Network


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

Hotspot


Dive into the research topics where Ilirjana Bajrami is active.

Publication


Featured researches published by Ilirjana Bajrami.


Clinical Cancer Research | 2013

BMN 673, a Novel and Highly Potent PARP1/2 Inhibitor for the Treatment of Human Cancers with DNA Repair Deficiency

Yuqiao Shen; Farah L. Rehman; Ying Feng; Julia Boshuizen; Ilirjana Bajrami; Richard Elliott; Bing Wang; Christopher J. Lord; Leonard Post; Alan Ashworth

Purpose: PARP1/2 inhibitors are a class of anticancer agents that target tumor-specific defects in DNA repair. Here, we describe BMN 673, a novel, highly potent PARP1/2 inhibitor with favorable metabolic stability, oral bioavailability, and pharmacokinetic properties. Experimental Design: Potency and selectivity of BMN 673 was determined by biochemical assays. Anticancer activity either as a single-agent or in combination with other antitumor agents was evaluated both in vitro and in xenograft cancer models. Results: BMN 673 is a potent PARP1/2 inhibitor (PARP1 IC50 = 0.57 nmol/L), but it does not inhibit other enzymes that we have tested. BMN 673 exhibits selective antitumor cytotoxicity and elicits DNA repair biomarkers at much lower concentrations than earlier generation PARP1/2 inhibitors (such as olaparib, rucaparib, and veliparib). In vitro, BMN 673 selectively targeted tumor cells with BRCA1, BRCA2, or PTEN gene defects with 20- to more than 200-fold greater potency than existing PARP1/2 inhibitors. BMN 673 is readily orally bioavailable, with more than 40% absolute oral bioavailability in rats when dosed in carboxylmethyl cellulose. Oral administration of BMN 673 elicited remarkable antitumor activity in vivo; xenografted tumors that carry defects in DNA repair due to BRCA mutations or PTEN deficiency were profoundly sensitive to oral BMN 673 treatment at well-tolerated doses in mice. Synergistic or additive antitumor effects were also found when BMN 673 was combined with temozolomide, SN38, or platinum drugs. Conclusion: BMN 673 is currently in early-phase clinical development and represents a promising PARP1/2 inhibitor with potentially advantageous features in its drug class. Clin Cancer Res; 19(18); 5003–15. ©2013 AACR.


Nature | 2012

Mosaic PPM1D mutations are associated with predisposition to breast and ovarian cancer

Elise Ruark; Katie Snape; Peter Humburg; Chey Loveday; Ilirjana Bajrami; Rachel Brough; Daniel Nava Rodrigues; Anthony Renwick; Sheila Seal; Emma Ramsay; Silvana Del Vecchio Duarte; Manuel A. Rivas; Margaret Warren-Perry; Anna Zachariou; Adriana Campion-Flora; Sandra Hanks; Anne Murray; Naser Ansari Pour; Jenny Douglas; Lorna Gregory; Andrew J. Rimmer; Neil Walker; Tsun-Po Yang; Julian Adlard; Julian Barwell; Jonathan Berg; Angela F. Brady; Carole Brewer; G Brice; Cyril Chapman

Improved sequencing technologies offer unprecedented opportunities for investigating the role of rare genetic variation in common disease. However, there are considerable challenges with respect to study design, data analysis and replication. Using pooled next-generation sequencing of 507 genes implicated in the repair of DNA in 1,150 samples, an analytical strategy focused on protein-truncating variants (PTVs) and a large-scale sequencing case–control replication experiment in 13,642 individuals, here we show that rare PTVs in the p53-inducible protein phosphatase PPM1D are associated with predisposition to breast cancer and ovarian cancer. PPM1D PTV mutations were present in 25 out of 7,781 cases versus 1 out of 5,861 controls (P = 1.12 × 10−5), including 18 mutations in 6,912 individuals with breast cancer (P = 2.42 × 10−4) and 12 mutations in 1,121 individuals with ovarian cancer (P = 3.10 × 10−9). Notably, all of the identified PPM1D PTVs were mosaic in lymphocyte DNA and clustered within a 370-base-pair region in the final exon of the gene, carboxy-terminal to the phosphatase catalytic domain. Functional studies demonstrate that the mutations result in enhanced suppression of p53 in response to ionizing radiation exposure, suggesting that the mutant alleles encode hyperactive PPM1D isoforms. Thus, although the mutations cause premature protein truncation, they do not result in the simple loss-of-function effect typically associated with this class of variant, but instead probably have a gain-of-function effect. Our results have implications for the detection and management of breast and ovarian cancer risk. More generally, these data provide new insights into the role of rare and of mosaic genetic variants in common conditions, and the use of sequencing in their identification.


Cancer Discovery | 2013

Histone H3.3 Mutations Drive Pediatric Glioblastoma through Upregulation of MYCN

Lynn Bjerke; Alan Mackay; Meera Nandhabalan; Anna Burford; Alexa Jury; Sergey Popov; Diana Carvalho; Kathryn R. Taylor; Maria Vinci; Ilirjana Bajrami; Imelda M. McGonnell; Christopher J. Lord; Rui M. Reis; Darren Hargrave; Alan Ashworth; Paul Workman; Chris Jones

UNLABELLED Children and young adults with glioblastoma (GBM) have a median survival rate of only 12 to 15 months, and these GBMs are clinically and biologically distinct from histologically similar cancers in older adults. They are defined by highly specific mutations in the gene encoding the histone H3.3 variant H3F3A , occurring either at or close to key residues marked by methylation for regulation of transcription—K27 and G34. Here, we show that the cerebral hemisphere-specific G34 mutation drives a distinct expression signature through differential genomic binding of the K36 trimethylation mark (H3K36me3). The transcriptional program induced recapitulates that of the developing forebrain, and involves numerous markers of stem-cell maintenance, cell-fate decisions, and self-renewal.Critically, H3F3A G34 mutations cause profound upregulation of MYCN , a potent oncogene that is causative of GBMs when expressed in the correct developmental context. This driving aberration is selectively targetable in this patient population through inhibiting kinases responsible for stabilization of the protein. SIGNIFICANCE We provide the mechanistic explanation for how the fi rst histone gene mutation inhuman disease biology acts to deliver MYCN, a potent tumorigenic initiator, into a stem-cell compartment of the developing forebrain, selectively giving rise to incurable cerebral hemispheric GBM. Using synthetic lethal approaches to these mutant tumor cells provides a rational way to develop novel and highly selective treatment strategies


Cancer Discovery | 2011

Functional viability profiles of breast cancer.

Rachel Brough; Jessica Frankum; David Sims; Alan Mackay; Ana M. Mendes-Pereira; Ilirjana Bajrami; Sara Costa-Cabral; Rumana Rafiq; Amar Ahmad; Maria Antonietta Cerone; Rachael Natrajan; Rachel Sharpe; Kai-Keen Shiu; Daniel Wetterskog; Konstantine J. Dedes; Maryou B. Lambros; Teeara Rawjee; Spiros Linardopoulos; Jorge S. Reis-Filho; Nicholas C. Turner; Christopher J. Lord; Alan Ashworth

UNLABELLED The design of targeted therapeutic strategies for cancer has largely been driven by the identification of tumor-specific genetic changes. However, the large number of genetic alterations present in tumor cells means that it is difficult to discriminate between genes that are critical for maintaining the disease state and those that are merely coincidental. Even when critical genes can be identified, directly targeting these is often challenging, meaning that alternative strategies such as exploiting synthetic lethality may be beneficial. To address these issues, we have carried out a functional genetic screen in >30 commonly used models of breast cancer to identify genes critical to the growth of specific breast cancer subtypes. In particular, we describe potential new therapeutic targets for PTEN-mutated cancers and for estrogen receptor-positive breast cancers. We also show that large-scale functional profiling allows the classification of breast cancers into subgroups distinct from established subtypes. SIGNIFICANCE Despite the wealth of molecular profiling data that describe breast tumors and breast tumor cell models, our understanding of the fundamental genetic dependencies in this disease is relatively poor. Using high-throughput RNA interference screening of a series of pharmacologically tractable genes, we have generated comprehensive functional viability profiles for a wide panel of commonly used breast tumor cell models. Analysis of these profiles identifies a series of novel genetic dependencies, including that of PTEN-null breast tumor cells upon mitotic checkpoint kinases, and provides a framework upon which additional dependencies and candidate therapeutic targets may be identified.


PLOS ONE | 2013

A Genetic Screen Using the PiggyBac Transposon in Haploid Cells Identifies Parp1 as a Mediator of Olaparib Toxicity

Stephen J. Pettitt; Farah L. Rehman; Ilirjana Bajrami; Rachel Brough; Fredrik Wallberg; Iwanka Kozarewa; Kerry Fenwick; Ioannis Assiotis; Lina Chen; James J. Campbell; Christopher J. Lord; Alan Ashworth

Genetic perturbation screens have the potential to dissect a wide range of cellular phenotypes. Such screens have historically been difficult in diploid mammalian cells. The recent derivation of haploid embryonic stem cells provides an opportunity to cause loss of function mutants with a random mutagen in a mammalian cell with a normal genetic background. We describe an approach to genetic screens that exploits the highly active piggyBac transposon in haploid mammalian cells. As an example of haploid transposon (HTP) screening, we apply this approach to identifying determinants of cancer drug toxicity and resistance. In a screen for 6-thioguanine resistance we recovered components of the DNA mismatch repair pathway, a known requirement for toxicity. In a further screen for resistance to the clinical poly(ADP-ribose) polymerase (PARP) inhibitor olaparib we recovered multiple Parp1 mutants. Our results show that olaparib toxicity to normal cells is mediated predominantly via Parp1, and suggest that the clinical side effects of olaparib may be on target. The transposon mutant libraries are stable and can be readily reused to screen other drugs. The screening protocol described has several advantages over other methods such as RNA interference: it is rapid and low cost, and mutations can be easily reverted to establish causality.


The Journal of Pathology | 2014

Characterization of the genomic features and expressed fusion genes in micropapillary carcinomas of the breast

Rachael Natrajan; Paul M. Wilkerson; Caterina Marchiò; Salvatore Piscuoglio; Charlotte K.Y. Ng; Patty Wai; Maryou B. Lambros; Eleftherios P. Samartzis; Konstantin J. Dedes; Jessica Frankum; Ilirjana Bajrami; Alicja Kopec; Alan Mackay; Roger A'Hern; Kerry Fenwick; Iwanka Kozarewa; Jarle Hakas; Costas Mitsopoulos; David Hardisson; Christopher J. Lord; Chandan Kumar-Sinha; Alan Ashworth; Britta Weigelt; Anna Sapino; Arul M. Chinnaiyan; Christopher A. Maher; Jorge S. Reis-Filho

Micropapillary carcinoma (MPC) is a rare histological special type of breast cancer, characterized by an aggressive clinical behaviour and a pattern of copy number aberrations (CNAs) distinct from that of grade‐ and oestrogen receptor (ER)‐matched invasive carcinomas of no special type (IC‐NSTs). The aims of this study were to determine whether MPCs are underpinned by a recurrent fusion gene(s) or mutations in 273 genes recurrently mutated in breast cancer. Sixteen MPCs were subjected to microarray‐based comparative genomic hybridization (aCGH) analysis and Sequenom OncoCarta mutation analysis. Eight and five MPCs were subjected to targeted capture and RNA sequencing, respectively. aCGH analysis confirmed our previous observations about the repertoire of CNAs of MPCs. Sequencing analysis revealed a spectrum of mutations similar to those of luminal B IC‐NSTs, and recurrent mutations affecting mitogen‐activated protein kinase family genes and NBPF10. RNA‐sequencing analysis identified 17 high‐confidence fusion genes, eight of which were validated and two of which were in‐frame. No recurrent fusions were identified in an independent series of MPCs and IC‐NSTs. Forced expression of in‐frame fusion genes (SLC2A1–FAF1 and BCAS4–AURKA) resulted in increased viability of breast cancer cells. In addition, genomic disruption of CDK12 caused by out‐of‐frame rearrangements was found in one MPC and in 13% of HER2‐positive breast cancers, identified through a re‐analysis of publicly available massively parallel sequencing data. In vitro analyses revealed that CDK12 gene disruption results in sensitivity to PARP inhibition, and forced expression of wild‐type CDK12 in a CDK12‐null cell line model resulted in relative resistance to PARP inhibition. Our findings demonstrate that MPCs are neither defined by highly recurrent mutations in the 273 genes tested, nor underpinned by a recurrent fusion gene. Although seemingly private genetic events, some of the fusion transcripts found in MPCs may play a role in maintenance of a malignant phenotype and potentially offer therapeutic opportunities.


Embo Molecular Medicine | 2012

Synthetic lethality of PARP and NAMPT inhibition in triple-negative breast cancer cells

Ilirjana Bajrami; Asha Kigozi; Antoinette van Weverwijk; Rachel Brough; Jessica Frankum; Christopher J. Lord; Alan Ashworth

PARP inhibitors have been proposed as a potential targeted therapy for patients with triple‐negative (ER‐, PR‐, HER2‐negative) breast cancers. However, it is as yet unclear as to whether single agent or combination therapy using PARP inhibitors would be most beneficial. To better understand the mechanisms that determine the response to PARP inhibitors, we investigated whether enzymes involved in metabolism of the PARP substrate, β‐NAD+, might alter the response to a clinical PARP inhibitor. Using an olaparib sensitization screen in a triple‐negative (TN) breast cancer model, we identified nicotinamide phosphoribosyltransferase (NAMPT) as a non‐redundant modifier of olaparib response. NAMPT is a rate‐limiting enzyme involved in the generation of the PARP substrate β‐NAD+ and the suppression of β‐NAD+ levels by NAMPT inhibition most likely explains these observations. Importantly, the combination of a NAMPT small molecule inhibitor, FK866, with olaparib inhibited TN breast tumour growth in vivo to a greater extent than either single agent alone suggesting that assessing NAMPT/PARP inhibitor combinations for the treatment of TN breast cancer may be warranted.


Cell Reports | 2016

Large-Scale Profiling of Kinase Dependencies in Cancer Cell Lines

James J. Campbell; Colm J. Ryan; Rachel Brough; Ilirjana Bajrami; Helen N. Pemberton; Irene Y. Chong; Sara Costa-Cabral; Jessica Frankum; Aditi Gulati; Harriet Holme; Rowan Miller; Sophie Postel-Vinay; Rumana Rafiq; Wenbin Wei; Chris T. Williamson; David A. Quigley; Joe E. Tym; Bissan Al-Lazikani; Tim Fenton; Rachael Natrajan; Sandra J. Strauss; Alan Ashworth; Christopher J. Lord

Summary One approach to identifying cancer-specific vulnerabilities and therapeutic targets is to profile genetic dependencies in cancer cell lines. Here, we describe data from a series of siRNA screens that identify the kinase genetic dependencies in 117 cancer cell lines from ten cancer types. By integrating the siRNA screen data with molecular profiling data, including exome sequencing data, we show how vulnerabilities/genetic dependencies that are associated with mutations in specific cancer driver genes can be identified. By integrating additional data sets into this analysis, including protein-protein interaction data, we also demonstrate that the genetic dependencies associated with many cancer driver genes form dense connections on functional interaction networks. We demonstrate the utility of this resource by using it to predict the drug sensitivity of genetically or histologically defined subsets of tumor cell lines, including an increased sensitivity of osteosarcoma cell lines to FGFR inhibitors and SMAD4 mutant tumor cells to mitotic inhibitors.


DNA Repair | 2013

Identification of miRNA modulators to PARP inhibitor response

Sari Neijenhuis; Ilirjana Bajrami; Rowan Miller; Christopher J. Lord; Alan Ashworth

Based on the principle of synthetic lethality, PARP inhibitors have been shown to be very effective in killing cells deficient in homologous recombination (HR), such as those bearing mutations in BRCA1/2. However, questions regarding their wider use persist and other determinants of responsiveness to PARP inhibitor remain to be fully explored. MicroRNAs (miRNAs) are small non-coding RNAs, which serve as post-transcriptional regulators of gene expression and are involved in a wide variety of cellular processes, including the DNA damage response (DDR). However, little is known about whether miRNAs might influence sensitivity to PARP inhibitors. To investigate this, we performed a high throughput miRNA mimetic screen, which identified several miRNAs whose over-expression results in sensitization to the clinical PARP inhibitor olaparib. In particular, our findings indicate that hsa-miR-107 and hsa-miR-222 regulate the DDR and sensitise tumour cells to olaparib by repressing expression of RAD51, thus impairing DSB repair by HR. Moreover, elevated expression of hsa-miR-107 has been observed in a subset of ovarian clear cell carcinomas, which correlates with PARP inhibitor sensitivity and reduced RAD51 expression. Taken together, these observations raise the possibility that these miRNAs could be used as biomarkers to identify patients that may benefit from treatment with PARP inhibitors.


The EMBO Journal | 2012

APRIN is a cell cycle specific BRCA2-interacting protein required for genome integrity and a predictor of outcome after chemotherapy in breast cancer

Rachel Brough; Ilirjana Bajrami; Radost Vatcheva; Rachael Natrajan; Jorge S. Reis-Filho; Christopher J. Lord; Alan Ashworth

Mutations in BRCA2 confer an increased risk of cancer development, at least in part because the BRCA2 protein is required for the maintenance of genomic integrity. Here, we use proteomic profiling to identify APRIN (PDS5B), a cohesion‐associated protein, as a BRCA2‐associated protein. After exposure of cells to hydroxyurea or aphidicolin, APRIN and other cohesin components associate with BRCA2 in early S‐phase. We demonstrate that APRIN expression is required for the normal response to DNA‐damaging agents, the nuclear localisation of RAD51 and BRCA2 and efficient homologous recombination. The clinical significance of these findings is indicated by the observation that the BRCA2/APRIN interaction is compromised by BRCA2 missense variants of previously unknown significance and that APRIN expression levels are associated with histological grade in breast cancer and the outcome of breast cancer patients treated with DNA‐damaging chemotherapy.

Collaboration


Dive into the Ilirjana Bajrami's collaboration.

Top Co-Authors

Avatar

Christopher J. Lord

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Alan Ashworth

University of California

View shared research outputs
Top Co-Authors

Avatar

Rachel Brough

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Jessica Frankum

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Helen N. Pemberton

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Rumana Rafiq

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Richard Elliott

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Rowan Miller

University College London

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Pettitt

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Colm J. Ryan

University College Dublin

View shared research outputs
Researchain Logo
Decentralizing Knowledge