Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Eur J Cancer ; 170: 196-208, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35671543

RESUMO

BACKGROUND: Childhood cancer is still a leading cause of death around the world. To improve outcomes, there is an urgent need for tailored treatment. The systematic evaluation of existing preclinical data can provide an overview of what is known and identify gaps in the current knowledge. Here, we applied the target actionability review (TAR) methodology to assess the strength and weaknesses of available scientific literature on CDK4/6 as a therapeutic target in paediatric solid and brain tumours by structured critical appraisal. METHODS: Using relevant search terms in PubMed, a list of original publications investigating CDK4/6 in paediatric solid tumour types was identified based on relevancy criteria. Each publication was annotated for the tumour type and categorised into separate proof-of-concept (PoC) data modules. Based on rubrics, quality and experimental outcomes were scored independently by two reviewers. A third reviewer evaluated and adjudicated score discrepancies. Scores for each PoC module were averaged for each tumour type and visualised in a heatmap matrix in the publicly available R2 data portal. RESULTS AND CONCLUSIONS: This CDK4/6 TAR, generated by analysis of 151 data entries from 71 publications, showed frequent genomic aberrations of CDK4/6 in rhabdomyosarcoma, osteosarcoma, high-grade glioma, medulloblastoma, and neuroblastoma. However, a clear correlation between CDK4/6 aberrations and compound efficacy is not coming forth from the literature. Our analysis indicates that several paediatric indications would need (further) preclinical evaluation to allow for better recommendations, especially regarding the dependence of tumours on CDK4/6, predictive biomarkers, resistance mechanisms, and combination strategies. Nevertheless, our TAR heatmap provides support for the relevance of CDK4/6 inhibition in Ewing sarcoma, medulloblastoma, malignant peripheral nerve sheath tumour and to a lesser extent neuroblastoma, rhabdomyosarcoma, rhabdoid tumour and high-grade glioma. The interactive heatmap is accessible through R2 [r2platform.com/TAR/CDK4_6].


Assuntos
Neoplasias Encefálicas , Neoplasias Cerebelares , Quinase 6 Dependente de Ciclina/metabolismo , Meduloblastoma , Neuroblastoma , Rabdomiossarcoma , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Criança , Quinase 4 Dependente de Ciclina , Humanos
2.
Eur J Cancer ; 162: 107-117, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34963094

RESUMO

BACKGROUND: Owing to the high numbers of paediatric cancer-related deaths, advances in therapeutic options for childhood cancer is a heavily studied field, especially over the past decade. Classical chemotherapy offers some therapeutic benefit but has proven long-term complications in survivors, and there is an urgent need to identify novel target-driven therapies. Replication stress is a major cause of genomic instability in cancer, triggering the stalling of the replication fork. Failure of molecular response by DNA damage checkpoints, DNA repair mechanisms and restarting the replication forks can exacerbate replication stress and initiate cell death pathways, thus presenting as a novel therapeutic target. To bridge the gap between preclinical evidence and clinical utility thereof, we apply the literature-driven systematic target actionability review methodology to published proof-of-concept (PoC) data related to the process of replication stress. METHODS: A meticulous PubMed literature search was performed to gather replication stress-related articles (published between 2014 and 2021) across 16 different paediatric solid tumour types. Articles that fulfilled inclusion criteria were uploaded into the R2 informatics platform [r2.amc.nl] and assessed by critical appraisal. Key evidence based on nine pre-established PoC modules was summarised, and scores based on the quality and outcome of each study were assigned by two separate reviewers. Articles with discordant modules/scores were re-scored by a third independent reviewer, and a final consensus score was agreed upon by adjudication between all three reviewers. To visualise the final scores, an interactive heatmap summarising the evidence and scores associated with each PoC module across all, including paediatric tumour types, were generated. RESULTS AND CONCLUSIONS: 145 publications related to targeting replication stress in paediatric tumours were systematically reviewed with an emphasis on DNA repair pathways and cell cycle checkpoint control. Although various targets in these pathways have been studied in these diseases to different extents, the results of this extensive literature search show that ATR, CHK1, PARP or WEE1 are the most promising targets using either single agents or in combination with chemotherapy or radiotherapy in neuroblastoma, osteosarcoma, high-grade glioma or medulloblastoma. Targeting these pathways in other paediatric malignancies may work as well, but here, the evidence was more limited. The evidence for other targets (such as ATM and DNA-PK) was also limited but showed promising results in some malignancies and requires more studies in other tumour types. Overall, we have created an extensive overview of targeting replication stress across 16 paediatric tumour types, which can be explored using the interactive heatmap on the R2 target actionability review platform [https://hgserver1.amc.nl/cgi-bin/r2/main.cgi?option=imi2_targetmap_v1].


Assuntos
Neoplasias Ósseas , Neoplasias Cerebelares , Meduloblastoma , Pontos de Checagem do Ciclo Celular , Criança , Reparo do DNA , Humanos
3.
Diana Rose E Ranoa; Robin L Holland; Fadi G Alnaji; Kelsie J Green; Leyi Wang; Richard L Fredrickson; Tong Wang; George N Wong; Johnny Uelmen; Sergei Maslov; Ahmed Elbanna; Zachary J Weiner; Alexei V Tkachenko; Hantao Zhang; Zhiru Liu; Sanjay J Patel; John M Paul; Nickolas P Vance; Joseph G Gulick; Sandeep P Satheesan; Isaac J Galvan; Andrew Miller; Joseph Grohens; Todd J Nelson; Mary P Stevens; P. Mark Hennessy; Robert C Parker; Edward Santos; Charles Brackett; Julie D Steinman; Melvin R Fenner Jr.; Kristin Dohrer; Kraig Wagenecht; Michael DeLorenzo; Laura Wilhelm-Barr; Brian R Brauer; Catherine Best-Popescu; Gary Durack; Nathan Wetter; David M Kranz; Jessica Breitbarth; Charlie Simpson; Julie A Pryde; Robin N Kaler; Chris Harris; Allison C Vance; Jodi L Silotto; Mark Johnson; Enrique Valera; Patricia K Anton; Lowa Mwilambwe; Stephen B Bryan; Deborah S Stone; Danita B Young; Wanda E Ward; John Lantz; John A Vozenilek; Rashid Bashir; Jeffrey S Moore; Mayank Garg; Julian C Cooper; Gillian Snyder; Michelle H Lore; Dustin L Yocum; Neal J Cohen; Jan E Novakofski; Melanie J Loots; Randy L Ballard; Mark Band; Kayla M Banks; Joseph D Barnes; Iuliana Bentea; Jessica Black; Jeremy Busch; Hannah Christensen; Abigail Conte; Madison Conte; Michael Curry; Jennifer Eardley; April Edwards; Therese Eggett; Judes Fleurimont; Delaney Foster; Bruce W Fouke; Nicholas Gallagher; Nicole Gastala; Scott A Genung; Declan Glueck; Brittani Gray; Andrew Greta; Robert M Healy; Ashley Hetrick; Arianna A Holterman; Nahed Ismail; Ian Jasenof; Patrick Kelly; Aaron Kielbasa; Teresa Kiesel; Lorenzo M Kindle; Rhonda L Lipking; Yukari C Manabe; Jade ? Mayes; Reubin McGuffin; Kenton G McHenry; Agha Mirza; Jada Moseley; Heba H Mostafa; Melody Mumford; Kathleen Munoz; Arika D Murray; Moira Nolan; Nil A Parikh; Andrew Pekosz; Janna Pflugmacher; Janise M Phillips; Collin Pitts; Mark C Potter; James Quisenberry; Janelle Rear; Matthew L Robinson; Edith Rosillo; Leslie N Rye; MaryEllen Sherwood; Anna Simon; Jamie M Singson; Carly Skadden; Tina H Skelton; Charlie Smith; Mary Stech; Ryan Thomas; Matthew A Tomaszewski; Erika A Tyburski; Scott Vanwingerden; Evette Vlach; Ronald S Watkins; Karriem Watson; Karen C White; Timothy L Killeen; Robert J Jones; Andreas C Cangellaris; Susan A Martinis; Awais Vaid; Christopher B Brooke; Joseph T Walsh; William C Sullivan; Rebecca L Smith; Nigel D Goldenfeld; Timothy M Fan; Paul J Hergenrother; Martin D Burke.
Preprint em Inglês | medRxiv | ID: ppmedrxiv-21261548

RESUMO

In the Fall of 2020, many universities saw extensive transmission of SARS-CoV-2 among their populations, threatening the health of students, faculty and staff, the viability of in-person instruction, and the health of surrounding communities.1, 2 Here we report that a multimodal "SHIELD: Target, Test, and Tell" program mitigated the spread of SARS-CoV-2 at a large public university, prevented community transmission, and allowed continuation of in-person classes amidst the pandemic. The program combines epidemiological modelling and surveillance (Target); fast and frequent testing using a novel and FDA Emergency Use Authorized low-cost and scalable saliva-based RT-qPCR assay for SARS-CoV-2 that bypasses RNA extraction, called covidSHIELD (Test); and digital tools that communicate test results, notify of potential exposures, and promote compliance with public health mandates (Tell). These elements were combined with masks, social distancing, and robust education efforts. In Fall 2020, we performed more than 1,000,000 covidSHIELD tests while keeping classrooms, laboratories, and many other university activities open. Generally, our case positivity rates remained less than 0.5%, we prevented transmission from our students to our faculty and staff, and data indicate that we had no spread in our classrooms or research laboratories. During this fall semester, we had zero COVID-19-related hospitalizations or deaths amongst our university community. We also prevented transmission from our university community to the surrounding Champaign County community. Our experience demonstrates that multimodal transmission mitigation programs can enable university communities to achieve such outcomes until widespread vaccination against COVID-19 is achieved, and provides a roadmap for how future pandemics can be addressed.

4.
Mol Cancer Ther ; 20(6): 1161-1172, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33850004

RESUMO

Neuroblastoma tumors frequently overexpress the anti-apoptotic protein B-cell lymphoma/leukemia 2 (BCL-2). We previously showed that treating BCL-2-dependent neuroblastoma cells with the BCL-2 inhibitor venetoclax results in apoptosis, but unfortunately partial therapy resistance is observed. The current study describes the identification of drugs capable of resensitizing venetoclax-resistant neuroblastoma cells to venetoclax. To examine these effects, venetoclax resistance was induced in BCL-2-dependent neuroblastoma cell lines KCNR and SJNB12 by continuous exposure to high venetoclax concentrations. Non-resistant and venetoclax-resistant neuroblastoma cell lines were exposed to a 209-compound library in the absence and presence of venetoclax to identify compounds that were more effective in the venetoclax-resistant cell lines under venetoclax pressure. Top hits were further validated in combination with venetoclax using BCL-2-dependent neuroblastoma model systems. Overall, high-throughput drug screening identified the MDM2 inhibitor idasanutlin as a promising resensitizing agent for venetoclax-resistant neuroblastoma cell lines. Idasanutlin treatment induced BAX-mediated apoptosis in venetoclax-resistant neuroblastoma cells in the presence of venetoclax, whereas it caused p21-mediated growth arrest in control cells. In vivo combination treatment showed tumor regression and superior efficacy over single-agent therapies in a BCL-2-dependent neuroblastoma cell line xenograft and a patient-derived xenograft. However, xenografts less dependent on BCL-2 were not sensitive to venetoclax-idasanutlin combination therapy. This study demonstrates that idasanutlin can overcome resistance to the BCL-2 inhibitor venetoclax in preclinical neuroblastoma model systems, which supports clinical development of a treatment strategy combining the two therapies.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Neuroblastoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-mdm2/uso terapêutico , Pirrolidinas/uso terapêutico , para-Aminobenzoatos/uso terapêutico , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-mdm2/farmacologia , Pirrolidinas/farmacologia , para-Aminobenzoatos/farmacologia
5.
Eur J Cancer ; 142: 1-9, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33190064

RESUMO

BACKGROUND: Despite intensive treatment protocols and recent advances, neuroblastomas still account for approximately 15% of all childhood cancer deaths. In contrast with adult cancers, p53 pathway inactivation in neuroblastomas is rarely caused by p53 mutation but rather by altered MDM2 or p14ARF expression. Moreover, neuroblastomas are characterised by high proliferation rates, frequently triggered by pRb pathway dysfunction due to aberrant expression of cyclin D1, CDK4 or p16INK4a. Simultaneous disturbance of these pathways can occur via co-amplification of MDM2 and CDK4 or homozygous deletion of CDKN2A, which encodes both p14ARF and p16INK4a. METHODS AND RESULTS: We examined whether both single and combined inhibition of MDM2 and CDK4/6 is effective in reducing neuroblastoma cell viability. In our panel of ten cell lines with a spectrum of aberrations in the p53 and pRb pathway, idasanutlin and abemaciclib were the most potent MDM2 and CDK4/6 inhibitors, respectively. No correlation was observed between the genetic background and response to the single inhibitors. We confirmed this lack of correlation in isogenic systems overexpressing MDM2 and/or CDK4. In addition, combined inhibition did not result in synergistic effects. Instead, abemaciclib diminished the pro-apoptotic effect of idasanutlin, leading to slightly antagonistic effects. In vivo treatment with idasanutlin and abemaciclib led to reduced tumour growth compared with single drug treatment, but no synergistic response was observed. CONCLUSION: We conclude that p53 and pRb pathway aberrations cannot be used as predictive biomarkers for neuroblastoma sensitivity to MDM2 and/or CDK4/6 inhibitors. Moreover, we advise to be cautious with combining these inhibitors in neuroblastomas.


Assuntos
Neuroblastoma/genética , Medicina de Precisão/métodos , Proteína Supressora de Tumor p53/metabolismo , Animais , Humanos , Camundongos , Neuroblastoma/patologia
6.
Eur J Cancer ; 130: 168-181, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32224415

RESUMO

BACKGROUND: Children with cancer are in urgent need of new therapies, as approximately 25% of patients experience a relapse and 20% succumb to their disease. Moreover, the majority of survivors suffer from clinically relevant health problems. Repurposing of targeted agents developed for adult indications could provide novel therapeutic options for paediatric cancer patients. To prioritise targeted drugs for paediatric clinical development, we applied a systematic review methodology to develop a Target Actionability Review (TAR) strategy. These TARs assess the strength and completeness of published preclinical proof-of-concept (PoC) data by structured critical appraisal of and summarising the available scientific literature for a specific target (pathway) and the associated drugs in paediatric tumours. METHODS: A sensitive literature search in PubMed was performed and relevant papers were identified. For each paper, the individual experimental findings were extracted, marked for paediatric tumour type and categorised into nine separate PoC data modules. Each experimental finding was scored for experimental outcome and quality independently by two reviewers; discrepancies were assessed by a third reviewer and resolved by adjudication. Scores corresponding to one PoC module were merged for each tumour type and visualised in a heat map matrix in the publicly available R2 data portal [r2.amc.nl]. RESULTS AND CONCLUSIONS: To test our TAR methodology, we conducted a pilot study on MDM2 and TP53. The heat map generated from analysis of 161 publications provides a rationale to support drug development in specific paediatric solid and brain tumour types. Furthermore, our review highlights tumour types where preclinical data are incomplete or lacking and for which additional preclinical testing is advisable.


Assuntos
Neoplasias/epidemiologia , Estudo de Prova de Conceito , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pediatria
7.
Neuro Oncol ; 19(12): 1607-1617, 2017 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-28482026

RESUMO

BACKGROUND: Embryonal tumor with multilayered rosettes (ETMR) is a rare and aggressive embryonal brain tumor that solely occurs in infants and young children and has only recently been recognized as a separate brain tumor entity in the World Health Organization classification for CNS tumors. Patients have a very dismal prognosis with a median survival of 12 months upon diagnosis despite aggressive treatment. The aim of this study was to develop novel treatment regimens in a preclinical drug screen in order to inform potentially more active clinical trial protocols. METHODS: We have carried out an in vitro and in vivo drug screen using the ETMR cell line BT183 and its xenograft model. Furthermore, we have generated the first patient-derived xenograft (PDX) model for ETMR and evaluated our top drug candidates in an in vitro drug screen using this model. RESULTS: BT183 cells are very sensitive to the topoisomerase inhibitors topotecan and doxorubicin, to the epigenetic agents decitabine and panobinostat, to actinomycin D, and to targeted drugs such as the polo-like kinase 1 (PLK1) inhibitor volasertib, the aurora kinase A inhibitor alisertib, and the mammalian target of rapamycin (mTOR) inhibitor MLN0128. In xenograft mice, monotherapy with topotecan, volasertib, and actinomycin D led to a temporary response in tumor growth and a significant increase in survival. Finally, using multi-agent treatment regimens of topotecan or doxorubicin combined with methotrexate and vincristine, the response in tumor growth and survival was further increased compared with mice receiving single treatments. CONCLUSIONS: We have identified several promising candidates for combination therapies in future clinical trials for ETMR patients.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Dactinomicina/farmacologia , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Tumores Neuroectodérmicos Primitivos/tratamento farmacológico , Pteridinas/farmacologia , Topotecan/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Pré-Escolar , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Embrionárias de Células Germinativas/patologia , Tumores Neuroectodérmicos Primitivos/patologia , Inibidores da Topoisomerase I/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Sci Rep ; 6: 30486, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27460417

RESUMO

The majority of low-grade and secondary high-grade gliomas carry heterozygous hotspot mutations in cytosolic isocitrate dehydrogenase 1 (IDH1) or the mitochondrial variant IDH2. These mutations mostly involve Arg132 in IDH1, and Arg172 or Arg140 in IDH2. Whereas IDHs convert isocitrate to alpha-ketoglutarate (α-KG) with simultaneous reduction of NADP(+) to NADPH, these IDH mutants reduce α-KG to D-2-hydroxyglutarate (D-2-HG) while oxidizing NADPH. D-2-HG is a proposed oncometabolite, acting via competitive inhibition of α-KG-dependent enzymes that are involved in metabolism and epigenetic regulation. However, much less is known about the implications of the metabolic stress, imposed by decreased α-KG and NADPH production, for tumor biology. We here present a novel heterozygous IDH1 mutation, IDH1(R314C), which was identified by targeted next generation sequencing of a high grade glioma from which a mouse xenograft model and a cell line were generated. IDH1(R314C) lacks isocitrate-to-α-KG conversion activity due to reduced affinity for NADP(+), and differs from the IDH1(R132) mutants in that it does not produce D-2-HG. Because IDH1(R314C) is defective in producing α-KG and NADPH, without concomitant production of the D-2-HG, it represents a valuable tool to study the effects of IDH1-dysfunction on cellular metabolism in the absence of this oncometabolite.


Assuntos
Astrocitoma/enzimologia , Astrocitoma/genética , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Isocitrato Desidrogenase/genética , Mutação/genética , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Glutaratos/metabolismo , Células HEK293 , Heterozigoto , Humanos , Isocitratos/metabolismo , Camundongos , NADP/metabolismo , Gradação de Tumores , Multimerização Proteica
9.
Int Immunol ; 13(8): 1031-42, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11470773

RESUMO

The generation of thymic NK1.1(+)alpha beta T (NKT) cells involves positive selection of cells enriched for V(alpha)14/V(beta)8 TCR by CD1d MHC class I molecules. However, it has not been determined whether positive selection is preceded by pre-TCR-dependent beta selection. Here we studied NKT cell development in CD3 signaling-deficient mice (CD3 zeta/eta(-/-) and/or p56(lck-/-)) and TCR alpha-deficient mice. In contrast to wild-type mice, NK1.1(+) thymocytes in CD3 signaling-deficient mice are approximately 10-fold reduced in number, do not exhibit V(alpha)14-J(alpha)281 rearrangements and fail to express alpha beta TCR at the cell surface. However, they exhibit TCR beta VDJ rearrangements and pre-T alpha mRNA, suggesting that they contain pre-NKT cells. Strikingly, pre-NKT cells of CD3 zeta/Lck double-deficient mice fail to express TCR beta mRNA and protein. Whereas in wild-type NKT cells TCR beta VDJ junctions are selected for productive V(beta)8 and against productive V(beta)5 rearrangements, V(beta)8 and V(beta)5 rearrangements are non-selected in pre-NKT cells of CD3 signaling-deficient mice. Thus, pre-NKT cell development in CD3 signaling-deficient mice is blocked after rearrangement of TCR beta VDJ genes but before expression of TCR beta proteins. Most NKT cells of TCR alpha-deficient mice exhibit cell surface gamma delta TCR. In contrast to pre-NKT cells of CD3 signaling-deficient mice, approximately 25% of NKT cells of TCR alpha-deficient mice exhibit intracellular TCR beta polypeptide chains. Moreover, both V(beta)8 and V(beta)5 families are selected for in-frame VDJ joints in the TCR beta(+) NKT cell subset of TCR alpha-deficient mice. The data suggest that CD3 signals regulate initial TCR beta VDJ gene expression prior to beta selection in developing pre-NKT cells.


Assuntos
Rearranjo Gênico da Cadeia beta dos Receptores de Antígenos dos Linfócitos T , Células Matadoras Naturais/metabolismo , Complexo Receptor-CD3 de Antígeno de Linfócitos T/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Citometria de Fluxo , Receptores de Hialuronatos/metabolismo , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Precursores de RNA/metabolismo , Complexo Receptor-CD3 de Antígeno de Linfócitos T/biossíntese , Complexo Receptor-CD3 de Antígeno de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T alfa-beta/deficiência
10.
J Immunol ; 165(7): 3663-72, 2000 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11034370

RESUMO

Cytotoxic T (Tc) cells deficient in perforin lyse Fas-negative targets after lengthy incubation periods. This process is independent of granzymes, and killing occurs via the Fas pathway for the following reasons. Interaction of perforin-deficient Tc cells with Fas-negative targets leads to an up-regulation of Fas that is dependent on Ag recognition, de novo synthesis, and transport of proteins to the target cell surface. Treatment of effectors with brefeldin A, but not with the exocytosis inhibitor concanamycin, inhibited this process. Lysis of targets is inhibited by anti-Fas Abs, soluble mouse Fas-Fc, and the caspase-cascade inhibitor, crm-A. Targets from Fas-mutant lpr mice are refractory to lysis, and Tc cells from mice deficient in Fas- and perforin-mediated lysis do not lyse Fas-negative targets. The possible relevance of this exocytosis-independent cytolytic process in the regulation of T cell activity and control of pathogens is discussed.


Assuntos
Apoptose/imunologia , Testes Imunológicos de Citotoxicidade , Exocitose/imunologia , Macrolídeos , Linfócitos T Citotóxicos/imunologia , Receptor fas/biossíntese , Animais , Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Brefeldina A/farmacologia , Técnicas de Cocultura , Cicloeximida/farmacologia , Testes Imunológicos de Citotoxicidade/métodos , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/genética , Dactinomicina/farmacologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Exocitose/efeitos dos fármacos , Exocitose/genética , Granzimas , Humanos , Imunossupressores/farmacologia , Isoantígenos/genética , Isoantígenos/imunologia , Células L/citologia , Células L/efeitos dos fármacos , Células L/imunologia , Leucemia L1210/imunologia , Leucemia L1210/patologia , Sarcoma de Mastócitos/imunologia , Sarcoma de Mastócitos/patologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Perforina , Proteínas Citotóxicas Formadoras de Poros , Serina Endopeptidases/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/patologia , Receptor fas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA