Network


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

Hotspot


Dive into the research topics where Rachael Natrajan is active.

Publication


Featured researches published by Rachael Natrajan.


Nature | 2009

Complex landscapes of somatic rearrangement in human breast cancer genomes.

Philip Stephens; David J. McBride; Meng-Lay Lin; Ignacio Varela; Erin Pleasance; Jared T. Simpson; Lucy Stebbings; Catherine Leroy; Sarah Edkins; Laura Mudie; Christopher Greenman; Mingming Jia; Calli Latimer; Jon Teague; King Wai Lau; John Burton; Michael A. Quail; Harold Swerdlow; Carol Churcher; Rachael Natrajan; Anieta M. Sieuwerts; John W.M. Martens; Daniel P. Silver; Anita Langerød; Hege G. Russnes; John A. Foekens; Jorge S. Reis-Filho; Laura J. van 't Veer; Andrea L. Richardson; Anne Lise Børresen-Dale

Multiple somatic rearrangements are often found in cancer genomes; however, the underlying processes of rearrangement and their contribution to cancer development are poorly characterized. Here we use a paired-end sequencing strategy to identify somatic rearrangements in breast cancer genomes. There are more rearrangements in some breast cancers than previously appreciated. Rearrangements are more frequent over gene footprints and most are intrachromosomal. Multiple rearrangement architectures are present, but tandem duplications are particularly common in some cancers, perhaps reflecting a specific defect in DNA maintenance. Short overlapping sequences at most rearrangement junctions indicate that these have been mediated by non-homologous end-joining DNA repair, although varying sequence patterns indicate that multiple processes of this type are operative. Several expressed in-frame fusion genes were identified but none was recurrent. The study provides a new perspective on cancer genomes, highlighting the diversity of somatic rearrangements and their potential contribution to cancer development.


Nature | 2008

Resistance to therapy caused by intragenic deletion in BRCA2.

Stacey L. Edwards; Rachel Brough; Christopher J. Lord; Rachael Natrajan; Radost Vatcheva; Douglas A. Levine; Jeff Boyd; Js Reis-Filho; Alan Ashworth

Cells with loss of BRCA2 function are defective in homologous recombination (HR) and are highly sensitive to inhibitors of poly(ADP-ribose) polymerase (PARP), which provides the basis for a new therapeutic approach. Here we show that resistance to PARP inhibition can be acquired by deletion of a mutation in BRCA2. We derived PARP-inhibitor-resistant (PIR) clones from the human CAPAN1 pancreatic cancer cell line, which carries the protein-truncating c.6174delT frameshift mutation. PIR clones could form DNA-damage-induced RAD51 nuclear foci and were able to limit genotoxin-induced genomic instability, both hallmarks of a competent HR pathway. New BRCA2 isoforms were expressed in the resistant lines as a result of intragenic deletion of the c.6174delT mutation and restoration of the open reading frame (ORF). Reconstitution of BRCA2-deficient cells with these revertant BRCA2 alleles rescued PARP inhibitor sensitivity and HR deficiency. Most of the deletions in BRCA2 were associated with small tracts of homology, and possibly arose from error-prone repair caused by BRCA2 deficiency. Similar ORF-restoring mutations were present in carboplatin-resistant ovarian tumours from c.6174delT mutation carriers. These observations have implications for understanding drug resistance in BRCA mutation carriers as well as in defining functionally important domains within BRCA2.


Cell Stem Cell | 2010

BRCA1 Basal-like Breast Cancers Originate from Luminal Epithelial Progenitors and Not from Basal Stem Cells

Gemma Molyneux; Felipe C. Geyer; Fiona-Ann Magnay; Afshan McCarthy; Howard Kendrick; Rachael Natrajan; Alan Mackay; Anita Grigoriadis; Andrew Tutt; Alan Ashworth; Js Reis-Filho; Matthew John Smalley

Breast cancers in BRCA1 mutation carriers frequently have a distinctive basal-like phenotype. It has been suggested that this results from an origin in basal breast epithelial stem cells. Here, we demonstrate that deleting Brca1 in mouse mammary epithelial luminal progenitors produces tumors that phenocopy human BRCA1 breast cancers. They also resemble the majority of sporadic basal-like breast tumors. However, directing Brca1 deficiency to basal cells generates tumors that express molecular markers of basal breast cancers but do not histologically resemble either human BRCA1 or the majority of sporadic basal-like breast tumors. These findings support a derivation of the majority of human BRCA1-associated and sporadic basal-like tumors from luminal progenitors rather than from basal stem cells. They also demonstrate that when target cells for transformation have the potential for phenotypic plasticity, tumor phenotypes may not directly reflect histogenesis. This has important implications for cancer prevention strategies.


Cancer Research | 2010

FGFR1 Amplification Drives Endocrine Therapy Resistance and Is a Therapeutic Target in Breast Cancer

Nicholas C. Turner; Alex Pearson; Rachel Sharpe; Maryou B. Lambros; Felipe C. Geyer; Maria A Lopez-Garcia; Rachael Natrajan; Caterina Marchiò; Elizabeth Iorns; Alan Mackay; Cheryl Gillett; Anita Grigoriadis; Andrew Tutt; Jorge S. Reis-Filho; Alan Ashworth

Amplification of fibroblast growth factor receptor 1 (FGFR1) occurs in approximately 10% of breast cancers and is associated with poor prognosis. However, it is uncertain whether overexpression of FGFR1 is causally linked to the poor prognosis of amplified cancers. Here, we show that FGFR1 overexpression is robustly associated with FGFR1 amplification in two independent series of breast cancers. Breast cancer cell lines with FGFR1 overexpression and amplification show enhanced ligand-dependent signaling, with increased activation of the mitogen-activated protein kinase and phosphoinositide 3-kinase-AKT signaling pathways in response to FGF2, but also show basal ligand-independent signaling, and are dependent on FGFR signaling for anchorage-independent growth. FGFR1-amplified cell lines show resistance to 4-hydroxytamoxifen, which is reversed by small interfering RNA silencing of FGFR1, suggesting that FGFR1 overexpression also promotes endocrine therapy resistance. FGFR1 signaling suppresses progesterone receptor (PR) expression in vitro, and likewise, amplified cancers are frequently PR negative, identifying a potential biomarker for FGFR1 activity. Furthermore, we show that amplified cancers have a high proliferative rate assessed by Ki67 staining and that FGFR1 amplification is found in 16% to 27% of luminal B-type breast cancers. Our data suggest that amplification and overexpression of FGFR1 may be a major contributor to poor prognosis in luminal-type breast cancers, driving anchorage-independent proliferation and endocrine therapy resistance.


Lancet Oncology | 2010

Breast cancer molecular profiling with single sample predictors: a retrospective analysis

Britta Weigelt; Alan Mackay; Roger A'Hern; Rachael Natrajan; David Sp Tan; Mitch Dowsett; Alan Ashworth; Jorge S. Reis-Filho

BACKGROUND Microarray expression profiling classifies breast cancer into five molecular subtypes: luminal A, luminal B, basal-like, HER2, and normal breast-like. Three microarray-based single sample predictors (SSPs) have been used to define molecular classification of individual samples. We aimed to establish agreement between these SSPs for identification of breast cancer molecular subtypes. METHODS Previously described microarray-based SSPs were applied to one in-house (n=53) and three publicly available (n=779) breast cancer datasets. Agreement was analysed between SSPs for the whole classification system and for the five molecular subtypes individually in each cohort. FINDINGS Fair-to-substantial agreement between every pair of SSPs in each cohort was recorded (kappa=0.238-0.740). Of the five molecular subtypes, only basal-like cancers consistently showed almost-perfect agreement (kappa>0.812). The proportion of cases classified as basal-like in each cohort was consistent irrespective of the SSP used; however, the proportion of each remaining molecular subtype varied substantially. Assignment of individual cases to luminal A, luminal B, HER2, and normal breast-like subtypes was dependent on the SSP used. The significance of associations with outcome of each molecular subtype, other than basal-like and luminal A, varied depending on SSP used. However, different SSPs produced broadly similar survival curves. INTERPRETATION Although every SSP identifies molecular subtypes with similar survival, they do not reliably assign the same patients to the same molecular subtypes. For molecular subtype classification to be incorporated into routine clinical practice and treatment decision making, stringent standardisation of methodologies and definitions for identification of breast cancer molecular subtypes is needed. FUNDING Breakthrough Breast Cancer, Cancer Research UK.


Nature Medicine | 2011

Functionally recurrent rearrangements of the MAST kinase and Notch gene families in breast cancer.

Dan R. Robinson; Shanker Kalyana-Sundaram; Yi Mi Wu; Sunita Shankar; Xuhong Cao; Bushra Ateeq; Irfan A. Asangani; Matthew K. Iyer; Christopher A. Maher; Catherine S. Grasso; Robert J. Lonigro; Michael J. Quist; Javed Siddiqui; Rohit Mehra; Xiaojun Jing; Thomas J. Giordano; Michael S. Sabel; Celina G. Kleer; Nallasivam Palanisamy; Rachael Natrajan; Maryou B. Lambros; Jorge S. Reis-Filho; Chandan Kumar-Sinha; Arul M. Chinnaiyan

Breast cancer is a heterogeneous disease that has a wide range of molecular aberrations and clinical outcomes. Here we used paired-end transcriptome sequencing to explore the landscape of gene fusions in a panel of breast cancer cell lines and tissues. We observed that individual breast cancers have a variety of expressed gene fusions. We identified two classes of recurrent gene rearrangements involving genes encoding microtubule-associated serine-threonine kinase (MAST) and members of the Notch family. Both MAST and Notch-family gene fusions have substantial phenotypic effects in breast epithelial cells. Breast cancer cell lines harboring Notch gene rearrangements are uniquely sensitive to inhibition of Notch signaling, and overexpression of MAST1 or MAST2 gene fusions has a proliferative effect both in vitro and in vivo. These findings show that recurrent gene rearrangements have key roles in subsets of carcinomas and suggest that transcriptome sequencing could identify individuals with rare, targetable gene fusions.


Modern Pathology | 2011

|[beta]|-Catenin pathway activation in breast cancer is associated with triple-negative phenotype but not with CTNNB1 mutation

Felipe C. Geyer; Magali Lacroix-Triki; Kay Savage; Monica Arnedos; Maryou B. Lambros; Alan Mackay; Rachael Natrajan; Jorge S. Reis-Filho

Aberrant β-catenin expression as determined by assessment of its subcellular localization constitutes a surrogate marker of Wnt signalling pathway activation and has been reported in a subset of breast cancers. The association of β-catenin/Wnt pathway activation with clinical outcome and the mechanisms leading to its activation in breast cancers still remain a matter of controversy. The aims of this study were to address the distribution of β-catenin expression in invasive breast cancers, the correlations between β-catenin expression and clinicopathological features and survival of breast cancer patients, and to determine whether aberrant β-catenin expression is driven by CTNNB1 (β-catenin encoding gene) activating mutations. Immunohistochemistry was performed on a tissue microarray containing 245 invasive breast carcinomas from uniformly treated patients, using two anti-β-catenin monoclonal antibodies. Selected samples were subjected to CTNNB1 exon 3 mutation analysis by direct gene sequencing. A good correlation between the two β-catenin antibodies was observed (Spearmans r >0.62, P<0.001). Respectively, 31 and 11% of the cases displayed lack/reduction of β-catenin membranous expression and nuclear accumulation. Complete lack of β-catenin expression was significantly associated with invasive lobular carcinoma histological type. Subgroup analysis of non-lobular cancers or non-lobular grade 3 carcinomas revealed that lack/reduction of β-catenin membranous expression and/or nuclear accumulation were significantly associated with oestrogen receptor negativity, absence of HER2 gene amplification and overexpression, lack/reduction of E-cadherin expression and tumours of triple-negative and basal-like phenotype. Univariate survival analysis revealed a significant association between β-catenin nuclear expression and shorter metastasis-free and overall survival in the whole cohort; however, β-catenin nuclear expression was not an independent predictor of outcome in multivariate analysis. No CTNNB1 mutations were identified in the 28 selected breast carcinomas analysed. In conclusion, β-catenin/Wnt pathway activation is preferentially found in triple-negative/basal-like breast carcinomas, is associated with poor clinical outcome and is unlikely to be driven by CTNNB1 mutations in breast cancer.


The Journal of Pathology | 2010

The molecular underpinning of lobular histological growth pattern: a genome‐wide transcriptomic analysis of invasive lobular carcinomas and grade‐ and molecular subtype‐matched invasive ductal carcinomas of no special type

Britta Weigelt; Felipe C. Geyer; Rachael Natrajan; Maria A Lopez-Garcia; Amar Ahmad; Kay Savage; Bas Kreike; Jorge S. Reis-Filho

Invasive lobular carcinoma (ILC) is the most frequent special type of breast cancer. The majority of these tumours are of low histological grade, express hormone receptors, and lack HER2 expression. The pleomorphic variant of ILCs (PLCs) is characterized by atypical cells with pleomorphic nuclei and is reported to have an aggressive clinical behaviour. Expression profiling studies have demonstrated that classic ILCs preferentially display a luminal phenotype, whereas PLCs may be of luminal, HER2 or molecular apocrine subtypes. The aims of this study were two‐fold: to determine the transcriptomic characteristics of lobular carcinomas and to define the genome‐wide transcriptomic differences between classic ILCs and PLCs. To define the transcriptomic characteristics of ILCs, minimizing the impact of histological grade and molecular subtype on the analysis, we subjected a series of grade‐ and molecular subtype‐matched ILCs and invasive ductal carcinomas (IDCs) to genome‐wide gene expression profiling using oligonucleotide microarrays. Hierarchical clustering analysis demonstrated that ILCs formed a separate cluster and a supervised analysis revealed that 5.8% of the transcriptionally regulated genes were significantly differentially expressed in ILCs compared to grade‐ and molecular subtype‐matched IDCs. ILCs displayed down‐regulation of E‐cadherin and of genes related to actin cytoskeleton remodelling, protein ubiquitin, DNA repair, cell adhesion, TGF‐beta signalling; and up‐regulation of transcription factors/immediate early genes, lipid/prostaglandin biosynthesis genes, and cell migration‐associated genes. Supervised analysis of classic ILCs and PLCs demonstrated that less than 0.1% of genes were significantly differentially expressed between these tumour subtypes. Our results demonstrate that ILCs differ from grade‐ and molecular subtype‐matched IDCs in the expression of genes related to cell adhesion, cell‐to‐cell signalling, and actin cytoskeleton signalling. However, classic ILCs and PLCs are remarkably similar at the molecular level and should be considered as part of a spectrum of lesions. 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 | 2010

PTEN Deficiency in Endometrioid Endometrial Adenocarcinomas Predicts Sensitivity to PARP Inhibitors

Konstantin J. Dedes; Daniel Wetterskog; Ana M. Mendes-Pereira; Rachael Natrajan; Maryou B. Lambros; Felipe C. Geyer; Radost Vatcheva; Kay Savage; Alan Mackay; Christopher J. Lord; Alan Ashworth; Jorge S. Reis-Filho

Endometrioid endometrial cancer cell lines harboring PTEN loss of function have impaired homologous recombination response to DNA double-strand breaks and show an exquisite sensitivity to PARP inhibition. Putting PARP Inhibitors on the Map Targeted drug therapies that interfere with specific oncoproteins in certain cancers like lung cancer and chronic myeloid leukemia have shown great promise, but designing targeted therapies for tumors that have lost a tumor suppressor protein has proved challenging. Building on the idea of targeting a complementary gene or pathway in tumors with an inactive tumor suppressor protein, Dedes and colleagues set out to tackle endometrial cancer. This cancer is the fourth most common malignancy in women and is very difficult to treat particularly in the later stages of the disease. The authors demonstrate that endometrial cancer cell lines that lack the tumor suppressor protein PTEN show defects in the repair of DNA damage and are consequently very sensitive to drugs that block poly(ADP) ribose polymerase (PARP), an enzyme critical for DNA repair. Given that such PARP inhibitors are currently in late-stage clinical trials for treating breast and ovarian cancer, this study opens the door to using PARP inhibitors for treating endometrial cancer. First the authors demonstrated that the majority of endometrial cancer cell lines that they analyzed indeed had lost the PTEN tumor suppressor protein. PTEN both regulates a major growth signaling pathway in cells (the PI3K-AKT-mTOR pathway) and has recently been shown to be important for maintaining genomic stability. Dedes and co-workers then showed that loss of PTEN rendered endometrial cancer cells unable to repair DNA double-strand breaks induced by ionizing radiation. Other types of tumors that cannot repair DNA damage, such as breast and ovarian cancer cells carrying mutations in the BRCA1 and BRCA2 genes, are acutely sensitive to PARP inhibitors. Dedes et al. reasoned that endometrial cancer cells lacking PTEN may also be sensitive to these drugs. This is exactly what they found when they treated their endometrial cancer cell lines with the potent PARP inhibitor, KU0058948. To demonstrate that it was loss of PTEN that rendered the cancer cells highly sensitive to the drug, they re-expressed PTEN in endometrial cancer cell lines lacking this tumor suppressor protein and demonstrated that these cancer cell lines were now able to repair DNA damage and thus were resistant to treatment with the PARP inhibitor. Given that 80% of endometrial cancers lack PTEN, treatment with PARP inhibitors may be an effective way to treat this disease. PTEN (phosphatase and tensin homolog) loss of function is the most common genetic aberration in endometrioid endometrial carcinomas. In addition to its well-described role in cell signaling, PTEN is involved in the maintenance of genomic stability. Loss of PTEN function causes defects in repair of DNA double-strand breaks by homologous recombination and, therefore, sensitizes cells to inhibition of the poly(adenosine diphosphate ribose) polymerase (PARP). Here, we determined the PTEN status of eight endometrioid endometrial carcinoma cell lines and correlated it with in vitro sensitivity to the PARP inhibitor KU0058948. PTEN-deficient cells showed a significantly greater sensitivity to KU0058948 than the two endometrioid endometrial carcinoma cell lines with wild-type PTEN. The cell lines lacking PTEN expression were unable to elicit a homologous recombination damage response as assayed by RAD51 focus function (a marker of competent homologous recombination DNA repair) upon irradiation and treatment with PARP inhibitors. PTEN silencing in PTEN wild-type Hec-1b cells resulted in reduced RAD51 foci formation after DNA damage and increased sensitivity to PARP inhibition. PTEN reexpression in PTEN-null cell lines resulted in enhanced RAD51 foci formation and in relative resistance to KU0058948. Given that up to 80% of endometrioid endometrial cancers lack PTEN expression, our results suggest that PARP inhibitors may be therapeutically useful for a subset of endometrioid endometrial cancers.

Collaboration


Dive into the Rachael Natrajan's collaboration.

Top Co-Authors

Avatar

Jorge S. Reis-Filho

Memorial Sloan Kettering Cancer Center

View shared research outputs
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

Maryou B. Lambros

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

Britta Weigelt

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Felipe C. Geyer

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Js Reis-Filho

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Kerry Fenwick

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge