EGFR-Inhibitor-basierte Erhaltungstherapie verlängert progressionsfreies Überleben

Gastro-News ◽  
2021 ◽  
Vol 8 (5) ◽  
pp. 62-62
Author(s):  
Claudia Schöllmann
2019 ◽  
Vol 10 (02) ◽  
pp. 78-78
Author(s):  
Alexander Kretzschmar

Männer mit metastasiertem Hormon-sensiblen Prostatakarzinom (mHSPC) erreichen ein signifikant höheres medianes progressionsfreies Überleben anhand radiologischer Befunde (rPFS) und haben eine höhere objektive Responserate, wenn sie zusätzlich zu einer Androgen-depravierenden Therapie (ADT) auch Enzalutamid erhalten (jeweils p < 0,0001). Dies zeigt die randomisierte, doppelblinde Phase-III-Studie ARCHES (NCT02677896), deren Ergebnisse auf dem Genitourinary Cancers Symposium (ASCO GU) vorgestellt wurden.


2011 ◽  
Vol 02 (06) ◽  
pp. 284-284
Author(s):  
Susanne Krome

Trotz einer aggressiven Chemotherapie ist die Prognose von Patienten mit fortgeschrittenen Melanomen ungünstig. Eine längere überlebenszeit und progressionsfreies überleben (PFS) hatten Patienten, die mit dem Antikörper Vemurafenib behandelt wurden. Auch Erkrankte mit besonderen Risikofaktoren profitierten.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi74-vi74
Author(s):  
Erin Smithberger ◽  
Abigail Shelton ◽  
Madison Butler ◽  
Alex Flores ◽  
Ryan Bash ◽  
...  

Abstract Glioblastoma (GBM) is an aggressive primary brain tumor with a poor survival rate. One of the most common molecular alterations seen in GBM is amplification and/or mutation of the Epidermal Growth Factor Receptor (EGFR), which has made it an attractive therapeutic target. However, several EGFR tyrosine kinase inhibitors have been tested clinically in GBM with minimal success. One reason for this lack of efficacy could be due to acute, adaptive resistance via alternative pathway activation. To investigate this mechanism of tumor resistance, we used RNA-seq and multiplex inhibitor bead/mass spectrometry (MIB-MS) to analyze the transcriptomes and kinomes of genetically engineered murine astrocytes with common GBM genotypes. We have previously shown that 38% of the expressed kinome varied among a panel of diverse nGEM astrocytes harboring Cdkn2a deletion (C) plus Pten deletion (CP), wild-type human EGFR (CE) or EGFRvIII (CEv3) overexpression or both EGFRvIII overexpression and Pten deletion (CEv3P). Although CE have a similar transcriptional profile to C cells at baseline, when treated with the EGFR inhibitor afatinib, CE respond more similarly to CEv3 cells. When cells containing endogenous murine EGFR (C and CP) are treated with afatinib, fewer than 0.5% of kinases showed differential expression. In cells with EGFR overexpression alone, more than 6% of kinases were differentially expressed upon afatinib treatment, including Ntrk3, Fgfr2 and 3, Lyn, Bmx, Epha2 and 5, Fn3k, a kinase involved in fructosamine processing, and Nrbp2, a kinase involved in regulation of apoptosis. This effect was blunted in cells lacking Pten in addition to having EGFRvIII (CEv3P), resulting in less than 2% of kinases being differentially expressed. The only kinase upregulated in all three EGFR-overexpressing cell types was Coq8a, which is involved in electron transport and response to DNA damage. Given this overlap in response, Coq8a could be a potential dual treatment target for GBM.


2021 ◽  
Vol 14 (2) ◽  
pp. 100961
Author(s):  
Fushun Fan ◽  
Minhua Zhou ◽  
Xiaolan Ye ◽  
Zhenxian Mo ◽  
Yaru Ma ◽  
...  

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Tao Liu ◽  
Xiujuan Han ◽  
Shutao Zheng ◽  
Qing Liu ◽  
Aerziguli Tuerxun ◽  
...  

Abstract Background Calmodulin1 (CALM1) has been identified as one of the overexpression genes in a variety of cancers and EGFR inhibitor have been widely used in clinical treatment but it is unknown whether CALM1 and epidermal growth factor receptor (EGFR) have a synergistic effect in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to explore the synergistic effects of knock-out CALM1 combined with EGFR inhibitor (Afatinib) and to elucidate the role of CALM1 in sensitizing the resistance to Afatinib in ESCC. Method Immunohistochemistry (IHC) and qRT-PCR were used to examine the expression of CALM1 and EGFR in ESCC tissues. Kaplan–Meier survival analysis was used to analyze the clinical and prognostic significance of CALM1 and EGFR expression in ESCC. Furthermore, to evaluate the biological function of CALM1 in ESCC, the latest gene editing technique CRISPR/Cas9(Clustered regularly interspaced short palindromic repeats)was applied to knockout CALM1 in ESCC cell lines KYSE150, Eca109 and TE-1. MTT, flow cytometry, Transwell migration, scratch wound-healing and colony formation assays were performed to assay the combined effect of knock-out CALM1 and EGFR inhibitor on ESCC cell proliferation and migration. In addition, nude mice xenograft model was used to observe the synergistic inhibition of knock-out CALM1 and Afatinib. Results Both CALM1 and EGFR were found to be significantly over-expressed in ESCC compared with paired normal control. Over-expressed CALM1 and EGFR were significantly associated with clinical stage, T classification and poor overall prognosis, respectively. In vitro, the combined effect of knock-out CALM1 mediated by the lentivirus and EGFR inhibitor was shown to be capable of inhibiting the proliferation, inducing cell cycle arrest at G1/S stage and increasing apoptosis of KYSE-150 and Eca109 cells; invasion and migration were also suppressed. In vivo, the results of tumor weight and total fluorescence were markedly reduced compared with the sgCtrl-infected group and sgCAML1 group. Conclusion Our data demonstrated that knock-out of CALM1 could sensitize ESCC cells to EGFR inhibitor, and it may exert oncogenic role via promotion of EMT. Taken together, CALM1 may be a tempting target to overcome Afatinib resistance.


Sign in / Sign up

Export Citation Format

Share Document