Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Pediatr Blood Cancer ; 71(3): e30821, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38148490

RESUMO

BACKGROUND AND AIMS: Nuclear protein of the testis (NUT) carcinoma (NC) is a rare and highly aggressive tumor defined by the presence of a somatic NUTM1 rearrangement, occurring mainly in adolescents and young adults. We analyzed the clinical and biological features of German pediatric patients (≤18 years) with NC. METHODS: This study describes the characteristics and outcome of 11 children with NC registered in the German Registry for Rare Pediatric Tumors (STEP). RESULTS: Eleven patients with a median age of 13.2 years (range 6.6-17.8) were analyzed. Malignant misdiagnoses were made in three patients. Thoracic/mediastinal tumors were found to be the primary in six patients, head/neck in four cases; one patient had multifocal tumor with an unknown primary. All patients presented with regional lymph node involvement, eight patients (72.7%) with distant metastases. Seven patients underwent surgery, eight radiotherapy with curative intent; polychemotherapy was administered in all patients. Novel treatment strategies including immunotherapy, targeted therapies, and virotherapy were applied in three patients. Median event-free survival and overall survival were 1.5 and 6.5 months, respectively. CONCLUSIONS: Every undifferentiated or poorly differentiated carcinoma should undergo testing for the specific rearrangement of NUTM1, in order to initiate an intense therapeutic regimen as early as possible. As in adults, only few pediatric patients with NC achieve prolonged survival. Thus, novel therapeutic strategies should be included and tested in clinical trials.


Assuntos
Carcinoma , Neoplasias Torácicas , Masculino , Adulto Jovem , Adolescente , Humanos , Criança , Proteínas de Neoplasias , Fatores de Transcrição , Testículo/patologia
2.
Clin Microbiol Rev ; 35(4): e0006222, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36069758

RESUMO

Phage therapy has become a hot topic in medical research due to the increasing prevalence of antibiotic-resistant bacteria strains. In the treatment of bacterial infections, bacteriophages have several advantages over antibiotics, including strain specificity, lack of serious side effects, and low development costs. However, scientists dismissed the clinical success of early clinical trials in the 1940s, slowing the adoption of this promising antibacterial application in Western countries. The current study used statistical methods commonly used in modern meta-analysis to reevaluate early 20th-century studies and compare them with clinical trials conducted in the last 20 years. Using a random effect model, the development of disease after treatment with or without phages was measured in odds ratios (OR) with 95% confidence intervals (CI). Based on the findings of 17 clinical trials conducted between 1921 and 1940, phage therapy was effective (OR = 0.21, 95% CI = 0.10 to 0.44, P value < 0.0001). The current study includes a topic review on modern clinical trials; four could be analyzed, indicating a noneffective therapy (OR = 2.84, 95% CI = 1.53 to 5.27, P value = 0.0009). The results suggest phage therapy was surprisingly less effective than standard treatments in resolving bacterial infections. However, the results were affected by the small sample set size. This work also contextualizes the development of phage therapy in the early 20th century and highlights the expansion of phage applications in the last few years. In conclusion, the current review shows phage therapy is no longer an underestimated tool in the treatment of bacterial infections.


Assuntos
Infecções Bacterianas , Bacteriófagos , Terapia por Fagos , Humanos , Terapia por Fagos/métodos , Infecções Bacterianas/terapia , Bactérias , Antibacterianos
3.
J Pediatr Hematol Oncol ; 45(4): 165-173, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36219702

RESUMO

BACKGROUND AND AIMS: Nuclear protein of the testis ( NUT ) carcinoma (NC) is a rare and highly aggressive tumor mainly occurring in adolescents and young adults, defined by the presence of a somatic NUTM1 rearrangement. The aim is to establish internationally harmonized consensus recommendations for the diagnosis and treatment of adolescents and young adults with NC in the framework of the European Reference Network for Paediatric Oncology. METHODS: The European Cooperative Study Group for Pediatric Rare Tumors developed recommendations according to the Consensus Conference Standard Operating procedure methodology and reviewed by external "experts." No evidence of level I to II exists. Recommendations were developed based on published prospective (level III), but more frequently retrospective series (level IV), case reports (level V), and personal expertise (level V). In addition, "strength" of recommendations were categorized by grading (grade A to E). RESULTS: Histology is mandatory for the diagnosis of NC, including immunolabeling with anti-NUT antibodies and molecular biology ( NUTM1 rearrangement) (level V; grade A). Treatment of NC usually combines aggressive approaches in multimodal regimens. Chemotherapy should be considered as first-line treatment (neoadjuvant vincristine-adriamycin-ifosfamide/cisplatin-adriamycin-ifsofamide or vincristine-doxorubicin-cyclophosphamide/ifosfamide-etoposide) for unresectable or metastatic tumor (ie, 3 courses), rapidly followed by local treatment (level IV; grade B). Referral to a specialized surgical oncology center is highly recommended (level V; grade A). In localized NC, a complete microscopic surgical resection should be attempted whenever and as soon as possible, followed by primary irradiation (60 to 70 Gy) and involved lymph nodes area (level IV; grade B). For head and neck tumors, a systematic neck dissection might be considered, even if N0 (level V; grade C). Adjuvant postirradiation chemotherapy is recommended, for a total of 9 to 12 courses (level IV; grade B). For first-line resected tumors, concomitant adjuvant chemotherapy to radiotherapy may be discussed (level IV; grade B). Targeted therapies and immunotherapeutic regimens should be delivered in the setting of prospective trials (level V; grade B). CONCLUSIONS: This project leads to a consensus strategy based on international experience with this very rare disease.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma , Adolescente , Criança , Humanos , Masculino , Adulto Jovem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma/tratamento farmacológico , Carcinoma/genética , Carcinoma/patologia , Cisplatino/administração & dosagem , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Etoposídeo/administração & dosagem , Ifosfamida/administração & dosagem , Terapia Neoadjuvante , Estudos Prospectivos , Estudos Retrospectivos , Vincristina/administração & dosagem
4.
Cancer Cell Int ; 22(1): 311, 2022 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-36221114

RESUMO

BACKGROUND: Immunotherapy with immune checkpoint inhibitors (ICI) has revolutionized cancer therapy. However, therapeutic targeting of inhibitory T cell receptors such as PD-1 not only initiates a broad immune response against tumors, but also causes severe adverse effects. An ideal future stratified immunotherapy would interfere with cancer-specific cell surface receptors only. METHODS: To identify such candidates, we profiled the surface receptors of the NCI-60 tumor cell panel via flow cytometry. The resulting surface receptor expression data were integrated into proteomic and transcriptomic NCI-60 datasets applying a sophisticated multiomics multiple co-inertia analysis (MCIA). This allowed us to identify surface profiles for skin, brain, colon, kidney, and bone marrow derived cell lines and cancer entity-specific cell surface receptor biomarkers for colon and renal cancer. RESULTS: For colon cancer, identified biomarkers are CD15, CD104, CD324, CD326, CD49f, and for renal cancer, CD24, CD26, CD106 (VCAM1), EGFR, SSEA-3 (B3GALT5), SSEA-4 (TMCC1), TIM1 (HAVCR1), and TRA-1-60R (PODXL). Further data mining revealed that CD106 (VCAM1) in particular is a promising novel immunotherapeutic target for the treatment of renal cancer. CONCLUSION: Altogether, our innovative multiomics analysis of the NCI-60 panel represents a highly valuable resource for uncovering surface receptors that could be further exploited for diagnostic and therapeutic purposes in the context of cancer immunotherapy.

5.
Future Oncol ; 2022 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-35818970

RESUMO

Despite advances in treatment, cancer remains a leading cause of death worldwide. Although treatment strategies are continually progressing, cancers have evolved many mechanisms for evading therapies and the host immune system. Oncolytic viruses (OVs) could provide a much-needed option for cancers that are resistant to existing treatments. OVs can be engineered to specifically target and kill cancer cells, while simultaneously triggering an immune response at the site of infection. This review will focus on the challenges of developing a successful OV and translation to clinical practice, discussing the innovative strategies that are being used to optimize the potential of OVs. Here, we will also explore the current clinical landscape and the prospects of OVs in early clinical development.


Oncolytic viruses (OVs) are viruses that may help destroy tumor cells. They work by selectively infecting and replicating within tumor cells, causing the cells to burst and release newly built viruses. These viruses infect nearby tumor cells, triggering the body's immune system to attack the tumor and any tumor cells that have spread throughout the body. Clinical trials have shown that OVs can destroy cancer cells that are resistant to standard therapies. OVs in combination with other cancer therapies can be more effective and there are over 100 clinical trials planned, ongoing or completed to investigate this approach. OVs are generally well tolerated, the most common treatment-related side effects include fever, aches and pains, and tiredness for 1­2 days. While only four OVs have been approved so far, there are more expected to come. Overall, OVs may provide a way to directly destroy tumors and turn on the immune system to destroy tumor cells throughout the body.

6.
Future Oncol ; 18(24): 2627-2638, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35699077

RESUMO

Patients with advanced, recurrent or metastatic cancer have poor prognosis despite treatment advancements. Vesicular stomatitis virus (VSV)-glycoprotein (GP; BI 1831169) is a chimeric VSV with its neurotropic glycoprotein G replaced by the non-neurotropic GP of the lymphocytic choriomeningitis virus. This live, recombinant oncolytic virus has demonstrated preclinical efficacy as a viral-based immunotherapy due to its interferon-dependent tumor specificity, potent oncolysis and stimulation of antitumor immune activity. Co-administration of the immune checkpoint inhibitor, ezabenlimab (BI 754091), alongside VSV-GP may synergistically enhance antitumor immune activity. Here, we describe the rationale and design of the first-in-human, phase I, dose-escalation study of VSV-GP alone and in combination with the immune checkpoint inhibitor ezabenlimab in patients with advanced, metastatic or relapsed and refractory solid tumors (NCT05155332).


There is a need to develop new treatments for people living with cancer. Immunotherapy is a type of medicine that works by helping the body's natural defenses, known as the immune system, to destroy cancer cells. There are different types of immunotherapies such as oncolytic viruses (OVs) and immune checkpoint inhibitors (ICIs). OVs are viruses that may help destroy cancer cells while leaving normal cells unharmed. They work by replicating within cancer cells; this causes them to burst and release more of the virus which then infects nearby cancer cells and activates the body's immune system. ICIs may be able to work together with OVs to amplify this effect. Vesicular stomatitis virus (VSV)-glycoprotein (GP) is a type of OV that has been shown to effectively destroy cancer cells in animal studies. This first-in-human study will investigate VSV-GP on its own and in combination with an ICI called ezabenlimab for the treatment of late-stage cancer or cancer that has spread to multiple parts of the body. Here, we describe the background and design of this study in progress which aims to find out if VSV-GP alone or in combination with ezabenlimab is effective against cancer, the suitable dose and if any side effects occur. Trial Registration Number: NCT05155332 (ClinicalTrials.gov).


Assuntos
Neoplasias , Terapia Viral Oncolítica , Vírus Oncolíticos , Anticorpos Monoclonais , Linhagem Celular Tumoral , Ensaios Clínicos Fase I como Assunto , Glicoproteínas , Humanos , Inibidores de Checkpoint Imunológico , Neoplasias/genética , Neoplasias/terapia , Vírus Oncolíticos/genética
7.
Molecules ; 26(18)2021 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-34577057

RESUMO

Resveratrol, a natural plant phytoalexin, is produced in response to fungal infection or- UV irradiation. It exists as an isomeric pair with cis- and trans-conformation. Whereas multiple physiological effects of the trans-form, including a pronounced anti-tumoral activity, are nowadays elucidated, much less knowledge exists concerning the cis-isomer. In our work, we analyzed the antiproliferative and cytotoxic properties of cis-resveratrol in four different human tumor entities in direct comparison to trans-resveratrol. We used human cell lines as tumor models for hepatocellular carcinoma (HCC; HepG2, Hep3B), colon carcinoma (HCT-116, HCT-116/p53(-/-)), pancreatic carcinoma (Capan-2, MiaPaCa-2), and renal cell carcinoma (A498, SN12C). Increased cytotoxicity in all investigated tumor cells was observed for the trans-isomer. To verify possible effects of the tumor suppressor p53 on resveratrol-induced cell death, we used wild type and p53-deleted or -mutated cell lines for every tested tumor entity. Applying viability and cytotoxicity assays, we demonstrated a differential, dose-dependent sensitivity towards cis- or trans-resveratrol among the respective tumor types.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Resveratrol , Proteína Supressora de Tumor p53 , Antineoplásicos , Apoptose/efeitos dos fármacos , Humanos
8.
BMC Cancer ; 20(1): 628, 2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32631270

RESUMO

BACKGROUND: Oncolytic virotherapy is an upcoming treatment option for many tumor entities. But so far, a first oncolytic virus only was approved for advanced stages of malignant melanomas. Neuroendocrine tumors (NETs) constitute a heterogenous group of tumors arising from the neuroendocrine system at diverse anatomic sites. Due to often slow growth rates and (in most cases) endocrine non-functionality, NETs are often detected only in a progressed metastatic situation, where therapy options are still severely limited. So far, immunotherapies and especially immunovirotherapies are not established as novel treatment modalities for NETs. METHODS: In this immunovirotherapy study, pancreatic NET (BON-1, QGP-1), lung NET (H727, UMC-11), as well as neuroendocrine carcinoma (NEC) cell lines (HROC-57, NEC-DUE1) were employed. The well characterized genetically engineered vaccinia virus GLV-1 h68, which has already been investigated in various clinical trials, was chosen as virotherapeutical treatment modality. RESULTS: Profound oncolytic efficiencies were found for NET/NEC tumor cells. Besides, NET/NEC tumor cell bound expression of GLV-1 h68-encoded marker genes was observed also. Furthermore, a highly efficient production of viral progenies was detected by sequential virus quantifications. Moreover, the mTOR inhibitor everolimus, licensed for treatment of metastatic NETs, was not found to interfere with GLV-1 h68 replication, making a combinatorial treatment of both feasible. CONCLUSIONS: In summary, the oncolytic vaccinia virus GLV-1 h68 was found to exhibit promising antitumoral activities, replication capacities and a potential for future combinatorial approaches in cell lines originating from neuroendocrine neoplasms. Based on these preliminary findings, virotherapeutic effects now have to be further evaluated in animal models for treatment of Neuroendocrine neoplasms (NENs).


Assuntos
Imunoterapia/métodos , Tumores Neuroendócrinos/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Vaccinia virus/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Terapia Combinada/métodos , Everolimo/farmacologia , Everolimo/uso terapêutico , Estudos de Viabilidade , Humanos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Tumores Neuroendócrinos/imunologia , Tumores Neuroendócrinos/patologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
9.
Int J Mol Sci ; 21(20)2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-33076270

RESUMO

Genetically modified vaccinia viruses (VACVs) have been shown to possess profound oncolytic capabilities. However, tumor cell resistance to VACVs may endanger broad clinical success. Using cell mass assays, viral replication studies, and fluorescence microscopy, we investigated primary resistance phenomena of cell lines of the NCI-60 tumor cell panel to GLV-1h94, a derivative of the Lister strain of VACV, which encodes the enzyme super cytosine deaminase (SCD) that converts the prodrug 5-fluorocytosine (5-FC) into the chemotherapeutic compound 5-fluorouracil (5-FU). After treatment with GLV-1h94 alone, only half of the cell lines were defined as highly susceptible to GLV-1h94-induced oncolysis. When adding 5-FC, 85% of the cell lines became highly susceptible to combinatorial treatment; none of the tested tumor cell lines exhibited a "high-grade resistance" pattern. Detailed investigation of the SCD prodrug system suggested that the cytotoxic effect of converted 5-FU is directed either against the cells or against the virus particles, depending on the balance between cell line-specific susceptibility to GLV-1h94-induced oncolysis and 5-FU sensitivity. The data provided by this work underline that cellular resistance against VACV-based virotherapy can be overcome by virus-encoded prodrug systems. Phase I/II clinical trials are recommended to further elucidate the enormous potential of this combination therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Engenharia Genética/métodos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vaccinia virus/genética , Antineoplásicos/toxicidade , Morte Celular , Linhagem Celular Tumoral , Terapia Combinada/métodos , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Flucitosina/farmacocinética , Fluoruracila/toxicidade , Humanos , Vírus Oncolíticos/enzimologia , Pró-Fármacos , Vaccinia virus/enzimologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
10.
BMC Cancer ; 19(1): 1172, 2019 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-31795974

RESUMO

BACKGROUND: In pediatric sarcomas, outcomes of established therapies still remain poor, especially due to high-grade resistances to chemotherapeutic compounds. Taking novel biological approaches into account, virotherapy was found to be efficient in many pediatric sarcoma types. Also NK cell therapy was denoted to represent a promising upcoming strategy for pediatric sarcoma patients. We here investigated a combinatorial approach employing oncolytic measles vaccine virotherapeutics (MeV) together with activated human NK cells (or PBMCs). METHODS: The human sarcoma cell lines A673 and HT1080 were used to evaluate the efficacy of this combinatorial treatment modality. Oncolysis was determined by measuring real-time cell proliferation using the xCELLigence RTCA SP system. Furthermore, expression of receptors on NK cells and the respective ligands on A673 cells was analyzed by flow cytometry. To measure the protein release of activated NK cells a LEGENDplex™ assay was performed. RESULTS: Monotherapy with MeV led to a time- and dose-dependent oncolytic reduction of A673 and HT1080 sarcoma tumor cell masses. Concurrently, such MeV infections did not change the expression of NK cell ligands MICA/B, ULBP1, 2, and 3, CD112, and CD155. As shown by real-time proliferation assays, infections of A673 and HT1080 sarcoma cells with MeV followed by co-culture with activated NK cells or PBMCs led to enhanced sarcoma cell destruction when compared to the respective monotherapies. In parallel, this dual therapy resulted in an increased release of granzymes, perforin, and granulysin from NK cells. In contrast, expression of activation and ontogenesis receptors on NK cells was not found to be altered after co-culture with MeV-infected A673 sarcoma cells. CONCLUSIONS: Taken together, the combined treatment strategy comprising oncolytic MeV and activated NK cells resulted in enhanced oncolysis of A673 and HT1080 cells when compared to the respective monotherapies. In parallel, we observed an increased release of NK cell activation markers upon co-culture with MeV-infected A673 human sarcoma cells. These results support the onset of clinical trials combining oncolytic virotherapy with NK cell based immunotherapies.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Vírus do Sarampo/fisiologia , Terapia Viral Oncolítica/métodos , Sarcoma/terapia , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Criança , Chlorocebus aethiops , Técnicas de Cocultura , Terapia Combinada , Humanos , Receptor de Morte Celular Programada 1/biossíntese , Receptor de Morte Celular Programada 1/imunologia , Sarcoma/imunologia , Sarcoma/virologia , Células Vero
11.
Neuroendocrinology ; 109(4): 346-361, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31280274

RESUMO

Metastatic neuroendocrine cancer still constitutes a palliative situation, lacking promising treatment options. Oncolytic virotherapy, a novel type of virus-based immunotherapy, lyses tumor cells using genetically engineered viruses thereby activating the immune system to induce an optimized antitumor response which could bring down tumor masses to a stage of minimal residual tumor disease. The oncolytic vector talimogene laherparepvec (T-VEC, herpes simplex virus [HSV] type 1) has already shown excellent safety profiles in clinical studies and has become the first ever FDA/EMA-approved oncolytic virus (OV). This work presents a first preclinical assessment of this state-of-the-art OV, using a panel of human neuroendocrine tumor/neuroendocrine carcinoma (NET/NEC) cell lines. Cytotoxicity, transgene expression, and viral replication patterns were studied. Furthermore, the antiproliferative activity was compared to the one of mTOR inhibitor Everolimus and also interactions between the OV and Everolimus were evaluated. Moreover, virostatic effects of ganciclovir (GCV) on replication of T-VEC were assessed and electron microscopic pictures were taken to comprehend viral envelopment and details of the replication cycle of T-VEC in human neuroendocrine cancer. It could be shown that T-VEC infects, replicates in, and lyses human NET/NEC cells exhibiting high oncolytic efficiencies already at quite low virus concentrations. Interestingly, Everolimus was not found to have any relevant impact on rates of viral replication, but no additive effects could be proved using a combinatorial therapy regimen. On the other hand, GCV was shown to be able to limit replication of T-VEC, thus establishing an important safety feature for future treatments of NET/NEC patients. Taken together, T-VEC opens up a promising novel treatment option for NET/NEC patients, warranting its further preclinical and clinical development.


Assuntos
Produtos Biológicos/uso terapêutico , Tumores Neuroendócrinos/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos , Antivirais/farmacologia , Linhagem Celular Tumoral , Terapia Combinada , Everolimo/uso terapêutico , Ganciclovir/farmacologia , Herpesvirus Humano 1 , Humanos , Transgenes/genética , Replicação Viral
12.
Cell Physiol Biochem ; 51(2): 543-556, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30458450

RESUMO

BACKGROUND/AIMS: Prenylnaringenins are natural prenylflavonoids with anticancer properties. However, the underlying mechanisms have not been elucidated yet. Here we report a novel mode of action of 6- and 8-prenylnaringenin (PN) on human melanoma cells: Inhibition of cellular histone deacetylases (HDACs). METHODS: We performed in silico and in vitro analyses using 6-PN or 8-PN to study a possible interaction of 6-PN or 8-PN with HDAC as well as Western blot and FACS analyses, real-time cell proliferation and cell viability assays to assess the impact of 6-PN and 8-PN on human metastatic melanoma cells. RESULTS: In silico, 6-PN and 8-PN fit into the binding pocket of HDAC2, 4, 7 and 8, binding to the zinc ion of their catalytic center that is essential for enzymatic activity. In vitro, 100 µmol/L of 6-PN or 8-PN inhibited all 11 conserved human HDAC of class I, II and IV. In clinical oncology HDAC inhibitors are currently investigated as new anticancer compounds. In line, treatment of SK-MEL-28 cells with 6-PN or 8-PN induced a hyperacetylation of histone complex H3 within 2 h. Further, 6-PN or 8-PN mediated a prominent, dose-dependent reduction of cellular proliferation and viability of SK-MEL-28 and BLM melanoma cells. This effect was apoptosis-independent and accompanied by down-regulation of mTOR-specific pS6 protein via pERK/pP90 in SK-MEL-28 cells. CONCLUSION: The identification of a broad inhibitory capacity of 6-PN and 8-PN for HDAC enzymes with antiproliferative effects on melanoma cells opens the perspective for clinical application as novel anti-melanoma drugs and the usage as innovative lead structures for chemical modification to enhance pharmacology or inhibitory activities.


Assuntos
Apoptose/efeitos dos fármacos , Flavanonas/farmacologia , Flavonoides/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Humulus/química , Acetilação/efeitos dos fármacos , Sítios de Ligação , Domínio Catalítico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavanonas/química , Flavanonas/isolamento & purificação , Flavonoides/química , Flavonoides/isolamento & purificação , Inibidores de Histona Desacetilases/química , Inibidores de Histona Desacetilases/isolamento & purificação , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Histonas/metabolismo , Humanos , Humulus/metabolismo , Melanoma/metabolismo , Melanoma/patologia , Simulação de Acoplamento Molecular , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/metabolismo , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo
13.
Plant Cell ; 27(11): 3175-89, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26530086

RESUMO

To secure their access to water, light, and nutrients, many plant species have developed allelopathic strategies to suppress competitors. To this end, they release into the rhizosphere phytotoxic substances that inhibit the germination and growth of neighbors. Despite the importance of allelopathy in shaping natural plant communities and for agricultural production, the underlying molecular mechanisms are largely unknown. Here, we report that allelochemicals derived from the common class of cyclic hydroxamic acid root exudates directly affect the chromatin-modifying machinery in Arabidopsis thaliana. These allelochemicals inhibit histone deacetylases both in vitro and in vivo and exert their activity through locus-specific alterations of histone acetylation and associated gene expression. Our multilevel analysis collectively shows how plant-plant interactions interfere with a fundamental cellular process, histone acetylation, by targeting an evolutionarily highly conserved class of enzymes.


Assuntos
Arabidopsis/crescimento & desenvolvimento , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Acetilação/efeitos dos fármacos , Arabidopsis/efeitos dos fármacos , Arabidopsis/enzimologia , Arabidopsis/genética , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Loci Gênicos , Herbicidas/farmacologia , Inibidores de Histona Desacetilases/química , Histonas/metabolismo , Modelos Biológicos , Oxazinas/química , Oxazinas/farmacologia , Feromônios/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética
14.
Acta Radiol ; 58(10): 1167-1173, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28084812

RESUMO

Background Implantation of a transjugular intrahepatic portosystemic shunt (TIPS) induces changes of liver perfusion. Purpose To determine the changes in arterial, portal venous, and total perfusion of the liver parenchyma induced by TIPS using the technique of volume perfusion computed tomography (VPCT) and compare results with invasively measured hepatic intravascular pressure values. Material and Methods VPCT quantification of liver perfusion was performed in 23 patients (mean age, 62.5 ± 8.8 years) with portal hypertension in the pre-TIPS and post-TIPS setting, respectively. A commercially available software package was used for post-processing, enabling separate calculation of the dual (arterial [ALP] and portal venous [PVP]) blood supply and additionally of the hepatic perfusion index (HPI) (HPI = ALP/(ALP + PVP)*100%). Invasive pressure measurements were performed during the intervention, before and after TIPS placement. Liver function tests performed before and after the procedure were compared. Results Mean decrease of pressure gradient through TIPS was 13.3 mmHg. Mean normal values for ALP, PVP, and total perfusion (ALP + PVP) before TIPS were 15.9, 37.7, and 53.5 mL/100 mL/min, respectively, mean HPI was 35.4%. After TIPS, ALP increased to a mean value of 37.7 mL/100 mL/min, PVP decreased (15.7 mL/100 mL/min, P < 0.05), whereas total perfusion remained unchanged (53.4 mL/100 mL/min, P = 0.97). HPI increased (71.9%; P < 0.05). No correlation between invasive pressure measurement and VPCT parameters was observed. After TIPS, liver function tests were found to worsen with a significant increase of bilirubin ( P < 0.05). Conclusion Following TIPS placement, ALP and HPI increased in all patients, whereas PVP markedly decreased. Interestingly, the magnitude of decrease in portosystemic pressure gradients was not found to correlate with VPCT parameters.


Assuntos
Tomografia Computadorizada de Feixe Cônico , Circulação Hepática , Fígado/irrigação sanguínea , Fígado/diagnóstico por imagem , Derivação Portossistêmica Transjugular Intra-Hepática , Idoso , Feminino , Humanos , Fígado/cirurgia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
15.
J Hepatol ; 65(2): 280-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26952006

RESUMO

BACKGROUND & AIMS: No established therapies for patients with hepatocellular carcinoma (HCC) and progression on first-line sorafenib treatment currently exist. This phase I/II trial investigated safety, pharmacokinetics and potential biomarkers of the histone deacetylase inhibitor resminostat and a combination therapy with resminostat and sorafenib. METHODS: Patients with HCC and radiologically confirmed progression on sorafenib were treated in an exploratory, multi-center, open-label, uncontrolled, non-randomized, parallel group phase I/II study. In the combination group (n=38) four dose levels ranged from daily 200 to 600mg resminostat plus 400 to 800mg sorafenib. The monotherapy group (n=19) received 600mg resminostat. RESULTS: 57 patients received treatment. Most common adverse events were gastrointestinal disorders, thrombocytopenia and fatigue. Median maximal histone deacetylase inhibition and highest increase in H4-acetylation matched Tmax of resminostat. Sorafenib or the Child-Pugh score did not affect typical pharmacokinetics characteristics of resminostat. Efficacy assessment as progression-free survival-rate after 6 treatment cycles (12weeks, primary endpoint) was 12.5% for resminostat and 62.5% for resminostat plus sorafenib. Median time to progression and overall survival were 1.8 and 4.1months for resminostat and 6.5 and 8.0months for the combination, respectively. Zinc finger protein 64 (ZFP64) baseline expression in blood cells was found to correlate with overall survival. CONCLUSIONS: The combination of sorafenib and resminostat in HCC patients was safe and showed early signs of efficacy. Sorafenib did not alter the pharmacokinetic profile of resminostat or its histone deacetylase inhibitory activity in vivo. A prognostic and potentially predictive role of ZFP64 for treatment with resminostat should be further investigated in HCC and possibly other cancer indications. LAY SUMMARY: No established therapy for patients with advanced hepatocellular carcinoma and progression under first-line systemic treatment with sorafenib currently exists. Epigenetic modulation by inhibition of histone deacetylases might be able to overcome therapy resistance. This exploratory phase I/II clinical study in patients with radiologically confirmed progression under first-line treatment with sorafenib investigated the histone deacetylases inhibitor resminostat as single agent or in combination with continued application of sorafenib. CLINICAL TRIAL REGISTRATION: The clinical trial has been registered at www.clinicaltrials.gov as NCT00943449.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Protocolos de Quimioterapia Combinada Antineoplásica , Humanos , Ácidos Hidroxâmicos , Niacinamida/análogos & derivados , Compostos de Fenilureia , Sorafenibe , Sulfonamidas
16.
Eur Radiol ; 26(12): 4277-4283, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27048535

RESUMO

OBJECTIVES: Establishment of transjugular intrahepatic portosystemic shunts (TIPS) constitutes a standard procedure in patients suffering from portal hypertension. The most difficult step in TIPS placement is blind puncture of the portal vein. This study aimed to evaluate three-dimensional mapping of portal vein branches and targeted puncture of the portal vein. METHODS: Twelve consecutive patients suffering from refractory ascites by liver cirrhosis were included in this retrospective study to evaluate feasibility, technical success and procedural time of C-arm CT-targeted puncture of the portal vein. As a control, 22 patients receiving TIPS placement with fluoroscopy-guided blind puncture were included to compare procedural time. RESULTS: Technical success could be obtained in 100 % of the study group (targeted puncture) and in 95.5 % of the control group (blind puncture). Appropriate, three-dimensional C-arm CT-guided mapping of the portal vein branches could be achieved in all patients. The median number of punctures in the C-arm CT-guided study group was 2 ± 1.3 punctures. Procedural time was significantly lower in the study group (14.8 ± 8.2 min) compared to the control group (32.6 ± 22.7 min) (p = 0.02). CONCLUSIONS: C-arm CT-guided portal vein mapping is technically feasible and a promising tool for TIPS placement resulting in a significant reduction of procedural time. KEY POINTS: • C-arm CT-mapping of the portal vein for 3D TIPS guidance is feasible. • Targeted punctures of the portal vein by C-arm CT reduce procedural time. • A decreased number of punctures could improve patient safety.


Assuntos
Hipertensão Portal/cirurgia , Imageamento Tridimensional/métodos , Derivação Portossistêmica Transjugular Intra-Hepática/métodos , Radiografia Intervencionista/métodos , Tomografia Computadorizada por Raios X/métodos , Adulto , Idoso , Estudos de Viabilidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Veia Porta/cirurgia , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/instrumentação , Resultado do Tratamento
17.
Nutr J ; 15(1): 47, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27142426

RESUMO

Natural killer (NK) cells as part of the innate immune system represent the first line of defence against (virus-) infected and malignantly transformed cells. The emerging field of nutritional immunology focuses on compounds featuring immune-modulating activities in particular on NK cells, which e.g. can be exploited for cancer prevention and treatment. The plant-based nutrition resveratrol is a ternary hydroxylated stilbene, which is present in many foods and beverages, respectively. In humans it comprises a large variety of distinct biological activities. Interestingly, resveratrol strongly modulates the immune response including the activity of NK cells. This review will give an overview on NK cell functions and summarize the resveratrol-mediated modulation thereof.


Assuntos
Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/prevenção & controle , Fenômenos Fisiológicos da Nutrição/imunologia , Estilbenos/farmacologia , Antineoplásicos Fitogênicos , Dieta , Humanos , Neoplasias/imunologia , Plantas Comestíveis/química , Resveratrol , Estilbenos/administração & dosagem , Estilbenos/farmacocinética
18.
Virol J ; 12: 37, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25889921

RESUMO

BACKGROUND/AIMS: Aim of this retrospective study was to analyze the efficacy, safety, and predictors of treatment success for first-generation-PI triple therapies, including either boceprevir or telaprevir, in a mono-centric "real-life" setting with respect to SVR 24. PATIENTS: 131 patients (102 patients telaprevir, 29 patients boceprevir) were treated. Of these, 33/131 patients were treatment naïve, 72/131 patients had been pretreated with PEG-IFN/RBV (PR) (thereof: 36 with non-response, 30 with relapse, 6 unknown), and 26/131 patients previously had received non-pegylated interferon. 96/131 patients were infected with HCV genotype 1b. 41/131 patients had liver cirrhosis. RESULTS: 95/131 (73%) patients achieved SVR 24. SVR rates for subgroups were: 26/33 (79%) for treatment naïve, 25/30 (83%) for PR-relapse, 20/36 (56%) for PR-non-response, 21/26 (81%) for non-PR pretreated patients, (26/41) 63% for patients with liver cirrhosis, 23/35 (66%) genotype 1a, 72/96 (75%) genotype 1b. Predictors of SVR 24 were eRVR and a negative viral load at PI-treatment week 4 (p < 0.0001), negative predictors were quantifiable HCV viral load at PI-treatment week 4 (p < 0.0001), baseline platelet count < 100/nl (p < 0.0001), and previous PR-non-response (p = 0.006). 33/131 (25%) patients discontinued treatment prematurely, of those 14/131 (11%) patients due to virological failure. Side effects were frequent (anemia 59/131 [45%], severe infections 6/131 [5%]). CONCLUSIONS: According to our SVR 24 results, efficacy of PI-based triple therapy in our "real-life" cohort is comparable to the large multi-centric clinical trials. Pronounced side effects are frequent during therapy and often need complex therapeutic interventions. Since new DAA are available, it is open to discussion, if first-generation PI-triple therapy is no longer indicated at all.


Assuntos
Antivirais/administração & dosagem , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Inibidores de Proteases/administração & dosagem , Adulto , Quimioterapia Combinada , Feminino , Hepacivirus/classificação , Hepacivirus/genética , Hepacivirus/fisiologia , Hepatite C Crônica/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/administração & dosagem , Prolina/administração & dosagem , Prolina/análogos & derivados , Estudos Retrospectivos
19.
Pol J Microbiol ; 64(1): 15-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26094311

RESUMO

BACKGROUND: Knowledge on HBV prevalence and genotype distribution in Europe still is hampered by lack of coherent data sampling, small numbers of patients studied so far, and also modern times migration which influences both parameters in a quite dynamic manner. To find out whether HBV prevalence and genotype distribution has undergone any significant changes over the past decades, we have analyzed our cohort of HBV patients. METHODS: Retrospective analysis of virological data and correlation with the epidemiological backgrounds of 408 chronically HBV-infected patients, followed in the year 2009 at Tübingen Virus Hepatitis Center, Germany. RESULTS: A background of migration was found in more than 80% of our HBV patients, displaying an origin from 41 different countries. Analysis of the genotypes revealed that genotype A predominated only among patients from Central Europe with 55.8% while genotype D, known to be most common worldwide, was most prevalent in patients born in Eastern and Southern Europe, Central Asia and Middle East, exhibiting a range from 81% to 94%. In Central Europe, genotype A was particularly seen in older patients as compared to genotype D that predominated in the younger patients. CONCLUSIONS: These data suggest that Central Europe is straight on its way to switch from genotype A to genotype D. One reason for this significant shift may be related to the ongoing European and global migration flow.


Assuntos
Vírus da Hepatite B/genética , Hepatite B Crônica/epidemiologia , Hepatite B Crônica/virologia , Adolescente , Adulto , Idoso , Europa (Continente)/epidemiologia , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Estudos Retrospectivos , Migrantes , Carga Viral , Adulto Jovem
20.
Wien Med Wochenschr ; 165(11-12): 258-61, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25925225

RESUMO

Flavonoids form a substantial group of secondary plant metabolites that display several health-promoting effects. Therefore, prenylflavonoids, a subclass of flavonoids, have attracted increasing attention. Here, we investigated the possible anti-cancer potential of 6-prenylnaringenin (6-PN) and 8-prenylnaringenin (8-PN), two prenylflavonoids present in hops and beer and demonstrate an unexpectedly pronounced, dose-dependent reduction of cellular proliferation of human PC-3 prostate cancer and UO.31 renal carcinoma cells upon treatment. Based on these findings 6-PN and 8-PN are currently further clinically evaluated in detail.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Flavanonas/farmacologia , Flavonoides/farmacologia , Humulus , Fitoterapia , Células Tumorais Cultivadas/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA