• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 53
  • 29
  • 13
  • 5
  • 4
  • 1
  • 1
  • 1
  • Tagged with
  • 108
  • 31
  • 30
  • 21
  • 19
  • 18
  • 15
  • 15
  • 15
  • 13
  • 12
  • 12
  • 12
  • 11
  • 11
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
51

Avaliação da metilação do gene TP53 e instabilidade genômica em ratos expostos a metionina e doxorrubicina / TP53 gene methylation and genomic instability in methionine and doxorubicin exposed rats

Cátia Lira do Amaral 08 February 2010 (has links)
O estado de metilação é suscetível a mudanças quando os organismos são expostos a agentes ambientais tais como componentes dos alimentos e medicamentos. Uma dieta rica em metionina (Met) poderia modular a concentração de S-adenosilmetionina (SAM) e S-adenosilhomocisteína (SAH) e alterar o estado de metilação da região promotora de genes supressores de tumores. Tanto a hipometilação global quanto a hipermetilação de genes específicos estão envolvidas na instabilidade genômica e poderiam resultar em dano ao DNA. Este estudo avalia se a dieta suplementada com Met associada a doxorrubicina (DXR), um fármaco antitumoral que induz espécies reativas, resulta em alterações no estado de metilação da região promotora do gene TP53, na razão SAM/SAH, na concentração de glutationa (GSH) e em dano ao DNA. Quarenta ratos machos Wistar foram separados em dois grupos: dieta suplementada com Met (ração comercial acrescida de 2% Met) e dieta controle (ração comercial) por seis semanas. Cada grupo foi subdividido em dois subgrupos que receberam DXR (1mg/Kg) ou solução salina intraperitoneal na terceira e sexta semanas de tratamento. Os rins e fígado foram utilizados para isolamento do DNA, determinação da concentração de SAM, SAH e GSH, e análise da instabilidade genômica. Todos os grupos apresentaram o mesmo estado de metilação da região promotora do gene TP53, determinado pelo método de análise de restrição combinada com bissulfito (COBRA). Este fato poderia ser explicado pelo índice de metilação (razão SAM/SAH) que permaneceu inalterado, possivelmente devido a uma adaptação do ciclo da Met que manteve a concentração de SAM. A depleção de GSH não ocorreu quando DXR foi associada a dieta suplementada com Met. Portanto, a suplementação com Met manteve a concentração de GSH em ratos tratados com DXR. A dieta suplementada com Met não induziu instabilidade genômica e não alterou o dano ao DNA induzido pela DXR. Em conclusão, DXR induz depleção de GSH que é inibida pela suplementação com Met. Entretanto, a mesma suplementação não previne a instabilidade genômica induzida pela DXR. A dieta suplementada com Met aumenta a concentração de SAH renal sem alterar a concentração de SAM e GSH. Tanto a dieta suplementada quanto a DXR não induzem hipermetilação na região promotora do gene TP53. / The DNA methylation status is susceptible to changes when organisms are exposed to environmental agents such as food components and drugs. A methionine-rich (Met) diet may modulate S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) concentrations, which could change the DNA methylation status in the promoter region of tumor suppressor genes. Global hipomethylation and gene-specific hipermethylation are involved in genomic instability and it could result in DNA damage. This study intends to evaluate if a Met-rich diet associated with doxorubicin (DXR), an antitumoral drug that induces reactive species, result in changes in the methylation status of the TP53 gene promoter, in the SAM/SAH ratio, in glutathione levels (GSH) and in DNA damage. Forty male Wistar rats were separated into two groups: Met-rich diet (standard chow plus 2% Met), and control diet (standard chow) for six weeks. Each group was subdivided into another two groups that received DXR (1mg/kg) or saline intraperitoneally in the third and sixth weeks of the experiment. The kidneys and the liver were removed for DNA isolation, SAM, SAH and GSH determination, and genomic instability assay. All groups showed the same unmethylated status in the TP53 promoter according to the Combined Bisulfite Restriction Analysis (COBRA). This could be explained by the fact that the methylation index (SAM/SAH ratio) remained unchanged, possibly because of an adaptive Met pathway that maintains SAM levels. GSH depletion did not occur when DXR was associated with the Met-rich diet. As a matter of fact, the Met-rich diet improved GSH concentration in DXR-treated rats. Met-rich diet did not induce genomic instability, and it did not alter DNA damage induced by DXR. In conclusion, DXR induces GSH depletion which is inhibited by Met supplementation. However, Met-rich diet may not prevent genomic instability induced by DXR. A Met-rich diet increases SAH levels; however, it does not change GSH and SAM levels. Neither Met supplementation nor DXR induced DNA hypermethylation in the TP53 gene promoter.
52

<em>TP53</em> as clinical marker in head and neck cancer

Peltonen, J. (Jenni) 04 October 2011 (has links)
Abstract The prognosis of patients with head and neck squamous cell carcinoma has improved only little during the last decades. Clinical markers for the biological aggressiveness of the cancer are few. The most reliable prognostic indicator is the stage of the disease. Research of the significance of the TP53 tumor suppressor gene as a predictive marker for prognosis and response to treatment in head and neck cancer has given discrepant results. One reason is probably the attempt to use p53 immunohistochemistry as a surrogate for TP53 mutations. However, in immunohistochemistry the protein is analyzed and thus the result does not usually correlate with TP53 mutations. The marker for prognosis of the response to treatment has to be reliable, and the analytical method needs to be both sensitive and specific. In addition to a sensitive method for TP53 mutation analysis the localization and quality of the mutations have to be analyzed to reveal the significance of the mutation on the function of the p53 protein. In this study, TP53 mutations were analyzed, using a sensitive PCR-SSCP method, in the tumors of patients with head and neck squamous cell carcinoma. The quality and localization of mutations were analyzed by sequencing. The frequency of the TP53 mutations and the effect on the function of the p53 protein were studied using IARC TP53 mutation database and literature. Correlation of TP53 mutations with chemical exposure and their significance on prognosis and response to radiation treatment was studied. In addition, the significance of cell cycle regulators cyclin D1, p16, p21 as potential markers of biological aggressiveness of tumors was studied. The results of this study showed that the patients carrying in their tumor TP53 mutations in the DNA binding region of the gene had been exposed to chemicals more than patients with no mutation or other types of mutations. These mutations also correlated with biological aggressiveness and prognosis and the response to radiation treatment. It was also shown that a combination of cyclin D1 and p16 analyzed by immunohistochemistry correlated with worse prognosis in head and neck cancer. / Tiivistelmä Pään ja kaulan alueen levyepiteelisyöpää sairastavien potilaiden ennuste ei ole juurikaan parantunut viime vuosikymmeninä. Syövän biologista aggressiivisuutta kuvaavia ennustetekijöitä on vähän, luotettavimpana niistä taudin levinneisyys. Tutkimustulokset TP53-kasvunrajoitegeenin merkityksestä prognostisena ja hoitovastetta kuvaavana, prediktiivisenä merkkiaineena pään ja kaulan alueen syövissä ovat ristiriitaisia. Tämä johtuu muun muassa siitä, että p53-immunohistokemiaa on yritetty käyttää TP53-mutaatioiden analysoimiseen. Immunohistokemiassa analysoidaan proteiinia, eikä tulos siksi yleensä korreloi TP53-mutaatioiden esiintymiseen. Prognostisen tai hoitovastetta ennustavan merkkiaineen tulee mitata haluttua asiaa luotettavasti, ja sen analysoimiseen tulee käyttää herkkää ja spesifistä menetelmää. Herkän mutaatioanalyysimenetelmän lisäksi mutaatioiden paikan ja laadun tarkempi analysoiminen on välttämätöntä, jotta saadaan selville mutaation merkitys p53 proteiinin toiminnalle. Tässä työssä tutkittiin pään ja kaulan alueen levyepiteelisyöpää sairastavien potilaiden kasvaimista TP53- mutaatioiden esiintymistä validoidulla, herkällä PCR-SSCP-menetelmällä. Mutaatioiden laatu ja sijainti geenissä analysoitiin sekvensoimalla. IARC:n TP53-mutaatiotietopankin ja kirjallisuuden tietojen avulla selvitettiin mutaatioiden yleisyys ja niiden vaikutus p53-proteiinin toimintaan. Tutkimuksessa selvitettiin TP53-mutaatioiden korrelaatiota kemialliseen altistumiseen ja niiden merkitystä potilaan ennusteeseen ja sädehoitovasteeseen. Lisäksi tutkittiin solusyklin säätelijöiden sykliini D1-, p16- ja p21-proteiinien merkitystä taudin biologiseen aggressiivisuuteen. Väitöskirjatutkimuksen tulokset osoittivat, että potilaat, joiden kasvaimessa oli TP53-mutaatio DNA-sitoutumisalueella, olivat altistuneet kemikaaleille enemmän kuin potilaat, joiden kasvaimissa oli toisenlaisia mutaatioita tai ei mutaatiota ollenkaan. Tutkimuksessa havaitut mutaatiot liittyivät taudin biologiseen aggressiivisuuteen ja huonoon ennusteeseen sekä heikompaan sädehoitovasteeseen. Lisäksi havaittiin, että sykliini D1- ja p16-proteiinin yhdistelmä immunohistokemiallisesti analysoituna korreloi huonoon ennusteeseen pään ja kaulan syövissä.
53

Caractérisation moléculaire des leucémies aigües myéloïdes avec dysmyélopoïèse / Molecular characterization of acute myeloid leukemia with myelodysplasia related changes

Devillier, Raynier 31 October 2014 (has links)
Les leucémies aiguës myéloïdes (LAM) avec dysplasie, identifiées par la classification OMS 2008 sous le nom de LAM-MRC (« AML with myelodysplasia-related changes »), sont actuellement définies par la présence de critères cliniques, cytologiques et cytogénétiques. Elles forment un groupe hétérogène tant sur le plan biologique que pronostique. Nous avons fait l'hypothèse que la caractérisation moléculaire des LAM-MRC pourrait permettre d'identifier des marqueurs spécifiques associés à ces pathologies et d'en distinguer différents sous-groupes. Nous avons mis en évidence que les LAM-MRC de risque cytogénétique intermédiaire présentent un profil mutationnel spécifique caractérisé par un taux élevé de mutation d'ASXL1 et une faible proportion de mutations de DNMT3A, NPM1 et FLT3. Les LAM-MRC de risque cytogénétique défavorable, essentiellement complexes et/ou monosomales, sont quant à elle associées aux mutations de TP53. Alors que les critères actuels des LAM-MRC ne permettent pas d'en stratifier le pronostic, nous avons montré que les mutations d'ASXL1 ou de TP53 sont des facteurs pronostics péjoratifs majeurs. Ainsi, une reclassification basée sur la présence de ces altérations moléculaires exclusives entre elles permettrait d'affiner le diagnostic et la stratification pronostique de ces maladies. Enfin, dans une stratégie de médecine personnalisée combinant le séquençage à haut débit à des tests de sensibilité thérapeutique in vitro, l'identification de tels marqueurs moléculaires permettraient de prédire la réponse aux traitements, de guider les choix thérapeutiques et d'orienter le développement de nouvelles drogues. / Acute myeloid leukemia (AML) with myelodysplasia-related changes (AML-MRC) as reported in the WHO 2008 classification are defined by the presence of clinical, morphological and cytogenetic criteria. AML-MRCs are heterogeneous diseases with prognostic heterogeneity. We hypothesized that molecular characterization of AML-MRC could identify specific molecular markers and disease subgroups. We showed that AML-MRCs with intermediate cytogenetic risk harbor a specific mutational profile characterized by a high frequency of ASXL1 mutations and a low incidence of DNMT3A, NPM1 and FLT3 mutations. Unfavorable cytogenetic risk AML-MRCs, especially due to complex and/or monosomal karyotypes, are associated with TP53 mutations. While WHO criteria do not stratify the prognosis of AML-MRC patients, we showed that the mutations of ASXL1 or TP53 are major poor prognostic factors. The criteria defining AML-MRC do not identify distinct clinical and biological subgroups and do not predict outcome of patients with AML-MRC. In contrast, ASXL1 and TP53-mutated AML identify two distinct biological subgroups of AML-MRC with very poor outcome. This molecular characterization could be useful to redefine AML-MRC in a future classification aiming at merging biological characterization and specific prognostic value. Finally, we showed that a personalized treatment approach combining next generation sequencing and in vitro drug screening could be useful to predict therapeutic response and to guide both treatment choices and new targeted drug developments.
54

Etude des mécanismes de résistances primaire et acquise aux inhibiteurs du récepteur de l’Epidermal Growth Factor dans le cancer bronchique non à petites cellules / Mechanisms of primary and acquired resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer

Cortot, Alexis 21 December 2010 (has links)
Les inhibiteurs de tyrosine kinase (ITK) du récepteur à l’Epidermal Growth Factor (EGFR) constituent un nouveau traitement du cancer bronchique non à petites cellules (CBNPC), particulièrement efficace chez les patients porteurs d’une mutation d’EGFR (EGFR M+). Néanmoins, certains de ces patients peuvent avoir une résistance primaire à ces traitements, et les autres développent inéluctablement une rechute correspondant au phénomène de résistance acquise. L’objectif de ce travail était d’étudier les mécanismes de résistances primaire et acquise chez les patients avec CBNPC EGFR M+.Nous avons montré que le statut mutationnel de KRAS était le même dans la tumeur primitive et les métastases dans la majorité des cas de CBNPC. Dans quelques cas cependant, le statut différait entre tumeur primitive et métastase, ce qui soulève la question d’une dissociation de la réponse aux ITK, les mutations de KRAS étant associées à une résistance primaire au traitement.Nous avons par ailleurs mis en évidence la fréquente inactivation de la voie p53, soit par mutation de TP53 soit par diminution d’expression de p14arf, dans les tumeurs EGFR M+, qui pourrait être une des causes des variations de réponse aux ITK chez les patients EGFR M+. Enfin, nous avons montré que la résistance à deux ITK de nouvelle génération, PF299804 et WZ4002, passait par un phénomène en deux étapes impliquant d’une part l’activation de la voie IGF1R, via la diminution de l’expression d’IGFBP3, et d’autre part celle de la voie des MAP kinases. Ces travaux pourraient déboucher sur des stratégies thérapeutiques innovantes chez les patients présentant une résistance acquise aux ITK de nouvelle génération. / Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors (TKI) provide clinical efficacy in Non Small Cell Lung Cancer (NSCLC) patients, especially in the presence of an EGFR mutation. However, some EGFR mutated patients display primary resistance to EGFR TKI, and the others will ultimately develop acquired resistance. We focused our work on mechanisms of primary and acquired resistance in EGFR mutated NSCLC.We first showed that KRAS mutational status is the same in primary NSCLC and matched metastases in most of the cases. However, in some patients, we found a discordant KRAS status between primary tumors and metastases, which could translate into a discordant response to EGFR TKI, since KRAS mutations are associated with resistance to EGFR TKI.We also showed that EGFR mutated tumors are associated in most of the cases with an inactivation of the p53 pathway, either through a TP53 mutation or through loss of expression of p14arf, which could account for some of the variations observed in the response to TKI in EGFR mutated patients.Last, we showed that acquired resistance to two new generation EGFR TKI, PF299804 and WZ4002, occurred through a multistep process involving activation of the IGF1R pathway through downregulation of IGFBP3 and activation of the MAP kinase pathway. These results provide new insights into the treatment of NSCLC in EGFR mutated patients with acquired resistance to new generation TKI.
55

Via Wnt/?-catenina em tumores adrenocorticais pediátricos / Wnt/?-catenin signaling pathway in childhood adrenocortical tumors

Letícia Ferro Leal 21 July 2011 (has links)
Introdução: Em crianças das regiões Sul e Sudeste do Brasil há uma incidência elevada de tumores adrenocorticais (TAC). Anormalidades da ?-catenina tem sido encontradas em TAC em adultos e sugerem a ativação da via Wnt/ -catenina nestes tumores. No entanto, não há estudos avaliando o papel desta via em casuísticas de TAC pediátricos. Objetivos: Avaliar o papel da via Wnt/catenina e mutações do gene CTNNB1 na tumorigênese adrenocortical pediátrica. Indivíduos, Material e Métodos: Foram avaliados 62 pacientes pediátricos com TAC oriundos de dois centros de referência. Controles: córtex adrenal de indivíduos jovens com morte acidental. Avaliou-se a presença de mutação nos genes TP53 e CTNNB1. A expressão de genes da via Wnt (CTNNB1, o ligante WNT4, os inibidores SFRP1, DKK3 e AXIN1, o fator de transcrição TCF7 e os genes-alvo MYC e WISP2) foi avaliada por qPCR, utilizando-se o método de 2-Ct. Adicionalmente, a expressão de proteínas da via Wnt/-catenina e P53 foi avaliada por imunoistoquímica. Avaliou-se a relação entre possíveis anormalidades moleculares com o fenótipo clínico e o desfecho. Resultados: A sobrevida geral foi maior em pacientes menores que 5 anos de idade (p<0.0001) e em pacientes com estágios tumorais menos avançados (p<0.0001). A mutação P53 p.R337H foi encontrada em 87% dos pacientes e não se associou com características clinicopatológicas ou desfecho. Mutações do gene CTNNB1 foram encontradas em 4/62 (6%) TAC, todos carreadores da mutação P53 p.R337H. Houve associação entre óbito e presença de mutações do gene CTNNB1 (p=0,02). Acúmulo difuso da -catenina foi observado em 71% dos TAC, a maioria sem mutações do CTNNB1. Comparados a adrenais normais, os TAC apresentaram aumento da expressão do RNAm de CTNNB1 (p=0.008) e diminuição da expressão de genes inibidores da via Wnt: DKK3 (p<0.0001), SFRP1 (p=0.05) e AXIN1 (p=0.04). Com relação aos genes-alvo da via Wnt/-catenina, TAC apresentaram expressão aumentada de WISP2 e baixa expressão de MYC. Maior sobrevida geral foi associada à expressão baixa de SFRP1 (p=0.01), WNT4 (p=0.004) e TCF7 (p<0.01). Conclusões: Em TAC pediátricos, mutações somáticas ativadoras do gene CTNNB1 são pouco freqüentes e parecem estar associadas à maior ocorrência de óbito. Mesmo na ausência de mutações do gene CTNNB1, estes tumores apresentaram acúmulo de -catenina e do gene-alvo WISP2 e expressão reduzida de inibidores da via Wnt (DKK3, SFRP1 e AXIN1). Estes dados demonstram evidências de anormalidades na via Wnt/-catenina em TAC pediátricos, mesmo na ausência de mutações do gene CTNNB1. É provável que outros eventos genéticos afetando a via Wnt/-catenina estejam envolvidos na tumorigênese adrenocortical pediátrica / Context: CTNNB1 mutations and activation of Wnt/-catenin pathway are frequent in adult adrenocortical tumors (ACTs) but data on childhood ACTs are lacking. Objective: To investigate Wnt/-catenin pathway abnormalities and CTNNB1 mutations in childhood ACTs. Patients and Methods: Clinicopathological findings and outcome of 62 childhood ACTs patients were analyzed regarding to CTNNB1/ -catenin mutations and to the expression of Wnt-related genes (CTNNB1, a Wnt ligand: WNT4, Wnt inhibitors: SFRP1, DKK3 and AXIN1, a transcription factor: TCF7, and target genes: MYC and WISP2) by qPCR and immunohistochemistry. Results: Overall survival (OS) was higher in patients younger than 5 years (p<0.0001) and associated with less advanced tumoral stage (p<0.0001). The p.R337H P53 mutation, found in 87% of the patients, was not associated with clinicopathological findings or outcome. CTNNB1 activating mutations were found in only 4/62 ACTs (6%), all of them harboring TP53 mutation. There was association between the presence of CTNNB1 mutation and death (p=0.02). Diffuse -catenin accumulation was found in 71% of ACTs, most of them without CTNNB1 mutation. CTNNB1 mutated ACTs presented weak/moderate -catenin accumulation. Compared to normal adrenals, ACTs presented increased expression of CTNNB1 (p=0.008) and underexpression of Wnt inhibitor genes: DKK3 (p<0.0001), SFRP1 (p=0.05) and AXIN1 (p=0.04). With regards to Wnt/-catenin target genes, ACTs presented lower expression of MYC but increased expression of WISP2. Higher overall survival was associated with underexpression of SFRP1 (p=0.01), WNT4 (p=0.004) and TCF7 (p<0.01). Conclusions: In childhood ACTs, CTNNB1 mutations are rare and appear to be associated with poor prognosis. Regardless of CTNNB1 mutations, these tumors presented reduced expression of Wnt inhibitor genes (DKK3, SFRP1 and AXIN1) and increased expression of CTNNB1 and a target gene, WISP2. Thus, besides CTNNB1 mutations, additional genetic events affecting the Wnt/-catenin pathway may be involved in childhood adrenocortical tumorigenesis.
56

Via Wnt/?-catenina em tumores adrenocorticais pediátricos / Wnt/?-catenin signaling pathway in childhood adrenocortical tumors

Leal, Letícia Ferro 21 July 2011 (has links)
Introdução: Em crianças das regiões Sul e Sudeste do Brasil há uma incidência elevada de tumores adrenocorticais (TAC). Anormalidades da ?-catenina tem sido encontradas em TAC em adultos e sugerem a ativação da via Wnt/ -catenina nestes tumores. No entanto, não há estudos avaliando o papel desta via em casuísticas de TAC pediátricos. Objetivos: Avaliar o papel da via Wnt/catenina e mutações do gene CTNNB1 na tumorigênese adrenocortical pediátrica. Indivíduos, Material e Métodos: Foram avaliados 62 pacientes pediátricos com TAC oriundos de dois centros de referência. Controles: córtex adrenal de indivíduos jovens com morte acidental. Avaliou-se a presença de mutação nos genes TP53 e CTNNB1. A expressão de genes da via Wnt (CTNNB1, o ligante WNT4, os inibidores SFRP1, DKK3 e AXIN1, o fator de transcrição TCF7 e os genes-alvo MYC e WISP2) foi avaliada por qPCR, utilizando-se o método de 2-Ct. Adicionalmente, a expressão de proteínas da via Wnt/-catenina e P53 foi avaliada por imunoistoquímica. Avaliou-se a relação entre possíveis anormalidades moleculares com o fenótipo clínico e o desfecho. Resultados: A sobrevida geral foi maior em pacientes menores que 5 anos de idade (p<0.0001) e em pacientes com estágios tumorais menos avançados (p<0.0001). A mutação P53 p.R337H foi encontrada em 87% dos pacientes e não se associou com características clinicopatológicas ou desfecho. Mutações do gene CTNNB1 foram encontradas em 4/62 (6%) TAC, todos carreadores da mutação P53 p.R337H. Houve associação entre óbito e presença de mutações do gene CTNNB1 (p=0,02). Acúmulo difuso da -catenina foi observado em 71% dos TAC, a maioria sem mutações do CTNNB1. Comparados a adrenais normais, os TAC apresentaram aumento da expressão do RNAm de CTNNB1 (p=0.008) e diminuição da expressão de genes inibidores da via Wnt: DKK3 (p<0.0001), SFRP1 (p=0.05) e AXIN1 (p=0.04). Com relação aos genes-alvo da via Wnt/-catenina, TAC apresentaram expressão aumentada de WISP2 e baixa expressão de MYC. Maior sobrevida geral foi associada à expressão baixa de SFRP1 (p=0.01), WNT4 (p=0.004) e TCF7 (p<0.01). Conclusões: Em TAC pediátricos, mutações somáticas ativadoras do gene CTNNB1 são pouco freqüentes e parecem estar associadas à maior ocorrência de óbito. Mesmo na ausência de mutações do gene CTNNB1, estes tumores apresentaram acúmulo de -catenina e do gene-alvo WISP2 e expressão reduzida de inibidores da via Wnt (DKK3, SFRP1 e AXIN1). Estes dados demonstram evidências de anormalidades na via Wnt/-catenina em TAC pediátricos, mesmo na ausência de mutações do gene CTNNB1. É provável que outros eventos genéticos afetando a via Wnt/-catenina estejam envolvidos na tumorigênese adrenocortical pediátrica / Context: CTNNB1 mutations and activation of Wnt/-catenin pathway are frequent in adult adrenocortical tumors (ACTs) but data on childhood ACTs are lacking. Objective: To investigate Wnt/-catenin pathway abnormalities and CTNNB1 mutations in childhood ACTs. Patients and Methods: Clinicopathological findings and outcome of 62 childhood ACTs patients were analyzed regarding to CTNNB1/ -catenin mutations and to the expression of Wnt-related genes (CTNNB1, a Wnt ligand: WNT4, Wnt inhibitors: SFRP1, DKK3 and AXIN1, a transcription factor: TCF7, and target genes: MYC and WISP2) by qPCR and immunohistochemistry. Results: Overall survival (OS) was higher in patients younger than 5 years (p<0.0001) and associated with less advanced tumoral stage (p<0.0001). The p.R337H P53 mutation, found in 87% of the patients, was not associated with clinicopathological findings or outcome. CTNNB1 activating mutations were found in only 4/62 ACTs (6%), all of them harboring TP53 mutation. There was association between the presence of CTNNB1 mutation and death (p=0.02). Diffuse -catenin accumulation was found in 71% of ACTs, most of them without CTNNB1 mutation. CTNNB1 mutated ACTs presented weak/moderate -catenin accumulation. Compared to normal adrenals, ACTs presented increased expression of CTNNB1 (p=0.008) and underexpression of Wnt inhibitor genes: DKK3 (p<0.0001), SFRP1 (p=0.05) and AXIN1 (p=0.04). With regards to Wnt/-catenin target genes, ACTs presented lower expression of MYC but increased expression of WISP2. Higher overall survival was associated with underexpression of SFRP1 (p=0.01), WNT4 (p=0.004) and TCF7 (p<0.01). Conclusions: In childhood ACTs, CTNNB1 mutations are rare and appear to be associated with poor prognosis. Regardless of CTNNB1 mutations, these tumors presented reduced expression of Wnt inhibitor genes (DKK3, SFRP1 and AXIN1) and increased expression of CTNNB1 and a target gene, WISP2. Thus, besides CTNNB1 mutations, additional genetic events affecting the Wnt/-catenin pathway may be involved in childhood adrenocortical tumorigenesis.
57

Identification de biomarqueurs de réponse à l'azacitidine dans les leucémies aigues myéloïdes du sujet âgé / Identification of biomarkers of response to azacitidine in older patients with acute myeloid leukemia

Bories, Pierre 26 October 2018 (has links)
Les leucémies aiguës myéloïdes (LAM) du sujet âgé sont les plus fréquentes des leucémies aiguës. Bien que de physiopathologie hétérogène, elles partagent un pronostic défavorable. L’azacitidine est devenue un des traitements de référence pour les patients jugés inéligibles pour une chimiothérapie intensive mais les critères de sélection des patients entre ces deux approches sont controversés. L’identification de biomarqueur prédictif de réponse à l’azacitidine doit permettre de rationnaliser ce choix thérapeutique. Les facteurs pronostiques classiques d’une cohorte de 334 patients atteints de LAM manquent de précision pour guider la meilleure stratégie pour un patient donné. A partir du séquençage de 224 patients traités par azacitidine, nous montrons un impact défavorable des mutations de TP53 sur la survie globale, quel que soit leur caractérisation fonctionnelle. Le séquençage des exomes de 49 patients selon leur réponse à l’azacitidine (26 répondeurs et 23 non répondeurs), suivi du re-séquençage ciblé de 4 polymorphismes chez 175 patients a montré un impact positif du polymorphisme rs7622799 de MECOM sur la survie globale sous azacitidine. / Elderly patients with acute myeloid leukemias (AML) represent the most frequent acute leukemias. Although they differ in their pathophysiology, they all share an adverse prognosis. Azacitidine has become one of the reference low-intensity frontline therapy for patients deemed unfit for intensive chemotherapy. Patients selection between these 2 options remains controversial. Predictive biomarkers of response to azacitidine should allowed to rationalize this decision making. Classical prognosis factors of a cohort of 334 newly diagnosed AML lack of precision to determine the optimum strategy for any individual patient. By sequencing of a 224-patients series of azacitidine-treated AML patients, we demonstrate an adverse impact of TP53 mutation on overall survival, irrespective of the functional characterization of p53 mutants. Exome sequencing of 49 patients with extreme phenotype as defined by their response under azacitidine (26 responders versus 23 non-responders), followed by targeted sequencing of 4 common polymorphisms in a validation set of 175 patients, showed a positif impact of MECOM rs7622799 on overall survival.
58

Efeito do tabagismo no perfil de metilação de DNA no promotor de genes MHL1, hTERT e TP53 em células epiteliais da mucosa bucal

Oliveira, Sabrina Rocha Luna de 27 February 2014 (has links)
Made available in DSpace on 2015-05-14T12:56:02Z (GMT). No. of bitstreams: 1 arquivototal.pdf: 1722534 bytes, checksum: d8557488d35ade21ba45951fd8412a2f (MD5) Previous issue date: 2014-02-27 / Coordenação de Aperfeiçoamento de Pessoal de Nível Superior / DNA methylation, characterized by the addition of a methyl group in cytosines within CpG dinucelotides can modified gene transcription, leading to decrease or even silence a gene. The ability of the environmental factors to induce epigenetic changes has been investigated and many studies have shown a relationship between them. Studies show that pesticides, metal ions, drugs, diet, alcohol dependence and smoking are associated with epigenetic changes. Smoking is often associated with the risk of cancer in various tissues and cardiovascular diseases, being considered the leading cause of preventable death. The MLH1 gene is related to the repair of badly paired bases of DNA (DNA mismatch repair (MMR)). The hTERT gene comprises the catalytic subunit of telomerase enzyme, which is considered a biological clock, a marker indicating that the cellular senescence can be installed inevitably form. The TP53 is a tumor suppressor gene and its hypermethylation is related to the development of various cancers. The aim of this work was to investigate the smoking habit influence on DNA methylation status in the promoter of cancer-related genes, MLH1, hTERT and TP53 in oral epithelial cells of healthy subjects. Samples of oral epithelium of smokers, nonsmokers and former smokers were collected by rinsing and DNA was extracted. After, DNA Methylation analysis was performed by Methylation Sensitive Restriction Enzymes, using two restriction enzymes, the HpaII and HhaI, which cleave different sites. Following the enzymatic digestion, DNA was amplified by PCR, subjected to electrophoresis on a 6% polyacrylamide gel and stained with silver nitrate. Statistical analysis was performed by Chi-square test at a significance level of 5%. The investigated CpG dinucleotides located at HhaI and HpaII sites in the MLH1 gene promoter were observed to be fully methylated in DNA majority samples from the smoker group and statistical differences were found between nonsmokers and smokers and between smokers and former smokers (p<0.05). The same was observed in the hTERT gene promoter at HhaI site (p<0.05) and for HpaII site the unmethylated condition was more frequent in smoker in comparison to nonsmokers (p<0.05). For TP53 no differences were found among groups (p>0.05) which the fully methylated condition was found to be an usual event in healthy oral epithelial cells. We conclude that smoking may induce changes in DNA methylation status in cancer-related genes, such as MLH1 and hTERT of healthy oral epithelial cells and the cessation of smoking reversed the process. / A metilação de DNA é uma modificação química na molécula de DNA, e consiste na presença de um radical metil em dinucleotídeos CpG, presente principalmente em regiões promotoras do gene. Uma das principais funções da metilação de DNA é regular a transcrição gênica, sendo que a presença do radical metil pode suprimir por completo a expressão gênica. Estudos mostram que o meio ambiente pode modular a metilação de DNA. Como exemplo de fatores ambientais temos: a radiação ultravioleta, agrotóxicos, dieta, fármacos, uso crônico do álcool e o hábito de fumar. O fumo é frequentemente associado ao risco de câncer em diversos tecidos e doenças cardiovasculares, sendo considerado a maior causa de morte evitável. O gene MLH1 está relacionado ao reparo de bases mal pareadas do DNA (DNA mismatch repair (MMR)). O gene hTERT compõe a subunidade catalítica da enzima telomerase, a qual é considerada um relógio biológico, um marcador que indica que a senescência celular poderá se instalar de forma inevitável. O TP53 é um gene supressor tumoral e sua hipermetilação está relacionada ao desenvolvimento de diversos tipos de câncer. Assim, o objetivo deste estudo foi investigar o efeito do tabagismo no perfil de metilação de DNA em genes relacionados ao câncer, MLH1, hTERT e TP53 em células da mucosa bucal de indivíduos saudáveis. Para tanto, amostras de epitélio da mucosa bucal de indivíduos fumantes, não fumantes e ex-fumantes foram coletadas por bochecho e o DNA dessas células foi extraído. Após esse processo, a análise de metilação de DNA foi feita utilizando o método de Digestão Enzimática Sensível à Metilação, utilizando-se de duas enzimas de restrição, a HhaI e a HpaII, as quais clivam sítios diferentes. Em seguida à digestão enzimática, DNA foi amplificado por PCR, submetido à eletroforese em gel de poliacrilamida a 6% e corado pelo nitrato de prata. A análise estatística foi realizada pelo Teste de Qui-Quadrado ao nível de significância de 5%. Os dinucleotídeos CpG localizados nos sítios HhaI e HpaII no promotor do gene MLH1 mostraram-se totalmente metilados na maioria dos indivíduos do grupo fumante e diferenças significativas foram observadas entre fumantes e não fumantes e entre fumantes e ex-fumantes (p<0,05). O mesmo foi observado para o sítio HhaI no promotor do gene hTERT (p<0,05) e para o sítio HpaII a condição não metilada foi mais frequente em fumantes em comparação com não fumantes (p<0,05). Para o gene TP53 não foram encontradas diferenças entre os grupos (p>0,05), sendo a condição totalmente metilada um evento usual das células saudáveis da mucosa bucal. Assim, concluímos que o fumo está associado a alterações no perfil de metilação de DNA em genes relacionados ao câncer, como MLH1 e hTERT em células epiteliais saudáveis da mucosa bucal e a cessação do hábito de fumar reverteu o processo.
59

Prevalência da mutação germinativa TP53 p.R337H em indivíduos com tumores do espectro da Síndrome de Li-Fraumeni

Giacomazzi, Juliana January 2012 (has links)
Introdução: Estudos prévios têm indicado que uma mutação germinativa específica no gene TP53, p.R337H está associada a efeito fundador no Brasil e é muito frequente entre certos tipos tumorais. Objetivos e Metodologia: O objetivo deste estudo foi verificar a prevalência da mutação germinativa TP53 p.R337H em indivíduos com tumores do espectro da Síndrome de Li-Fraumeni (SLF) e/ou Li- Fraumeni-Like (LFL) divididos em diferentes grupos: (1) mulheres com câncer de mama (CM) subdivididas em 2 subgrupos: (a) com critérios para síndromes de predisposição hereditária ao câncer, exceto para SLF/LFL; (b) não selecionadas para história familiar (HF), diagnosticadas antes dos 45 e após 55 anos de idade em diferentes regiões do país; (2) mulheres com tumores phyllodes da mama, diagnosticadas em diferentes idades e não selecionadas para HF da doença nas regiões Sul e Sudeste; (3) crianças com tumores do espectro da SLF/LFL, não selecionadas para HF e atendidas em um Serviço de Oncologia Pediátrica do Sul do país. Adicionalmente, foi investigada a presença do haplótipo fundador brasileiro nos portadores da mutação TP53 p.R337H. No grupo 3 foi ainda avaliada a prevalência de critérios para SLF/LFL e a presença de outras mutações germinativas no gene TP53. Resultados: No subgrupo 1A foram incluídas 59 mulheres das quais 2 eram portadoras da mutação TP53 p.R337H, ambas com HF exclusivamente para a Síndrome de Prediposição Hereditária ao Câncer de Mama e Ovário no momento do recrutamento. No subrupo 1B foram incluídas 815 mulheres (403 com CM N45 anos e 412 casos com CM O 55 anos) provenientes de 25 diferentes Estados do Brasil. A mutação foi encontrada em 8,6% (70/815) dos casos, sendo observada diferença significativa na frequência de acordo com o centro de recrutamento. Em todos centros, a mutação foi mais frequente em mulheres com CM antes dos 45 anos. Análise do tecido tumoral de 15 portadoras evidenciou perda de heterozigosidade (LOH) em apenas 2 casos. Os tumores de mama de mulheres portadoras apresentavam mais frequentemente alguma expressão de HER2 (1+, 2+ ou 3+). No grupo 2, foram incluídas 148 mulheres com tumores phyllodes da mama e 8 (5,4%) apresentaram a mutação, sendo mais frequente entre os tumores malignos (3/13; 23%) quando comparada aos benignos (5/128; 3.4%). No grupo 3 foram incluídas 292 crianças diagnosticadas com tumores do espectro da SLF/LFL, das quais 25,3% tinham critérios para SLF/LFL. O sequenciamento completo e pesquisa de rearranjos gênicos de TP53 em 48 das crianças com câncer e critérios estritos para LFL evidenciou apenas um caso com uma mutação clássica – p.G245S. A mutação TP53 p.R337H foi identificada somente entre os casos com carcinoma adrenocortical (9/11; 81,8%) e de plexo coróide (2/2; 100%), correspondendo a 3,8% da amostra total. Um dos casos de carcinoma adrenocortical apresentou a mutação TP53 p.R337H em homozigose. Todos tumores de portadores analisados (n=8) apresentaram LOH. Conclusão: Conclui-se que a mutação germinativa TP53 p.R337H ocorre em mulheres brasileiras com CM em diferentes faixas etárias, independente da HF de câncer. Em mulheres com fenótipo de CM hereditário sem critérios para SLF/LFL a mesma também foi observada. A freqüência da mutação foi variável de acordo com o centro de recrutamento, sendo identificada em todas as regiões brasileiras. Os tumores de mama de pacientes portadoras da mutação têm um perfil peculiar, com baixa frequência de LOH e presença de alguma expressão de HER2. A mutação p.R337H ocorre em tumores phyllodes da mama benignos e especialmente malignos, confirmando a hipótese prévia de associação de mutações germinativas em TP53 com estes tumores. Entre crianças com tumores do espectro SLF/LFL de um centro de referência no Sul do Brasil, um percentual significativo tem indicação para teste de mutações em TP53, no entanto, na maioria destes casos não se identifica mutação germinativa no gene. A mutação TP53 p.R337H ocorre em elevada frequência na linhagem germinativa de crianças com carcinomas adrenocortical e de plexo coróide, como descrito em outras regiões brasileiras. Por fim, o haplótipo fundador foi identificado em todos portadores da mutação analisados, incluindo 3 casos das regiões Norte, Nordeste e Centro-Oeste do país. A mutação TP53 p.R337H está associada a efeito fundador no Brasil e tem importante contribuição na gênese de tumores de mama, bem como de carcinomas adrenocortical e de plexo coróide no Sul do país. / Introduction: Previous studies have reported that a specific germline mutation in TP53, p.R337H, is associated with a founder effect and is common among certain tumor types. Objectives and Methodology: Assess the prevalence of TP53 p.R337H germline mutation in subjects with tumors of the Li-Fraumeni/ Li-Fraumeni- Like (LFS/LFL) spectrum divided into different groups: (1) women diagnosed with breast cancer (BC) subdivided into 2 subgroups: (a) with criteria for hereditary predisposition syndromes, except for LFS/LFL (b) unselected for family history (FH) diagnosed at or before 45 years and at or after 55 years recruited in different regions of the country; (2) women with phyllodes breast tumors, diagnosed at different ages and not selected for FH of the disease from the South and Southeast of Brazil; (3) children with tumors of the LFS/LFL spectrum and not selected for FH of the disease from a Pediatric Oncology Service in South Brazil. In addition, presence of the founder Brazilian haplotype in mutation carriers identified in groups 1, 2 and 3 was assessed. Finally, in group 3, prevalence of LFS/LFL criteria and presence of other germline TP53 mutations were investigated. Results: In subgroup 1A, 59 women were included and two of these were mutation carriers, both had a FH consistent only with Hereditary Breast and Ovarian Cancer Syndrome at recruitment. In subgroup 1B, 815 women (403 cases diagnosed N 45 years and 412 cases, O 55 years) were included from 25 different Brazilian States. The TP53 p.R377H mutation was found in 8.6% (70/815), and significant differences in mutation frequency were observed according to recruitment center. In all centers the mutation was more frequent in women diagnosed with BC at or before 45 years. Analysis of tumor tissue of 15 carriers showed loss of heterozygosity (LOH) in only 2 cases. Breast tumors of carriers showed, more frequently, some expression of HER2. In group 2 were included 148 women with breast phyllodes tumors and 8 (5.4%) were p.R337H carriers, which was most common among malignant phyllodes tumors (3/13, 23%) when compared to benign (5/128, 3.4%). In the group 3, 292 children with tumors of the LFS/LFL spectrum were included, and 25.3% of these had criteria for LFS/LFL. TP53 gene sequencing and rearrangement testing in 48 children with the more strict criteria for the syndrome identified a classic germline mutation, p.G245S, in only one proband. The TP53 p.R337H mutation was identified only among patients with adrenocortical carcinoma (9/11, 81.8%) and choroid plexus carcinoma (2/2, 100%), corresponding to 3.8% of the total sample. One of the patients with adrenocortical carcinoma was homozygous mutant. All tumors of TP53 p.R37H carriers analyzed (n=8) showed LOH. Conclusion: We conclude that the TP53 p.R337H germline mutation occurs in Brazilian women with BC in different age groups, regardless of BC FH. In women with hereditary BC without criteria for LFS/LFL criteria, it was also observed. The mutation frequency varied according to the recruiting center and carriers were identified in all Brazilian regions. Breast tumors of carriers have a peculiar profile, with low frequency of LOH and the presence of HER2 expression. The mutation was present, also, in benign and especially malignant phyllodes tumors, confirming the previous hypothesis of an association between germline mutations in the TP53 gene and these specific tumors. Among children with tumors of the LFS/LFL spectrum from a Oncopediatrics referral center in South Brazil, a significant percentage fulfills criteria for TP53 mutation testing, however, in most of these cases, no germline mutations were identified. The TP53 p.R337H mutation occurs in high frequency in the germline of children with adrenocortical and choroid plexus carcinomas, as described in other Brazilian regions. Finally the founder haplotype was identified in all mutation carriers analyzed, including 3 cases from the Northern, Northeastern and Midwestern Brazilian regions. The TP53 p.R337H mutation is associated with a founder effect in Brazil and has an important contribution in the genesis of breast tumors in the country, as well as adrenocortical and choroid plexus carcinomas in the Southern region of the country.
60

Etude moléculaire de l'évolution clonalede TP53 des Syndromes Myélodysplasiques avec del(5q) : conséquences sur la résistance au traitement et la progression du cancer / Molecular Analysis of clonal evolutions in hematological malignancies, including mutations of TP53 : consequences on therapeutic resistance and cancer progression

Lode, Laurence 29 November 2017 (has links)
La protéine p53 (« Gardien du génome ») doit être altérée pour que le cancer puisse se développer. Les nombreuses thérapies anti-cancéreuses disponibles sont très efficaces mais la réponse clinique est souvent transitoire et les cancers disséminés rechutent ou progressent du fait de l'évolution de sous-populations cancéreuses résistantes au traitement, impliquant souvent TP53 qui est le gène le plus muté dans les formes agressives de nombreux cancers. Nous l’avons étudié dans la leucémie lymphoïde chronique (LLC) et les syndromes myélodysplasiques avec délétion 5q (SMD del(5q)). Grâce à l’étude rétrospective longitudinale de 40 patients atteints de SMD del(5q), nous avons généré des données de NGS ciblé et montré que le statut mutationnel de TP53 au diagnostic ne permettait pas de prédire la progression tumorale, contrairement à ce qui avait été publié précédemment (Jädersten et al., JCO 2011). Nous avons montré que c’était l’évolution clonale du gène TP53 qui était l’élément clé de la progression des SMD del(5q). Nous avons observé de nombreuses émergences de clones mutés entre le stade diagnostique et un stade ultérieur de la maladie, toujours après initiation du traitement par lénalidomide.Le lénalidomide a été approuvé comme nouveau traitement spécifique et très efficace contre l’anémie liée aux SMD del(5q), permettant à la plupart des patients d’être indépendants des transfusions sanguines. Le lénalidomide permet souvent d’éradiquer le clone tumoral porteur de l’anomalie génétique del(5q) isolée, induisant une rémission clinique. Malheureusement, cette rémission est courte avec une durée médiane de 2 ans, puis, dans environ 1 cas sur 2, survient une transformation en leucémie aiguë secondaire de pronostic péjoratif.Nous avons étudié un possible lien entre le traitement par lénalidomide et l’évolution clonale de TP53 par annotation clinico-bio-thérapeutiques des résultats de séquençage de TP53 chez les 24 patients dont les échantillons séquentiels avaient été analysés. Dans notre étude, les patients avec progression tumorale (dont 10 évolutions clonales de TP53 et 1 évolution clonale de RUNX1) avaient reçu une dose cumulée de lénalidomide supérieure à celle reçue par les patients dont la tumeur était restée stable (p = 0.036). Nous avons observé chez plusieurs patients que l’éradication de la tumeur n’était pas utile à l’amélioration de la qualité de vie des patients. La non-éradication semblait même permettre un maintien de l’équilibre clonal et une compétition entre les différents sous-clones de la tumeur, résistants ou non au lénalidomide.Nous discutons de l’évolution de l'écologie de la tumeur au cours du traitement, i.e., l’évolution de ses interactions avec son micro-environnement qui se modifie après chaque nouvelle dose de traitement. Un modèle évolutif dit théorie de la thérapie adaptative, développée récemment remet en question les protocoles conventionnels de thérapie anti-cancéreuse qui préconisent souvent d'administrer la dose maximale tolérée par le patient (Gatenby, 2009). Elle suggère que la dose minimale efficace présenterait l’avantage de ne pas éradiquer les cellules cancéreuses sensibles au traitement pour qu'elles restent en compétition avec les cellules cancéreuses résistantes et limiter la progression ou la rechute. Nous suggérons de prendre en compte également la diminution des effets indésirables pour le patient, améliorant ainsi sa qualité de vie, et enfin la diminution des dépenses de santé pour la collectivité. A ce jour, peu d’études cliniques évaluent l’intérêt de l’adoption de tels protocoles de thérapie adaptative.Néanmoins, des modèles in vivo (xénogreffes) et in silico (modèles statistiques) ont permis d’analyser la dynamique évolutive des populations tumorales en fonction du traitement reçu. Ces modèles prédisent que la survie de l’hôte peut être maximisée par la mise en place d’une thérapie adaptative. / P53 protein is named «guardian of the genome » because it must be altered to let cancer grow.TP53 is the most mutated gene in agressive cancers.Numerous systemic therapies are successful for treatment of disseminated cancers. However, clinical response is often transient, and cancer undergo relapse or progression due to emergence of resistant populations. These latter often harbour TP53 mutations. We studied TP53 in chronic lymphoid leukemia (CLL) and lower-risk myelodysplastic syndroms with del(5q), MDS del(5q). We conducted a retrospective longitudinal study in 40 patients suffering from MDS del(5q). We obtained targeted NGS data showing that TP53 mutational status at diagnosis could not predict tumor progression, by contrast with previously published data (Jädersten et al., JCO 2011). We show that TP53 clonal evolution is the key feature of tumor progression in MDS del(5q). We observed numerous mutated sub-clones emerging between diagnosis and follow-up. In our study, this emergence always followed onset of lenalidomide treatment. Lenalidomide was recently approved as a new therapy specifically improving anemia in patients with MDS del(5q). It allows most patients to become red-blood-cells-transfusion independent. Lenalidomide often eradicates the major tumor clone harbouring the isolated genetic abnormality deletion (5q) and allows clinical remission. Unfortunately, this remission is short (median, 2 years) and is followed, in 1 case out of 2, by a secondary acute leukemic transformation with a very poor prognosis.We studied the issue of a possible link between lenalidomide therapy and TP53 clonal evolution by annotating TP53 sequencing results with acute biological, clinical and therapeutic features in the 24 patients with sequential samples analyzed. In our study, patients with tumor progression (10 TP53 clonal evolution and 1 RUNX1 clonal evolution) were given a higher cumulative dose of lenalidomide compared to patients with stable disease (p = 0.036). Similarly to « adaptive therapy theory »(Gatenby 2009), we observed that eradication of the tumor wasn’t useful for improvement of quality of life. Absence of eradication might even allow to maintain a clonal equilibrium and a clonal competition between the distinct tumor sub-clones, resistant to lenalidomide or not, and therefore maintain stable disease.This theory of adaptive therapy questions the classical protocols of treatments against cancer, in which the maximal tolerated dose is preferred to the minimal effective dose. The latter might however slow down cancer progression or cancer relapse, with decreased side effects in patients, and decreased health costs.To date, few clinical trials (if any) questions such protocols of adaptive therapy. However, in vivo experiments (xenografts) and in silico statistical models allowed to study evolutionary dynamics of tumor sub-populations with and without therapy.The models predict that host survival can be maximized if “treatment-for-cure strategy” is replaced by “treatment-for-stability.” Specifically, the models predict that an optimal treatment strategy will modulate therapy to maintain a stable population of chemosensitive cells that can, in turn, suppress the growth of resistant populations under normal tumor conditions, Dr Gatenby said.

Page generated in 0.0831 seconds