Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kerry Fenwick is active.

Publication


Featured researches published by Kerry Fenwick.


Clinical Cancer Research | 2006

FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas

Jorge S. Reis-Filho; Peter T. Simpson; Nicholas C. Turner; Maryou Ballo Lambros; Chris Jones; Alan Mackay; Anita Grigoriadis; David Sarrió; Kay Savage; Tim Dexter; Marjan Iravani; Kerry Fenwick; Barbara L. Weber; David Hardisson; Fernando Schmitt; José Palacios; Sunil R. Lakhani; Alan Ashworth

Purpose: Classic lobular carcinomas (CLC) account for 10% to 15% of all breast cancers. At the genetic level, CLCs show recurrent physical loss of chromosome16q coupled with the lack of E-cadherin (CDH1 gene) expression. However, little is known about the putative therapeutic targets for these tumors. The aim of this study was to characterize CLCs at the molecular genetic level and identify putative therapeutic targets. Experimental Design: We subjected 13 cases of CLC to a comprehensive molecular analysis including immunohistochemistry for E-cadherin, estrogen and progesterone receptors, HER2/neu and p53; high-resolution comparative genomic hybridization (HR-CGH); microarray-based CGH (aCGH); and fluorescent and chromogenic in situ hybridization for CCND1 and FGFR1. Results: All cases lacked the expression of E-cadherin, p53, and HER2, and all but one case was positive for estrogen receptors. HR-CGH revealed recurrent gains on 1q and losses on 16q (both, 85%). aCGH showed a good agreement with but higher resolution and sensitivity than HR-CGH. Recurrent, high level gains at 11q13 (CCND1) and 8p12-p11.2 were identified in seven and six cases, respectively, and were validated with in situ hybridization. Examination of aCGH and the gene expression profile data of the cell lines, MDA-MB-134 and ZR-75-1, which harbor distinct gains of 8p12-p11.2, identified FGFR1 as a putative amplicon driver of 8p12-p11.2 amplification in MDA-MB-134. Inhibition of FGFR1 expression using small interfering RNA or a small-molecule chemical inhibitor showed that FGFR1 signaling contributes to the survival of MDA-MB-134 cells. Conclusions: Our findings suggest that receptor FGFR1 inhibitors may be useful as therapeutics in a subset of CLCs.


The Journal of Pathology | 2005

Pleomorphic lobular carcinoma of the breast: role of comprehensive molecular pathology in characterization of an entity

Jorge S. Reis-Filho; Peter T. Simpson; Chris Jones; Dawn Steele; Alan Mackay; Marjan Iravani; Kerry Fenwick; Haukur Valgeirsson; Maryou B. Lambros; Alan Ashworth; José Palacios; Fernando Schmitt; Sunil R. Lakhani

Immunohistochemical analysis of E‐cadherin has changed the way lobular neoplasia is perceived. It has helped to classify difficult cases of carcinoma in situ with indeterminate features and led to the identification of new variants of lobular carcinoma. Pleomorphic lobular carcinoma (PLC) and pleomorphic lobular carcinoma in situ (PLCIS), recently described variants of invasive and in situ classic lobular carcinoma, are reported to be associated with more aggressive clinical behaviour. Although PLC/PLCIS show morphological features of classic lobular neoplasia and lack E‐cadherin expression, it is still unclear whether these lesions evolve through the same genetic pathway as lobular carcinomas or are high‐grade ductal neoplasms that have lost E‐cadherin. Here we have analysed a case of extensive PLCIS and invasive PLC associated with areas of E‐cadherin‐negative carcinoma in situ with indeterminate features, using immunohistochemistry, chromogenic in situ hybridization, high‐resolution comparative genomic hybridization (CGH) and array‐based CGH. We observed that all lesions lacked E‐cadherin and β‐catenin and showed gain of 1q and loss of 16q, features that are typical of lobular carcinomas but are not seen in high‐grade ductal lesions. In addition, amplifications of c‐myc and HER2 were detected in the pleomorphic components, which may account for the high‐grade features in this case and the reported aggressive clinical behaviour of these lesions. Taken together, these data suggest that at least some PLCs may evolve from the same precursor or through the same genetic pathway as classic lobular carcinomas. Copyright


Blood | 2012

Intraclonal heterogeneity and distinct molecular mechanisms characterize the development of t(4;14) and t(11;14) myeloma.

Brian A. Walker; Christopher P. Wardell; Lorenzo Melchor; Sanna Hulkki; Nicola E. Potter; David C. Johnson; Kerry Fenwick; Iwanka Kozarewa; David Gonzalez; Christopher J. Lord; Alan Ashworth; Faith E. Davies; Gareth J. Morgan

We have used whole exome sequencing to compare a group of presentation t(4;14) with t(11;14) cases of myeloma to define the mutational landscape. Each case was characterized by a median of 24.5 exonic nonsynonymous single-nucleotide variations, and there was a consistently higher number of mutations in the t(4;14) group, but this number did not reach statistical significance. We show that the transition and transversion rates in the 2 subgroups are similar, suggesting that there was no specific mechanism leading to mutation differentiating the 2 groups. Only 3% of mutations were seen in both groups, and recurrently mutated genes include NRAS, KRAS, BRAF, and DIS3 as well as DNAH5, a member of the axonemal dynein family. The pattern of mutation in each group was distinct, with the t(4;14) group being characterized by deregulation of chromatin organization, actin filament, and microfilament movement. Recurrent RAS pathway mutations identified subclonal heterogeneity at a mutational level in both groups, with mutations being present as either dominant or minor subclones. The presence of subclonal diversity was confirmed at a single-cell level using other tumor-acquired mutations. These results are consistent with a distinct molecular pathogenesis underlying each subgroup and have important impacts on targeted treatment strategies. The Medical Research Council Myeloma IX trial is registered under ISRCTN68454111.


The Journal of Pathology | 2009

Does chromosome 17 centromere copy number predict polysomy in breast cancer? A fluorescence in situ hybridization and microarray-based CGH analysis

Caterina Marchiò; Maryou B. Lambros; Patrizia Gugliotta; Ludovica Verdun di Cantogno; C. Botta; Barbara Pasini; David Sp Tan; Alan Mackay; Kerry Fenwick; Narinder Tamber; Gianni Bussolati; Alan Ashworth; Jorge S. Reis-Filho; Anna Sapino

Approximately 8% of breast cancers show increased copy numbers of chromosome 17 centromere (CEP17) by fluorescence in situ hybridization (FISH) (ie average CEP17 >3.0 per nucleus). Currently, this pattern is believed to represent polysomy of chromosome 17. HER2‐amplified cancers have been shown to harbour complex patterns of genetic aberrations of chromosome 17, in particular involving its long arm. We hypothesized that aberrant copy numbers of CEP17 in FISH assays may not necessarily represent true chromosome 17 polysomy. Eighteen randomly selected CEP17 polysomic cases and a control group of ten CEP17 disomic cases, as defined by dual‐colour FISH, were studied by microarray‐based comparative genomic hybridization (aCGH), which was performed on microdissected samples using a 32K tiling‐path bacterial artificial chromosome microarray platform. Additional FISH probes were employed for SMS (17p11.2) and RARA (17q21.2) genes, as references for chromosome 17 copy number. Microarray‐based comparative genomic hybridization revealed that 11 out of the 18 polysomic cases harboured gains of 17q with involvement of the centromere, one displayed 17q gain sparing the centromeric region, and only one could be defined as polysomic. The remaining five cases displayed amplification of the centromeric region. Among these, one case, showing score 2+ by immunohistochemistry and 8.5 HER2 mean copy number, was classified as not amplified by HER2/CEP17 ratio and as amplified by HER2/SMS ratio. Our results suggest that true chromosome 17 polysomy is likely to be a rare event in breast cancer and that CEP17 copy number greater than 3.0 in FISH analysis is frequently related to gain or amplification of the centromeric region. Larger studies investigating the genetic profiles of CEP17 polysomic cases are warranted. Copyright


Clinical Cancer Research | 2009

Tiling Path Genomic Profiling of Grade 3 Invasive Ductal Breast Cancers

Rachael Natrajan; Maryou B. Lambros; Socorro María Rodríguez-Pinilla; Gema Moreno-Bueno; David Sp Tan; Caterina Marchiò; Radost Vatcheva; Sydonia Rayter; Betania Mahler-Araujo; Laura G. Fulford; Daniela Hungermann; Alan Mackay; Anita Grigoriadis; Kerry Fenwick; Narinder Tamber; David Hardisson; Andrew Tutt; José Palacios; Christopher J. Lord; Horst Buerger; Alan Ashworth; Jorge S. Reis-Filho

Purpose: To characterize the molecular genetic profiles of grade 3 invasive ductal carcinomas of no special type using high-resolution microarray-based comparative genomic hybridization (aCGH) and to identify recurrent amplicons harboring putative therapeutic targets associated with luminal, HER-2, and basal-like tumor phenotypes. Experimental Design: Ninety-five grade 3 invasive ductal carcinomas of no special type were classified into luminal, HER-2, and basal-like subgroups using a previously validated immunohistochemical panel. Tumor samples were microdissected and subjected to aCGH using a tiling path 32K BAC array platform. Selected regions of recurrent amplification were validated by means of in situ hybridization. Expression of genes pertaining to selected amplicons was investigated using quantitative real-time PCR and gene silencing was done using previously validated short hairpin RNA constructs. Results: We show that basal-like and HER-2 tumors are characterized by “sawtooth” and “firestorm” genetic patterns, respectively, whereas luminal cancers were more heterogeneous. Apart from confirming known amplifications associated with basal-like (1q21, 10p, and 12p), luminal (8p12, 11q13, and 11q14), and HER-2 (17q12) cancers, we identified previously unreported recurrent amplifications associated with each molecular subgroup: 19q12 in basal-like, 1q32.1 in luminal, and 14q12 in HER-2 cancers. PPM1D gene amplification (17q23.2) was found in 20% and 8% of HER-2 and luminal cancers, respectively. Silencing of PPM1D by short hairpin RNA resulted in selective loss of viability in tumor cell lines harboring the 17q23.2 amplification. Conclusions: Our results show the power of aCGH analysis in unraveling the genetic profiles of specific subgroups of cancer and for the identification of novel therapeutic targets.


Science Translational Medicine | 2015

Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer.

Gaia Schiavon; Sarah Hrebien; Isaac Garcia-Murillas; Rosalind J. Cutts; Alex Pearson; Noelia Tarazona; Kerry Fenwick; Iwanka Kozarewa; Elena Lopez-Knowles; Ricardo Ribas; Ashutosh Nerurkar; Peter Osin; Sarat Chandarlapaty; Lesley-Ann Martin; Mitch Dowsett; Ian E. Smith; Nicholas C. Turner

ESR1 mutations evolve during the treatment of metastatic breast cancer. An evolving problem A large number of breast cancers express the estrogen receptor, making them susceptible to hormonal treatments. Unfortunately, these tumors can develop mutations in the estrogen receptor gene (ESR1) and become resistant to hormonal therapies that were previously effective. Schiavon et al. used three independent cohorts of breast cancer patients to demonstrate that these mutations only evolved in cases where hormonal therapy was started late in the course of the disease, after development of metastasis, and not during the initial course of treatment. If these findings are confirmed in prospective clinical trials, then they will explain why starting hormonal treatment early decreases the risk of subsequent resistance to hormonal therapy. Acquired ESR1 mutations are a major mechanism of resistance to aromatase inhibitors (AIs). We developed ultra high–sensitivity multiplex digital polymerase chain reaction assays for ESR1 mutations in circulating tumor DNA (ctDNA) and investigated the clinical relevance and origin of ESR1 mutations in 171 women with advanced breast cancer. ESR1 mutation status in ctDNA showed high concordance with contemporaneous tumor biopsies and was accurately assessed in samples shipped at room temperature in preservative tubes. ESR1 mutations were found exclusively in estrogen receptor–positive breast cancer patients previously exposed to AI. Patients with ESR1 mutations had a substantially shorter progression-free survival on subsequent AI-based therapy [hazard ratio, 3.1; 95% confidence interval (CI), 1.9 to 23.1; P = 0.0041]. ESR1 mutation prevalence differed markedly between patients who were first exposed to AI during the adjuvant and metastatic settings [5.8% (3 of 52) versus 36.4% (16 of 44), respectively; P = 0.0002]. In an independent cohort, ESR1 mutations were identified in 0% (0 of 32; 95% CI, 0 to 10.9) tumor biopsies taken after progression on adjuvant AI. In a patient with serial sampling, ESR1 mutation was selected during metastatic AI therapy to become the dominant clone in the cancer. ESR1 mutations can be robustly identified with ctDNA analysis and predict for resistance to subsequent AI therapy. ESR1 mutations are rarely acquired during adjuvant AI but are commonly selected by therapy for metastatic disease, providing evidence that mechanisms of resistance to targeted therapy may be substantially different between the treatment of micrometastatic and overt metastatic cancer.


Science Translational Medicine | 2011

Identification of a Disease-Defining Gene Fusion in Epithelioid Hemangioendothelioma

Munir R. Tanas; Andrea Sboner; Andre M. Oliveira; Michele R. Erickson-Johnson; Jessica Hespelt; Philip J. Hanwright; John G. Flanagan; Yuling Luo; Kerry Fenwick; Rachael Natrajan; Costas Mitsopoulos; Marketa Zvelebil; Benjamin Hoch; Sharon W. Weiss; Maria Debiec-Rychter; Raf Sciot; Robert B. West; Alexander J. Lazar; Alan Ashworth; Jorge S. Reis-Filho; Christopher J. Lord; Mark Gerstein; Mark A. Rubin; Brian P. Rubin

A newly identified gene fusion defines the vascular cancer epithelioid hemangioendothelioma and encodes a chimeric transcription factor. FISHing for a Gene Fusion Mother was right: There is a time and place for everything. And at the molecular level, inappropriate behavior can have consequences much more severe than being grounded. Using an unbiased deep-sequencing approach coupled with traditional chromosomal karyotyping, Tanas et al. now describe the genes involved in a fusion event that defines epithelioid hemangioendothelioma (EHE), a rare vascular cancer. This genetic aberration may instigate the bad behavior—an improper transcriptional program in endothelial cells. A rare sarcoma, EHE is difficult to diagnose because it shares many characteristics with normal endothelial cells and resembles other abnormal vascular neoplasms, such as epithelioid hemangioma, a benign condition, and epithelioid angiosarcoma, an aggressive vascular cancer. Treatment for patients with localized EHE includes surgical removal, when possible, or liver transplantation in the case of hepatic involvement, and there is no treatment for metastatic disease. To aid in diagnosis and decipher the pathological processes behind this mysterious cancer, researchers and clinicians need a defining biomarker for EHE. Traditional cytogenetic techniques for identifying the genes involved in a genetic translocation are labor-intensive, especially for a rare cancer for which no cell lines are available. So, Tanas et al. took a shortcut; the authors combined cytogenetic methods with deep transcriptome sequencing, which they used to search in an unbiased way for the product of the t(1;3)(p36;q25) chromosomal translocation characteristic of EHE. The translocation involved two genes, WWTR1, which encodes a transcriptional coactivator that is highly expressed in endothelial cells, and CAMTA1, a DNA binding transcriptional regulatory protein that is normally expressed during brain development. The WWTR1/CAMTA1 gene fusion contains the strong endothelial cell promoter of WWTR1, which may drive the inappropriate expression of a protein-encoding fragment of CAMTA1 in endothelial cells. The authors suggest that this promoter switch initiates an ill-suited and ill-timed transcriptional program that may play a role in cancer biology. If this is the case, then the chimeric WWTR1/CAMTA1 transcription factor may represent a therapeutic target for EHE-specific drugs. To aid in disease diagnosis, Tanas et al. also devised a sensitive and specific fluorescence in situ hybridization assay to detect the EHE translocation. Together, these tools should teach researchers about the biology and prognosis of this rare cancer and eventually help bring the bad behavior under control. Integrating transcriptomic sequencing with conventional cytogenetics, we identified WWTR1 (WW domain–containing transcription regulator 1) (3q25) and CAMTA1 (calmodulin-binding transcription activator 1) (1p36) as the two genes involved in the t(1;3)(p36;q25) chromosomal translocation that is characteristic of epithelioid hemangioendothelioma (EHE), a vascular sarcoma. This WWTR1/CAMTA1 gene fusion is under the transcriptional control of the WWTR1 promoter and encodes a putative chimeric transcription factor that joins the amino terminus of WWTR1, a protein that is highly expressed in endothelial cells, in-frame to the carboxyl terminus of CAMTA1, a protein that is normally expressed only in brain. Thus, CAMTA1 expression is activated inappropriately through a promoter-switch mechanism. The gene fusion is present in virtually all EHEs tested but is absent from all other vascular neoplasms, demonstrating it to be a disease-defining genetic alteration. A sensitive and specific break-apart fluorescence in situ hybridization assay was also developed to detect the translocation and will assist in the evaluation of this diagnostically challenging neoplasm. The chimeric WWTR1/CAMTA1 transcription factor may represent a therapeutic target for EHE and offers the opportunity to shed light on the functions of two poorly characterized proteins.


Laboratory Investigation | 2008

Genomic analysis of the HER2/TOP2A amplicon in breast cancer and breast cancer cell lines.

Edurne Arriola; Caterina Marchiò; David Sp Tan; Suzanne Drury; Maryou B. Lambros; Rachael Natrajan; Socorro María Rodríguez-Pinilla; Alan Mackay; Narinder Tamber; Kerry Fenwick; Chris Jones; Mitch Dowsett; Alan Ashworth; Jorge S. Reis-Filho

HER2 and TOP2A are targets for the therapeutic agents trastuzumab and anthracyclines and are frequently amplified in breast cancers. The aims of this study were to provide a detailed molecular genetic analysis of the 17q12–q21 amplicon in breast cancers harbouring HER2/TOP2A co-amplification and to investigate additional recurrent co-amplifications in HER2/TOP2A-co-amplified cancers. In total, 15 breast cancers with HER2 amplification, 10 of which also harboured TOP2A amplification, as defined by chromogenic in situ hybridisation, and 6 breast cancer cell lines known to be amplified for HER2 were subjected to high-resolution microarray-based comparative genomic hybridisation analysis. This revealed that the genomes of 12 cases were characterised by at least one localised region of clustered, relatively narrow peaks of amplification, with each cluster confined to a single chromosome arm (ie ‘firestorm’ pattern) and 3 cases displayed many narrow segments of duplication and deletion affecting the vast majority of chromosomes (ie ‘sawtooth’ pattern). The smallest region of amplification (SRA) on 17q12 in the whole series extended from 34.73 to 35.48 Mb, and encompassed HER2 but not TOP2A. In HER2/TOP2A-co-amplified samples, the SRA extended from 34.73 to 36.54 Mb, spanning a region of ∼1.8 Mb. Apart from HER2 and TOP2A, this region encompassed four additional genes whose expression levels as defined by quantitative real-time PCR are significantly higher in HER2/TOP2A-co-amplified vs HER2-amplified breast cancers: CASC3, CDC6, RARA and SMARCE1. Of the cell lines studied, SKBR3 and UACC812 showed HER2/TOP2A co-amplification. In conclusion, this is the first detailed genome-wide characterisation of HER2/TOP2A-amplified breast cancers; cell lines were identified that can be used to model these cancers in vitro. The 17q12 amplicon is complex and harbours multiple genes that may be associated with breast cancer development and progression, and potentially exploitable as therapeutic targets.


The Journal of Pathology | 2008

Molecular profiling pleomorphic lobular carcinomas of the breast: evidence for a common molecular genetic pathway with classic lobular carcinomas

Peter T. Simpson; Jorge S. Reis-Filho; Mb Lambros; Chris Jones; Dawn Steele; Alan Mackay; Marjan Iravani; Kerry Fenwick; Tim Dexter; A. Jones; Lynne Reid; L. Da Silva; Sandra J. Shin; David Hardisson; Alan Ashworth; Fernando Schmitt; José Palacios; Sunil R. Lakhani

Pleomorphic lobular carcinomas (PLC) of the breast display histological features associated with classic invasive lobular carcinoma (ILC), yet they also exhibit more conspicuous nuclear atypia and pleomorphism, and an aggressive clinical behaviour. From a breast cancer progression perspective, it is unclear whether PLC is a variant of ILC or is a high‐grade invasive ductal carcinoma (IDC) that has lost E‐cadherin. The molecular features of 26 PLC were studied using immunohistochemistry [oestrogen receptor (ER), progesterone receptor (PR), HER2, p53 and E‐cadherin], 0.9 Mb resolution, microarray‐based comparative genomic hybridization (aCGH), fluorescent (FISH) and chromogenic (CISH) in situ hybridization and loss of heterozygosity. Comparative analysis was performed with aCGH data from PLC with classic ILC (16 cases) and high grade IDC (35 cases). PLCs were frequently ER‐ and PR‐positive, E‐cadherin‐negative and occasionally HER2‐ and p53‐positive. Recurrent copy number changes identified by aCGH included gains on 1q, 8q, 11q, 12q, 16p and 17q and losses on 8p, 11q, 13q, 16q and Xq. Highly recurrent 1q+ (100% of cases), 16p+ (93%), 11q− (53%) and 16q− (93%) and evidence of the der(1;16)/der(16)t(1;16) rearrangement, as detected by FISH, suggested that PLC had a ‘lobular genotype’. Focal amplifications were evident at 8p12‐p11, 8q24, 11q13.1‐q14.1, 12q14, 17q12 and 20q13. Loss of BRCA2 was detected in 40% of PLC by LOH. Comparative analysis of aCGH data suggested the molecular features of PLC (ER/PR‐positive, E‐cadherin‐negative, 1q+, 11q−, 16p+ and 16q−) were more closely related to those of ILC than IDC, implicating an overlapping developmental pathway for these lobular tumour types. Molecular alterations found in PLC that are more typical of high‐grade IDC than ILC (p53 and HER2 positivity, 8q+, 17q24‐q25+, 13q− and amplification of 8q24, 12q14, 17q12 and 20q13) are likely to drive the high‐grade and more aggressive biology of PLC. Copyright


The Journal of Pathology | 2006

Array CGH profiling of favourable histology Wilms tumours reveals novel gains and losses associated with relapse

Rachael Natrajan; Richard D. Williams; Sandra Hing; Alan Mackay; Jorge S. Reis-Filho; Kerry Fenwick; Marjan Iravani; Haukur Valgeirsson; Anita Grigoriadis; Cordelia Langford; Oliver M. Dovey; Simon G. Gregory; Barbara L. Weber; Alan Ashworth; Paul E. Grundy; Kathy Pritchard-Jones; Chris Jones

Despite the excellent survival of Wilms tumour patients treated with multimodality therapy, approximately 15% will suffer from tumour relapse, where response rates are markedly reduced. We have carried out microarray‐based comparative genomic hybridisation on a series of 76 Wilms tumour samples, enriched for cases which recurred, to identify changes in DNA copy number associated with clinical outcome. Using 1Mb‐spaced genome‐wide BAC arrays, the most significantly different genomic changes between favourable histology tumours that did (n = 37), and did not (n = 39), subsequently relapse were gains on 1q, and novel deletions at 12q24 and 18q21. Further relapse‐associated loci included losses at 1q32.1, 2q36.3‐2q37.1, and gain at 13q31. 1q gains correlated strongly with loss of 1p and/or 16q. In 3 of 11 cases with concurrent 1p−/1q+, a breakpoint was identified at 1p13. Multiple low‐level sub‐megabase gains along the length of 1q were identified using chromosome 1 tiling‐path arrays. One such recurrent region at 1q22‐q23.1 included candidate genes RAB25, NES, CRABP2, HDGF and NTRK1, which were screened for mRNA expression using quantitative RT‐PCR. These data provide a high‐resolution catalogue of genomic copy number changes in relapsing favourable histology Wilms tumours. Copyright

Collaboration


Dive into the Kerry Fenwick's collaboration.

Top Co-Authors

Avatar

Alan Ashworth

University of California

View shared research outputs
Top Co-Authors

Avatar

Alan Mackay

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Christopher J. Lord

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Iwanka Kozarewa

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jorge S. Reis-Filho

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rachael Natrajan

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Chris Jones

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Ioannis Assiotis

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Maryou B. Lambros

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Costas Mitsopoulos

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge