higher in the gel group (group 1) than in the solution group (group 2) at 1, 3, and 7 days after therapy (1510, 1224, and 1069 pg/mg for group 1 vs 598, 382, and 287 pg/mg for group 2; P = .007, P = .005, and P = .0002, respectively). Laboratory findings showed renal insufficiency in the animals of the solution group at 1 week, with values of 98 mmol/L versus 7.7 mmol/L for urea and 410 mumol/L versus 43 mumol/L for creatinine (P = .02 and P = .01, respectively), which was confirmed by means of pathologic analysis. CONCLUSIONS: Intrapleural administration of cisplatin with the carrier Vivostat significantly provides sustained higher platinum concentrations up to 1 week in tissue in comparison with application of cisplatin solution without conferring systemic toxicity in this model.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Portadores de Fármacos , Adhesivo de Tejido de Fibrina , Mesotelioma/tratamiento farmacológico , Neoplasias Pleurales/tratamiento farmacológico , Administración Tópica , Animales , Inmunocompetencia , Mesotelioma/sangre , Mesotelioma/química , Platino (Metal)/análisis , Pleura , Neoplasias Pleurales/sangre , Neoplasias Pleurales/química , Ratas , Ratas Endogámicas F344
10.
Br J Cancer ; 88(11): 1785-92, 2003 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-12771996

RESUMEN

Mistletoe extracts are used as alternative cancer treatment in addition to standard chemotherapy and radiation treatment and have an immunostimulatory and pain-relieving effect. A direct antitumour effect of mistletoe extracts against tumour cells of lymphoid origin has been linked to the D-galactoside-specific mistletoe lectin I. In this study, we investigated the cellular effect of bacterially expressed, recombinant mistletoe lectin alone or in combination with ionising radiation in a genetically defined p53-wild-type and p53-deficient E1A/ras-transformed murine tumour cells system. Downregulation of the proliferative activity and cell killing by recombinant mistletoe lectin occurred in a clear dose response (0.1-1 ng ml(-1)). Induction of apoptosis was p53-independent, but apoptosis-associated factor-1-dependent. Cellular treatment with lectin in combination with ionising radiation resulted in both p53-wild-type and p53-deficient tumour cells in an at least additive, antiproliferative effect and enhanced activation of caspase-3. Combined treatment with ionising radiation and lectin revealed a similar cytotoxic effect in human, p53-mutated adenocarcinoma cells. Thus, recombinant mistletoe lectin alone and in combination with ionising radiation bypasses often prevalent apoptotic deficiencies in treatment-resistant tumour cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Lectinas de Plantas/farmacología , Preparaciones de Plantas/farmacología , Proteínas de Plantas , Toxinas Biológicas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Anexina A5/metabolismo , Factor Apoptótico 1 Activador de Proteasas , Western Blotting , Caspasa 3 , Caspasas/metabolismo , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Fibroblastos/metabolismo , Genes ras , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/efectos de la radiación , Ratones , Muérdago/química , Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Mutación/genética , Proteínas/genética , Proteínas/metabolismo , Radiación Ionizante , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 2 , Sensibilidad y Especificidad , Células Tumorales Cultivadas
11.
Curr Med Chem Anticancer Agents ; 2(6): 727-42, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12678723

RESUMEN

DNA double strand breaks are the pivotal cellular damage induced by ionizing radiation. A plethora of molecular and cellular processes are activated as part of the cellular stress response that result in cell cycle arrest and induction of the DNA-repair machinery to restore the damage of DNA or to activate a cell death program. However ionizing radiation also initiates signal transduction cascades that are generated at cellular sites distant from and independent of DNA-damage. These signaling processes are similar to hormone activated growth factor receptor controlled signal transduction cascades and represent interesting targets for anticancer treatment modalities combining ionizing radiation with molecular defined pharmacological compounds. Activation of these signal transduction cascades upon irradiation or upregulation of growth factor mediated pathways due to oncogene-transformation often contribute to an acquired or inherent treatment resistance in malignant cells. Therefore pharmacological compounds inhibiting specific key-entities of these signal transduction cascades potentially sensitize for radiation induced cell death. Here we describe current preclinical concepts of combined treatment strategies with locoregional-applied ionizing radiation and molecular defined signal transduction inhibitors to overcome a high treatment threshold in tumor cells.


Asunto(s)
Neoplasias/radioterapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ligandos , Neoplasias/tratamiento farmacológico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Radioterapia/efectos adversos , Radioterapia/métodos
12.
Br J Cancer ; 85(12): 2010-6, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11747347

RESUMEN

The vascular endothelial growth factor (VEGF) receptor is a major target for anti-angiogenesis-based cancer treatment. Here we report the treatment effect of ionizing radiation in combination with the novel orally bioavailable VEGF receptor tyrosine kinase inhibitor PTK787/ZK222584 on endothelial cell proliferation in vitro and with tumour xenografts in vivo. Combined treatment of human umbilical vein endothelial cells with increasing doses of PTK787/ZK222584 and ionizing radiation abrogated VEGF-dependent proliferation in a dose-dependent way, but inhibition of endothelial cell proliferation was not due to apoptosis induction. In vivo, a combined treatment regimen of PTK787/ZK222584 (4 x 100 mg/kg) during 4 consecutive days in combination with ionizing radiation (4 x 3 Gy) exerted a substantial tumour growth delay for radiation-resistant p53-dysfunctional tumour xenografts derived from SW480 colon adenocarcinoma cells while each treatment modality alone had only a minimal effect on tumour size and neovascularization. SW480 tumours from animals that received a combined treatment regimen, displayed not only an extended tumour growth delay but also a significant decrease in the number of microvessels in the tumour xenograft. These results support the model of a cooperative anti-tumoral effect of angiogenesis inhibitor and irradiation and show that the orally bioavailable VEGF receptor tyrosine kinase inhibitor PTK787/ZK222584 is suitable for combination therapy with irradiation.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/uso terapéutico , Proteínas de Neoplasias/antagonistas & inhibidores , Neovascularización Patológica/tratamiento farmacológico , Ftalazinas/uso terapéutico , Piridinas , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/patología , Adenocarcinoma/radioterapia , Administración Oral , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Neoplasias del Colon/radioterapia , Terapia Combinada , Endotelio Vascular/citología , Endotelio Vascular/efectos de la radiación , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/radioterapia , Ftalazinas/administración & dosificación , Ftalazinas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Radioterapia Adyuvante , Receptores de Factores de Crecimiento Endotelial Vascular , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Res ; 61(22): 8203-10, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11719451

RESUMEN

Activation of the phosphatidylinositol 3'-kinase (PI3K)/Akt survival pathway protects against apoptotic stress stimuli. Therefore, compounds that down-regulate this pathway are of clinical interest for single and combined anticancer treatment modalities. Here we demonstrate that the cytotoxic effect of the protein kinase C (PKC)-inhibitor N-benzoylated staurosporine (PKC412) is mediated via the PI3K/Akt pathway. Dose-dependent down-regulation of the proliferative activity, activation of the apoptotic machinery, and cell killing by PKC412 (0-1 microM) in Rat1a-fibroblasts and H-ras-oncogene-transformed fibroblasts correlated with a decrease of Akt phosphorylation and a reduced phosphorylation of the endogenous Akt-substrate GSK3-alpha. Expression of the dominant-active myristoylated form of Akt abrogated this cytotoxic effect of PKC412. Experiments with Apaf-1-deficient cells revealed that PKC412-induced cytotoxicity depends on an intact apoptosome but that the decrease of Akt phosphorylation is not attributable to apoptosis execution. Comparative experiments indicate that PKC412 and the parent-compound staurosporine down-regulate this survival pathway upstream or at the level of Akt but by a different mechanism than the PI3K-inhibitor LY294002. Furthermore, inhibition of this pathway by PKC412 is relevant for sensitization to ionizing radiation. These results demonstrate the specific role of this signaling pathway for the PKC412-mediated down-regulation of an apoptotic threshold and its cytotoxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal/efectos de los fármacos , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Animales , Línea Celular Transformada , Transformación Celular Neoplásica/genética , Cromonas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Genes ras/fisiología , Humanos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Ratas , Transducción de Señal/fisiología , Células Tumorales Cultivadas
14.
Int J Radiat Oncol Biol Phys ; 49(2): 561-7, 2001 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11173155

RESUMEN

In many human hematologic and solid malignancies, intrinsic or acquired treatment resistance remains a major obstacle for successful cancer therapy. The molecular understanding of how tumor cells respond to chemotherapy and ionizing radiation is rapidly evolving. Induction of programmed cell death, apoptosis, is one important strategy for successful cancer therapy. This has been shown convincingly for oncogene-transformed normal cells as well as tumor cells of lymphoid origin. However, the relevance of apoptosis in solid human malignancies is less clear. Loss of apoptosis might be linked to specific mutations in the often tissue-specific apoptotic pathways due to aberrations in the stress-related signal transduction cascades. Restoration of a dysfunctional apoptotic program in cancer tissue where apoptosis has been identified as an important mechanism for tissue homeostasis is one rational approach for innovative cancer therapy. In this review, we focus on the relevance of the tumor suppressor p53 for apoptosis-induction and successful cancer therapy outlining the importance of an intact caspase machinery for apoptosis execution. Strategies are discussed to overcome treatment resistance and a high apoptotic threshold in human malignancies where apoptosis is the dominant mode of cell death and the status of p53 is an important determinant for apoptosis induction.


Asunto(s)
Apoptosis/genética , Caspasas/fisiología , Genes p53/fisiología , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Proteínas de Neoplasias/fisiología , Neoplasias/terapia , Adenoviridae/genética , Animales , Apoptosis/fisiología , Ciclo Celular/fisiología , Reparación del ADN , Activación Enzimática , Inhibidores Enzimáticos/uso terapéutico , Genes p53/genética , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados , Neoplasias/genética , Neoplasias/radioterapia , Estaurosporina/uso terapéutico
15.
Cancer Res ; 61(2): 732-8, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11212276

RESUMEN

The cellular response to ionizing radiation is governed by the DNA-damage recognition process but is also modulated by cytoplasmic signal transduction cascades that are part of the cellular stress response. Growth-promoting protein kinase C activity antagonizes irradiation-induced cell death, and, therefore, protein kinase C inhibitors might be potent radiosensitizers. The antiproliferative and radiosensitizing effect of the novel N-benzoylated staurosporine analogue PKC412 was tested in vitro against genetically defined p53-wild type (+/+) and p53-deficient (-/-) murine fibrosarcoma cells and in vivo against radioresistant p53-/- murine fibrosarcoma and human colon adenocarcinoma tumor xenograft (SW480, p53-mutated). PKC412 sensitized both p53+/+ and p53-/- tumor cells in vitro and in vivo for treatment with ionizing radiation but with a different mechanism of radiosensitization depending on the p53 status. In p53+/+, cells combined treatment with PKC412 and ionizing radiation drastically induced apoptotic cell death, whereas no apoptosis induction could be observed in p53-deficient cells in vitro and in histological tumor sections. Combined treatment resulted in an increased G2 cell cycle distribution in p53-/- cells at PKC412 concentrations that did not alter cell cycle distribution when applied alone. In vivo, a minimal treatment regimen during 4 consecutive days of PKC412 (4 x 100 mg/kg) in combination with ionizing radiation (4 x 3 Gy) exerted a substantial tumor growth delay for both p53-disfunctional tumor xenografts and showed that the clinically relevant protein kinase C inhibitor PKC412 is a promising new radiosensitizer with a potentially broad therapeutic window.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Proteína p53 Supresora de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Terapia Combinada , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Genotipo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neoplasias Experimentales/radioterapia , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cell Growth Differ ; 11(9): 491-9, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11007454

RESUMEN

Caspases are a family of cysteine proteases that constitute the apoptotic cell death machinery. We report the importance of the cytochrome c-mediated caspase-9 death pathway for radiosensitization by the protein kinase C (PKC) inhibitors staurosporine (STP) and PKC-412. In our genetically defined tumor cells, treatment with low doses of STP or the conventional PKC-specific inhibitor PKC-412 in combination with irradiation (5 Gy) potently reduced viability, enhanced mitochondrial cytochrome c release into the cytosol, and specifically stimulated the initiator caspase-9. Whereas treatment with each agent alone had a minimal effect, combined treatment resulted in enhanced caspase-3 activation. This was prevented by broad-range and specific caspase-9 inhibitors and absent in caspase-9-deficient cells. The tumor suppressor p53 was required for apoptosis induction by combined treatment but was dispensable for dose-dependent STP-induced caspase activation. These results demonstrate the requirement for an intact caspase-9 pathway for apoptosis-based radiosensitization by PKC inhibitors and show that STP induces apoptosis independent of p53.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Caspasas/efectos de los fármacos , Caspasas/efectos de la radiación , Grupo Citocromo c/efectos de los fármacos , Grupo Citocromo c/efectos de la radiación , Proteína Quinasa C/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Estaurosporina/análogos & derivados , Animales , Apoptosis/fisiología , Caspasa 9 , Caspasas/metabolismo , Grupo Citocromo c/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/radioterapia , Proteína Quinasa C/metabolismo , Tolerancia a Radiación/fisiología , Estaurosporina/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/efectos de la radiación , Proteína p53 Supresora de Tumor/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
Anticancer Drug Des ; 15(1): 17-28, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10888033

RESUMEN

The staurosporine derivative PKC412 was originally identified as an inhibitor of protein kinase C (PKC) and subsequently shown to inhibit other kinases including the kinase insert domain receptor (KDR) (vascular endothelial growth factor receptor, VEGF-R2), the receptor of platelet-derived growth factor, and the receptor for the stem cell factor, c-kit. PKC412 showed a broad antiproliferative activity against various tumor and normal cell lines in vitro, and was able to reverse the Pgp-mediated multidrug resistance of tumor cells in vitro. Exposure of cells to PKC412 resulted in a dose-dependent increase in the G2/M phase of the cell cycle concomitant with increased polyploidy, apoptosis and enhanced sensitivity to ionizing radiation. PKC412 displayed a potent antitumor activity as single agent and was able to potentiate the antitumor activity of some of the clinically used cytotoxins (Taxol and doxorubicin) in vivo. The combined treatment of PKC412 with loco-regional ionizing irradiation showed significant antitumor activity against tumors which are resistant to both ionizing radiation and chemotherapeutic agents (dysfunctional p53). The finding that PKC412 is an inhibitor of the VEGF-mediated cellular signaling via inhibition of KDR and PKC in vitro is consistent with the in vivo inhibition of VEGF-dependent angiogenesis in a growth factor implant model. Orally administered PKC412 also strongly inhibited retinal neovascularization as well as laser-induced choroidal neovascularization in murine models. In summary, PKC412 may suppress tumor growth by inhibiting tumor angiogenesis in addition to directly-inhibiting tumor cell proliferation via its effects on PKC and/or other protein kinases. PKC412 is currently in Phase I clinical trials for treatment of advanced cancer as well as for the treatment of ischemic retinopathy.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Neoplasias/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Estaurosporina/análogos & derivados , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Proteínas Sanguíneas/metabolismo , Ciclo Celular/efectos de los fármacos , Ensayos Clínicos Fase I como Asunto , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Humanos , Ratones , Neoplasias/patología , Unión Proteica , Estaurosporina/metabolismo , Estaurosporina/farmacología , Estaurosporina/uso terapéutico , Células Tumorales Cultivadas
18.
Radiother Oncol ; 55(2): 95-9, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10799720

RESUMEN

INTRODUCTION: The incidence of ductal carcinoma of the breast (DCIS) increased in Europe and the US up to 10-fold over the last 20 years ¿8. This could be linked to more vigorous screening mammography, as well as changes of histopathologic and diagnostic criteria for breast lesions during the last decades ¿31,26. Optimal screening for DCIS, the diagnostic procedures and best treatment is still controversial. For many DCIS patients lumpectomy and adjuvant radiotherapy are a valid treatment option. There is need for better prognostic factors in DCIS, which indicate the need for therapy and tailor the intensity of treatment. Recently prognostic factors based on clinical and pathological findings for DCIS were established and are currently validated. Molecular mechanisms involved in DCIS formation, DCIS progression to invasive breast cancer, and predicting DCIS treatment response are rapidly emerging ¿46. RESULTS AND CONCLUSION: Here we discuss some of the known molecular mechanisms of DCIS and how they could be further exploited as prognostic factors for screening and tailoring DCIS therapy. This review will summarize relevant molecular mechanism of DCIS carcinogenesis including dysregulation of the cell cycle clock and changes of the apoptotic threshold. In particular, recently published molecular and cellular abnormalities in DCIS, potentially relevant for treatment decision, will be discussed.


Asunto(s)
Neoplasias de la Mama/radioterapia , Carcinoma in Situ/radioterapia , Carcinoma Ductal de Mama/radioterapia , Apoptosis/efectos de la radiación , Femenino , Humanos , Pronóstico , Receptores de Factores de Crecimiento/análisis , Proteína p53 Supresora de Tumor/fisiología
19.
Br J Cancer ; 80(5-6): 693-8, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10360645

RESUMEN

p53 mutations are among the most common genetic alterations in human cancer and are frequently described in intrinsic or acquired radio- and chemotherapy resistance. Radiation-induced cell kill is not only mediated by DNA damage but also by the activation of signal transduction cascades generated at the plasma membrane like the sphingomyelin pathway. We used genetically defined wild-type p53 or p53-deficient mouse fibrosarcoma cells to investigate the p53-dependence of tumour response upon activation of the sphingomyelin pathway. Treatment of the tumour cells with neutral sphingomyelinase drastically reduced the amount of wild-type p53 fibrosarcoma cell proliferation over 72 h in a clear dose-response (0.2-1.0 U ml(-1) nSMase). Sphingomyelinase had no effect on cell proliferation in tumour cells lacking p53. Similarly, cell proliferation was abolished by C2-ceramide (5-20 microM) only in wild-type p53 cells. FACS-analysis revealed that C2-ceramide induced massive p53-dependent apoptosis (40-50% after 12-24 h) and cell cycle analysis showed a transient G1 arrest in p53-deficient tumour cells 12-24 h after C2-ceramide exposure. These results suggest that ceramide-induced apoptosis in tumour cells can be dependent on the status of p53 and imply that p53 is also important for stress-induced apoptotic signal transduction cascades generated at the plasma membrane.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fibrosarcoma/genética , Fibrosarcoma/patología , Esfingosina/análogos & derivados , Proteína p53 Supresora de Tumor/fisiología , Animales , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Fibrosarcoma/tratamiento farmacológico , Inhibidores de Crecimiento/farmacología , Ratones , Ratones Noqueados , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingomielina Fosfodiesterasa/farmacología , Esfingomielinas/fisiología , Esfingosina/farmacología , Células Tumorales Cultivadas
20.
Cell Growth Differ ; 10(3): 141-6, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10099827

RESUMEN

Negative regulation of E2F-1 DNA binding function by cyclin A kinase represents part of an S-phase checkpoint control system that, when activated, leads to apoptosis. In this study, we examined the cellular sensitivity and resistance of isogenic mouse fibrosarcoma cell lines, differing primarily in their p53 status, to ectopic expression of wild-type (wt) E2F-1 and cyclin A kinase binding-defective mutants of it. We found that E2F-1 (wt) potently affected the survival of p53+/+ tumor cells but not that of p53-/- cells. In contrast, expression of cyclin A kinase binding-defective E2F-1 species interfered with cell survival of fibrosarcoma cells irrespective of their p53 status. Finally, expression of E2F-1 (wt) in p53-/- fibrosarcoma cells enhanced the cytotoxic effect of ionizing radiation in vitro and in vivo in a mouse tumor model. These results suggest that E2F-1-dependent activation of an S-phase checkpoint is p53 independent and that E2F-1 possesses radiosensitizing properties in the absence of p53.


Asunto(s)
Proteínas Portadoras , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Fibrosarcoma/metabolismo , Genes p53 , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Transformada , Supervivencia Celular , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Fibrosarcoma/genética , Expresión Génica , Ratones , Ratones Desnudos , Inhibidores de la Síntesis de la Proteína/metabolismo , Puromicina/metabolismo , Tolerancia a Radiación/fisiología , Proteína 1 de Unión a Retinoblastoma , Factores de Tiempo , Factor de Transcripción DP1 , Factores de Transcripción/farmacología , Proteína p53 Supresora de Tumor/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...