Network


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

Hotspot


Dive into the research topics where Benjamin Haibe-Kains is active.

Publication


Featured researches published by Benjamin Haibe-Kains.


Clinical Cancer Research | 2007

Strong Time Dependence of the 76-Gene Prognostic Signature for Node-Negative Breast Cancer Patients in the TRANSBIG Multicenter Independent Validation Series

Christine Desmedt; Fanny Piette; Sherene Loi; Yixin Wang; Françoise Lallemand; Benjamin Haibe-Kains; Giuseppe Viale; Mauro Delorenzi; Yi Zhang; Mahasti Saghatchian d'Assignies; Jonas Bergh; Rosette Lidereau; P. Ellis; Adrian L. Harris; J.G.M. Klijn; John A. Foekens; Fatima Cardoso; Martine Piccart; Marc Buyse; Christos Sotiriou

Purpose: Recently, a 76-gene prognostic signature able to predict distant metastases in lymph node–negative (N−) breast cancer patients was reported. The aims of this study conducted by TRANSBIG were to independently validate these results and to compare the outcome with clinical risk assessment. Experimental Design: Gene expression profiling of frozen samples from 198 N− systemically untreated patients was done at the Bordet Institute, blinded to clinical data and independent of Veridex. Genomic risk was defined by Veridex, blinded to clinical data. Survival analyses, done by an independent statistician, were done with the genomic risk and adjusted for the clinical risk, defined by Adjuvant! Online. Results: The actual 5- and 10-year time to distant metastasis were 98% (88-100%) and 94% (83-98%), respectively, for the good profile group and 76% (68-82%) and 73% (65-79%), respectively, for the poor profile group. The actual 5- and 10-year overall survival were 98% (88-100%) and 87% (73-94%), respectively, for the good profile group and 84% (77-89%) and 72% (63-78%), respectively, for the poor profile group. We observed a strong time dependence of this signature, leading to an adjusted hazard ratio of 13.58 (1.85-99.63) and 8.20 (1.10-60.90) at 5 years and 5.11 (1.57-16.67) and 2.55 (1.07-6.10) at 10 years for time to distant metastasis and overall survival, respectively. Conclusion: This independent validation confirmed the performance of the 76-gene signature and adds to the growing evidence that gene expression signatures are of clinical relevance, especially for identifying patients at high risk of early distant metastases.


Nature Communications | 2014

Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach.

Hugo J.W.L. Aerts; Emmanuel Rios Velazquez; R. Leijenaar; Chintan Parmar; Patrick Grossmann; S. Carvalho; Sara Cavalho; Johan Bussink; René Monshouwer; Benjamin Haibe-Kains; D. Rietveld; Frank Hoebers; C. René Leemans; Andre Dekker; John Quackenbush; Robert J. Gillies; Philippe Lambin

Human cancers exhibit strong phenotypic differences that can be visualized noninvasively by medical imaging. Radiomics refers to the comprehensive quantification of tumour phenotypes by applying a large number of quantitative image features. Here we present a radiomic analysis of 440 features quantifying tumour image intensity, shape and texture, which are extracted from computed tomography data of 1,019 patients with lung or head-and-neck cancer. We find that a large number of radiomic features have prognostic power in independent data sets of lung and head-and-neck cancer patients, many of which were not identified as significant before. Radiogenomics analysis reveals that a prognostic radiomic signature, capturing intratumour heterogeneity, is associated with underlying gene-expression patterns. These data suggest that radiomics identifies a general prognostic phenotype existing in both lung and head-and-neck cancer. This may have a clinical impact as imaging is routinely used in clinical practice, providing an unprecedented opportunity to improve decision-support in cancer treatment at low cost.


Journal of Clinical Oncology | 2007

Definition of Clinically Distinct Molecular Subtypes in Estrogen Receptor–Positive Breast Carcinomas Through Genomic Grade

Sherene Loi; Benjamin Haibe-Kains; Christine Desmedt; Françoise Lallemand; Andrew Tutt; Cheryl Gillet; Paul Ellis; Adrian L. Harris; Jonas Bergh; John A. Foekens; J.G.M. Klijn; Denis Larsimont; Marc Buyse; Gianluca Bontempi; Mauro Delorenzi; Martine Piccart; Christos Sotiriou

PURPOSE A number of microarray studies have reported distinct molecular profiles of breast cancers (BC), such as basal-like, ErbB2-like, and two to three luminal-like subtypes. These were associated with different clinical outcomes. However, although the basal and the ErbB2 subtypes are repeatedly recognized, identification of estrogen receptor (ER) -positive subtypes has been inconsistent. Therefore, refinement of their molecular definition is needed. MATERIALS AND METHODS We have previously reported a gene expression grade index (GGI), which defines histologic grade based on gene expression profiles. Using this algorithm, we assigned ER-positive BC to either high-or low-genomic grade subgroups and compared these with previously reported ER-positive molecular classifications. As further validation, we classified 666 ER-positive samples into subtypes and assessed their clinical outcome. RESULTS Two ER-positive molecular subgroups (high and low genomic grade) could be defined using the GGI. Despite tracking a single biologic pathway, these were highly comparable to the previously described luminal A and B classification and significantly correlated to the risk groups produced using the 21-gene recurrence score. The two subtypes were associated with statistically distinct clinical outcome in both systemically untreated and tamoxifen-treated populations. CONCLUSION The use of genomic grade can identify two clinically distinct ER-positive molecular subtypes in a simple and highly reproducible manner across multiple data sets. This study emphasizes the important role of proliferation-related genes in predicting prognosis in ER-positive BC.


Clinical Cancer Research | 2008

Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes.

Christine Desmedt; Benjamin Haibe-Kains; Pratyaksha Wirapati; Marc Buyse; Denis Larsimont; Gianluca Bontempi; Mauro Delorenzi; Martine Piccart; Christos Sotiriou

Purpose: Recently, several prognostic gene expression signatures have been identified; however, their performance has never been evaluated according to the previously described molecular subtypes based on the estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2), and their biological meaning has remained unclear. Here we aimed to perform a comprehensive meta-analysis integrating both clinicopathologic and gene expression data, focusing on the main molecular subtypes. Experimental Design: We developed gene expression modules related to key biological processes in breast cancer such as tumor invasion, immune response, angiogenesis, apoptosis, proliferation, and ER and HER2 signaling, and then analyzed these modules together with clinical variables and several prognostic signatures on publicly available microarray studies (>2,100 patients). Results: Multivariate analysis showed that in the ER+/HER2− subgroup, only the proliferation module and the histologic grade were significantly associated with clinical outcome. In the ER−/HER2− subgroup, only the immune response module was associated with prognosis, whereas in the HER2+ tumors, the tumor invasion and immune response modules displayed significant association with survival. Proliferation was identified as the most important component of several prognostic signatures, and their performance was limited to the ER+/HER2− subgroup. Conclusions: Although proliferation is the strongest parameter predicting clinical outcome in the ER+/HER2− subtype and the common denominator of most prognostic gene signatures, immune response and tumor invasion seem to be the main molecular processes associated with prognosis in the ER−/HER2− and HER2+ subgroups, respectively. These findings may help to define new clinicogenomic models and to identify new therapeutic strategies in the specific molecular subgroups.


Journal of Clinical Investigation | 2013

CD4+ follicular helper T cell infiltration predicts breast cancer survival

Chunyan Gu-Trantien; Sherene Loi; Soizic Garaud; Carole Equeter; Myriam Libin; Alexandre de Wind; Marie Ravoet; Hélène Le Buanec; Catherine Sibille; Germain Manfouo-Foutsop; Isabelle Veys; Benjamin Haibe-Kains; Sandeep Singhal; Stefan Michiels; Françoise Rothé; Roberto Salgado; Hugues Duvillier; Michail Ignatiadis; Christine Desmedt; Dominique Bron; Denis Larsimont; Martine Piccart; Christos Sotiriou; Karen Willard-Gallo

CD4⁺ T cells are critical regulators of immune responses, but their functional role in human breast cancer is relatively unknown. The goal of this study was to produce an image of CD4⁺ T cells infiltrating breast tumors using limited ex vivo manipulation to better understand the in vivo differences associated with patient prognosis. We performed comprehensive molecular profiling of infiltrating CD4⁺ T cells isolated from untreated invasive primary tumors and found that the infiltrating T cell subpopulations included follicular helper T (Tfh) cells, which have not previously been found in solid tumors, as well as Th1, Th2, and Th17 effector memory cells and Tregs. T cell signaling pathway alterations included a mixture of activation and suppression characterized by restricted cytokine/chemokine production, which inversely paralleled lymphoid infiltration levels and could be reproduced in activated donor CD4⁺ T cells treated with primary tumor supernatant. A comparison of extensively versus minimally infiltrated tumors showed that CXCL13-producing CD4⁺ Tfh cells distinguish extensive immune infiltrates, principally located in tertiary lymphoid structure germinal centers. An 8-gene Tfh signature, signifying organized antitumor immunity, robustly predicted survival or preoperative response to chemotherapy. Our identification of CD4⁺ Tfh cells in breast cancer suggests that they are an important immune element whose presence in the tumor is a prognostic factor.


BMC Genomics | 2008

Predicting prognosis using molecular profiling in estrogen receptor-positive breast cancer treated with tamoxifen

Sherene Loi; Benjamin Haibe-Kains; Christine Desmedt; Pratyaksha Wirapati; Françoise Lallemand; Andrew Tutt; Cheryl Gillet; Paul Ellis; K Ryder; James F. Reid; Maria Grazia Daidone; Marco A. Pierotti; Els M. J. J. Berns; Maurice P.H.M. Jansen; John A. Foekens; Mauro Delorenzi; Gianluca Bontempi; Martine Piccart; Christos Sotiriou

BackgroundEstrogen receptor positive (ER+) breast cancers (BC) are heterogeneous with regard to their clinical behavior and response to therapies. The ER is currently the best predictor of response to the anti-estrogen agent tamoxifen, yet up to 30–40% of ER+BC will relapse despite tamoxifen treatment. New prognostic biomarkers and further biological understanding of tamoxifen resistance are required. We used gene expression profiling to develop an outcome-based predictor using a training set of 255 ER+ BC samples from women treated with adjuvant tamoxifen monotherapy. We used clusters of highly correlated genes to develop our predictor to facilitate both signature stability and biological interpretation. Independent validation was performed using 362 tamoxifen-treated ER+ BC samples obtained from multiple institutions and treated with tamoxifen only in the adjuvant and metastatic settings.ResultsWe developed a gene classifier consisting of 181 genes belonging to 13 biological clusters. In the independent set of adjuvantly-treated samples, it was able to define two distinct prognostic groups (HR 2.01 95%CI: 1.29–3.13; p = 0.002). Six of the 13 gene clusters represented pathways involved in cell cycle and proliferation. In 112 metastatic breast cancer patients treated with tamoxifen, one of the classifier components suggesting a cellular inflammatory mechanism was significantly predictive of response.ConclusionWe have developed a gene classifier that can predict clinical outcome in tamoxifen-treated ER+ BC patients. Whilst our study emphasizes the important role of proliferation genes in prognosis, our approach proposes other genes and pathways that may elucidate further mechanisms that influence clinical outcome and prediction of response to tamoxifen.


Proceedings of the National Academy of Sciences of the United States of America | 2010

PIK3CA mutations associated with gene signature of low mTORC1 signaling and better outcomes in estrogen receptor–positive breast cancer

Sherene Loi; Benjamin Haibe-Kains; Samira Majjaj; Françoise Lallemand; Virginie Durbecq; Denis Larsimont; Ana M. Gonzalez-Angulo; Lajos Pusztai; W. Fraser Symmans; Alberto Bardelli; Paul Ellis; Andrew Tutt; Cheryl Gillett; Bryan T. Hennessy; Gordon B. Mills; Wayne A. Phillips; Martine Piccart; Terence P. Speed; Grant A. McArthur; Christos Sotiriou

PIK3CA mutations are reported to be present in approximately 25% of breast cancer (BC), particularly the estrogen receptor–positive (ER+) and HER2-overexpressing (HER2+) subtypes, making them one of the most common genetic aberrations in BC. In experimental models, these mutations have been shown to activate AKT and induce oncogenic transformation, and hence these lesions have been hypothesized to render tumors highly sensitive to therapeutic PI3K/mTOR inhibition. By analyzing gene expression and protein data from nearly 1,800 human BCs, we report that a PIK3CA mutation–associated gene signature (PIK3CA-GS) derived from exon 20 (kinase domain) mutations was able to predict PIK3CA mutation status in two independent datasets, strongly suggesting a characteristic set of gene expression–induced changes. However, in ER+/HER2− BC despite pathway activation, PIK3CA mutations were associated with a phenotype of relatively low mTORC1 signaling and a good prognosis with tamoxifen monotherapy. The relationship between clinical outcome and the PIK3CA-GS was also assessed. Although the PIK3CA-GS was not associated with prognosis in ER− and HER2+ BC, it could identify better clinical outcomes in ER+/HER2− disease. In ER+ BC cell lines, PIK3CA mutations were also associated with sensitivity to tamoxifen. These findings could have important implications for the treatment of PIK3CA-mutant BCs and the development of PI3K/mTOR inhibitors.


Nature Methods | 2014

Similarity network fusion for aggregating data types on a genomic scale

Bo Wang; Aziz M. Mezlini; Feyyaz Demir; Marc Fiume; Zhuowen Tu; Michael Brudno; Benjamin Haibe-Kains; Anna Goldenberg

Recent technologies have made it cost-effective to collect diverse types of genome-wide data. Computational methods are needed to combine these data to create a comprehensive view of a given disease or a biological process. Similarity network fusion (SNF) solves this problem by constructing networks of samples (e.g., patients) for each available data type and then efficiently fusing these into one network that represents the full spectrum of underlying data. For example, to create a comprehensive view of a disease given a cohort of patients, SNF computes and fuses patient similarity networks obtained from each of their data types separately, taking advantage of the complementarity in the data. We used SNF to combine mRNA expression, DNA methylation and microRNA (miRNA) expression data for five cancer data sets. SNF substantially outperforms single data type analysis and established integrative approaches when identifying cancer subtypes and is effective for predicting survival.


Nature | 2013

Inconsistency in large pharmacogenomic studies

Benjamin Haibe-Kains; Nehme El-Hachem; Nicolai Juul Birkbak; Andrew C. Jin; Andrew H. Beck; Hugo J.W.L. Aerts; John Quackenbush

Two large-scale pharmacogenomic studies were published recently in this journal. Genomic data are well correlated between studies; however, the measured drug response data are highly discordant. Although the source of inconsistencies remains uncertain, it has potential implications for using these outcome measures to assess gene–drug associations or select potential anticancer drugs on the basis of their reported results.


Nature Medicine | 2010

Amplification of LAPTM4B and YWHAZ contributes to chemotherapy resistance and recurrence of breast cancer

Yang Li; Lihua Zou; Qiyuan Li; Benjamin Haibe-Kains; Ruiyang Tian; Yan-Yan Li; Christine Desmedt; Christos Sotiriou; Zoltan Szallasi; J. Dirk Iglehart; Andrea L. Richardson; Zhigang C. Wang

Adjuvant chemotherapy for breast cancer after surgery has effectively lowered metastatic recurrence rates. However, a considerable proportion of women suffer recurrent cancer at distant metastatic sites despite adjuvant treatment. Identification of the genes crucial for tumor response to specific chemotherapy drugs is a challenge but is necessary to improve outcomes. By using integrated genomics, we identified a small number of overexpressed and amplified genes from chromosome 8q22 that were associated with early disease recurrence despite anthracycline-based adjuvant chemotherapy. We confirmed the association in an analysis of multiple independent cohorts. SiRNA-mediated knockdown of either of two of these genes, the antiapoptotic gene YWHAZ and a lysosomal gene LAPTM4B, sensitized tumor cells to anthracyclines, and overexpression of either of the genes induced anthracycline resistance. Overexpression of LAPTM4B resulted in sequestration of the anthracycline doxorubicin, delaying its appearance in the nucleus. Overexpression of these two genes was associated with poor tumor response to anthracycline treatment in a neoadjuvant chemotherapy trial in women with primary breast cancer. Our results suggest that 8q22 amplification and overexpression of LAPTM4B and YWHAZ contribute to de novo chemoresistance to anthracyclines and are permissive for metastatic recurrence. Overexpression of these two genes may predict anthracycline resistance and influence selection of chemotherapy.

Collaboration


Dive into the Benjamin Haibe-Kains's collaboration.

Top Co-Authors

Avatar

Christos Sotiriou

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Christine Desmedt

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Gianluca Bontempi

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Sherene Loi

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Martine Piccart

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Denis Larsimont

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martine Piccart-Gebhart

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Françoise Lallemand

Université libre de Bruxelles

View shared research outputs
Top Co-Authors

Avatar

Michail Ignatiadis

Université libre de Bruxelles

View shared research outputs
Researchain Logo
Decentralizing Knowledge