11 research outputs found

    Mutasjonstesting ved ikke-smĂĄcellet lungekreft

    Get PDF
    Bakgrunn. Epidermal vekstfaktorreseptor (EGFR) tyrosinkinasehemmere (EGFR-TKI) er en relativt ny klasse legemidler til behandling av ikke-småcellet lungekreft. Den nasjonale faggruppen for lungekreft, Norsk Lunge Cancer Gruppe, anbefaler at pasienter med ikke-småcellet lungekreft testes for mutasjoner i EGFRgenet. Vi rapporterer her erfaringene som er gjort etter at slik testing ble innført i Norge i 2010. Materiale og metode. Opplysninger om hvor mange som er testet, kjønnsfordeling, histopatologiske data og analyseresultater er samlet inn fra de molekylærpatologiske laboratorier ved universitetssykehusene i Tromsø, Trondheim, Bergen og Oslo for perioden mai 2010 til mai 2011. Resultater. 1 058 pasienter med lungekreft ble testet for mutasjoner i EGFRgenet i denne perioden, hvilket svarer til ca. halvdelen av alle som fikk diagnosen ikke-småcellet lungekreft. Mutasjon ble påvist hos 123 pasienter (11,6 %). Det var en høyere andel mutasjonspositive kvinner enn menn (17,6 % mot 6,3 %, p < 0,001), og lavere andel ved plateepitelkarsinom enn ved andre histopatologiske undertyper (3,0 % mot 12,9 %, p = 0,001). Av 80 cytologiske prøver var ni (11,3 %) positive. Fortolkning. På bakgrunn av den relativt høye mutasjonsfrekvensen og et ikke ubetydelig antall positive i plateepitelkarsinomgruppen, anbefaler vi videreføring av mutasjonstesting av alle pasienter med ikke-småcellet lungekreft

    Quality assurance of human papillomavirus (HPV) testing in the implementation of HPV primary screening in Norway: an inter-laboratory reproducibility study

    No full text
    Abstract Background Human papillomavirus (HPV) testing as primary screening for cervical cancer is currently being implemented in Norway in a randomized controlled fashion, involving three laboratories. As part of the quality assurance programme of the implementation, an evaluation of the inter-laboratory reproducibility of the HPV test was initiated, to ensure satisfactory HPV test reliability in all three laboratories. Methods The HPV test used is the cobas 4800 HPV Test, detecting 14 high-risk types with individual HPV genotype results for HPV16 and HPV18. In addition to the three laboratories involved in the implementation, the Norwegian HPV reference laboratory was included as a fourth comparative laboratory. A stratified sample of 500 cervical liquid based cytology (LBC) samples was used in the evaluation, with an aim towards a high-risk HPV positivity of ~25%. Samples were collected at one laboratory, anonymized, aliquoted, and distributed to the other laboratories. Results Comparison of the test results of all four laboratories revealed a 95.6% agreement, an 86.3% positive agreement and a kappa value of 0.94 (95% CI 0.92–0.97). For negative cytology specimens, there was a 95.8% overall agreement, a 67.4% positive agreement, and a kappa value of 0.88 (95% CI 0.80–0.93). For abnormal cytology specimens, there was a 95.8% overall agreement, a 95.5% positive agreement, and a kappa value of 0.86 (95% CI 0.71–0.97). Conclusions The study showed a high inter-laboratory reproducibility of HPV testing, implying satisfactory user performance and reliability in the laboratories involved in the implementation project. This is important knowledge and we recommend similar studies always to be performed prior to the introduction of new screening routines

    Tumor versus Stromal Cells in Culture—Survival of the Fittest?

    Get PDF
    <div><p>Two of the signature genetic events that occur in human gliomas, <i>EGFR</i> amplification and <i>IDH</i> mutation, are poorly represented in experimental models <i>in vitro</i>. <i>EGFR</i> amplification, for example, occurs in 40 to 50% of GBM, and yet, <i>EGFR</i> amplification is rarely preserved in cell cultures derived from human tumors. To analyze the fate of <i>EGFR</i> amplified and <i>IDH</i> mutated cells in culture, we followed the development over time of cultures derived from human xenografts in nude rats enriched for tumor cells with <i>EGFR</i> amplification and of cultures derived from patient samples with <i>IDH</i> mutations, in serum monolayer and spheroid suspension culture, under serum and serum free conditions. We observed under serum monolayer conditions, that nestin positive or nestin and SMA double positive rat stromal cells outgrew <i>EGFR</i> amplified tumor cells, while serum spheroid cultures preserved tumor cells with <i>EGFR</i> amplification. Serum free suspension culture exhibited a more variable cell composition in that the resultant cell populations were either predominantly nestin/SOX2 co-expressing rat stromal cells or human tumor cells, or a mixture of both. The selection for nestin/SMA positive stromal cells under serum monolayer conditions was also consistently observed in human oligodendrogliomas and oligoastrocytomas with <i>IDH</i> mutations. Our results highlight for the first time that serum monolayer conditions can select for stromal cells instead of tumor cells in certain brain tumor subtypes. This result has an important impact on the establishment of new tumor cell cultures from brain tumors and raises the question of the proper conditions for the growth of the tumor cell populations of interest.</p></div

    Characterization of rat stromal cells <i>in vitro</i> and <i>in vivo</i>.

    No full text
    <p><b>(a)</b> Rat stromal cells from P8 NBM, and P8 and P6 monolayer cultures were stained with antibodies against GFAP, beta-tubulin III, SOX2 and SMA. Rat cells from P8 NBM cultures are positive for SOX2 and negative for SMA, while monolayer cells are negative for SOX2 and positive for SMA. <b>(b)</b> Xenografts were stained with antibodies against rat nestin, SOX2 and SMA. Rat nestin is positive on tumor vessels and single cells in human <i>EGFR</i> amplified GBM xenografts <i>in vivo</i>. Double staining for rat nestin and SOX2 shows rat nestin+/SOX- vessels and rat nestin+/SOX2+ single cells (arrow). SMA is positive on vessels. Scale bars 50 µm.</p

    Rat stromal cells or a combination of tumor and stromal cells have a growth advantage in serum free cultures.

    No full text
    <p><b>(a)</b> FISH with an <i>EGFR</i>/Chromosome 7 probe in red and green, respectively, and immunohistochemical staining with antibodies against EGFR. In P8 cultures, both EGFR amplification and the chromosome 7 probe are not detectable after two months. In P6 cultures, EGFR amplification is preserved in spheroids (dotted line), while both EGFR amplification and the chromosome 7 probe are not detectable in single cells. EGFR expression is lost in P8 cultures, while it is still detectable at a low expression level in spheroids from P6 cultures. Quantification of EGFR expressing cells in three random high power (400×) microscopic view fields (HPF) in each group. Asterix indicates 0%. Values represent mean ± s.d. <b>(b)</b> Immunohistochemical staining with antibodies against human-specific and rat-specific nestin. In P8 cultures, rat-nestin positive stromal cells have a growth advantage over human cells. In P6 cultures, a mix of human and rat cells survive. Human cells form spheroids, while single cells are rat cells. Scale bars 50 µm. Quantification of rat and human nestin expressing cells in three random high power (400×) microscopic view fields (HPF) in each group. Asterix indicates 0%. Values represent mean ± s.d.</p

    Genomic profiles are normal in monolayer cell populations derived from human oligodendrogliomas.

    No full text
    <p><b>(a)</b> Sanger sequencing demonstrates that the <i>IDH1</i> mutation of the primary tumor is not preserved in the derived monolayer cell population. <b>(b)</b> Immunostaining with antibodies against IDH1 R132H confirms that monolayer cells are negative and the primary biopsy strongly positive. <b>(c)</b> aCGH profiles for the derived cell populations and the matching human oligodendroglioma are shown. Data is shown in log<sub>2</sub> space across the genome where log<sub>2</sub> 0 (at the midline) indicates the normal diploid copy number. Asterisks (*) highlight 1p and 19q losses that are typical of oligodendroglioma.</p

    EGFR amplification and expression is preserved in serum spheroid cultures and “lost” in monolayer cultures.

    No full text
    <p>Cultured spheroids or single cells were coagulated, fixed with 4% formalin, and embedded in paraffin. FISH with an <i>EGFR</i>/Chromosome 7 probe in red and green, respectively, and immunohistochemical staining with antibodies against EGFR. In monolayer culture, both EGFR amplification and the chromosome 7 probe are not detectable after two months in cultures from both xenografts. Immunhistochemical staining for EGFR is also negative. Scale bars 50 µm. Quantification of EGFR expressing cells in three random high power (400×) microscopic view fields (HPF) in each group. Asterix indicates 0%. Values represent mean ± s.d.</p

    Rat stromal cells have a selective growth advantage over EGFR amplified cells in serum monolayer culture.

    No full text
    <p>Cultured spheroids or single cells were coagulated, fixed with 4% formalin, and embedded in paraffin. Immunohistochemical staining was performed with antibodies against human-specific and rat-specific nestin. After two months, nestin-expressing rat cells take over at expense of human tumor cells in serum monolayer culture. As expected, nestin-expressing human cells are predominant in serum spheroid cultures. Scale bars 50 µm. Quantification of human and rat nestin expressing cells in three random high power (400×) microscopic view fields (HPF) in each group. Asterix indicates 0%. Values represent mean ± s.d.</p

    Characterization of stromal cells derived from human oligodendroglioma <i>in vitro</i> and <i>in vivo</i>.

    No full text
    <p><b>(a)</b> Human stromal cells from oligodendrogliomas, cultured under monolayer conditions are positive for nestin and SMA. Only few single cells are positive for GFAP. <b>(b)</b> Nestin is localized to reactive astrocytes and the vasculature in human oligodendroglioma samples <i>in vivo</i>. SMA is positive on the vasculature. Scale bars 50 µm.</p
    corecore