scholarly journals Detecting Gene Copy Number Fluctuations in Tumor Cells by Microarray Analysis of Genomic Representations

2000 ◽  
Vol 10 (11) ◽  
pp. 1726-1736 ◽  
Author(s):  
R. Lucito
2008 ◽  
Vol 123 (4) ◽  
pp. 817-825 ◽  
Author(s):  
Samuel Myllykangas ◽  
Siina Junnila ◽  
Arto Kokkola ◽  
Reija Autio ◽  
Ilari Scheinin ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2868-2868 ◽  
Author(s):  
Pedro Farrajota Neves Da Silva ◽  
Vassilis Atsaves ◽  
Nikolaos Tsesmetzis ◽  
Georgia Kokaraki ◽  
Anders Österborg ◽  
...  

Abstract Introduction: The PD-1 / PD-L1 interaction is a crucial immune checkpoint in cancer, which has been targeted by monoclonal antibodies currently used in clinical trials. The genetic basis and transcriptional regulation of PD-L1 has been previously investigated in ALK+ and ALK- anaplastic large cell lymphomas (ALCL) by our group (Atsaves et al, Leukemia 2017, (7):1633-1637), however, the regulatory mechanisms of PD-1/PD-L1 immune checkpoint in other T-cell lymphomas are not yet known. PD-L1 expression in neoplastic cells has been associated with worse clinical outcome in other hematologic malignancies including Hodgkin lymphoma, diffuse large B-cell lymphoma (DLBCL) and plasma cell myeloma, however, the clinical significance of PD-L1 expression in neoplastic cells and in the immune cells of the tumor microenvironment in PTCLs has not been studied to date. In this study, we investigated PD-L1 gene copy number alterations using a genome-wide approach and diagnostic biopsies. We also assessed for the expression patterns of PD-L1 in the neoplastic cells and histiocytes in PTCL tumors as well as for their possible associations with clinical outcome. Methods: The PTCL study group included 64 patients (median age 62 years; 61% males, various histologic types), diagnosed and treated at Karolinska University Hospital (Sweden). All tissue samples were obtained prior to therapy. PD-L1 expression was assessed by immunohistochemistry performed on tissue microarrays or full tissue sections using a PD-L1 monoclonal antibody (SP263, Ventana). At least 500 lymphoma cells were counted to calculate the percentage of PDL1-positive tumor cells. PD-L1 expression in histiocytes was scored as "high" or "low" (arbitrary) based on the number of positive infiltrating histiocytes. Overall survival (OS) defined as time from diagnosis to death or last follow-up was chosen as the endpoint for outcome. Survival analyses were performed using the Kaplan-Meier method (log-rank test) and Cox hazards models. In addition, a pilot series of 24 PTCLs were analysed by Oncoscan (Affymetrics) for PD-L1 gene copy number alterations and the results were confirmed by digital q-RT PCR at the mRNA level. Genomic DNA and total RNA were extracted from diagnostic formalin-fixed, paraffin-embedded (FFPE) biopsies using standard protocols. Associations between PD-L1 expression and PTCL type were statistically analysed with Fisher's exact, chi-square, Mann-Whitney and Kruskal-Wallis tests. Survival analysis was performed using the Kaplan-Meier method (log rank test) and Cox proportional hazards model. Results: Whole genome analysis using Oncoscan (Affymetrics) showed low-level chromosomal gains in 2 (8.3%) PTCLs, which were confirmed by digital q-RT PCR that demonstrated slightly increased PD-L1 mRNA levels (PD-L1/reference gene <2). No correlation of the gene copy number findings with PD-L1 protein expression assessed by immunohistochemistry was seen. Expression of PD-L1 was observed in 92% ALK+ ALCL, 57% ALK- ALCL, 38% PTCL, NOS and 18% angioimmunoblastic T-cell lymphomas (AILT) and less frequently in other PTCL types (p=0.0019). After a median follow up of 66 months, PD-L1 expression by the lymphoma cells was associated with superior OS in the entire study group and in the subgroup of PTCL other than ALK+ ALCL (p<0.0001, logrank). By contrast, high PD-L1 expression in histiocytes was associated with inferior OS (p<0.0001, logrank). The PTCL patients with PD-L1-positive tumor cells and PD-L1-low expression in histiocytes showed a 5-year OS 71% as compared to 24% for the PTCLs with PD-L1-negative tumor cells and PD-L1-high expression in histiocytes (p=0.0002, logrank) and this difference remained highly significant in the subgroup of PTCL other than ALK+ ALCL (p=0.0005, logrank). Both the PD-L1 negativity in lymphoma cells (p=0.0007) and PD-L1-high expression in histiocytes (p=0.04) were independent adverse prognostic factors in the multivariable analysis (Cox model). Conclusions: Copy number alterations of the PD-L1 gene (9p24.1 locus) are uncommon in PTCL subtypes and therefore, PD-L1 gene expression is likely regulated at the transcriptional of post-translational levels in T-cell lymphomas. PD-L1 negativity in the lymphoma cells and high PD-L1 expression in histiocytes are strong adverse prognostic factors in PTCL, however, larger series are needed to confirm these associations in each histologic type of PTCL. Disclosures Österborg: Pharmacyclics: Research Funding; Janssen: Research Funding; Gilead: Consultancy, Research Funding; Abbvie: Research Funding; Beigene: Research Funding.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 10569-10569
Author(s):  
N. Personeni ◽  
G. De Hertogh ◽  
S. Störkel ◽  
M. Debiec-Rychter ◽  
K. Geboes ◽  
...  

10569 Background: Recent studies suggest that increased epidermal growth factor receptor (EGFR) gene copy number assessed by in situ hybridization predicts response to cetuximab in patients (pts) with advanced colorectal cancer (mCRC). Additionally, preclinical data indicate that HER2, a member of the EGFR family, can modulate the efficacy of anti-EGFR monoclonal antibodies. Methods: We assessed EGFR and HER2 gene copies by fluorescent in situ hybridization (FISH) on paraffin-embedded samples from 70 pts with mCRC treated with cetuximab alone or with irinotecan. FISH was assessed either on primary tumors (55 pts) or on metastases (15 pts) according to two parameters: the absolute copy number of genes and chromosome (chr) centromeres, and their frequencies in 100 tumor cells. A sensitivity analysis was performed and fitted to outcome data in an attempt to define relevant cutoffs. EGFR protein expression by immunohistochemistry was deemed positive with =10% tumor cells being stained. Multiple paired samples originating from different tumor sources were analyzed whenever available (27 pts). Results: The overall response rate was 25%. Prevalent FISH patterns were disomy (8%) and balanced polysomy (90%) for EGFR gene and chr 7. EGFR amplification was seen in two pts (2%), of which only one responded. HER2 amplification was seen in two of 54 pts (3%), both experiencing stable disease. Average EGFR copies and frequency of tumor cells with >2 copies respectively ranged from 1.6 to 4.0 and from 10% to 90%, thus reflecting a substantial tumor heterogeneity. Despite assessing multiple centile cutoffs of gene copy numbers and their frequencies in tumor cells, we found no association between EGFR and HER2 copy numbers and objective response, time to progression, and overall survival. Analysis of paired samples did not improve the predictive value of EGFR copies by FISH. Excluding EGFR amplification, associated to a strong (3+) EGFR staining, we found no correlation between EGFR copy number and protein expression. Conclusions: Neither EGFR nor HER2 copies by FISH, nor EGFR expression, are predictors of outcome in mCRC pts treated with cetuximab. FISH might still play a role in screening EGFR and HER2 amplification, but cost-effectiveness is debatable. [Table: see text]


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 8045-8045 ◽  
Author(s):  
Adam Platt ◽  
Paul Elvin ◽  
John Morten ◽  
Qunsheng Ji ◽  
Emma Donald ◽  
...  

8045 Background: The prevalence of the tumorigenic KIF5B:RET fusion gene in NSCLC tumors has been estimated at 0.2–6% (Jiu et al 2012; Lipson et al 2012). We retrospectively analyzed tumor samples from 4 Phase III NSCLC trials of vandetanib, a TKI that selectively targets RET, VEGFR and EGFR signaling, to determine the prevalence of RET fusions and other RET biomarkers, and any potential association with outcome to vandetanib (V). Methods: The studies evaluated were ZODIAC (NCT00312377; docetaxel ± V 100mg), ZEAL (NCT00418886; pemetrexed ± V 100mg), ZEPHYR (NCT00404924; V 300mg vs placebo) and ZEST (NCT00364351; V 300mg vs erlotinib). RET biomarkers evaluated included RET fusions (including KIF5B:RET) and RET gene copy number (assessed by a 4-probe FISH assay), as well as RET protein expression (by IHC). Results: Of 4089 patients randomized across the 4 studies, 1291 and 1234 had tumor samples available for FISH and IHC analysis, respectively, with evaluable data obtained for 944 and 1102. RET fusions (in >10% of tumor cells) were detected in 7 of 944 samples (vandetanib, n=3; comparator, n=4), at a prevalence of 0.7% (95% CI, 0.3–1.5%). None of the 3 vandetanib-treated RET fusion-positive patients had an objective RECIST response, although there was radiologic evidence of tumor shrinkage in 2. Overall, 2.8% (n=26) of samples had RET amplification (innumerable RET clusters, or ≥7 copies in >10% tumor cells), 8.1% (n=76) had lower RET gene copy number gain (4–6 copies in ≥40% tumor cells) and 8.3% (n=92) were RET expression positive (signal intensity ++ or +++ in >10% of tumor cells). There was no difference in ORR between vandetanib and comparator for the RET amplification-positive subset (both 8.3% [1/12]), the RET copy number gain subset (9.8% [4/41] vs 9.1% [3/33], respectively) or the RET protein expression-positive subset (15.2% [7/46] and 13.6% [6/44], respectively). Conclusions: The prevalence of RET fusions was estimated at 0.7%. There were too few vandetanib-treated patients with RET fusions to make any firm conclusion regarding association with efficacy. Evidence from the other RET biomarkers tested suggested that these do not infer a differential advantage in patients treated with vandetanib. Clinical trial information: NCT00312377; NCT00418886; NCT00404924.


2019 ◽  
Vol 237 ◽  
pp. 10-18
Author(s):  
Paweł Krawczyk ◽  
Anna Grenda ◽  
Kamila Wojas-Krawczyk ◽  
Marcin Nicoś ◽  
Tomasz Kucharczyk ◽  
...  

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 7078-7078
Author(s):  
Anne S. Tsao ◽  
Nusrat Harun ◽  
Junya Fujimoto ◽  
Vikki Devito ◽  
J. Jack Lee ◽  
...  

7078 Background: PDGF/PDGFR pathway has been implicated in malignant pleural mesothelioma (MPM) carcinogenesis. We sought to evaluate the incidence of PDGFRB gene copy number gain (CNG) and PDGFR pathway protein expression by immunohistochemistry (IHC) in the tumor cell cytoplasm, membrane, nucleus, and stroma, and correlate it to patient clinical outcome. Methods: 88 archived tumor blocks from resected MPM with full clinical information were used to perform the analyses. IHC biomarkers for PDGFRα,β and p-PDGFRβ, and fluorescence in situ hybridization were performed for analysis of PDGFRB gene CNG. Spearman’s rank correlation, Wilcoxon rank-sum test or Kruskal-Wallis test, BLiP plots, and Kaplan-Meier method were used to assess the biomarkers and their correlation to clinical outcome. Results: There were several correlations identified between the IHC biomarkers; however, none associated with patient demographics or histology subtype, with the exception of high cytoplasmic PDGFRα occurring in patients with no prior known asbestos exposure (p=0.029). In the CNG analysis, PDGFRB gene CNG in > 10% of tumor cells had lower cytoplasmic p-PDGFRβ (p=0.029), while PDGFRB gene CNG in > 40% of tumor cells had a higher cytoplasmic PDGFRβ (p=0.04). PDGFRB gene CNG status did not associate with patient demographics or tumor characteristics. Patients with PDGFRB CNG > 40% of tumor cells had an improved relapse-free survival (RFS) [HR 0.25 (95% CI 0.09, 0.72), p=0.0096]. In the patients with PDGFRB CNG > 40% of cells, the addition of chemotherapy appeared to also improve RFS (p=0.017). In the multi-covariate analyses for RFS, there was no association with any IHC biomarker. In the overall survival (OS) analysis, having PDGFRB gene CNG > 40% of tumor cells correlated with an improved OS [HR 0.32 (95% CI 0.11, 0.89), p=0.029] and the addition of peri-operative chemotherapy led to a trend towards improved OS (p=0.089). Conclusions: PDGFRB CNG > 40% of tumor cells is a potential prognostic biomarker in surgically resected MPM tumors. Adding chemotherapy to patients with PDGFRB CNG > 40% of cells improved RFS and led to a trend towards improved OS. Future validation of this biomarker in prospective trials is needed.


Sign in / Sign up

Export Citation Format

Share Document