Dr Syed Haider

Team Leader: Breast Cancer Research Bioinformatics Group

Email: [email protected]

Location: Chelsea

Headshot of Syed Haider

Email: [email protected]

Location: Chelsea
Dr Haider has expertise in developing computational approaches for the discovery of target genes of breast cancer risk loci, and biomarkers of disease outcome and therapy response. His team employs a range of data science techniques to prioritise candidate genes by prosecuting multi-modal bulk and single-cell molecular profiles. His team also enables bioinformatics research and development in the breast division encompassing both hypothesis-driven and hypothesis-generating translational research.

Types of Publications

Journal articles

Mohd Nafi, S.N. Generali, D. Kramer-Marek, G. Gijsen, M. Strina, C. Cappelletti, M. Andreis, D. Haider, S. Li, J.-.L. Bridges, E. Capala, J. Ioannis, R. Harris, A.L. Kong, A (2014) Nuclear HER4 mediates acquired resistance to trastuzumab and is associated with poor outcome in HER2 positive breast cancer.. Show Abstract full text

The role of HER4 in breast cancer is controversial and its role in relation to trastuzumab resistance remains unclear. We showed that trastuzumab treatment and its acquired resistance induced HER4 upregulation, cleavage and nuclear translocation. However, knockdown of HER4 by specific siRNAs increased trastuzumab sensitivity and reversed its resistance in HER2 positive breast cancer cells. Preventing HER4 cleavage by a γ-secretase inhibitor and inhibiting HER4 tyrosine kinase activity by neratinib decreased trastuzumab-induced HER4 nuclear translocation and enhanced trastuzumab response. There was also increased nuclear HER4 staining in the tumours from BT474 xenograft mice and human patients treated with trastuzumab. Furthermore, nuclear HER4 predicted poor clinical response to trastuzumab monotherapy in patients undergoing a window study and was shown to be an independent poor prognostic factor in HER2 positive breast cancer. Our data suggest that HER4 plays a key role in relation to trastuzumab resistance in HER2 positive breast cancer. Therefore, our study provides novel findings that HER4 activation, cleavage and nuclear translocation influence trastuzumab sensitivity and resistance in HER2 positive breast cancer. Nuclear HER4 could be a potential prognostic and predictive biomarker and understanding the role of HER4 may provide strategies to overcome trastuzumab resistance in HER2 positive breast cancer.

Nafi, S. Generali, D. Kramer-Marek, G. Gijsen, M. Strina, C. Cappelletti, M. Andreis, D. Haider, S. Li, J.L. Bridges, E. Capala, J. Ioannis, R. Harris, A.L. Kong, A (2014) Nuclear HER4 mediates acquired resistance to trastuzumab and is associated with poor outcome in HER2 positive breast cancer.. Show Abstract full text

The role of HER4 in breast cancer is controversial and its role in relation to trastuzumab resistance remains unclear. We showed that trastuzumab treatment and its acquired resistance induced HER4 upregulation, cleavage and nuclear translocation. However, knockdown of HER4 by specific siRNAs increased trastuzumab sensitivity and reversed its resistance in HER2 positive breast cancer cells. Preventing HER4 cleavage by a γ-secretase inhibitor and inhibiting HER4 tyrosine kinase activity by neratinib decreased trastuzumab-induced HER4 nuclear translocation and enhanced trastuzumab response. There was also increased nuclear HER4 staining in the tumours from BT474 xenograft mice and human patients treated with trastuzumab. Furthermore, nuclear HER4 predicted poor clinical response to trastuzumab monotherapy in patients undergoing a window study and was shown to be an independent poor prognostic factor in HER2 positive breast cancer. Our data suggest that HER4 plays a key role in relation to trastuzumab resistance in HER2 positive breast cancer. Therefore, our study provides novel findings that HER4 activation, cleavage and nuclear translocation influence trastuzumab sensitivity and resistance in HER2 positive breast cancer. Nuclear HER4 could be a potential prognostic and predictive biomarker and understanding the role of HER4 may provide strategies to overcome trastuzumab resistance in HER2 positive breast cancer.

Leszczynska, K.B. Foskolou, I.P. Abraham, A.G. Anbalagan, S. Tellier, C. Haider, S. Span, P.N. O'Neill, E.E. Buffa, F.M. Hammond, E.M (2015) Hypoxia-induced p53 modulates both apoptosis and radiosensitivity via AKT.. Show Abstract full text

Restoration of hypoxia-induced apoptosis in tumors harboring p53 mutations has been proposed as a potential therapeutic strategy; however, the transcriptional targets that mediate hypoxia-induced p53-dependent apoptosis remain elusive. Here, we demonstrated that hypoxia-induced p53-dependent apoptosis is reliant on the DNA-binding and transactivation domains of p53 but not on the acetylation sites K120 and K164, which, in contrast, are essential for DNA damage-induced, p53-dependent apoptosis. Evaluation of hypoxia-induced transcripts in multiple cell lines identified a group of genes that are hypoxia-inducible proapoptotic targets of p53, including inositol polyphosphate-5-phosphatase (INPP5D), pleckstrin domain-containing A3 (PHLDA3), sulfatase 2 (SULF2), B cell translocation gene 2 (BTG2), cytoplasmic FMR1-interacting protein 2 (CYFIP2), and KN motif and ankyrin repeat domains 3 (KANK3). These targets were also regulated by p53 in human cancers, including breast, brain, colorectal, kidney, bladder, and melanoma cancers. Downregulation of these hypoxia-inducible targets associated with poor prognosis, suggesting that hypoxia-induced apoptosis contributes to p53-mediated tumor suppression and treatment response. Induction of p53 targets, PHLDA3, and a specific INPP5D transcript mediated apoptosis in response to hypoxia through AKT inhibition. Moreover, pharmacological inhibition of AKT led to apoptosis in the hypoxic regions of p53-deficient tumors and consequently increased radiosensitivity. Together, these results identify mediators of hypoxia-induced p53-dependent apoptosis and suggest AKT inhibition may improve radiotherapy response in p53-deficient tumors.

Oliemuller, E. Kogata, N. Bland, P. Kriplani, D. Daley, F. Haider, S. Shah, V. Sawyer, E.J. Howard, B.A (2017) SOX11 promotes invasive growth and ductal carcinoma in situ progression.. Show Abstract full text

Here, we show that SOX11, an embryonic mammary marker that is normally silent in postnatal breast cells, is expressed in many oestrogen receptor-negative preinvasive ductal carcinoma in situ (DCIS) lesions. Mature mammary epithelial cells engineered to express SOX11 showed alterations in progenitor cell populations, including an expanded basal-like population with increased aldehyde dehydrogenase (ALDH) activity, and increased mammosphere-forming capacity. DCIS.com cells engineered to express SOX11 showed increased ALDH activity, which is a feature of cancer stem cells. The CD44+/CD24-/ALDH+ cell population was increased in DCIS.com cells that expressed SOX11. Upregulating SOX11 expression in DCIS.com cells led to increased invasive growth both in vitro and when they were injected intraductally in a mouse model of DCIS that recapitulates human disease. Invasive lesions formed sooner and tumour growth was augmented in vivo, suggesting that SOX11 contributes to the progression of DCIS to invasive breast cancer. We identified potential downstream effectors of SOX11 during both microinvasive and invasive tumour growth stages, including several with established links to regulation of progenitor cell function and prenatal developmental growth. Our findings suggest that SOX11 is a potential biomarker for DCIS lesions containing cells harbouring distinct biological features that are likely to progress to invasive breast cancer. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Naidoo, K. Wai, P.T. Maguire, S.L. Daley, F. Haider, S. Kriplani, D. Campbell, J. Mirza, H. Grigoriadis, A. Tutt, A. Moseley, P.M. Abdel-Fatah, T.M.A. Chan, S.Y.T. Madhusudan, S. Rhaka, E.A. Ellis, I.O. Lord, C.J. Yuan, Y. Green, A.R. Natrajan, R (2018) Evaluation of CDK12 Protein Expression as a Potential Novel Biomarker for DNA Damage Response-Targeted Therapies in Breast Cancer.. Show Abstract full text

Disruption of Cyclin-Dependent Kinase 12 (<i>CDK12</i>) is known to lead to defects in DNA repair and sensitivity to platinum salts and PARP1/2 inhibitors. However, <i>CDK12</i> has also been proposed as an oncogene in breast cancer. We therefore aimed to assess the frequency and distribution of CDK12 protein expression by IHC in independent cohorts of breast cancer and correlate this with outcome and genomic status. We found that 21% of primary unselected breast cancers were CDK12 high, and 10.5% were absent, by IHC. CDK12 positivity correlated with HER2 positivity but was not an independent predictor of breast cancer-specific survival taking HER2 status into account; however, absent CDK12 protein expression significantly correlated with a triple-negative phenotype. Interestingly, CDK12 protein absence was associated with reduced expression of a number of DDR proteins including ATR, Ku70/Ku80, PARP1, DNA-PK, and γH2AX, suggesting a novel mechanism of CDK12-associated DDR dysregulation in breast cancer. Our data suggest that diagnostic IHC quantification of CDK12 in breast cancer is feasible, with CDK12 absence possibly signifying defective DDR function. This may have important therapeutic implications, particularly for triple-negative breast cancers. <i>Mol Cancer Ther; 17(1); 306-15. ©2017 AACR</i>.

Baxter, J.S. Leavy, O.C. Dryden, N.H. Maguire, S. Johnson, N. Fedele, V. Simigdala, N. Martin, L.-.A. Andrews, S. Wingett, S.W. Assiotis, I. Fenwick, K. Chauhan, R. Rust, A.G. Orr, N. Dudbridge, F. Haider, S. Fletcher, O (2018) Capture Hi-C identifies putative target genes at 33 breast cancer risk loci.. Show Abstract full text

Genome-wide association studies (GWAS) have identified approximately 100 breast cancer risk loci. Translating these findings into a greater understanding of the mechanisms that influence disease risk requires identification of the genes or non-coding RNAs that mediate these associations. Here, we use Capture Hi-C (CHi-C) to annotate 63 loci; we identify 110 putative target genes at 33 loci. To assess the support for these target genes in other data sources we test for associations between levels of expression and SNP genotype (eQTLs), disease-specific survival (DSS), and compare them with somatically mutated cancer genes. 22 putative target genes are eQTLs, 32 are associated with DSS and 14 are somatically mutated in breast, or other, cancers. Identifying the target genes at GWAS risk loci will lead to a greater understanding of the mechanisms that influence breast cancer risk and prognosis.

Aleksic, T. Verrill, C. Bryant, R.J. Han, C. Worrall, A.R. Brureau, L. Larré, S. Higgins, G.S. Fazal, F. Sabbagh, A. Haider, S. Buffa, F.M. Cole, D. Macaulay, V.M (2017) IGF-1R associates with adverse outcomes after radical radiotherapy for prostate cancer.. Show Abstract full text

<h4>Background</h4>Activated type 1 insulin-like growth factor receptors (IGF-1Rs) undergo internalisation and nuclear translocation, promoting cell survival. We previously reported that IGF-1R inhibition delays DNA damage repair, sensitising prostate cancer cells to ionising radiation. Here we tested the clinical relevance of these findings.<h4>Methods</h4>We assessed associations between IGF-1R and clinical outcomes by immunohistochemistry in diagnostic biopsies of 136 men treated with 55-70 Gy external beam radiotherapy for prostate cancer, comparing results with publicly available transcriptional data in surgically treated patients.<h4>Results</h4>Following radiotherapy, overall recurrence-free survival was shorter in patients whose tumours contained high total, cytoplasmic and internalised (nuclear/cytoplasmic) IGF-1R. High total IGF-1R associated with high primary Gleason grade and risk of metastasis, and cytoplasmic and internalised IGF-1R with biochemical recurrence, which includes patients experiencing local recurrence within the radiation field indicating radioresistance. In multivariate analysis, cytoplasmic, internalised and total IGF-1R were independently associated with risk of overall recurrence, and cytoplasmic IGF-1R was an independent predictor of biochemical recurrence post radiotherapy. Insulin-like growth factor receptors expression did not associate with biochemical recurrence after radical prostatectomy.<h4>Conclusions</h4>These data reveal increased risk of post-radiotherapy recurrence in men whose prostate cancers contain high levels of total or cytoplasmic IGF-1R.

Brough, R. Gulati, A. Haider, S. Kumar, R. Campbell, J. Knudsen, E. Pettitt, S.J. Ryan, C.J. Lord, C.J (2018) Identification of highly penetrant Rb-related synthetic lethal interactions in triple negative breast cancer.. Show Abstract full text

Although defects in the RB1 tumour suppressor are one of the more common driver alterations found in triple-negative breast cancer (TNBC), therapeutic approaches that exploit this have not been identified. By integrating molecular profiling data with data from multiple genetic perturbation screens, we identified candidate synthetic lethal (SL) interactions associated with RB1 defects in TNBC. We refined this analysis by identifying the highly penetrant effects, reasoning that these would be more robust in the face of molecular heterogeneity and would represent more promising therapeutic targets. A significant proportion of the highly penetrant RB1 SL effects involved proteins closely associated with RB1 function, suggesting that this might be a defining characteristic. These included nuclear pore complex components associated with the MAD2 spindle checkpoint protein, the kinase and bromodomain containing transcription factor TAF1, and multiple components of the SCF<sup>SKP</sup> Cullin F box containing complex. Small-molecule inhibition of SCF<sup>SKP</sup> elicited an increase in p27<sup>Kip</sup> levels, providing a mechanistic rationale for RB1 SL. Transcript expression of SKP2, a SCF<sup>SKP</sup> component, was elevated in RB1-defective TNBCs, suggesting that in these tumours, SKP2 activity might buffer the effects of RB1 dysfunction.

Lord, S.R. Cheng, W.-.C. Liu, D. Gaude, E. Haider, S. Metcalf, T. Patel, N. Teoh, E.J. Gleeson, F. Bradley, K. Wigfield, S. Zois, C. McGowan, D.R. Ah-See, M.-.L. Thompson, A.M. Sharma, A. Bidaut, L. Pollak, M. Roy, P.G. Karpe, F. James, T. English, R. Adams, R.F. Campo, L. Ayers, L. Snell, C. Roxanis, I. Frezza, C. Fenwick, J.D. Buffa, F.M. Harris, A.L (2018) Integrated Pharmacodynamic Analysis Identifies Two Metabolic Adaption Pathways to Metformin in Breast Cancer.. Show Abstract full text

Late-phase clinical trials investigating metformin as a cancer therapy are underway. However, there remains controversy as to the mode of action of metformin in tumors at clinical doses. We conducted a clinical study integrating measurement of markers of systemic metabolism, dynamic FDG-PET-CT, transcriptomics, and metabolomics at paired time points to profile the bioactivity of metformin in primary breast cancer. We show metformin reduces the levels of mitochondrial metabolites, activates multiple mitochondrial metabolic pathways, and increases 18-FDG flux in tumors. Two tumor groups are identified with distinct metabolic responses, an OXPHOS transcriptional response (OTR) group for which there is an increase in OXPHOS gene transcription and an FDG response group with increased 18-FDG uptake. Increase in proliferation, as measured by a validated proliferation signature, suggested that patients in the OTR group were resistant to metformin treatment. We conclude that mitochondrial response to metformin in primary breast cancer may define anti-tumor effect.

Chabanon, R.M. Muirhead, G. Krastev, D.B. Adam, J. Morel, D. Garrido, M. Lamb, A. Hénon, C. Dorvault, N. Rouanne, M. Marlow, R. Bajrami, I. Cardeñosa, M.L. Konde, A. Besse, B. Ashworth, A. Pettitt, S.J. Haider, S. Marabelle, A. Tutt, A.N. Soria, J.-.C. Lord, C.J. Postel-Vinay, S (2019) PARP inhibition enhances tumor cell-intrinsic immunity in ERCC1-deficient non-small cell lung cancer.. Show Abstract full text

The cyclic GMP-AMP synthase/stimulator of IFN genes (cGAS/STING) pathway detects cytosolic DNA to activate innate immune responses. Poly(ADP-ribose) polymerase inhibitors (PARPi) selectively target cancer cells with DNA repair deficiencies such as those caused by BRCA1 mutations or ERCC1 defects. Using isogenic cell lines and patient-derived samples, we showed that ERCC1-defective non-small cell lung cancer (NSCLC) cells exhibit an enhanced type I IFN transcriptomic signature and that low ERCC1 expression correlates with increased lymphocytic infiltration. We demonstrated that clinical PARPi, including olaparib and rucaparib, have cell-autonomous immunomodulatory properties in ERCC1-defective NSCLC and BRCA1-defective triple-negative breast cancer (TNBC) cells. Mechanistically, PARPi generated cytoplasmic chromatin fragments with characteristics of micronuclei; these were found to activate cGAS/STING, downstream type I IFN signaling, and CCL5 secretion. Importantly, these effects were suppressed in PARP1-null TNBC cells, suggesting that this phenotype resulted from an on-target effect of PARPi on PARP1. PARPi also potentiated IFN-γ-induced PD-L1 expression in NSCLC cell lines and in fresh patient tumor cells; this effect was enhanced in ERCC1-deficient contexts. Our data provide a preclinical rationale for using PARPi as immunomodulatory agents in appropriately molecularly selected populations.

Morotti, M. Bridges, E. Valli, A. Choudhry, H. Sheldon, H. Wigfield, S. Gray, N. Zois, C.E. Grimm, F. Jones, D. Teoh, E.J. Cheng, W.-.C. Lord, S. Anastasiou, D. Haider, S. McIntyre, A. Goberdhan, D.C.I. Buffa, F. Harris, A.L (2019) Hypoxia-induced switch in SNAT2/SLC38A2 regulation generates endocrine resistance in breast cancer.. Show Abstract full text

Tumor hypoxia is associated with poor patient outcomes in estrogen receptor-α-positive (ERα<sup>+</sup>) breast cancer. Hypoxia is known to affect tumor growth by reprogramming metabolism and regulating amino acid (AA) uptake. Here, we show that the glutamine transporter, SNAT2, is the AA transporter most frequently induced by hypoxia in breast cancer, and is regulated by hypoxia both in vitro and in vivo in xenografts. SNAT2 induction in MCF7 cells was also regulated by ERα, but it became predominantly a hypoxia-inducible factor 1α (HIF-1α)-dependent gene under hypoxia. Relevant to this, binding sites for both HIF-1α and ERα overlap in SNAT2's <i>cis</i>-regulatory elements. In addition, the down-regulation of SNAT2 by the ER antagonist fulvestrant was reverted in hypoxia. Overexpression of SNAT2 in vitro to recapitulate the levels induced by hypoxia caused enhanced growth, particularly after ERα inhibition, in hypoxia, or when glutamine levels were low. SNAT2 up-regulation in vivo caused complete resistance to antiestrogen and, partially, anti-VEGF therapies. Finally, high SNAT2 expression levels correlated with hypoxia profiles and worse outcome in patients given antiestrogen therapies. Our findings show a switch in the regulation of SNAT2 between ERα and HIF-1α, leading to endocrine resistance in hypoxia. Development of drugs targeting SNAT2 may be of value for a subset of hormone-resistant breast cancer.

Fox, N.S. Haider, S. Harris, A.L. Boutros, P.C (2019) Landscape of transcriptomic interactions between breast cancer and its microenvironment.. Show Abstract full text

Solid tumours comprise mixtures of tumour cells (TCs) and tumour-adjacent cells (TACs), and the intricate interconnections between these diverse populations shape the tumour's microenvironment. Despite this complexity, clinical genomic profiling is typically performed from bulk samples, without distinguishing TCs from TACs. To better understand TC-TAC interactions, we computationally distinguish their transcriptomes in 1780 primary breast tumours. We show that TC and TAC mRNA abundances are divergently associated with clinical phenotypes, including tumour subtypes and patient survival. These differences reflect distinct responses of TCs and TACs to specific somatic driver mutations, particularly TP53. These data further elucidate how the molecular interplay between breast tumours and their microenvironment drives aggressive tumour phenotypes.

Barrow-McGee, R. Procter, J. Owen, J. Woodman, N. Lombardelli, C. Kothari, A. Kovacs, T. Douek, M. George, S. Barry, P.A. Ramsey, K. Gibson, A. Buus, R. Holgersen, E. Natrajan, R. Haider, S. Shattock, M.J. Gillett, C. Tutt, A.N. Pinder, S.E. Naidoo, K (2020) Real-time ex vivo perfusion of human lymph nodes invaded by cancer (REPLICANT): a feasibility study.. Show Abstract full text

Understanding how breast cancer (BC) grows in axillary lymph nodes (ALNs), and refining how therapies might halt that process, is clinically important. However, modelling the complex ALN microenvironment is difficult, and no human models exist at present. We harvested ALNs from ten BC patients, and perfused them at 37 °C ex vivo for up to 24 h. Controlled autologous testing showed that ALNs remain viable after 24 h of ex vivo perfusion: haematoxylin and eosin-stained histological appearance and proliferation (by Ki67 immunohistochemistry) did not change significantly over time for any perfused ALN compared with a control from time-point zero. Furthermore, targeted gene expression analysis (NanoString PanCancer IO360 panel) showed that only 21/750 genes were differentially expressed between control and perfused ALNs (|log<sub>2</sub> FC| > 1 and q < 0.1): none were involved in apoptosis and metabolism, but rather all 21 genes were involved in immune function and angiogenesis. During perfusion, tissue acid-base balance remained stable. Interestingly, the flow rate increased (p < 0.001) in cancer-replaced (i.e. metastasis occupied more than 90% of the surface area on multiple levels) compared to cancer-free nodes (i.e. nodes with no metastasis on multiple sections). CXCL11 transcripts were significantly more abundant in cancer-replaced nodes, while CXCL12 transcripts were significantly more abundant in cancer-free nodes. These cytokines were also detected in the circulating perfusate. Monoclonal antibodies (nivolumab and trastuzumab) were administered into a further three ALNs to confirm perfusion efficacy. These drugs saturated the nodes; nivolumab even induced cancer cell death. Normothermic ALN perfusion is not only feasible but sustains the tumour microenvironment ex vivo for scientific investigation. This model could facilitate the identification of actionable immuno-oncology targets. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Favara, D.M. Zois, C.E. Haider, S. Pires, E. Sheldon, H. McCullagh, J. Banham, A.H. Harris, A.L (2019) ADGRL4/ELTD1 Silencing in Endothelial Cells Induces ACLY and SLC25A1 and Alters the Cellular Metabolic Profile.. Show Abstract full text

Adhesion G Protein-Coupled Receptor L4 (ADGRL4/ELTD1) is an endothelial cell adhesion G protein-coupled receptor (aGPCR) which regulates physiological and tumour angiogenesis, providing an attractive target for anti-cancer therapeutics. To date, ADGRL4/ELTD1's full role and mechanism of function within endothelial biology remains unknown, as do its ligand(s). In this study, ADGRL4/ELTD1 silencing, using two independent small interfering RNAs (siRNAs), was performed in human umbilical vein endothelial cells (HUVECS) followed by transcriptional profiling, target gene validation, and metabolomics using liquid chromatography-mass spectrometry in order to better characterise ADGRL4/ELTD1's role in endothelial cell biology. We show that ADGRL4/ELTD1 silencing induced expression of the cytoplasmic metabolic regulator ATP Citrate Lyase (ACLY) and the mitochondria-to-cytoplasm citrate transporter Solute Carrier Family 25 Member 1 (SLC25A1) but had no apparent effect on pathways downstream of ACLY (fatty acid and cholesterol synthesis or acetylation). Silencing induced KIT expression and affected the Notch signalling pathway, upregulating Delta Like Canonical Notch Ligand 4 (DLL4) and suppressing Jagged Canonical Notch Ligand 1 (<i>JAG1</i>) and Hes Family BHLH Transcription Factor 2 (<i>HES2</i>). The effect of ADGRL4/ELTD1 silencing on the cellular metabolic profile was modest but several metabolites were significantly affected. Cis-aconitic acid, uridine diphosphate (UDP)-glucoronate, fructose 2,6-diphosphate, uridine 5-diphosphate, and aspartic acid were all elevated as a result of silencing and phosphocreatine, N-acetylglutamic acid, taurine, deoxyadenosine triphosphate, and cytidine monophosphate were depleted. Metabolic pathway analysis implicated ADGRL4/ELTD1 in pyrimidine, amino acid, and sugar metabolism. In summary, this study shows that ADGRL4/ELTD1 impacts core components of endothelial metabolism and regulates genes involved in endothelial differentiation/homeostasis and Notch signalling.

Lord, S.R. Collins, J.M. Cheng, W.-.C. Haider, S. Wigfield, S. Gaude, E. Fielding, B.A. Pinnick, K.E. Harjes, U. Segaran, A. Jha, P. Hoefler, G. Pollak, M.N. Thompson, A.M. Roy, P.G. English, R. Adams, R.F. Frezza, C. Buffa, F.M. Karpe, F. Harris, A.L (2020) Transcriptomic analysis of human primary breast cancer identifies fatty acid oxidation as a target for metformin.. Show Abstract full text

<h4>Background</h4>Epidemiological studies suggest that metformin may reduce the incidence of cancer in patients with diabetes and multiple late phase clinical trials assessing the potential of repurposing this drug are underway. Transcriptomic profiling of tumour samples is an excellent tool to understand drug bioactivity, identify candidate biomarkers and assess for mechanisms of resistance to therapy.<h4>Methods</h4>Thirty-six patients with untreated primary breast cancer were recruited to a window study and transcriptomic profiling of tumour samples carried out before and after metformin treatment.<h4>Results</h4>Multiple genes that regulate fatty acid oxidation were upregulated at the transcriptomic level and there was a differential change in expression between two previously identified cohorts of patients with distinct metabolic responses. Increase in expression of a mitochondrial fatty oxidation gene composite signature correlated with change in a proliferation gene signature. In vitro assays showed that, in contrast to previous studies in models of normal cells, metformin reduces fatty acid oxidation with a subsequent accumulation of intracellular triglyceride, independent of AMPK activation.<h4>Conclusions</h4>We propose that metformin at clinical doses targets fatty acid oxidation in cancer cells with implications for patient selection and drug combinations.<h4>Clinical trial registration</h4>NCT01266486.

ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, (2020) Pan-cancer analysis of whole genomes.. Show Abstract full text

Cancer is driven by genetic change, and the advent of massively parallel sequencing has enabled systematic documentation of this variation at the whole-genome scale<sup>1-3</sup>. Here we report the integrative analysis of 2,658 whole-cancer genomes and their matching normal tissues across 38 tumour types from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). We describe the generation of the PCAWG resource, facilitated by international data sharing using compute clouds. On average, cancer genomes contained 4-5 driver mutations when combining coding and non-coding genomic elements; however, in around 5% of cases no drivers were identified, suggesting that cancer driver discovery is not yet complete. Chromothripsis, in which many clustered structural variants arise in a single catastrophic event, is frequently an early event in tumour evolution; in acral melanoma, for example, these events precede most somatic point mutations and affect several cancer-associated genes simultaneously. Cancers with abnormal telomere maintenance often originate from tissues with low replicative activity and show several mechanisms of preventing telomere attrition to critical levels. Common and rare germline variants affect patterns of somatic mutation, including point mutations, structural variants and somatic retrotransposition. A collection of papers from the PCAWG Consortium describes non-coding mutations that drive cancer beyond those in the TERT promoter<sup>4</sup>; identifies new signatures of mutational processes that cause base substitutions, small insertions and deletions and structural variation<sup>5,6</sup>; analyses timings and patterns of tumour evolution<sup>7</sup>; describes the diverse transcriptional consequences of somatic mutation on splicing, expression levels, fusion genes and promoter activity<sup>8,9</sup>; and evaluates a range of more-specialized features of cancer genomes<sup>8,10-18</sup>.

Crespo-Rodriguez, E. Bergerhoff, K. Bozhanova, G. Foo, S. Patin, E.C. Whittock, H. Buus, R. Haider, S. Muirhead, G. Thway, K. Newbold, K. Coffin, R.S. Vile, R.G. Kim, D. McLaughlin, M. Melcher, A.A. Harrington, K.J. Pedersen, M (2020) Combining BRAF inhibition with oncolytic herpes simplex virus enhances the immune-mediated antitumor therapy of BRAF-mutant thyroid cancer.. Show Abstract full text

<h4>Background</h4>The aggressive clinical behavior of poorly differentiated and anaplastic thyroid cancers (PDTC and ATC) has proven challenging to treat, and survival beyond a few months from diagnosis is rare. Although 30%-60% of these tumors contain mutations in the <i>BRAF</i> gene, inhibitors designed specifically to target oncogenic BRAF have shown limited and only short-lasting therapeutic benefits as single agents, thus highlighting the need for improved treatment strategies, including novel combinations.<h4>Methods</h4>Using a BRAF<sup>V600E</sup>-driven mouse model of ATC, we investigated the therapeutic efficacy of the combination of BRAF inhibition and oncolytic herpes simplex virus (oHSV). Analyses of samples from tumor-bearing mice were performed to immunologically characterize the effects of different treatments. These immune data were used to inform the incorporation of immune checkpoint inhibitors into triple combination therapies.<h4>Results</h4>We characterized the immune landscape in vivo following BRAF inhibitor treatment and detected only modest immune changes. We, therefore, hypothesized that the addition of oncolytic virotherapy to BRAF inhibition in thyroid cancer would create a more favorable tumor immune microenvironment, boost the inflammatory status of tumors and improve BRAF inhibitor therapy. First, we showed that thyroid cancer cells were susceptible to infection with oHSV and that this process was associated with activation of the immune tumor microenvironment in vivo. Next, we showed improved therapeutic responses when combining oHSV and BRAF inhibition in vivo, although no synergistic effects were seen in vitro, further confirming that the dominant effect of oHSV in this context was likely immune-mediated. Importantly, both gene and protein expression data revealed an increase in activation of T cells and natural killer (NK) cells in the tumor in combination-treated samples. The benefit of combination oHSV and BRAF inhibitor therapy was abrogated when T cells or NK cells were depleted in vivo. In addition, we showed upregulation of PD-L1 and CTLA-4 following combined treatment and demonstrated that blockade of the PD-1/PD-L1 axis or CTLA-4 further improved combination therapy.<h4>Conclusions</h4>The combination of oHSV and BRAF inhibition significantly improved survival in a mouse model of ATC by enhancing immune-mediated antitumor effects, and triple combination therapies, including either PD-1 or CTLA-4 blockade, further improved therapy.

Yeow, Z.Y. Lambrus, B.G. Marlow, R. Zhan, K.H. Durin, M.-.A. Evans, L.T. Scott, P.M. Phan, T. Park, E. Ruiz, L.A. Moralli, D. Knight, E.G. Badder, L.M. Novo, D. Haider, S. Green, C.M. Tutt, A.N.J. Lord, C.J. Chapman, J.R. Holland, A.J (2020) Targeting TRIM37-driven centrosome dysfunction in 17q23-amplified breast cancer.. Show Abstract full text

Genomic instability is a hallmark of cancer, and has a central role in the initiation and development of breast cancer<sup>1,2</sup>. The success of poly-ADP ribose polymerase inhibitors in the treatment of breast cancers that are deficient in homologous recombination exemplifies the utility of synthetically lethal genetic interactions in the treatment of breast cancers that are driven by genomic instability<sup>3</sup>. Given that defects in homologous recombination are present in only a subset of breast cancers, there is a need to identify additional driver mechanisms for genomic instability and targeted strategies to exploit these defects in the treatment of cancer. Here we show that centrosome depletion induces synthetic lethality in cancer cells that contain the 17q23 amplicon, a recurrent copy number aberration that defines about 9% of all primary breast cancer tumours and is associated with high levels of genomic instability<sup>4-6</sup>. Specifically, inhibition of polo-like kinase 4 (PLK4) using small molecules leads to centrosome depletion, which triggers mitotic catastrophe in cells that exhibit amplicon-directed overexpression of TRIM37. To explain this effect, we identify TRIM37 as a negative regulator of centrosomal pericentriolar material. In 17q23-amplified cells that lack centrosomes, increased levels of TRIM37 block the formation of foci that comprise pericentriolar material-these foci are structures with a microtubule-nucleating capacity that are required for successful cell division in the absence of centrosomes. Finally, we find that the overexpression of TRIM37 causes genomic instability by delaying centrosome maturation and separation at mitotic entry, and thereby increases the frequency of mitotic errors. Collectively, these findings highlight TRIM37-dependent genomic instability as a putative driver event in 17q23-amplified breast cancer and provide a rationale for the use of centrosome-targeting therapeutic agents in treating these cancers.

Pettitt, S.J. Frankum, J.R. Punta, M. Lise, S. Alexander, J. Chen, Y. Yap, T.A. Haider, S. Tutt, A.N.J. Lord, C.J (2020) Clinical <i>BRCA1/2</i> Reversion Analysis Identifies Hotspot Mutations and Predicted Neoantigens Associated with Therapy Resistance.. Show Abstract full text

Reversion mutations in <i>BRCA1</i> or <i>BRCA2</i> are associated with resistance to PARP inhibitors and platinum. To better understand the nature of these mutations, we collated, codified, and analyzed more than 300 reversions. This identified reversion "hotspots" and "deserts" in regions encoding the <i>N</i> and <i>C</i> terminus, respectively, of BRCA2, suggesting that pathogenic mutations in these regions may be at higher or lower risk of reversion. Missense and splice-site pathogenic mutations in <i>BRCA1/2</i> also appeared less likely to revert than truncating mutations. Most reversions were <100 bp deletions. Although many deletions exhibited microhomology, this was not universal, suggesting that multiple DNA-repair processes cause reversion. Finally, we found that many reversions were predicted to encode immunogenic neopeptides, suggesting a route to the treatment of reverted disease. As well as providing a freely available database for the collation of future reversion cases, these observations have implications for how drug resistance might be managed in <i>BRCA</i>-mutant cancers. SIGNIFICANCE: Reversion mutations in <i>BRCA</i> genes are a major cause of clinical platinum and PARP inhibitor resistance. This analysis of all reported clinical reversions suggests that the position of <i>BRCA2</i> mutations affects the risk of reversion. Many reversions are also predicted to encode tumor neoantigens, providing a potential route to targeting resistance.<i>This article is highlighted in the In This Issue feature, p. 1426</i>.

Chong, I.Y. Starling, N. Rust, A. Alexander, J. Aronson, L. Llorca-Cardenosa, M. Chauhan, R. Chaudry, A. Kumar, S. Fenwick, K. Assiotis, I. Matthews, N. Begum, R. Wotherspoon, A. Terlizzo, M. Watkins, D. Chau, I. Lord, C.J. Haider, S. Rao, S. Cunningham, D (2021) The Mutational Concordance of Fixed Formalin Paraffin Embedded and Fresh Frozen Gastro-Oesophageal Tumours Using Whole Exome Sequencing.. Show Abstract full text

1.<h4>Background</h4>The application of massively parallel sequencing has led to the identification of aberrant druggable pathways and somatic mutations within therapeutically relevant genes in gastro-oesophageal cancer. Given the widespread use of formalin-fixed paraffin-embedded (FFPE) samples in the study of this disease, it would be beneficial, especially for the purposes of biomarker evaluation, to assess the concordance between comprehensive exome-wide sequencing data from archival FFPE samples originating from a prospective clinical study and those derived from fresh-frozen material. 2.<h4>Methods</h4>We analysed whole-exome sequencing data to define the mutational concordance of 16 matched fresh-frozen and FFPE gastro-oesophageal tumours (<i>N</i> = 32) from a prospective clinical study. We assessed DNA integrity prior to sequencing and then identified coding mutations in genes that have previously been implicated in other cancers. In addition, we calculated the mutant-allele heterogeneity (MATH) for these samples. 3.<h4>Results</h4>Although there was increased degradation of DNA in FFPE samples compared with frozen samples, sequencing data from only two FFPE samples failed to reach an adequate mapping quality threshold. Using a filtering threshold of mutant read counts of at least ten and a minimum of 5% variant allele frequency (VAF) we found that there was a high median mutational concordance of 97% (range 80.1-98.68%) between fresh-frozen and FFPE gastro-oesophageal tumour-derived exomes. However, the majority of FFPE tumours had higher mutant-allele heterogeneity (MATH) scores when compared with corresponding frozen tumours (<i>p</i> < 0.001), suggesting that FFPE-based exome sequencing is likely to over-represent tumour heterogeneity in FFPE samples compared to fresh-frozen samples. Furthermore, we identified coding mutations in 120 cancer-related genes, including those associated with chromatin remodelling and Wnt/β-catenin and Receptor Tyrosine Kinase signalling. 4.<h4>Conclusions</h4>These data suggest that comprehensive genomic data can be generated from exome sequencing of selected DNA samples extracted from archival FFPE gastro-oesophageal tumour tissues within the context of prospective clinical trials.

Peck, B. Bland, P. Mavrommati, I. Muirhead, G. Cottom, H. Wai, P.T. Maguire, S.L. Barker, H.E. Morrison, E. Kriplani, D. Yu, L. Gibson, A. Falgari, G. Brennan, K. Farnie, G. Buus, R. Marlow, R. Novo, D. Knight, E. Guppy, N. Kolarevic, D. Susnjar, S. Milijic, N.M. Naidoo, K. Gazinska, P. Roxanis, I. Pancholi, S. Martin, L.-.A. Holgersen, E.M. Cheang, M.C.U. Noor, F. Postel-Vinay, S. Quinn, G. McDade, S. Krasny, L. Huang, P. Daley, F. Wallberg, F. Choudhary, J.S. Haider, S. Tutt, A.N. Natrajan, R (2021) 3D Functional Genomics Screens Identify CREBBP as a Targetable Driver in Aggressive Triple-Negative Breast Cancer.. Show Abstract full text

Triple-negative breast cancers (TNBC) are resistant to standard-of-care chemotherapy and lack known targetable driver gene alterations. Identification of novel drivers could aid the discovery of new treatment strategies for this hard-to-treat patient population, yet studies using high-throughput and accurate models to define the functions of driver genes in TNBC to date have been limited. Here, we employed unbiased functional genomics screening of the 200 most frequently mutated genes in breast cancer, using spheroid cultures to model <i>in vivo</i>-like conditions, and identified the histone acetyltransferase CREBBP as a novel tumor suppressor in TNBC. CREBBP protein expression in patient tumor samples was absent in 8% of TNBCs and at a high frequency in other tumors, including squamous lung cancer, where CREBBP-inactivating mutations are common. In TNBC, CREBBP alterations were associated with higher genomic heterogeneity and poorer patient survival and resulted in upregulation and dependency on a FOXM1 proliferative program. Targeting FOXM1-driven proliferation indirectly with clinical CDK4/6 inhibitors (CDK4/6i) selectively impaired growth in spheroids, cell line xenografts, and patient-derived models from multiple tumor types with CREBBP mutations or loss of protein expression. In conclusion, we have identified CREBBP as a novel driver in aggressive TNBC and identified an associated genetic vulnerability in tumor cells with alterations in CREBBP and provide a preclinical rationale for assessing CREBBP alterations as a biomarker of CDK4/6i response in a new patient population. SIGNIFICANCE: This study demonstrates that CREBBP genomic alterations drive aggressive TNBC, lung cancer, and lymphomas and may be selectively treated with clinical CDK4/6 inhibitors.

Holgersen, E.M. Gillespie, A. Leavy, O.C. Baxter, J.S. Zvereva, A. Muirhead, G. Johnson, N. Sipos, O. Dryden, N.H. Broome, L.R. Chen, Y. Kozin, I. Dudbridge, F. Fletcher, O. Haider, S (2021) Identifying high-confidence capture Hi-C interactions using CHiCANE.. Show Abstract full text

The ability to identify regulatory interactions that mediate gene expression changes through distal elements, such as risk loci, is transforming our understanding of how genomes are spatially organized and regulated. Capture Hi-C (CHi-C) is a powerful tool to delineate such regulatory interactions. However, primary analysis and downstream interpretation of CHi-C profiles remains challenging and relies on disparate tools with ad-hoc input/output formats and specific assumptions for statistical modeling. Here we present a data processing and interaction calling toolkit (CHiCANE), specialized for the analysis and meaningful interpretation of CHi-C assays. In this protocol, we demonstrate applications of CHiCANE to region capture Hi-C (rCHi-C) and promoter capture Hi-C (pCHi-C) libraries, followed by quality assessment of interaction peaks, as well as downstream analysis specific to rCHi-C and pCHi-C to aid functional interpretation. For a typical rCHi-C/pCHi-C dataset this protocol takes up to 3 d for users with a moderate understanding of R programming and statistical concepts, although this is dependent on dataset size and compute power available. CHiCANE is freely available at https://cran.r-project.org/web/packages/chicane .

Jungwirth, U. van Weverwijk, A. Evans, R.J. Jenkins, L. Vicente, D. Alexander, J. Gao, Q. Haider, S. Iravani, M. Isacke, C.M (2021) Impairment of a distinct cancer-associated fibroblast population limits tumour growth and metastasis.. Show Abstract full text

Profiling studies have revealed considerable phenotypic heterogeneity in cancer-associated fibroblasts (CAFs) present within the tumour microenvironment, however, functional characterisation of different CAF subsets is hampered by the lack of specific markers defining these populations. Here we show that genetic deletion of the Endo180 (MRC2) receptor, predominantly expressed by a population of matrix-remodelling CAFs, profoundly limits tumour growth and metastasis; effects that can be recapitulated in 3D co-culture assays. This impairment results from a CAF-intrinsic contractility defect and reduced CAF viability, which coupled with the lack of phenotype in the normal mouse, demonstrates that upregulated Endo180 expression by a specific, potentially targetable CAF subset is required to generate a supportive tumour microenvironment. Further, characterisation of a tumour subline selected via serial in vivo passage for its ability to overcome these stromal defects provides important insight into, how tumour cells adapt to a non-activated stroma in the early stages of metastatic colonisation.

Zatreanu, D. Robinson, H.M.R. Alkhatib, O. Boursier, M. Finch, H. Geo, L. Grande, D. Grinkevich, V. Heald, R.A. Langdon, S. Majithiya, J. McWhirter, C. Martin, N.M.B. Moore, S. Neves, J. Rajendra, E. Ranzani, M. Schaedler, T. Stockley, M. Wiggins, K. Brough, R. Sridhar, S. Gulati, A. Shao, N. Badder, L.M. Novo, D. Knight, E.G. Marlow, R. Haider, S. Callen, E. Hewitt, G. Schimmel, J. Prevo, R. Alli, C. Ferdinand, A. Bell, C. Blencowe, P. Bot, C. Calder, M. Charles, M. Curry, J. Ekwuru, T. Ewings, K. Krajewski, W. MacDonald, E. McCarron, H. Pang, L. Pedder, C. Rigoreau, L. Swarbrick, M. Wheatley, E. Willis, S. Wong, A.C. Nussenzweig, A. Tijsterman, M. Tutt, A. Boulton, S.J. Higgins, G.S. Pettitt, S.J. Smith, G.C.M. Lord, C.J (2021) Polθ inhibitors elicit BRCA-gene synthetic lethality and target PARP inhibitor resistance.. Show Abstract full text

To identify approaches to target DNA repair vulnerabilities in cancer, we discovered nanomolar potent, selective, low molecular weight (MW), allosteric inhibitors of the polymerase function of DNA polymerase Polθ, including ART558. ART558 inhibits the major Polθ-mediated DNA repair process, Theta-Mediated End Joining, without targeting Non-Homologous End Joining. In addition, ART558 elicits DNA damage and synthetic lethality in BRCA1- or BRCA2-mutant tumour cells and enhances the effects of a PARP inhibitor. Genetic perturbation screening revealed that defects in the 53BP1/Shieldin complex, which cause PARP inhibitor resistance, result in in vitro and in vivo sensitivity to small molecule Polθ polymerase inhibitors. Mechanistically, ART558 increases biomarkers of single-stranded DNA and synthetic lethality in 53BP1-defective cells whilst the inhibition of DNA nucleases that promote end-resection reversed these effects, implicating these in the synthetic lethal mechanism-of-action. Taken together, these observations describe a drug class that elicits BRCA-gene synthetic lethality and PARP inhibitor synergy, as well as targeting a biomarker-defined mechanism of PARPi-resistance.

Baxter, J.S. Johnson, N. Tomczyk, K. Gillespie, A. Maguire, S. Brough, R. Fachal, L. Michailidou, K. Bolla, M.K. Wang, Q. Dennis, J. Ahearn, T.U. Andrulis, I.L. Anton-Culver, H. Antonenkova, N.N. Arndt, V. Aronson, K.J. Augustinsson, A. Becher, H. Beckmann, M.W. Behrens, S. Benitez, J. Bermisheva, M. Bogdanova, N.V. Bojesen, S.E. Brenner, H. Brucker, S.Y. Cai, Q. Campa, D. Canzian, F. Castelao, J.E. Chan, T.L. Chang-Claude, J. Chanock, S.J. Chenevix-Trench, G. Choi, J.-.Y. Clarke, C.L. NBCS Collaborators, . Colonna, S. Conroy, D.M. Couch, F.J. Cox, A. Cross, S.S. Czene, K. Daly, M.B. Devilee, P. Dörk, T. Dossus, L. Dwek, M. Eccles, D.M. Ekici, A.B. Eliassen, A.H. Engel, C. Fasching, P.A. Figueroa, J. Flyger, H. Gago-Dominguez, M. Gao, C. García-Closas, M. García-Sáenz, J.A. Ghoussaini, M. Giles, G.G. Goldberg, M.S. González-Neira, A. Guénel, P. Gündert, M. Haeberle, L. Hahnen, E. Haiman, C.A. Hall, P. Hamann, U. Hartman, M. Hatse, S. Hauke, J. Hollestelle, A. Hoppe, R. Hopper, J.L. Hou, M.-.F. kConFab Investigators, . ABCTB Investigators, . Ito, H. Iwasaki, M. Jager, A. Jakubowska, A. Janni, W. John, E.M. Joseph, V. Jung, A. Kaaks, R. Kang, D. Keeman, R. Khusnutdinova, E. Kim, S.-.W. Kosma, V.-.M. Kraft, P. Kristensen, V.N. Kubelka-Sabit, K. Kurian, A.W. Kwong, A. Lacey, J.V. Lambrechts, D. Larson, N.L. Larsson, S.C. Le Marchand, L. Lejbkowicz, F. Li, J. Long, J. Lophatananon, A. Lubiński, J. Mannermaa, A. Manoochehri, M. Manoukian, S. Margolin, S. Matsuo, K. Mavroudis, D. Mayes, R. Menon, U. Milne, R.L. Mohd Taib, N.A. Muir, K. Muranen, T.A. Murphy, R.A. Nevanlinna, H. O'Brien, K.M. Offit, K. Olson, J.E. Olsson, H. Park, S.K. Park-Simon, T.-.W. Patel, A.V. Peterlongo, P. Peto, J. Plaseska-Karanfilska, D. Presneau, N. Pylkäs, K. Rack, B. Rennert, G. Romero, A. Ruebner, M. Rüdiger, T. Saloustros, E. Sandler, D.P. Sawyer, E.J. Schmidt, M.K. Schmutzler, R.K. Schneeweiss, A. Schoemaker, M.J. Shah, M. Shen, C.-.Y. Shu, X.-.O. Simard, J. Southey, M.C. Stone, J. Surowy, H. Swerdlow, A.J. Tamimi, R.M. Tapper, W.J. Taylor, J.A. Teo, S.H. Teras, L.R. Terry, M.B. Toland, A.E. Tomlinson, I. Truong, T. Tseng, C.-.C. Untch, M. Vachon, C.M. van den Ouweland, A.M.W. Wang, S.S. Weinberg, C.R. Wendt, C. Winham, S.J. Winqvist, R. Wolk, A. Wu, A.H. Yamaji, T. Zheng, W. Ziogas, A. Pharoah, P.D.P. Dunning, A.M. Easton, D.F. Pettitt, S.J. Lord, C.J. Haider, S. Orr, N. Fletcher, O (2021) Functional annotation of the 2q35 breast cancer risk locus implicates a structural variant in influencing activity of a long-range enhancer element.. Show Abstract full text

A combination of genetic and functional approaches has identified three independent breast cancer risk loci at 2q35. A recent fine-scale mapping analysis to refine these associations resulted in 1 (signal 1), 5 (signal 2), and 42 (signal 3) credible causal variants at these loci. We used publicly available in silico DNase I and ChIP-seq data with in vitro reporter gene and CRISPR assays to annotate signals 2 and 3. We identified putative regulatory elements that enhanced cell-type-specific transcription from the IGFBP5 promoter at both signals (30- to 40-fold increased expression by the putative regulatory element at signal 2, 2- to 3-fold by the putative regulatory element at signal 3). We further identified one of the five credible causal variants at signal 2, a 1.4 kb deletion (esv3594306), as the likely causal variant; the deletion allele of this variant was associated with an average additional increase in IGFBP5 expression of 1.3-fold (MCF-7) and 2.2-fold (T-47D). We propose a model in which the deletion allele of esv3594306 juxtaposes two transcription factor binding regions (annotated by estrogen receptor alpha ChIP-seq peaks) to generate a single extended regulatory element. This regulatory element increases cell-type-specific expression of the tumor suppressor gene IGFBP5 and, thereby, reduces risk of estrogen receptor-positive breast cancer (odds ratio = 0.77, 95% CI 0.74-0.81, p = 3.1 × 10<sup>-31</sup>).

Khalique, S. Nash, S. Mansfield, D. Wampfler, J. Attygale, A. Vroobel, K. Kemp, H. Buus, R. Cottom, H. Roxanis, I. Jones, T. von Loga, K. Begum, D. Guppy, N. Ramagiri, P. Fenwick, K. Matthews, N. Hubank, M.J.F. Lord, C.J. Haider, S. Melcher, A. Banerjee, S. Natrajan, R (2021) Quantitative Assessment and Prognostic Associations of the Immune Landscape in Ovarian Clear Cell Carcinoma.. Show Abstract full text

Ovarian clear cell carcinoma (OCCC) is a rare subtype of epithelial ovarian cancer characterised by a high frequency of loss-of-function <i>ARID1A</i> mutations and a poor response to chemotherapy. Despite their generally low mutational burden, an intratumoural T cell response has been reported in a subset of OCCC, with <i>ARID1A</i> purported to be a biomarker for the response to the immune checkpoint blockade independent of micro-satellite instability (MSI). However, assessment of the different immune cell types and spatial distribution specifically within OCCC patients has not been described to date. Here, we characterised the immune landscape of OCCC by profiling a cohort of 33 microsatellite stable OCCCs at the genomic, gene expression and histological level using targeted sequencing, gene expression profiling using the NanoString targeted immune panel, and multiplex immunofluorescence to assess the spatial distribution and abundance of immune cell populations at the protein level. Analysis of these tumours and subsequent independent validation identified an immune-related gene expression signature associated with risk of recurrence of OCCC. Whilst histological quantification of tumour-infiltrating lymphocytes (TIL, Salgado scoring) showed no association with the risk of recurrence or <i>ARID1A</i> mutational status, the characterisation of TILs via multiplexed immunofluorescence identified spatial differences in immunosuppressive cell populations in OCCC. Tumour-associated macrophages (TAM) and regulatory T cells were excluded from the vicinity of tumour cells in low-risk patients, suggesting that high-risk patients have a more immunosuppressive microenvironment. We also found that TAMs and cytotoxic T cells were also excluded from the vicinity of tumour cells in <i>ARID1A</i>-mutated OCCCs compared to <i>ARID1A</i> wild-type tumours, suggesting that the exclusion of these immune effectors could determine the host response of <i>ARID1A</i>-mutant OCCCs to therapy. Overall, our study has provided new insights into the immune landscape and prognostic associations in OCCC and suggest that tailored immunotherapeutic approaches may be warranted for different subgroups of OCCC patients.

Sheldon, H. Alexander, J. Bridges, E. Moreira, L. Reilly, S. Ang, K.H. Wang, D. Lin, S. Haider, S. Banham, A.H. Harris, A.L (2021) ELTD1 Activation Induces an Endothelial-EMT Transition to a Myofibroblast Phenotype.. Show Abstract full text

ELTD1 is expressed in endothelial and vascular smooth muscle cells and has a role in angiogenesis. It has been classified as an adhesion GPCR, but as yet, no ligand has been identified and its function remains unknown. To establish its role, ELTD1 was overexpressed in endothelial cells. Expression and consequently ligand independent activation of ELTD1 results in endothelial-mesenchymal transistion (EndMT) with a loss of cell-cell contact, formation of stress fibres and mature focal adhesions and an increased expression of smooth muscle actin. The effect was pro-angiogenic, increasing Matrigel network formation and endothelial sprouting. RNA-Seq analysis after the cells had undergone EndMT revealed large increases in chemokines and cytokines involved in regulating immune response. Gene set enrichment analysis of the data identified a number of pathways involved in myofibroblast biology suggesting that the endothelial cells had undergone a type II EMT. This type of EMT is involved in wound repair and is closely associated with inflammation implicating ELTD1 in these processes.

Bajrami, I. Walker, C. Krastev, D.B. Weekes, D. Song, F. Wicks, A.J. Alexander, J. Haider, S. Brough, R. Pettitt, S.J. Tutt, A.N.J. Lord, C.J (2021) Sirtuin inhibition is synthetic lethal with BRCA1 or BRCA2 deficiency.. Show Abstract full text

PARP enzymes utilise NAD<sup>+</sup> as a co-substrate for their enzymatic activity. Inhibition of PARP1 is synthetic lethal with defects in either BRCA1 or BRCA2. In order to assess whether other genes implicated in NAD<sup>+</sup> metabolism were synthetic lethal with BRCA1 or BRCA2 gene defects, we carried out a genetic screen, which identified a synthetic lethality between BRCA1 and genetic inhibition of either of two sirtuin (SIRT) enzymes, SIRT1 or SIRT6. This synthetic lethal interaction was replicated using small-molecule SIRT inhibitors and was associated with replication stress and increased cellular PARylation, in contrast to the decreased PARylation associated with BRCA-gene/PARP inhibitor synthetic lethality. SIRT/BRCA1 synthetic lethality was reversed by genetic ablation of either PARP1 or the histone PARylation factor-coding gene HPF1, implicating PARP1/HPF1-mediated serine ADP-ribosylation as part of the mechanistic basis of this synthetic lethal effect. These observations suggest that PARP1/HPF1-mediated serine ADP-ribosylation, when driven by SIRT inhibition, can inadvertently inhibit the growth of BRCA-gene mutant cells.

Romualdo Cardoso, S. Gillespie, A. Haider, S. Fletcher, O (2022) Functional annotation of breast cancer risk loci: current progress and future directions.. Show Abstract full text

Genome-wide association studies coupled with large-scale replication and fine-scale mapping studies have identified more than 150 genomic regions that are associated with breast cancer risk. Here, we review efforts to translate these findings into a greater understanding of disease mechanism. Our review comes in the context of a recently published fine-scale mapping analysis of these regions, which reported 352 independent signals and a total of 13,367 credible causal variants. The vast majority of credible causal variants map to noncoding DNA, implicating regulation of gene expression as the mechanism by which functional variants influence risk. Accordingly, we review methods for defining candidate-regulatory sequences, methods for identifying putative target genes and methods for linking candidate-regulatory sequences to putative target genes. We provide a summary of available data resources and identify gaps in these resources. We conclude that while much work has been done, there is still much to do. There are, however, grounds for optimism; combining statistical data from fine-scale mapping with functional data that are more representative of the normal "at risk" breast, generated using new technologies, should lead to a greater understanding of the mechanisms that influence an individual woman's risk of breast cancer.

Fitzpatrick, A. Iravani, M. Mills, A. Childs, L. Alaguthurai, T. Clifford, A. Garcia-Murillas, I. Van Laere, S. Dirix, L. Harries, M. Okines, A. Turner, N.C. Haider, S. Tutt, A.N.J. Isacke, C.M (2022) Assessing CSF ctDNA to Improve Diagnostic Accuracy and Therapeutic Monitoring in Breast Cancer Leptomeningeal Metastasis.. Show Abstract full text

<h4>Purpose</h4>Cerebrospinal fluid (CSF) cytology is the gold standard diagnostic test for breast cancer leptomeningeal metastasis (BCLM), but has impaired sensitivity, often necessitating repeated lumbar puncture to confirm or refute diagnosis. Further, there is no quantitative response tool to assess response or progression during BCLM treatment.<h4>Experimental design</h4>Facing the challenge of working with small-volume samples and the lack of common recurrent mutations in breast cancers, cell-free DNA was extracted from the CSF and plasma of patients undergoing investigation for BCLM (n = 30). ctDNA fraction was assessed by ultra-low-pass whole genome sequencing (ulpWGS), which does not require prior tumor sequencing.<h4>Results</h4>In this proof-of-concept study, ctDNA was detected (fraction ≥0.10) in the CSF of all 24 patients with BCLM+ (median ctDNA fraction, 0.57), regardless of negative cytology or borderline MRI imaging, whereas CSF ctDNA was not detected in the six patients with BCLM- (median ctDNA fraction 0.03, P < 0.0001). Plasma ctDNA was only detected in patients with extracranial disease progression or who had previously received whole brain radiotherapy. ctDNA fraction was highly concordant with mutant allele fraction measured by tumor mutation-specific ddPCR assays (r = 0.852; P < 0.0001). During intrathecal treatment, serial monitoring (n = 12 patients) showed that suppression of CSF ctDNA fraction was associated with longer BCLM survival (P = 0.034), and rising ctDNA fraction was detectable up to 12 weeks before clinical progression.<h4>Conclusions</h4>Measuring ctDNA fraction by ulpWGS is a quantitative marker demonstrating potential for timely and accurate BCLM diagnosis and therapy response monitoring, with the ultimate aim to improve management of this poor-prognosis patient group.

Alexander, J. Mariani, O. Meaudre, C. Fuhrmann, L. Xiao, H. Naidoo, K. Gillespie, A. Roxanis, I. Vincent-Salomon, A. Haider, S. Natrajan, R (2022) Assessment of the Molecular Heterogeneity of E-Cadherin Expression in Invasive Lobular Breast Cancer.. Show Abstract full text

Mutations and loss of E-cadherin protein expression define the vast majority of invasive lobular carcinomas. In a subset of these cases, the heterogeneous expression of E-cadherin is observed either as wild-type (strong membranous) expression or aberrant expression (cytoplasmic expression). However, it is unclear as to whether the two components would be driven by distinct genetic or epigenetic alterations. Here, we used whole genome DNA sequencing and methylation array profiling of two separately dissected components of nine invasive lobular carcinomas with heterogeneous E-cadherin expression. E-cadherin negative and aberrant/positive components of E-cadherin heterogeneous tumours showed a similar mutational, copy number and promoter methylation repertoire, suggesting they arise from a common ancestor, as opposed to the collision of two independent tumours. We found that the majority of E-cadherin heterogeneous tumours harboured <i>CDH1</i> mutations in both the E-cadherin negative and aberrant/positive components together with somatic mutations in additional driver genes known to be enriched in both pure invasive carcinomas of no special type and invasive lobular breast cancers, whereas these were less commonly observed in <i>CDH1</i> wild-type tumours. <i>CDH1</i> mutant tumours also exhibited a higher mutation burden as well as increased presence of APOBEC-dependent mutational signatures 2 and 13 compared to <i>CDH1</i> wild-type tumours. Together, our results suggest that regardless of E-cadherin protein expression, tumours showing heterogeneous expression of E-cadherin should be considered as part of the spectrum of invasive lobular breast cancers.

Gómez-Cuadrado, L. Bullock, E. Mabruk, Z. Zhao, H. Souleimanova, M. Noer, P.R. Turnbull, A.K. Oxvig, C. Bertos, N. Byron, A. Dixon, J.M. Park, M. Haider, S. Natrajan, R. Sims, A.H. Brunton, V.G (2022) Characterisation of the Stromal Microenvironment in Lobular Breast Cancer.. Show Abstract full text

Invasive lobular carcinoma (ILC) is the second most common histological subtype of breast cancer, and it exhibits a number of clinico-pathological characteristics distinct from the more common invasive ductal carcinoma (IDC). We set out to identify alterations in the tumor microenvironment (TME) of ILC. We used laser-capture microdissection to separate tumor epithelium from stroma in 23 ER+ ILC primary tumors. Gene expression analysis identified 45 genes involved in regulation of the extracellular matrix (ECM) that were enriched in the non-immune stroma of ILC, but not in non-immune stroma from ER+ IDC or normal breast. Of these, 10 were expressed in cancer-associated fibroblasts (CAFs) and were increased in ILC compared to IDC in bulk gene expression datasets, with <i>PAPPA</i> and <i>TIMP2</i> being associated with better survival in ILC but not IDC. <i>PAPPA,</i> a gene involved in IGF-1 signaling, was the most enriched in the stroma compared to the tumor epithelial compartment in ILC. Analysis of <i>PAPPA</i>- and <i>IGF1</i>-associated genes identified a paracrine signaling pathway, and active PAPP-A was shown to be secreted from primary CAFs. This is the first study to demonstrate molecular differences in the TME between ILC and IDC identifying differences in matrix organization and growth factor signaling pathways.

Stevenson, J. Barrow-McGee, R. Yu, L. Paul, A. Mansfield, D. Owen, J. Woodman, N. Natrajan, R. Haider, S. Gillett, C. Tutt, A. Pinder, S.E. Choudary, J. Naidoo, K (2021) Proteomics of REPLICANT perfusate detects changes in the metastatic lymph node microenvironment.. Show Abstract full text

In breast cancer (BC), detecting low volumes of axillary lymph node (ALN) metastasis pre-operatively is difficult and novel biomarkers are needed. We recently showed that patient-derived ALNs can be sustained ex-vivo using normothermic perfusion. We now compare reactive (tumour-free; n = 5) and macrometastatic (containing tumour deposits >2 mm; n = 4) ALNs by combining whole section multiplex immunofluorescence with TMT-labelled LC-MS/MS of the circulating perfusate. Macrometastases contained significantly fewer B cells and T cells (CD4<sup>+</sup>/CD8<sup>+</sup>/regulatory) than reactive nodes (p = 0.02). Similarly, pathway analysis of the perfusate proteome (119/1453 proteins significantly differentially expressed) showed that immune function was diminished in macrometastases in favour of 'extracellular matrix degradation'; only 'neutrophil degranulation' was preserved. Qualitative comparison of the perfusate proteome to that of node-positive pancreatic and prostatic adenocarcinoma also highlighted 'neutrophil degranulation' as a contributing factor to nodal metastasis. Thus, metastasis-induced changes in the REPLICANT perfusate proteome are detectable, and could facilitate biomarker discovery.

Sipos, O. Tovey, H. Quist, J. Haider, S. Nowinski, S. Gazinska, P. Kernaghan, S. Toms, C. Maguire, S. Orr, N. Linn, S.C. Owen, J. Gillett, C. Pinder, S.E. Bliss, J.M. Tutt, A. Cheang, M.C.U. Grigoriadis, A (2021) Assessment of structural chromosomal instability phenotypes as biomarkers of carboplatin response in triple negative breast cancer: the TNT trial.. Show Abstract full text

<h4>Background</h4>In the TNT trial of triple negative breast cancer (NCT00532727), germline BRCA1/2 mutations were present in 28% of carboplatin responders. We assessed quantitative measures of structural chromosomal instability (CIN) to identify a wider patient subgroup within TNT with preferential benefit from carboplatin over docetaxel.<h4>Patients and methods</h4>Copy number aberrations (CNAs) were established from 135 formalin-fixed paraffin-embedded primary carcinomas using Illumina OmniExpress SNP-arrays. Seven published [allelic imbalanced CNA (AiCNA); allelic balanced CNA (AbCNA); copy number neutral loss of heterozygosity (CnLOH); number of telomeric allelic imbalances (NtAI); BRCA1-like status; percentage of genome altered (PGA); homologous recombination deficiency (HRD) scores] and two novel [Shannon diversity index (SI); high-level amplifications (HLAMP)] CIN-measurements were derived. HLAMP was defined based on the presence of at least one of the top 5% amplified cytobands located on 1q, 8q and 10p. Continuous CIN-measurements were divided into tertiles. All nine CIN-measurements were used to analyse objective response rate (ORR) and progression-free survival (PFS).<h4>Results</h4>Patients with tumours without HLAMP had a numerically higher ORR and significantly longer PFS in the carboplatin (C) than in the docetaxel (D) arm [56% (C) versus 29% (D), P<sub>HLAMP,quiet</sub> = 0.085; PFS 6.1 months (C) versus 4.1 months (D), P<sub>interaction/HLAMP</sub> = 0.047]. In the carboplatin arm, patients with tumours showing intermediate telomeric NtAI and AiCNA had higher ORR [54% (C) versus 20% (D), P<sub>NtAI,intermediate</sub> = 0.03; 62% (C) versus 33% (D), P<sub>AiCNA,intermediate</sub> = 0.076]. Patients with high AiCNA and PGA had shorter PFS in the carboplatin arm [3.4 months (high) versus 5.7 months (low/intermediate); and 3.8 months (high) versus 5.6 months (low/intermediate), respectively; P<sub>interaction/AiCNA</sub> = 0.027, P<sub>adj.interaction/AiCNA</sub> = 0.125 and P<sub>interaction/PGA</sub> = 0.053, P<sub>adj.interaction/PGA</sub> = 0.176], whilst no difference was observed in the docetaxel arm.<h4>Conclusions</h4>Patients with tumours lacking HLAMP and demonstrating intermediate CIN-measurements formed a subgroup benefitting from carboplatin relative to docetaxel treatment within the TNT trial. This suggests a complex and paradoxical relationship between the extent of genomic instability in primary tumours and treatment response in the metastatic setting.

Cappello, P. Blaser, H. Gorrini, C. Lin, D.C.C. Elia, A.J. Wakeham, A. Haider, S. Boutros, P.C. Mason, J.M. Miller, N.A. Youngson, B. Done, S.J. Mak, T.W (2014) Role of Nek2 on centrosome duplication and aneuploidy in breast cancer cells.. Show Abstract full text

Breast cancer is the most common solid tumor and the second most common cause of death in women. Despite a large body of literature and progress in breast cancer research, many molecular aspects of this complex disease are still poorly understood, hindering the design of specific and effective therapeutic strategies. To identify the molecules important in breast cancer progression and metastasis, we tested the in vivo effects of inhibiting the functions of various kinases and genes involved in the regulation/modulation of the cytoskeleton by downregulating them in mouse PyMT mammary tumor cells and human breast cancer cell lines. These kinases and cytoskeletal regulators were selected based on their prognostic values for breast cancer patient survival. PyMT tumor cells, in which a selected gene was stably knocked down were injected into the tail veins of mice, and the formation of tumors in the lungs was monitored. One of the several genes found to be important for tumor growth in the lungs was NIMA-related kinases 2 (Nek2), a cell cycle-related protein kinase. Furthermore, Nek2 was also important for tumor growth in the mammary fat pad. In various human breast cancer cell lines, Nek2 knockdown induced aneuploidy and cell cycle arrest that led to cell death. Significantly, the breast cancer cell line most sensitive to Nek2 depletion was of the triple negative breast cancer subtype. Our data indicate that Nek2 has a pivotal role in breast cancer growth at primary and secondary sites, and thus may be an attractive and novel therapeutic target for this disease.

Gazinska, P. Milton, C. Iacovacci, J. Ward, J. Buus, R. Alaguthurai, T. Graham, R. Akarca, A. Lips, E. Naidoo, K. Wesseling, J. Marafioti, T. Cheang, M. Gillett, C. Wu, Y. Khan, A. Melcher, A. Salgado, R. Dowsett, M. Tutt, A. Roxanis, I. Haider, S. Irshad, S (2022) Dynamic Changes in the NK-, Neutrophil-, and B-cell Immunophenotypes Relevant in High Metastatic Risk Post Neoadjuvant Chemotherapy-Resistant Early Breast Cancers.. Show Abstract full text

<h4>Purpose</h4>To identify potential immune targets in post-neoadjuvant chemotherapy (NAC)-resistant triple-negative breast cancer (TNBC) and ER+HER2- breast cancer disease.<h4>Experimental design</h4>Following pathology review, 153 patients were identified as having residual cancer burden (RCB) II/III disease (TNBC n = 80; ER+HER2-n = 73). Baseline pre-NAC samples were available for evaluation for 32 of 80 TNBC and 36 of 73 ER+HER2- cases. Bright-field hematoxylin and eosin assessment allowed for tumor-infiltrating lymphocyte (TIL) evaluation in all cases. Multiplexed immunofluorescence was used to identify the abundance and distribution of immune cell subsets. Levels of checkpoints including PD-1/PD-L1 expression were also quantified. Findings were then validated using expression profiling of cancer and immune-related genes. Cytometry by time-of-flight characterized the dynamic changes in circulating immune cells with NAC.<h4>Results</h4>RCB II/III TNBC and ER+HER2- breast cancer were immunologically "cold" at baseline and end of NAC. Although the distribution of immune cell subsets across subtypes was similar, the mRNA expression profiles were both subtype- and chemotherapy-specific. TNBC RCB II/III disease was enriched with genes related to neutrophil degranulation, and displayed strong interplay across immune and cancer pathways. We observed similarities in the dynamic changes in B-cell biology following NAC irrespective of subtype. However, NAC induced changes in the local and circulating tumor immune microenvironment (TIME) that varied by subtype and response. Specifically, in TNBC residual disease, we observed downregulation of stimulatory (CD40/OX40L) and inhibitory (PD-L1/PD-1) receptor expression and an increase in NK cell populations (especially non-cytolytic, exhausted CD56dimCD16-) within both the local TIME and peripheral white cell populations.<h4>Conclusions</h4>This study identifies several potential immunologic pathways in residual disease, which may be targeted to benefit high-risk patients.

Llorca-Cardenosa, M.J. Aronson, L.I. Krastev, D.B. Nieminuszczy, J. Alexander, J. Song, F. Dylewska, M. Broderick, R. Brough, R. Zimmermann, A. Zenke, F.T. Gurel, B. Riisnaes, R. Ferreira, A. Roumeliotis, T. Choudhary, J. Pettitt, S.J. de Bono, J. Cervantes, A. Haider, S. Niedzwiedz, W. Lord, C.J. Chong, I.Y (2022) SMG8/SMG9 Heterodimer Loss Modulates SMG1 Kinase to Drive ATR Inhibitor Resistance.. Show Abstract full text

Gastric cancer represents the third leading cause of global cancer mortality and an area of unmet clinical need. Drugs that target the DNA damage response, including ATR inhibitors (ATRi), have been proposed as novel targeted agents in gastric cancer. Here, we sought to evaluate the efficacy of ATRi in preclinical models of gastric cancer and to understand how ATRi resistance might emerge as a means to identify predictors of ATRi response. A positive selection genome-wide CRISPR-Cas9 screen identified candidate regulators of ATRi resistance in gastric cancer. Loss-of-function mutations in either SMG8 or SMG9 caused ATRi resistance by an SMG1-mediated mechanism. Although ATRi still impaired ATR/CHK1 signaling in SMG8/9-defective cells, other characteristic responses to ATRi exposure were not seen, such as changes in ATM/CHK2, γH2AX, phospho-RPA, or 53BP1 status or changes in the proportions of cells in S- or G2-M-phases of the cell cycle. Transcription/replication conflicts (TRC) elicited by ATRi exposure are a likely cause of ATRi sensitivity, and SMG8/9-defective cells exhibited a reduced level of ATRi-induced TRCs, which could contribute to ATRi resistance. These observations suggest ATRi elicits antitumor efficacy in gastric cancer but that drug resistance could emerge via alterations in the SMG8/9/1 pathway.<h4>Significance</h4>These findings reveal how cancer cells acquire resistance to ATRi and identify pathways that could be targeted to enhance the overall effectiveness of these inhibitors.

Jenkins, L. Jungwirth, U. Avgustinova, A. Iravani, M. Mills, A. Haider, S. Harper, J. Isacke, C.M (2022) Cancer-Associated Fibroblasts Suppress CD8+ T-cell Infiltration and Confer Resistance to Immune-Checkpoint Blockade.. Show Abstract full text

Immune-checkpoint blockade (ICB) promotes antitumor immune responses and can result in durable patient benefit. However, response rates in breast cancer patients remain modest, stimulating efforts to discover novel treatment options. Cancer-associated fibroblasts (CAF) represent a major component of the breast tumor microenvironment and have known immunosuppressive functions in addition to their well-established roles in directly promoting tumor growth and metastasis. Here we utilized paired syngeneic mouse mammary carcinoma models to show that CAF abundance is associated with insensitivity to combination αCTLA4 and αPD-L1 ICB. CAF-rich tumors exhibited an immunologically cold tumor microenvironment, with transcriptomic, flow cytometric, and quantitative histopathologic analyses demonstrating a relationship between CAF density and a CD8+ T-cell-excluded tumor phenotype. The CAF receptor Endo180 (Mrc2) is predominantly expressed on myofibroblastic CAFs, and its genetic deletion depleted a subset of αSMA-expressing CAFs and impaired tumor progression in vivo. The addition of wild-type, but not Endo180-deficient, CAFs in coimplantation studies restricted CD8+ T-cell intratumoral infiltration, and tumors in Endo180 knockout mice exhibited increased CD8+ T-cell infiltration and enhanced sensitivity to ICB compared with tumors in wild-type mice. Clinically, in a trial of melanoma patients, high MRC2 mRNA levels in tumors were associated with a poor response to αPD-1 therapy, highlighting the potential benefits of therapeutically targeting a specific CAF subpopulation in breast and other CAF-rich cancers to improve clinical responses to immunotherapy.<h4>Significance</h4>Paired syngeneic models help unravel the interplay between CAF and tumor immune evasion, highlighting the benefits of targeting fibroblast subpopulations to improve clinical responses to immunotherapy.

Turrell, F.K. Orha, R. Guppy, N.J. Gillespie, A. Guelbert, M. Starling, C. Haider, S. Isacke, C.M (2023) Age-associated microenvironmental changes highlight the role of PDGF-C in ER<sup>+</sup> breast cancer metastatic relapse.. Show Abstract full text

Patients with estrogen receptor (ER)-positive breast cancer are at risk of metastatic relapse for decades after primary tumor resection and treatment, a consequence of dormant disseminated tumor cells (DTCs) reawakening at secondary sites. Here we use syngeneic ER<sup>+</sup> mouse models in which DTCs display a dormant phenotype in young mice but accelerated metastatic outgrowth in an aged or fibrotic microenvironment. In young mice, low-level Pdgfc expression by ER<sup>+</sup> DTCs is required for their maintenance in secondary sites but is insufficient to support development of macrometastases. By contrast, the platelet-derived growth factor (PDGF)-C<sup>hi</sup> environment of aging or fibrotic lungs promotes DTC proliferation and upregulates tumor cell Pdgfc expression stimulating further stromal activation, events that can be blocked by pharmacological inhibition of PDGFRα or with a PDGF-C-blocking antibody. These results highlight the role of the changing microenvironment in regulating DTC outgrowth and the opportunity to target PDGF-C signaling to limit metastatic relapse in ER<sup>+</sup> breast cancer.

Tovey, H. Sipos, O. Parker, J.S. Hoadley, K.A. Quist, J. Kernaghan, S. Kilburn, L. Salgado, R. Loi, S. Kennedy, R.D. Roxanis, I. Gazinska, P. Pinder, S.E. Bliss, J. Perou, C.M. Haider, S. Grigoriadis, A. Tutt, A. Cheang, M.C.U (2023) Integrated Multimodal Analyses of DNA Damage Response and Immune Markers as Predictors of Response in Metastatic Triple-Negative Breast Cancer in the TNT Trial (NCT00532727).. Show Abstract full text

<h4>Purpose</h4>The TNT trial (NCT00532727) showed no evidence of carboplatin superiority over docetaxel in metastatic triple-negative breast cancer (mTNBC), but carboplatin benefit was observed in the germline BRCA1/2 mutation subgroup. Broader response-predictive biomarkers are needed. We explored the predictive ability of DNA damage response (DDR) and immune markers.<h4>Experimental design</h4>Tumor-infiltrating lymphocytes were evaluated for 222 of 376 patients. Primary tumors (PT) from 186 TNT participants (13 matched recurrences) were profiled using total RNA sequencing. Four transcriptional DDR-related and 25 immune-related signatures were evaluated. We assessed their association with objective response rate (ORR) and progression-free survival (PFS). Conditional inference forest clustering was applied to integrate multimodal data. The biology of subgroups was characterized by 693 gene expression modules and other markers.<h4>Results</h4>Transcriptional DDR-related biomarkers were not predictive of ORR to either treatment overall. Changes from PT to recurrence were demonstrated; in chemotherapy-naïve patients, transcriptional DDR markers separated carboplatin responders from nonresponders (P values = 0.017; 0.046). High immune infiltration was associated with docetaxel ORR (interaction P values < 0.05). Six subgroups were identified; the immune-enriched cluster had preferential docetaxel response [62.5% (D) vs. 29.4% (C); P = 0.016]. The immune-depleted cluster had preferential carboplatin response [8.0% (D) vs. 40.0% (C); P = 0.011]. DDR-related subgroups were too small to assess ORR.<h4>Conclusions</h4>High immune features predict docetaxel response, and high DDR signature scores predict carboplatin response in treatment-naïve mTNBC. Integrating multimodal DDR and immune-related markers identifies subgroups with differential treatment sensitivity. Treatment options for patients with immune-low and DDR-proficient tumors remains an outstanding need. Caution is needed using PT-derived transcriptional signatures to direct treatment in mTNBC, particularly DDR-related markers following prior chemotherapy.

Bland, P. Saville, H. Wai, P.T. Curnow, L. Muirhead, G. Nieminuszczy, J. Ravindran, N. John, M.B. Hedayat, S. Barker, H.E. Wright, J. Yu, L. Mavrommati, I. Read, A. Peck, B. Allen, M. Gazinska, P. Pemberton, H.N. Gulati, A. Nash, S. Noor, F. Guppy, N. Roxanis, I. Pratt, G. Oldreive, C. Stankovic, T. Barlow, S. Kalirai, H. Coupland, S.E. Broderick, R. Alsafadi, S. Houy, A. Stern, M.-.H. Pettit, S. Choudhary, J.S. Haider, S. Niedzwiedz, W. Lord, C.J. Natrajan, R (2023) SF3B1 hotspot mutations confer sensitivity to PARP inhibition by eliciting a defective replication stress response.. Show Abstract full text

SF3B1 hotspot mutations are associated with a poor prognosis in several tumor types and lead to global disruption of canonical splicing. Through synthetic lethal drug screens, we identify that SF3B1 mutant (SF3B1<sup>MUT</sup>) cells are selectively sensitive to poly (ADP-ribose) polymerase inhibitors (PARPi), independent of hotspot mutation and tumor site. SF3B1<sup>MUT</sup> cells display a defective response to PARPi-induced replication stress that occurs via downregulation of the cyclin-dependent kinase 2 interacting protein (CINP), leading to increased replication fork origin firing and loss of phosphorylated CHK1 (pCHK1; S317) induction. This results in subsequent failure to resolve DNA replication intermediates and G<sub>2</sub>/M cell cycle arrest. These defects are rescued through CINP overexpression, or further targeted by a combination of ataxia-telangiectasia mutated and PARP inhibition. In vivo, PARPi produce profound antitumor effects in multiple SF3B1<sup>MUT</sup> cancer models and eliminate distant metastases. These data provide the rationale for testing the clinical efficacy of PARPi in a biomarker-driven, homologous recombination proficient, patient population.

Zelceski, A. Francica, P. Lingg, L. Mutlu, M. Stok, C. Liptay, M. Alexander, J. Baxter, J.S. Brough, R. Gulati, A. Haider, S. Raghunandan, M. Song, F. Sridhar, S. Forment, J.V. O'Connor, M.J. Davies, B.R. van Vugt, M.A.T.M. Krastev, D.B. Pettitt, S.J. Tutt, A.N.J. Rottenberg, S. Lord, C.J (2023) MND1 and PSMC3IP control PARP inhibitor sensitivity in mitotic cells.. Show Abstract full text

The PSMC3IP-MND1 heterodimer promotes meiotic D loop formation before DNA strand exchange. In genome-scale CRISPR-Cas9 mutagenesis and interference screens in mitotic cells, depletion of PSMC3IP or MND1 causes sensitivity to poly (ADP-Ribose) polymerase inhibitors (PARPi) used in cancer treatment. PSMC3IP or MND1 depletion also causes ionizing radiation sensitivity. These effects are independent of PSMC3IP/MND1's role in mitotic alternative lengthening of telomeres. PSMC3IP- or MND1-depleted cells accumulate toxic RAD51 foci in response to DNA damage, show impaired homology-directed DNA repair, and become PARPi sensitive, even in cells lacking both BRCA1 and TP53BP1. Epistasis between PSMC3IP-MND1 and BRCA1/BRCA2 defects suggest that abrogated D loop formation is the cause of PARPi sensitivity. Wild-type PSMC3IP reverses PARPi sensitivity, whereas a PSMC3IP p.Glu201del mutant associated with D loop defects and ovarian dysgenesis does not. These observations suggest that meiotic proteins such as MND1 and PSMC3IP have a greater role in mitotic DNA repair.

Thakur, S. Haider, S. Natrajan, R (2023) Implications of tumour heterogeneity on cancer evolution and therapy resistance: lessons from breast cancer.. Show Abstract full text

Tumour heterogeneity is pervasive amongst many cancers and leads to disease progression, and therapy resistance. In this review, using breast cancer as an exemplar, we focus on the recent advances in understanding the interplay between tumour cells and their microenvironment using single cell sequencing and digital spatial profiling technologies. Further, we discuss the utility of lineage tracing methodologies in pre-clinical models of breast cancer, and how these are being used to unravel new therapeutic vulnerabilities and reveal biomarkers of breast cancer progression. © 2023 The Pathological Society of Great Britain and Ireland.

Perkins, D.W. Steiner, I. Haider, S. Robertson, D. Buus, R. O'Leary, L. Isacke, C.M (2024) Therapy-induced normal tissue damage promotes breast cancer metastasis.. Show Abstract full text

Disseminated tumor cells frequently exhibit a period of dormancy, rendering them chemotherapy insensitive; conversely, the systemic delivery of chemotherapies can result in normal tissue damage. Using multiple mouse and human breast cancer models, we demonstrate that prior chemotherapy administration enhances metastatic colonization and outgrowth. <i>In vitro</i>, chemotherapy-treated fibroblasts display a pro-tumorigenic senescence-associated secretory phenotype (SASP) and are effectively eliminated by targeting the anti-apoptotic protein BCL-xL. <i>In vivo</i>, chemotherapy treatment induces SASP expression in normal tissues; however, the accumulation of senescent cells is limited, and BCL-xL inhibitors are unable to reduce chemotherapy-enhanced metastasis. This likely reflects that chemotherapy-exposed stromal cells do not enter a BCL-xL-dependent phenotype or switch their dependency to other anti-apoptotic BCL-2 family members. This study highlights the role of the metastatic microenvironment in controlling outgrowth of disseminated tumor cells and the need to identify additional approaches to limit the pro-tumorigenic effects of therapy-induced normal tissue damage.

Harvey-Jones, E. Raghunandan, M. Robbez-Masson, L. Magraner-Pardo, L. Alaguthurai, T. Yablonovitch, A. Yen, J. Xiao, H. Brough, R. Frankum, J. Song, F. Yeung, J. Savy, T. Gulati, A. Alexander, J. Kemp, H. Starling, C. Konde, A. Marlow, R. Cheang, M. Proszek, P. Hubank, M. Cai, M. Trendell, J. Lu, R. Liccardo, R. Ravindran, N. Llop-Guevara, A. Rodriguez, O. Balmana, J. Lukashchuk, N. Dorschner, M. Drusbosky, L. Roxanis, I. Serra, V. Haider, S. Pettitt, S.J. Lord, C.J. Tutt, A.N.J (2024) Longitudinal profiling identifies co-occurring BRCA1/2 reversions, TP53BP1, RIF1 and PAXIP1 mutations in PARP inhibitor-resistant advanced breast cancer.. Show Abstract full text

<h4>Background</h4>Resistance to therapies that target homologous recombination deficiency (HRD) in breast cancer limits their overall effectiveness. Multiple, preclinically validated, mechanisms of resistance have been proposed, but their existence and relative frequency in clinical disease are unclear, as is how to target resistance.<h4>Patients and methods</h4>Longitudinal mutation and methylation profiling of circulating tumour (ct)DNA was carried out in 47 patients with metastatic BRCA1-, BRCA2- or PALB2-mutant breast cancer treated with HRD-targeted therapy who developed progressive disease-18 patients had primary resistance and 29 exhibited response followed by resistance. ctDNA isolated at multiple time points in the patient treatment course (before, on-treatment and at progression) was sequenced using a novel >750-gene intron/exon targeted sequencing panel. Where available, matched tumour biopsies were whole exome and RNA sequenced and also used to assess nuclear RAD51.<h4>Results</h4>BRCA1/2 reversion mutations were present in 60% of patients and were the most prevalent form of resistance. In 10 cases, reversions were detected in ctDNA before clinical progression. Two new reversion-based mechanisms were identified: (i) intragenic BRCA1/2 deletions with intronic breakpoints; and (ii) intragenic BRCA1/2 secondary mutations that formed novel splice acceptor sites, the latter being confirmed by in vitro minigene reporter assays. When seen before commencing subsequent treatment, reversions were associated with significantly shorter time to progression. Tumours with reversions retained HRD mutational signatures but had functional homologous recombination based on RAD51 status. Although less frequent than reversions, nonreversion mechanisms [loss-of-function (LoF) mutations in TP53BP1, RIF1 or PAXIP1] were evident in patients with acquired resistance and occasionally coexisted with reversions, challenging the notion that singular resistance mechanisms emerge in each patient.<h4>Conclusions</h4>These observations map the prevalence of candidate drivers of resistance across time in a clinical setting, information with implications for clinical management and trial design in HRD breast cancers.

Fitzpatrick, A. Iravani, M. Mills, A. Vicente, D. Alaguthurai, T. Roxanis, I. Turner, N.C. Haider, S. Tutt, A.N.J. Isacke, C.M (2023) Genomic profiling and pre-clinical modelling of breast cancer leptomeningeal metastasis reveals acquisition of a lobular-like phenotype.. Show Abstract full text

Breast cancer leptomeningeal metastasis (BCLM), where tumour cells grow along the lining of the brain and spinal cord, is a devastating development for patients. Investigating this metastatic site is hampered by difficulty in accessing tumour material. Here, we utilise cerebrospinal fluid (CSF) cell-free DNA (cfDNA) and CSF disseminated tumour cells (DTCs) to explore the clonal evolution of BCLM and heterogeneity between leptomeningeal and extracranial metastatic sites. Somatic alterations with potential therapeutic actionability were detected in 81% (17/21) of BCLM cases, with 19% detectable in CSF cfDNA only. BCLM was enriched in genomic aberrations in adherens junction and cytoskeletal genes, revealing a lobular-like breast cancer phenotype. CSF DTCs were cultured in 3D to establish BCLM patient-derived organoids, and used for the successful generation of BCLM in vivo models. These data reveal that BCLM possess a unique genomic aberration profile and highlight potential cellular dependencies in this hard-to-treat form of metastatic disease.

Baxter, J.S. Brough, R. Krastev, D.B. Song, F. Sridhar, S. Gulati, A. Alexander, J. Roumeliotis, T.I. Kozik, Z. Choudhary, J.S. Haider, S. Pettitt, S.J. Tutt, A.N.J. Lord, C.J (2024) Cancer-associated FBXW7 loss is synthetic lethal with pharmacological targeting of CDC7.. Show Abstract full text

The F-box and WD repeat domain containing 7 (FBXW7) tumour suppressor gene encodes a substrate-recognition subunit of Skp, cullin, F-box (SCF)-containing complexes. The tumour-suppressive role of FBXW7 is ascribed to its ability to drive ubiquitination and degradation of oncoproteins. Despite this molecular understanding, therapeutic approaches that target defective FBXW7 have not been identified. Using genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 screens, focussed RNA-interference screens and whole and phospho-proteome mass spectrometry profiling in multiple FBXW7 wild-type and defective isogenic cell lines, we identified a number of FBXW7 synthetic lethal targets, including proteins involved in the response to replication fork stress and proteins involved in replication origin firing, such as cell division cycle 7-related protein kinase (CDC7) and its substrate, DNA replication complex GINS protein SLD5 (GINS4). The CDC7 synthetic lethal effect was confirmed using small-molecule inhibitors. Mechanistically, FBXW7/CDC7 synthetic lethality is dependent upon the replication factor telomere-associated protein RIF1 (RIF1), with RIF1 silencing reversing the FBXW7-selective effects of CDC7 inhibition. The delineation of FBXW7 synthetic lethal effects we describe here could serve as the starting point for subsequent drug discovery and/or development in this area.