Dr Stephen Pettitt

Senior Staff Scientist and Deputy Group Leader:

Phone: 020 7153 5322

Email: [email protected]

Also on:  stveep

Phone: 020 7153 5322

Email: [email protected]

Also on:  stveep

Biography and research overview

Dr Stephen Pettitt is a Staff Scientist and Deputy Group Leader of the Gene Function Group. He uses experimental genetic approaches to investigate the function of genes that are mutated in cancer. Cancer cells can have thousands of mutations in their DNA when compared to normal cells, making it difficult to know which mutated genes are responsible for their different characteristics. Using an experimental approach where mutations are introduced individually removes some of this complexity associated with analysing cancer genomes.

Dr Pettitt uses CRISPR mutagenesis, where an enzyme that cuts DNA can be programmed to introduce mutations at a site specified by a short RNA molecule that binds to the enzyme and guides it to its DNA target site. This has a wide variety of uses in research, such as in genetic screens, where each of the roughly 20,000 genes is mutated individually using a different guide RNA. This results in a “library” of cells containing mutants for each gene – if any of these mutants show different properties, it is likely that this is due to the mutated gene.

One property that Dr Pettitt is particularly interested in is drug sensitivity or resistance – if the genetic profiles that are responsible for these responses can be identified, this information can be used to better target cancer drugs. Dr Pettitt is particularly interested in drugs targeting the DNA damage response, such as PARP inhibitors. He has used CRISPR mutant libraries to isolate PARP inhibitor resistant cells, identifying several new genes that result in PARP inhibitor resistance when mutated. He has also developed techniques for dense CRISPR mutagenesis, focused on a single gene, to map resistance-causing mutations in fine detail.

In parallel with his laboratory studies, Dr Pettitt analyses DNA sequencing data from patients involved in clinical trials of new agents to see if predictions of drug resistance or sensitivity from lab experiments can be informative in the clinic. These studies also generate new hypotheses to be tested in the lab.

Pettitt SJ et al., A Genetic Screen Using the PiggyBac Transposon in Haploid Cells Identifies Parp1 as a Mediator of Olaparib Toxicity. PloS One (2013) 8: e61520, doi:10.1371/journal.pone.0061520.

Pettitt, SJ, Krastev, DB et al., Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. bioRxiv, 203224 (2017)

Types of Publications

Journal articles

Pettitt, S.J. Rehman, F.L. Bajrami, I. Brough, R. Wallberg, F. Kozarewa, I. Fenwick, K. Assiotis, I. Chen, L. Campbell, J. Lord, C.J. Ashworth, A (2013) A genetic screen using the PiggyBac transposon in haploid cells identifies Parp1 as a mediator of olaparib toxicity.. Show Abstract full text

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.

Pettitt, S.J. Liang, Q. Rairdan, X.Y. Moran, J.L. Prosser, H.M. Beier, D.R. Lloyd, K.C. Bradley, A. Skarnes, W.C (2009) Agouti C57BL/6N embryonic stem cells for mouse genetic resources.. Show Abstract full text

We report the characterization of a highly germline competent C57BL/6N mouse embryonic stem cell line, JM8. To simplify breeding schemes, the dominant agouti coat color gene was restored in JM8 cells by targeted repair of the C57BL/6 nonagouti mutation. These cells provide a robust foundation for large-scale mouse knockout programs that aim to provide a public resource of targeted mutations in the C57BL/6 genetic background.

Li, M.A. Pettitt, S.J. Eckert, S. Ning, Z. Rice, S. Cadiñanos, J. Yusa, K. Conte, N. Bradley, A (2013) The piggyBac transposon displays local and distant reintegration preferences and can cause mutations at noncanonical integration sites.. Show Abstract full text

The DNA transposon piggyBac is widely used as a tool in mammalian experimental systems for transgenesis, mutagenesis, and genome engineering. We have characterized genome-wide insertion site preferences of piggyBac by sequencing a large set of integration sites arising from transposition from two separate genomic loci and a plasmid donor in mouse embryonic stem cells. We found that piggyBac preferentially integrates locally to the excision site when mobilized from a chromosomal location and identified other nonlocal regions of the genome with elevated insertion frequencies. piggyBac insertions were associated with expressed genes and markers of open chromatin structure and were excluded from heterochromatin. At the nucleotide level, piggyBac prefers to insert into TA-rich regions within a broader GC-rich context. We also found that piggyBac can insert into sites other than its known TTAA insertion site at a low frequency (2%). Such insertions introduce mismatches that are repaired with signatures of host cell repair pathways. Transposons could be mobilized from plasmids with the observed noncanonical flanking regions, indicating that piggyBac could generate point mutations in the genome.

Pettitt, S.J. Rehman, F.L. Bajrami, I. Pemberton, H. Brough, R. Kozarewa, I. Lord, C.J. Ashworth, A (2013) A transposon-based genetic screen in haploid mouse embryonic stem cells identifies Parp1 as a major mediator of olaparib toxicity. full text
Li, M.A. Pettitt, S.J. Yusa, K. Bradley, A (2010) Genome-wide forward genetic screens in mouse ES cells.. Show Abstract full text

Mouse embryonic stem (ES) cells are an attractive model system for investigating mammalian biology. Their relatively stable genome and high amenability to genome modification enables the generation of large-scale mutant libraries, which can be subsequently used for phenotype-driven genetic screens. While retroviral vectors have traditionally been used to generate insertional mutations in ES cells, their severe distribution-bias in the mammalian genome substantially limits genome-wide mutagenesis. The recent development of the DNA transposon piggyBac offers an efficient and highly versatile alternative for achieving more unbiased mutagenesis. Furthermore, heterozygous mutations created by insertional mutagens can be converted in parallel to homozygosity by using Blm-deficient ES cells, allowing genome-wide loss-of-function screens to be conducted. In this chapter, we describe the principles underpinning genetic screens in mouse ES cells with examples of previously successful screens. Protocols are provided for piggyBac transposon-mediated mutagenesis, production of the corresponding homozygous mutants in a Blm-deficient genetic background, and methods for mapping and validation of mutations recovered from screens of such libraries.

Huang, Y. Pettitt, S.J. Guo, G. Liu, G. Li, M.A. Yang, F. Bradley, A (2012) Isolation of homozygous mutant mouse embryonic stem cells using a dual selection system.. Show Abstract full text

Obtaining random homozygous mutants in mammalian cells for forward genetic studies has always been problematic due to the diploid genome. With one mutation per cell, only one allele of an autosomal gene can be disrupted, and the resulting heterozygous mutant is unlikely to display a phenotype. In cells with a genetic background deficient for the Bloom's syndrome helicase, such heterozygous mutants segregate homozygous daughter cells at a low frequency due to an elevated rate of crossover following mitotic recombination between homologous chromosomes. We constructed DNA vectors that are selectable based on their copy number and used these to isolate these rare homozygous mutant cells independent of their phenotype. We use the piggyBac transposon to limit the initial mutagenesis to one copy per cell, and select for cells that have increased the transposon copy number to two or more. This yields homozygous mutants with two allelic mutations, but also cells that have duplicated the mutant chromosome and become aneuploid during culture. On average, 26% of the copy number gain events occur by the mitotic recombination pathway. We obtained homozygous cells from 40% of the heterozygous mutants tested. This method can provide homozygous mammalian loss-of-function mutants for forward genetic applications.

Pettitt, S.J. Tan, E.-.P. Yusa, K (2015) piggyBac transposon-based insertional mutagenesis in mouse haploid embryonic stem cells.. Show Abstract full text

Forward genetic screening is a powerful non-hypothesis-driven approach to unveil the molecular mechanisms and pathways underlying phenotypes of interest. In this approach, a genome-wide mutant library is first generated and then screened for a phenotype of interest. Subsequently, genes responsible for the phenotype are identified. There have been a number of successful screens in yeasts, Caenorhabditis elegans and Drosophila. These model organisms all allow loss-of-function mutants to be generated easily on a genome-wide scale: yeasts have a haploid stage in their reproductive cycles and the latter two organisms have short generation times, allowing mutations to be systematically bred to homozygosity. However, in mammals, the diploid genome and long generation time have always hampered rapid and efficient production of homozygous mutant cells and animals. The recent discovery of several haploid mammalian cell lines promises to revolutionize recessive genetic screens in mammalian cells. In this protocol, we describe an overview of insertional mutagenesis, focusing on DNA transposons, and provide a method for an efficient generation of genome-wide mutant libraries using mouse haploid embryonic stem cells.

Pettitt, S.J. Krastev, D.B. Pemberton, H.N. Fontebasso, Y. Frankum, J. Rehman, F.L. Brough, R. Song, F. Bajrami, I. Rafiq, R. Wallberg, F. Kozarewa, I. Fenwick, K. Armisen-Garrido, J. Swain, A. Gulati, A. Campbell, J. Ashworth, A. Lord, C.J (2017) Genome-wide barcoded transposon screen for cancer drug sensitivity in haploid mouse embryonic stem cells.. Show Abstract full text

We describe a screen for cellular response to drugs that makes use of haploid embryonic stem cells. We generated ten libraries of mutants with piggyBac gene trap transposon integrations, totalling approximately 100,000 mutant clones. Random barcode sequences were inserted into the transposon vector to allow the number of cells bearing each insertion to be measured by amplifying and sequencing the barcodes. These barcodes were associated with their integration sites by inverse PCR. We exposed these libraries to commonly used cancer drugs and profiled changes in barcode abundance by Ion Torrent sequencing in order to identify mutations that conferred sensitivity. Drugs tested included conventional chemotherapeutics as well as targeted inhibitors of topoisomerases, poly(ADP-ribose) polymerase (PARP), Hsp90 and WEE1.

Nikkilä, J. Kumar, R. Campbell, J. Brandsma, I. Pemberton, H.N. Wallberg, F. Nagy, K. Scheer, I. Vertessy, B.G. Serebrenik, A.A. Monni, V. Harris, R.S. Pettitt, S.J. Ashworth, A. Lord, C.J (2017) Elevated APOBEC3B expression drives a kataegic-like mutation signature and replication stress-related therapeutic vulnerabilities in p53-defective cells.. Show Abstract full text

<h4>Background</h4>Elevated APOBEC3B expression in tumours correlates with a kataegic pattern of localised hypermutation. We assessed the cellular phenotypes associated with high-level APOBEC3B expression and the influence of p53 status on these phenotypes using an isogenic system.<h4>Methods</h4>We used RNA interference of p53 in cells with inducible APOBEC3B and assessed DNA damage response (DDR) biomarkers. The mutational effects of APOBEC3B were assessed using whole-genome sequencing. In vitro small-molecule inhibitor sensitivity profiling was used to identify candidate therapeutic vulnerabilities.<h4>Results</h4>Although APOBEC3B expression increased the incorporation of genomic uracil, invoked DDR biomarkers and caused cell cycle arrest, inactivation of p53 circumvented APOBEC3B-induced cell cycle arrest without reversing the increase in genomic uracil or DDR biomarkers. The continued expression of APOBEC3B in p53-defective cells not only caused a kataegic mutational signature but also caused hypersensitivity to small-molecule DDR inhibitors (ATR, CHEK1, CHEK2, PARP, WEE1 inhibitors) as well as cisplatin/ATR inhibitor and ATR/PARP inhibitor combinations.<h4>Conclusions</h4>Although loss of p53 might allow tumour cells to tolerate elevated APOBEC3B expression, continued expression of this enzyme might impart a number of therapeutic vulnerabilities upon tumour cells.

Dréan, A. Williamson, C.T. Brough, R. Brandsma, I. Menon, M. Konde, A. Garcia-Murillas, I. Pemberton, H.N. Frankum, J. Rafiq, R. Badham, N. Campbell, J. Gulati, A. Turner, N.C. Pettitt, S.J. Ashworth, A. Lord, C.J (2017) Modeling Therapy Resistance in <i>BRCA1/2</i>-Mutant Cancers.. Show Abstract full text

Although PARP inhibitors target <i>BRCA1</i>- or <i>BRCA2</i>-mutant tumor cells, drug resistance is a problem. PARP inhibitor resistance is sometimes associated with the presence of secondary or "revertant" mutations in <i>BRCA1</i> or <i>BRCA2</i> Whether secondary mutant tumor cells are selected for in a Darwinian fashion by treatment is unclear. Furthermore, how PARP inhibitor resistance might be therapeutically targeted is also poorly understood. Using CRISPR mutagenesis, we generated isogenic tumor cell models with secondary <i>BRCA1</i> or <i>BRCA2</i> mutations. Using these in heterogeneous <i>in vitro</i> culture or <i>in vivo</i> xenograft experiments in which the clonal composition of tumor cell populations in response to therapy was monitored, we established that PARP inhibitor or platinum salt exposure selects for secondary mutant clones in a Darwinian fashion, with the periodicity of PARP inhibitor administration and the pretreatment frequency of secondary mutant tumor cells influencing the eventual clonal composition of the tumor cell population. In xenograft studies, the presence of secondary mutant cells in tumors impaired the therapeutic effect of a clinical PARP inhibitor. However, we found that both PARP inhibitor-sensitive and PARP inhibitor-resistant <i>BRCA2</i> mutant tumor cells were sensitive to AZD-1775, a WEE1 kinase inhibitor. In mice carrying heterogeneous tumors, AZD-1775 delivered a greater therapeutic benefit than olaparib treatment. This suggests that despite the restoration of some <i>BRCA1</i> or <i>BRCA2</i> gene function in "revertant" tumor cells, vulnerabilities still exist that could be therapeutically exploited. <i>Mol Cancer Ther; 16(9); 2022-34. ©2017 AACR</i>.

Bajrami, I. Marlow, R. van de Ven, M. Brough, R. Pemberton, H.N. Frankum, J. Song, F. Rafiq, R. Konde, A. Krastev, D.B. Menon, M. Campbell, J. Gulati, A. Kumar, R. Pettitt, S.J. Gurden, M.D. Cardenosa, M.L. Chong, I. Gazinska, P. Wallberg, F. Sawyer, E.J. Martin, L.-.A. Dowsett, M. Linardopoulos, S. Natrajan, R. Ryan, C.J. Derksen, P.W.B. Jonkers, J. Tutt, A.N.J. Ashworth, A. Lord, C.J (2018) E-Cadherin/ROS1 Inhibitor Synthetic Lethality in Breast Cancer.. Show Abstract full text

The cell adhesion glycoprotein E-cadherin (CDH1) is commonly inactivated in breast tumors. Precision medicine approaches that exploit this characteristic are not available. Using perturbation screens in breast tumor cells with CRISPR/Cas9-engineered <i>CDH1</i> mutations, we identified synthetic lethality between E-cadherin deficiency and inhibition of the tyrosine kinase ROS1. Data from large-scale genetic screens in molecularly diverse breast tumor cell lines established that the E-cadherin/ROS1 synthetic lethality was not only robust in the face of considerable molecular heterogeneity but was also elicited with clinical ROS1 inhibitors, including foretinib and crizotinib. ROS1 inhibitors induced mitotic abnormalities and multinucleation in E-cadherin-defective cells, phenotypes associated with a defect in cytokinesis and aberrant p120 catenin phosphorylation and localization. <i>In vivo</i>, ROS1 inhibitors produced profound antitumor effects in multiple models of E-cadherin-defective breast cancer. These data therefore provide the preclinical rationale for assessing ROS1 inhibitors, such as the licensed drug crizotinib, in appropriately stratified patients.<b>Significance:</b> E-cadherin defects are common in breast cancer but are currently not targeted with a precision medicine approach. Our preclinical data indicate that licensed ROS1 inhibitors, including crizotinib, should be repurposed to target E-cadherin-defective breast cancers, thus providing the rationale for the assessment of these agents in molecularly stratified phase II clinical trials. <i>Cancer Discov; 8(4); 498-515. ©2018 AACR.</i><i>This article is highlighted in the In This Issue feature, p. 371</i>.

Pettitt, S.J. Lord, C.J (2018) PARP inhibitors and breast cancer: highlights and hang-ups. full text
Noordermeer, S.M. Adam, S. Setiaputra, D. Barazas, M. Pettitt, S.J. Ling, A.K. Olivieri, M. Álvarez-Quilón, A. Moatti, N. Zimmermann, M. Annunziato, S. Krastev, D.B. Song, F. Brandsma, I. Frankum, J. Brough, R. Sherker, A. Landry, S. Szilard, R.K. Munro, M.M. McEwan, A. Goullet de Rugy, T. Lin, Z.-.Y. Hart, T. Moffat, J. Gingras, A.-.C. Martin, A. van Attikum, H. Jonkers, J. Lord, C.J. Rottenberg, S. Durocher, D (2018) The shieldin complex mediates 53BP1-dependent DNA repair.. Show Abstract full text

53BP1 is a chromatin-binding protein that regulates the repair of DNA double-strand breaks by suppressing the nucleolytic resection of DNA termini<sup>1,2</sup>. This function of 53BP1 requires interactions with PTIP<sup>3</sup> and RIF1<sup>4-9</sup>, the latter of which recruits REV7 (also known as MAD2L2) to break sites<sup>10,11</sup>. How 53BP1-pathway proteins shield DNA ends is currently unknown, but there are two models that provide the best potential explanation of their action. In one model the 53BP1 complex strengthens the nucleosomal barrier to end-resection nucleases<sup>12,13</sup>, and in the other 53BP1 recruits effector proteins with end-protection activity. Here we identify a 53BP1 effector complex, shieldin, that includes C20orf196 (also known as SHLD1), FAM35A (SHLD2), CTC-534A2.2 (SHLD3) and REV7. Shieldin localizes to double-strand-break sites in a 53BP1- and RIF1-dependent manner, and its SHLD2 subunit binds to single-stranded DNA via OB-fold domains that are analogous to those of RPA1 and POT1. Loss of shieldin impairs non-homologous end-joining, leads to defective immunoglobulin class switching and causes hyper-resection. Mutations in genes that encode shieldin subunits also cause resistance to poly(ADP-ribose) polymerase inhibition in BRCA1-deficient cells and tumours, owing to restoration of homologous recombination. Finally, we show that binding of single-stranded DNA by SHLD2 is critical for shieldin function, consistent with a model in which shieldin protects DNA ends to mediate 53BP1-dependent DNA repair.

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.

Pettitt, S.J. Krastev, D.B. Brandsma, I. Dréan, A. Song, F. Aleksandrov, R. Harrell, M.I. Menon, M. Brough, R. Campbell, J. Frankum, J. Ranes, M. Pemberton, H.N. Rafiq, R. Fenwick, K. Swain, A. Guettler, S. Lee, J.-.M. Swisher, E.M. Stoynov, S. Yusa, K. Ashworth, A. Lord, C.J (2018) Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance.. Show Abstract full text

Although PARP inhibitors (PARPi) target homologous recombination defective tumours, drug resistance frequently emerges, often via poorly understood mechanisms. Here, using genome-wide and high-density CRISPR-Cas9 "tag-mutate-enrich" mutagenesis screens, we identify close to full-length mutant forms of PARP1 that cause in vitro and in vivo PARPi resistance. Mutations both within and outside of the PARP1 DNA-binding zinc-finger domains cause PARPi resistance and alter PARP1 trapping, as does a PARP1 mutation found in a clinical case of PARPi resistance. This reinforces the importance of trapped PARP1 as a cytotoxic DNA lesion and suggests that PARP1 intramolecular interactions might influence PARPi-mediated cytotoxicity. PARP1 mutations are also tolerated in cells with a pathogenic BRCA1 mutation where they result in distinct sensitivities to chemotherapeutic drugs compared to other mechanisms of PARPi resistance (BRCA1 reversion, 53BP1, REV7 (MAD2L2) mutation), suggesting that the underlying mechanism of PARPi resistance that emerges could influence the success of subsequent therapies.

Krastev, D.B. Pettitt, S.J. Campbell, J. Song, F. Tanos, B.E. Stoynov, S.S. Ashworth, A. Lord, C.J (2018) Coupling bimolecular PARylation biosensors with genetic screens to identify PARylation targets.. Show Abstract full text

Poly (ADP-ribose)ylation is a dynamic protein modification that regulates multiple cellular processes. Here, we describe a system for identifying and characterizing PARylation events that exploits the ability of a PBZ (PAR-binding zinc finger) protein domain to bind PAR with high-affinity. By linking PBZ domains to bimolecular fluorescent complementation biosensors, we developed fluorescent PAR biosensors that allow the detection of temporal and spatial PARylation events in live cells. Exploiting transposon-mediated recombination, we integrate the PAR biosensor en masse into thousands of protein coding genes in living cells. Using these PAR-biosensor "tagged" cells in a genetic screen we carry out a large-scale identification of PARylation targets. This identifies CTIF (CBP80/CBP20-dependent translation initiation factor) as a novel PARylation target of the tankyrase enzymes in the centrosomal region of cells, which plays a role in the distribution of the centrosomal satellites.

Holme, H. Gulati, A. Brough, R. Fleuren, E.D.G. Bajrami, I. Campbell, J. Chong, I.Y. Costa-Cabral, S. Elliott, R. Fenton, T. Frankum, J. Jones, S.E. Menon, M. Miller, R. Pemberton, H.N. Postel-Vinay, S. Rafiq, R. Selfe, J.L. von Kriegsheim, A. Munoz, A.G. Rodriguez, J. Shipley, J. van der Graaf, W.T.A. Williamson, C.T. Ryan, C.J. Pettitt, S. Ashworth, A. Strauss, S.J. Lord, C.J (2018) Chemosensitivity profiling of osteosarcoma tumour cell lines identifies a model of BRCAness.. Show Abstract full text

Osteosarcoma (OS) is an aggressive sarcoma, where novel treatment approaches are required. Genomic studies suggest that a subset of OS, including OS tumour cell lines (TCLs), exhibit genomic loss of heterozygosity (LOH) patterns reminiscent of BRCA1 or BRCA2 mutant tumours. This raises the possibility that PARP inhibitors (PARPi), used to treat BRCA1/2 mutant cancers, could be used to target OS. Using high-throughput drug sensitivity screening we generated chemosensitivity profiles for 79 small molecule inhibitors, including three clinical PARPi. Drug screening was performed in 88 tumour cell lines, including 18 OS TCLs. This identified known sensitivity effects in OS TCLs, such as sensitivity to FGFR inhibitors. When compared to BRCA1/2 mutant TCLs, OS TCLs, with the exception of LM7, were PARPi resistant, including those with previously determined BRCAness LoH profiles. Post-screen validation experiments confirmed PARPi sensitivity in LM7 cells as well as a defect in the ability to form nuclear RAD51 foci in response to DNA damage. LM7 provides one OS model for the study of PARPi sensitivity through a potential defect in RAD51-mediated DNA repair. The drug sensitivity dataset we generated in 88 TCLs could also serve as a resource for the study of drug sensitivity effects in OS.

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.

Khalique, S. Pettitt, S.J. Kelly, G. Tunariu, N. Natrajan, R. Banerjee, S. Lord, C.J (2020) Longitudinal analysis of a secondary BRCA2 mutation using digital droplet PCR.. Show Abstract full text

Development of resistance to platinum and poly(ADP-ribose) polymerase inhibitors via secondary BRCA gene mutations that restore functional homologous recombination has been observed in a number of cancer types. Here we report a case of somatic BRCA2 mutation in a patient with high grade serous ovarian carcinoma. A secondary mutation predicted to restore the BRCA2 open reading frame was detected at low frequency (2.3%) in whole exome sequencing of a peritoneal biopsy at disease progression after treatment that included carboplatin and olaparib. We used digital droplet PCR (ddPCR) to verify the presence and frequency of this mutation in the biopsy sample at progression and also used this approach to assess the presence of the secondary mutation in preceding biopsies at diagnosis and first relapse. We found no evidence for the secondary mutation being present prior to the final progression biopsy, suggesting that this mutation was acquired late in the course of treatment. ddPCR provides a sensitive and specific technique to investigate the presence of low frequency mutations in a time series of biopsies.

Barazas, M. Annunziato, S. Pettitt, S.J. de Krijger, I. Ghezraoui, H. Roobol, S.J. Lutz, C. Frankum, J. Song, F.F. Brough, R. Evers, B. Gogola, E. Bhin, J. van de Ven, M. van Gent, D.C. Jacobs, J.J.L. Chapman, R. Lord, C.J. Jonkers, J. Rottenberg, S (2018) The CST Complex Mediates End Protection at Double-Strand Breaks and Promotes PARP Inhibitor Sensitivity in BRCA1-Deficient Cells.. Show Abstract full text

Selective elimination of BRCA1-deficient cells by inhibitors of poly(ADP-ribose) polymerase (PARP) is a prime example of the concept of synthetic lethality in cancer therapy. This interaction is counteracted by the restoration of BRCA1-independent homologous recombination through loss of factors such as 53BP1, RIF1, and REV7/MAD2L2, which inhibit end resection of DNA double-strand breaks (DSBs). To identify additional factors involved in this process, we performed CRISPR/SpCas9-based loss-of-function screens and selected for factors that confer PARP inhibitor (PARPi) resistance in BRCA1-deficient cells. Loss of members of the CTC1-STN1-TEN1 (CST) complex were found to cause PARPi resistance in BRCA1-deficient cells in vitro and in vivo. We show that CTC1 depletion results in the restoration of end resection and that the CST complex may act downstream of 53BP1/RIF1. These data suggest that, in addition to its role in protecting telomeres, the CST complex also contributes to protecting DSBs from end resection.

Jones, S.E. Fleuren, E.D.G. Frankum, J. Konde, A. Williamson, C.T. Krastev, D.B. Pemberton, H.N. Campbell, J. Gulati, A. Elliott, R. Menon, M. Selfe, J.L. Brough, R. Pettitt, S.J. Niedzwiedz, W. van der Graaf, W.T.A. Shipley, J. Ashworth, A. Lord, C.J (2017) ATR Is a Therapeutic Target in Synovial Sarcoma.. Show Abstract full text

Synovial sarcoma (SS) is an aggressive soft-tissue malignancy characterized by expression of SS18-SSX fusions, where treatment options are limited. To identify therapeutically actionable genetic dependencies in SS, we performed a series of parallel, high-throughput small interfering RNA (siRNA) screens and compared genetic dependencies in SS tumor cells with those in >130 non-SS tumor cell lines. This approach revealed a reliance of SS tumor cells upon the DNA damage response serine/threonine protein kinase ATR. Clinical ATR inhibitors (ATRi) elicited a synthetic lethal effect in SS tumor cells and impaired growth of SS patient-derived xenografts. Oncogenic SS18-SSX family fusion genes are known to alter the composition of the BAF chromatin-remodeling complex, causing ejection and degradation of wild-type SS18 and the tumor suppressor SMARCB1. Expression of oncogenic SS18-SSX fusion proteins caused profound ATRi sensitivity and a reduction in SS18 and SMARCB1 protein levels, but an SSX18-SSX1 Δ71-78 fusion containing a C-terminal deletion did not. ATRi sensitivity in SS was characterized by an increase in biomarkers of replication fork stress (increased γH2AX, decreased replication fork speed, and increased R-loops), an apoptotic response, and a dependence upon cyclin E expression. Combinations of cisplatin or PARP inhibitors enhanced the antitumor cell effect of ATRi, suggesting that either single-agent ATRi or combination therapy involving ATRi might be further assessed as candidate approaches for SS treatment. <i>Cancer Res; 77(24); 7014-26. ©2017 AACR</i>.

Pettitt, S.J. Lord, C.J (2019) Dissecting PARP inhibitor resistance with functional genomics.. Show Abstract full text

The poly-(ADP-ribose) polymerase (PARP) inhibitor (PARPi) olaparib was the first licenced cancer drug that targeted an inherited form of cancer, namely ovarian cancers caused by germline BRCA1 or BRCA2 gene mutations. Multiple different PARPi have now been approved for use in a wider group of gynaecological cancers as well as for the treatment of BRCA-gene mutant breast cancer. Despite these advances, resistance to PARPi is a common clinical phenotype. Understanding, at the molecular level, how tumour cells respond to PARPi has the potential to inform how these drugs should be used clinically and since the discovery of this drug class, multiple different functional genomic strategies have been employed to dissect PARPi sensitivity and resistance. These have included genetic perturbation via classical gene targeting, gene silencing by siRNA or shRNA or transposon mutagenesis techniques. Recently, CRISPR-Cas9-based mutagenesis has greatly expanded the available range of relevant preclinical models and the precision of mutagenesis. Here, we review how these approaches have been used either in low-throughput, hypothesis-testing experiments or in the setting of large, hypothesis-generating, genetic screens aimed at understanding the molecular basis of PARPi sensitivity and resistance.

Chong, I.Y. Aronson, L. Bryant, H. Gulati, A. Campbell, J. Elliott, R. Pettitt, S. Wilkerson, P. Lambros, M.B. Reis-Filho, J.S. Ramessur, A. Davidson, M. Chau, I. Cunningham, D. Ashworth, A. Lord, C.J (2018) Mapping genetic vulnerabilities reveals BTK as a novel therapeutic target in oesophageal cancer.. Show Abstract full text

<h4>Objective</h4>Oesophageal cancer is the seventh most common cause of cancer-related death worldwide. Disease relapse is frequent and treatment options are limited.<h4>Design</h4>To identify new biomarker-defined therapeutic approaches for patients with oesophageal cancer, we integrated the genomic profiles of 17 oesophageal tumour-derived cell lines with drug sensitivity data from small molecule inhibitor profiling, identifying drug sensitivity effects associated with cancer driver gene alterations. We also interrogated recently described RNA interference screen data for these tumour cell lines to identify candidate genetic dependencies or vulnerabilities that could be exploited as therapeutic targets.<h4>Results</h4>By integrating the genomic features of oesophageal tumour cell lines with siRNA and drug screening data, we identified a series of candidate targets in oesophageal cancer, including a sensitivity to inhibition of the kinase BTK in <i>MYC</i> amplified oesophageal tumour cell lines. We found that this genetic dependency could be elicited with the clinical BTK/ERBB2 kinase inhibitor, ibrutinib. In both <i>MYC</i> and <i>ERBB2</i> amplified tumour cells, ibrutinib downregulated ERK-mediated signal transduction, cMYC Ser-62 phosphorylation and levels of MYC protein, and elicited G<sub>1</sub> cell cycle arrest and apoptosis, suggesting that this drug could be used to treat biomarker-selected groups of patients with oesophageal cancer.<h4>Conclusions</h4>BTK represents a novel candidate therapeutic target in oesophageal cancer that can be targeted with ibrutinib. On the basis of this work, a proof-of-concept phase II clinical trial evaluating the efficacy of ibrutinib in patients with <i>MYC</i> and/or <i>ERBB2</i> amplified advanced oesophageal cancer is currently underway (NCT02884453).<h4>Trial registration number</h4>NCT02884453; Pre-results.

Zandarashvili, L. Langelier, M.-.F. Velagapudi, U.K. Hancock, M.A. Steffen, J.D. Billur, R. Hannan, Z.M. Wicks, A.J. Krastev, D.B. Pettitt, S.J. Lord, C.J. Talele, T.T. Pascal, J.M. Black, B.E (2020) Structural basis for allosteric PARP-1 retention on DNA breaks.. Show Abstract full text

The success of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors (PARPi) to treat cancer relates to their ability to trap PARP-1 at the site of a DNA break. Although different forms of PARPi all target the catalytic center of the enzyme, they have variable abilities to trap PARP-1. We found that several structurally distinct PARPi drive PARP-1 allostery to promote release from a DNA break. Other inhibitors drive allostery to retain PARP-1 on a DNA break. Further, we generated a new PARPi compound, converting an allosteric pro-release compound to a pro-retention compound and increasing its ability to kill cancer cells. These developments are pertinent to clinical applications where PARP-1 trapping is either desirable or undesirable.

Yap, T.A. Kristeleit, R. Michalarea, V. Pettitt, S.J. Lim, J.S.J. Carreira, S. Roda, D. Miller, R. Riisnaes, R. Miranda, S. Figueiredo, I. Rodrigues, D.N. Ward, S. Matthews, R. Parmar, M. Turner, A. Tunariu, N. Chopra, N. Gevensleben, H. Turner, N.C. Ruddle, R. Raynaud, F.I. Decordova, S. Swales, K.E. Finneran, L. Hall, E. Rugman, P. Lindemann, J.P.O. Foxley, A. Lord, C.J. Banerji, U. Plummer, R. Basu, B. Lopez, J.S. Drew, Y. de Bono, J.S (2020) Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with <i>BRCA1/2</i>- and Non-<i>BRCA1/2</i>-Mutant Cancers.. Show Abstract full text

Preclinical studies have demonstrated synergy between PARP and PI3K/AKT pathway inhibitors in <i>BRCA1</i> and <i>BRCA2</i> (<i>BRCA1/2)</i>-deficient and <i>BRCA1/2</i>-proficient tumors. We conducted an investigator-initiated phase I trial utilizing a prospective intrapatient dose- escalation design to assess two schedules of capivasertib (AKT inhibitor) with olaparib (PARP inhibitor) in 64 patients with advanced solid tumors. Dose expansions enrolled germline <i>BRCA1/2</i>-mutant tumors, or <i>BRCA1/2</i> wild-type cancers harboring somatic DNA damage response (DDR) or PI3K-AKT pathway alterations. The combination was well tolerated. Recommended phase II doses for the two schedules were: olaparib 300 mg twice a day with either capivasertib 400 mg twice a day 4 days on, 3 days off, or capivasertib 640 mg twice a day 2 days on, 5 days off. Pharmacokinetics were dose proportional. Pharmacodynamic studies confirmed phosphorylated (p) GSK3β suppression, increased pERK, and decreased BRCA1 expression. Twenty-five (44.6%) of 56 evaluable patients achieved clinical benefit (RECIST complete response/partial response or stable disease ≥ 4 months), including patients with tumors harboring germline <i>BRCA1/2</i> mutations and <i>BRCA1/2</i> wild-type cancers with or without DDR and PI3K-AKT pathway alterations. SIGNIFICANCE: In the first trial to combine PARP and AKT inhibitors, a prospective intrapatient dose- escalation design demonstrated safety, tolerability, and pharmacokinetic-pharmacodynamic activity and assessed predictive biomarkers of response/resistance. Antitumor activity was observed in patients harboring tumors with germline <i>BRCA1/2</i> mutations and <i>BRCA1/2</i> wild-type cancers with or without somatic DDR and/or PI3K-AKT pathway alterations.<i>This article is highlighted in the In This Issue feature, p. 1426</i>.

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>.

Chabanon, R.M. Morel, D. Eychenne, T. Colmet-Daage, L. Bajrami, I. Dorvault, N. Garrido, M. Meisenberg, C. Lamb, A. Ngo, C. Hopkins, S.R. Roumeliotis, T.I. Jouny, S. Hénon, C. Kawai-Kawachi, A. Astier, C. Konde, A. Del Nery, E. Massard, C. Pettitt, S.J. Margueron, R. Choudhary, J.S. Almouzni, G. Soria, J.-.C. Deutsch, E. Downs, J.A. Lord, C.J. Postel-Vinay, S (2021) PBRM1 Deficiency Confers Synthetic Lethality to DNA Repair Inhibitors in Cancer.. Show Abstract full text

Inactivation of <i>Polybromo 1</i> (<i>PBRM1</i>), a specific subunit of the PBAF chromatin remodeling complex, occurs frequently in cancer, including 40% of clear cell renal cell carcinomas (ccRCC). To identify novel therapeutic approaches to targeting PBRM1-defective cancers, we used a series of orthogonal functional genomic screens that identified PARP and ATR inhibitors as being synthetic lethal with <i>PBRM1</i> deficiency. The PBRM1/PARP inhibitor synthetic lethality was recapitulated using several clinical PARP inhibitors in a series of <i>in vitro</i> model systems and <i>in vivo</i> in a xenograft model of ccRCC. In the absence of exogenous DNA damage, PBRM1-defective cells exhibited elevated levels of replication stress, micronuclei, and R-loops. PARP inhibitor exposure exacerbated these phenotypes. Quantitative mass spectrometry revealed that multiple R-loop processing factors were downregulated in PBRM1-defective tumor cells. Exogenous expression of the R-loop resolution enzyme RNase H1 reversed the sensitivity of PBRM1-deficient cells to PARP inhibitors, suggesting that excessive levels of R-loops could be a cause of this synthetic lethality. PARP and ATR inhibitors also induced cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) innate immune signaling in PBRM1-defective tumor cells. Overall, these findings provide the preclinical basis for using PARP inhibitors in PBRM1-defective cancers. SIGNIFICANCE: This study demonstrates that PARP and ATR inhibitors are synthetic lethal with the loss of PBRM1, a PBAF-specific subunit, thus providing the rationale for assessing these inhibitors in patients with PBRM1-defective cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/11/2888/F1.large.jpg.

Carreira, S. Porta, N. Arce-Gallego, S. Seed, G. Llop-Guevara, A. Bianchini, D. Rescigno, P. Paschalis, A. Bertan, C. Baker, C. Goodall, J. Miranda, S. Riisnaes, R. Figueiredo, I. Ferreira, A. Pereira, R. Crespo, M. Gurel, B. Nava Rodrigues, D. Pettitt, S.J. Yuan, W. Serra, V. Rekowski, J. Lord, C.J. Hall, E. Mateo, J. de Bono, J.S (2021) Biomarkers Associating with PARP Inhibitor Benefit in Prostate Cancer in the TOPARP-B Trial.. Show Abstract full text

PARP inhibitors are approved for treating advanced prostate cancers (APC) with various defective DNA repair genes; however, further studies to clinically qualify predictive biomarkers are warranted. Herein we analyzed TOPARP-B phase II clinical trial samples, evaluating whole-exome and low-pass whole-genome sequencing and IHC and IF assays evaluating <i>ATM</i> and RAD51 foci (testing homologous recombination repair function). <i>BRCA1/2</i> germline and somatic pathogenic mutations associated with similar benefit from olaparib; greater benefit was observed with homozygous <i>BRCA2</i> deletion. Biallelic, but not monoallelic, <i>PALB2</i> deleterious alterations were associated with clinical benefit. In the ATM cohort, loss of ATM protein by IHC was associated with a better outcome. RAD51 foci loss identified tumors with biallelic <i>BRCA</i> and <i>PALB2</i> alterations while most <i>ATM</i>- and <i>CDK12</i>-altered APCs had higher RAD51 foci levels. Overall, APCs with homozygous <i>BRCA2</i> deletion are exceptional responders; <i>PALB2</i> biallelic loss and loss of ATM IHC expression associated with clinical benefit. SIGNIFICANCE: Not all APCs with DNA repair defects derive similar benefit from PARP inhibition. Most benefit was seen among patients with <i>BRCA2</i> homozygous deletions, biallelic loss of <i>PALB2</i>, and loss of ATM protein. Loss of RAD51 foci, evaluating homologous recombination repair function, was found primarily in tumors with biallelic <i>BRCA1/2</i> and <i>PALB2</i> alterations.<i>This article is highlighted in the In This Issue feature, p. 2659</i>.

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.

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.

Krastev, D.B. Li, S. Sun, Y. Wicks, A.J. Hoslett, G. Weekes, D. Badder, L.M. Knight, E.G. Marlow, R. Pardo, M.C. Yu, L. Talele, T.T. Bartek, J. Choudhary, J.S. Pommier, Y. Pettitt, S.J. Tutt, A.N.J. Ramadan, K. Lord, C.J (2022) The ubiquitin-dependent ATPase p97 removes cytotoxic trapped PARP1 from chromatin.. Show Abstract full text

Poly (ADP-ribose) polymerase (PARP) inhibitors elicit antitumour activity in homologous recombination-defective cancers by trapping PARP1 in a chromatin-bound state. How cells process trapped PARP1 remains unclear. Using wild-type and a trapping-deficient PARP1 mutant combined with rapid immunoprecipitation mass spectrometry of endogenous proteins and Apex2 proximity labelling, we delineated mass spectrometry-based interactomes of trapped and non-trapped PARP1. These analyses identified an interaction between trapped PARP1 and the ubiquitin-regulated p97 ATPase/segregase. We found that following trapping, PARP1 is SUMOylated by PIAS4 and subsequently ubiquitylated by the SUMO-targeted E3 ubiquitin ligase RNF4, events that promote recruitment of p97 and removal of trapped PARP1 from chromatin. Small-molecule p97-complex inhibitors, including a metabolite of the clinically used drug disulfiram (CuET), prolonged PARP1 trapping and enhanced PARP inhibitor-induced cytotoxicity in homologous recombination-defective tumour cells and patient-derived tumour organoids. Together, these results suggest that p97 ATPase plays a key role in the processing of trapped PARP1 and the response of tumour cells to PARP inhibitors.

Tarantino, D. Walker, C. Weekes, D. Pemberton, H. Davidson, K. Torga, G. Frankum, J. Mendes-Pereira, A.M. Prince, C. Ferro, R. Brough, R. Pettitt, S.J. Lord, C.J. Grigoriadis, A. Nj Tutt, A (2022) Functional screening reveals HORMAD1-driven gene dependencies associated with translesion synthesis and replication stress tolerance.. Show Abstract full text

HORMAD1 expression is usually restricted to germline cells, but it becomes mis-expressed in epithelial cells in ~60% of triple-negative breast cancers (TNBCs), where it is associated with elevated genomic instability (1). HORMAD1 expression in TNBC is bimodal with HORMAD1-positive TNBC representing a biologically distinct disease group. Identification of HORMAD1-driven genetic dependencies may uncover novel therapies for this disease group. To study HORMAD1-driven genetic dependencies, we generated a SUM159 cell line model with doxycycline-inducible HORMAD1 that replicated genomic instability phenotypes seen in HORMAD1-positive TNBC (1). Using small interfering RNA screens, we identified candidate genes whose depletion selectively inhibited the cellular growth of HORMAD1-expressing cells. We validated five genes (ATR, BRIP1, POLH, TDP1 and XRCC1), depletion of which led to reduced cellular growth or clonogenic survival in cells expressing HORMAD1. In addition to the translesion synthesis (TLS) polymerase POLH, we identified a HORMAD1-driven dependency upon additional TLS polymerases, namely POLK, REV1, REV3L and REV7. Our data confirms that out-of-context somatic expression of HORMAD1 can lead to genomic instability and reveals that HORMAD1 expression induces dependencies upon replication stress tolerance pathways, such as translesion synthesis. Our data also suggest that HORMAD1 expression could be a patient selection biomarker for agents targeting replication stress.

Serra, V. Wang, A.T. Castroviejo-Bermejo, M. Polanska, U.M. Palafox, M. Herencia-Ropero, A. Jones, G.N. Lai, Z. Armenia, J. Michopoulos, F. Llop-Guevara, A. Brough, R. Gulati, A. Pettitt, S.J. Bulusu, K.C. Nikkilä, J. Wilson, Z. Hughes, A. Wijnhoven, P.W.G. Ahmed, A. Bruna, A. Gris-Oliver, A. Guzman, M. Rodríguez, O. Grueso, J. Arribas, J. Cortés, J. Saura, C. Lau, A. Critchlow, S. Dougherty, B. Caldas, C. Mills, G.B. Barrett, J.C. Forment, J.V. Cadogan, E. Lord, C.J. Cruz, C. Balmaña, J. O'Connor, M.J (2022) Identification of a Molecularly-Defined Subset of Breast and Ovarian Cancer Models that Respond to WEE1 or ATR Inhibition, Overcoming PARP Inhibitor Resistance.. Show Abstract full text

<h4>Purpose</h4>PARP inhibitors (PARPi) induce synthetic lethality in homologous recombination repair (HRR)-deficient tumors and are used to treat breast, ovarian, pancreatic, and prostate cancers. Multiple PARPi resistance mechanisms exist, most resulting in restoration of HRR and protection of stalled replication forks. ATR inhibition was highlighted as a unique approach to reverse both aspects of resistance. Recently, however, a PARPi/WEE1 inhibitor (WEE1i) combination demonstrated enhanced antitumor activity associated with the induction of replication stress, suggesting another approach to tackling PARPi resistance.<h4>Experimental design</h4>We analyzed breast and ovarian patient-derived xenoimplant models resistant to PARPi to quantify WEE1i and ATR inhibitor (ATRi) responses as single agents and in combination with PARPi. Biomarker analysis was conducted at the genetic and protein level. Metabolite analysis by mass spectrometry and nucleoside rescue experiments ex vivo were also conducted in patient-derived models.<h4>Results</h4>Although WEE1i response was linked to markers of replication stress, including STK11/RB1 and phospho-RPA, ATRi response associated with ATM mutation. When combined with olaparib, WEE1i could be differentiated from the ATRi/olaparib combination, providing distinct therapeutic strategies to overcome PARPi resistance by targeting the replication stress response. Mechanistically, WEE1i sensitivity was associated with shortage of the dNTP pool and a concomitant increase in replication stress.<h4>Conclusions</h4>Targeting the replication stress response is a valid therapeutic option to overcome PARPi resistance including tumors without an underlying HRR deficiency. These preclinical insights are now being tested in several clinical trials where the PARPi is administered with either the WEE1i or the ATRi.

Yap, T.A. O'Carrigan, B. Penney, M.S. Lim, J.S. Brown, J.S. de Miguel Luken, M.J. Tunariu, N. Perez-Lopez, R. Rodrigues, D.N. Riisnaes, R. Figueiredo, I. Carreira, S. Hare, B. McDermott, K. Khalique, S. Williamson, C.T. Natrajan, R. Pettitt, S.J. Lord, C.J. Banerji, U. Pollard, J. Lopez, J. de Bono, J.S (2020) Phase I Trial of First-in-Class ATR Inhibitor M6620 (VX-970) as Monotherapy or in Combination With Carboplatin in Patients With Advanced Solid Tumors.. Show Abstract full text

<h4>Purpose</h4>Preclinical studies demonstrated that ATR inhibition can exploit synthetic lethality (eg, in cancer cells with impaired compensatory DNA damage responses through ATM loss) as monotherapy and combined with DNA-damaging drugs such as carboplatin.<h4>Patients and methods</h4>This phase I trial assessed the ATR inhibitor M6620 (VX-970) as monotherapy (once or twice weekly) and combined with carboplatin (carboplatin on day 1 and M6620 on days 2 and 9 in 21-day cycles). Primary objectives were safety, tolerability, and maximum tolerated dose; secondary objectives included pharmacokinetics and antitumor activity; exploratory objectives included pharmacodynamics in timed paired tumor biopsies.<h4>Results</h4>Forty patients were enrolled; 17 received M6620 monotherapy, which was safe and well tolerated. The recommended phase II dose (RP2D) for once- or twice-weekly administration was 240 mg/m<sup>2</sup>. A patient with metastatic colorectal cancer harboring molecular aberrations, including ATM loss and an <i>ARID1A</i> mutation, achieved RECISTv1.1 complete response and maintained this response, with a progression-free survival of 29 months at last assessment. Twenty-three patients received M6620 with carboplatin, with mechanism-based hematologic toxicities at higher doses, requiring dose delays and reductions. The RP2D for combination therapy was M6620 90 mg/m<sup>2</sup> with carboplatin AUC5. A patient with advanced germline <i>BRCA1</i> ovarian cancer achieved RECISTv1.1 partial response and Gynecologic Cancer Intergroup CA125 response despite being platinum refractory and PARP inhibitor resistant. An additional 15 patients had RECISTv1.1 stable disease as best response. Pharmacokinetics were dose proportional and exceeded preclinical efficacious levels. Pharmacodynamic studies demonstrated substantial inhibition of phosphorylation of CHK1, the downstream ATR substrate.<h4>Conclusion</h4>To our knowledge, this report is the first of an ATR inhibitor as monotherapy and combined with carboplatin. M6620 was well tolerated, with target engagement and preliminary antitumor responses observed.

Hewitt, G. Borel, V. Segura-Bayona, S. Takaki, T. Ruis, P. Bellelli, R. Lehmann, L.C. Sommerova, L. Vancevska, A. Tomas-Loba, A. Zhu, K. Cooper, C. Fugger, K. Patel, H. Goldstone, R. Schneider-Luftman, D. Herbert, E. Stamp, G. Brough, R. Pettitt, S. Lord, C.J. West, S.C. Ahel, I. Ahel, D. Chapman, J.R. Deindl, S. Boulton, S.J (2021) Defective ALC1 nucleosome remodeling confers PARPi sensitization and synthetic lethality with HRD.. Show Abstract full text

Chromatin is a barrier to efficient DNA repair, as it hinders access and processing of certain DNA lesions. ALC1/CHD1L is a nucleosome-remodeling enzyme that responds to DNA damage, but its precise function in DNA repair remains unknown. Here we report that loss of ALC1 confers sensitivity to PARP inhibitors, methyl-methanesulfonate, and uracil misincorporation, which reflects the need to remodel nucleosomes following base excision by DNA glycosylases but prior to handover to APEX1. Using CRISPR screens, we establish that ALC1 loss is synthetic lethal with homologous recombination deficiency (HRD), which we attribute to chromosome instability caused by unrepaired DNA gaps at replication forks. In the absence of ALC1 or APEX1, incomplete processing of BER intermediates results in post-replicative DNA gaps and a critical dependence on HR for repair. Hence, targeting ALC1 alone or as a PARP inhibitor sensitizer could be employed to augment existing therapeutic strategies for HRD cancers.

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>).

Sumanasuriya, S. Seed, G. Parr, H. Christova, R. Pope, L. Bertan, C. Bianchini, D. Rescigno, P. Figueiredo, I. Goodall, J. Fowler, G. Flohr, P. Mehra, N. Neeb, A. Rekowski, J. Eisenberger, M. Sartor, O. Oudard, S. Geffriaud-Ricouard, C. Ozatilgan, A. Chadjaa, M. Macé, S. Lord, C. Baxter, J. Pettitt, S. Lambros, M. Sharp, A. Mateo, J. Carreira, S. Yuan, W. de Bono, J.S (2021) Elucidating Prostate Cancer Behaviour During Treatment via Low-pass Whole-genome Sequencing of Circulating Tumour DNA.. Show Abstract full text

<h4>Background</h4>Better blood tests to elucidate the behaviour of metastatic castration-resistant prostate cancer (mCRPC) are urgently needed to drive therapeutic decisions. Plasma cell-free DNA (cfDNA) comprises normal and circulating tumour DNA (ctDNA). Low-pass whole-genome sequencing (lpWGS) of ctDNA can provide information on mCRPC behaviour.<h4>Objective</h4>To validate and clinically qualify plasma lpWGS for mCRPC.<h4>Design, setting, and participants</h4>Plasma lpWGS data were obtained for mCRPC patients consenting to optional substudies of two prospective phase 3 trials (FIRSTANA and PROSELICA). In FIRSTANA, chemotherapy-naïve patients were randomised to treatment with docetaxel (75 mg/m<sup>2</sup>) or cabazitaxel (20 or 25 mg/m<sup>2</sup>). In PROSELICA, patients previously treated with docetaxel were randomised to 20 or 25 mg/m<sup>2</sup> cabazitaxel. lpWGS data were generated from 540 samples from 188 mCRPC patients acquired at four different time points (screening, cycle 1, cycle 4, and end of study). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: lpWGS data for ctDNA were evaluated for prognostic, response, and tumour genomic measures. Associations with response and survival data were determined for tumour fraction. Genomic biomarkers including large-scale transition (LST) scores were explored in the context of prior treatments.<h4>Results and limitations</h4>Plasma tumour fraction was prognostic for overall survival in univariable and stratified multivariable analyses (hazard ratio 1.75, 95% confidence interval 1.08-2.85; p = 0.024) and offered added value compared to existing biomarkers (C index 0.722 vs 0.709; p = 0.021). Longitudinal changes were associated with drug response. PROSELICA samples were enriched for LSTs (p = 0.029) indicating genomic instability, and this enrichment was associated with prior abiraterone and enzalutamide treatment but not taxane or radiation therapy. Higher LSTs were correlated with losses of RB1/RNASEH2B, independent of BRCA2 loss.<h4>Conclusions</h4>Plasma lpWGS of ctDNA describes CRPC behaviour, providing prognostic and response data of clinical relevance. The added prognostic value of the ctDNA fraction over established biomarkers should be studied further.<h4>Patient summary</h4>We studied tumour DNA in blood samples from patients with prostate cancer. We found that levels of tumour DNA in blood were indicative of disease prognosis, and that changes after treatment could be detected. We also observed a "genetic scar" in the results that was associated with certain previous treatments. This test allows an assessment of tumour activity that can complement existing tests, offer insights into drug response, and detect clinically relevant genetic changes.

Baxter, J.S. Zatreanu, D. Pettitt, S.J. Lord, C.J (2022) Resistance to DNA repair inhibitors in cancer.. Show Abstract full text

The DNA damage response (DDR) represents a complex network of proteins which detect and repair DNA damage, thereby maintaining the integrity of the genome and preventing the transmission of mutations and rearranged chromosomes to daughter cells. Faults in the DDR are a known driver and hallmark of cancer. Furthermore, inhibition of DDR enzymes can be used to treat the disease. This is exemplified by PARP inhibitors (PARPi) used to treat cancers with defects in the homologous recombination DDR pathway. A series of novel DDR targets are now also under pre-clinical or clinical investigation, including inhibitors of ATR kinase, WRN helicase or the DNA polymerase/helicase Polθ (Pol-Theta). Drug resistance is a common phenomenon that impairs the overall effectiveness of cancer treatments and there is already some understanding of how resistance to PARPi occurs. Here, we discuss how an understanding of PARPi resistance could inform how resistance to new drugs targeting the DDR emerges. We also discuss potential strategies that could limit the impact of these therapy resistance mechanisms in cancer.

Brunton, H. Caligiuri, G. Cunningham, R. Upstill-Goddard, R. Bailey, U.-.M. Garner, I.M. Nourse, C. Dreyer, S. Jones, M. Moran-Jones, K. Wright, D.W. Paulus-Hock, V. Nixon, C. Thomson, G. Jamieson, N.B. McGregor, G.A. Evers, L. McKay, C.J. Gulati, A. Brough, R. Bajrami, I. Pettitt, S.J. Dziubinski, M.L. Barry, S.T. Grützmann, R. Brown, R. Curry, E. Glasgow Precision Oncology Laboratory, . Australian Pancreatic Cancer Genome Initiative, . Pajic, M. Musgrove, E.A. Petersen, G.M. Shanks, E. Ashworth, A. Crawford, H.C. Simeone, D.M. Froeling, F.E.M. Lord, C.J. Mukhopadhyay, D. Pilarsky, C. Grimmond, S.E. Morton, J.P. Sansom, O.J. Chang, D.K. Bailey, P.J. Biankin, A.V (2020) HNF4A and GATA6 Loss Reveals Therapeutically Actionable Subtypes in Pancreatic Cancer.. Show Abstract full text

Pancreatic ductal adenocarcinoma (PDAC) can be divided into transcriptomic subtypes with two broad lineages referred to as classical (pancreatic) and squamous. We find that these two subtypes are driven by distinct metabolic phenotypes. Loss of genes that drive endodermal lineage specification, HNF4A and GATA6, switch metabolic profiles from classical (pancreatic) to predominantly squamous, with glycogen synthase kinase 3 beta (GSK3β) a key regulator of glycolysis. Pharmacological inhibition of GSK3β results in selective sensitivity in the squamous subtype; however, a subset of these squamous patient-derived cell lines (PDCLs) acquires rapid drug tolerance. Using chromatin accessibility maps, we demonstrate that the squamous subtype can be further classified using chromatin accessibility to predict responsiveness and tolerance to GSK3β inhibitors. Our findings demonstrate that distinct patterns of chromatin accessibility can be used to identify patient subgroups that are indistinguishable by gene expression profiles, highlighting the utility of chromatin-based biomarkers for patient selection in the treatment of PDAC.

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.

Rouleau-Turcotte, É. Krastev, D.B. Pettitt, S.J. Lord, C.J. Pascal, J.M (2022) Captured snapshots of PARP1 in the active state reveal the mechanics of PARP1 allostery.. Show Abstract full text

PARP1 rapidly detects DNA strand break damage and allosterically signals break detection to the PARP1 catalytic domain to activate poly(ADP-ribose) production from NAD<sup>+</sup>. PARP1 activation is characterized by dynamic changes in the structure of a regulatory helical domain (HD); yet, there are limited insights into the specific contributions that the HD makes to PARP1 allostery. Here, we have determined crystal structures of PARP1 in isolated active states that display specific HD conformations. These captured snapshots and biochemical analysis illustrate HD contributions to PARP1 multi-domain and high-affinity interaction with DNA damage, provide novel insights into the mechanics of PARP1 allostery, and indicate how HD active conformations correspond to alterations in the catalytic region that reveal the active site to NAD<sup>+</sup>. Our work deepens the understanding of PARP1 catalytic activation, the dynamics of the binding site of PARP inhibitor compounds, and the mechanisms regulating PARP1 retention on DNA damage.

Dreyer, S.B. Upstill-Goddard, R. Paulus-Hock, V. Paris, C. Lampraki, E.-.M. Dray, E. Serrels, B. Caligiuri, G. Rebus, S. Plenker, D. Galluzzo, Z. Brunton, H. Cunningham, R. Tesson, M. Nourse, C. Bailey, U.-.M. Jones, M. Moran-Jones, K. Wright, D.W. Duthie, F. Oien, K. Evers, L. McKay, C.J. McGregor, G.A. Gulati, A. Brough, R. Bajrami, I. Pettitt, S. Dziubinski, M.L. Candido, J. Balkwill, F. Barry, S.T. Grutzmann, R. Rahib, L. Johns, A. Pajic, M. Froeling, F.E.M. Beer, P. Musgrove, E.A. Petersen, G.M. Ashworth, A. Frame, M.C. Crawford, H.C. Simeone, D.M. Lord, C. Mukhopadhyay, D. Pilarsky, C. Tuveson, D.A. Cooke, S.L. Jamieson, N.B. Morton, J.P. Sansom, O.J. Bailey, P.J. Biankin, A. Chang, D.K (2021) Targeting DNA Damage Response and Replication Stress in Pancreatic Cancer.
Pettitt, S.J. Shao, N. Zatreanu, D. Frankum, J. Bajrami, I. Brough, R. Krastev, D.B. Roumeliotis, T.I. Choudhary, J.S. Lorenz, S. Rust, A. de Bono, J.S. Yap, T.A. Tutt, A.N.J. Lord, C.J (2023) A HUWE1 defect causes PARP inhibitor resistance by modulating the BRCA1-∆11q splice variant.. Show Abstract full text

Although PARP inhibitors (PARPi) now form part of the standard-of-care for the treatment of homologous recombination defective cancers, de novo and acquired resistance limits their overall effectiveness. Previously, overexpression of the BRCA1-∆11q splice variant has been shown to cause PARPi resistance. How cancer cells achieve increased BRCA1-∆11q expression has remained unclear. Using isogenic cells with different BRCA1 mutations, we show that reduction in HUWE1 leads to increased levels of BRCA1-∆11q and PARPi resistance. This effect is specific to cells able to express BRCA1-∆11q (e.g. BRCA1 exon 11 mutant cells) and is not seen in BRCA1 mutants that cannot express BRCA1-∆11q, nor in BRCA2 mutant cells. As well as increasing levels of BRCA1-∆11q protein in exon 11 mutant cells, HUWE1 silencing also restores RAD51 nuclear foci and platinum salt resistance. HUWE1 catalytic domain mutations were also seen in a case of PARPi resistant, BRCA1 exon 11 mutant, high grade serous ovarian cancer. These results suggest how elevated levels of BRCA1-∆11q and PARPi resistance can be achieved, identify HUWE1 as a candidate biomarker of PARPi resistance for assessment in future clinical trials and illustrate how some PARPi resistance mechanisms may only operate in patients with particular BRCA1 mutations.

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.

Ryan, C.J. Devakumar, L.P.S. Pettitt, S.J. Lord, C.J (2023) Complex synthetic lethality in cancer.. Show Abstract full text

The concept of synthetic lethality has been widely applied to identify therapeutic targets in cancer, with varying degrees of success. The standard approach normally involves identifying genetic interactions between two genes, a driver and a target. In reality, however, most cancer synthetic lethal effects are likely complex and also polygenic, being influenced by the environment in addition to involving contributions from multiple genes. By acknowledging and delineating this complexity, we describe in this article how the success rate in cancer drug discovery and development could be improved.

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.

Francis, J.C. Capper, A. Rust, A.G. Ferro, K. Ning, J. Yuan, W. de Bono, J. Pettitt, S.J. Swain, A (2024) Identification of genes that promote PI3K pathway activation and prostate tumour formation.. Show Abstract full text

We have performed a functional in vivo mutagenesis screen to identify genes that, when altered, cooperate with a heterozygous Pten mutation to promote prostate tumour formation. Two genes, Bzw2 and Eif5a2, which have been implicated in the process of protein translation, were selected for further validation. Using prostate organoid models, we show that either Bzw2 downregulation or EIF5A2 overexpression leads to increased organoid size and in vivo prostate growth. We show that both genes impact the PI3K pathway and drive a sustained increase in phospho-AKT expression, with PTEN protein levels reduced in both models. Mechanistic studies reveal that EIF5A2 is directly implicated in PTEN protein translation. Analysis of patient datasets identified EIF5A2 amplifications in many types of human cancer, including the prostate. Human prostate cancer samples in two independent cohorts showed a correlation between increased levels of EIF5A2 and upregulation of a PI3K pathway gene signature. Consistent with this, organoids with high levels of EIF5A2 were sensitive to AKT inhibitors. Our study identified novel genes that promote prostate cancer formation through upregulation of the PI3K pathway, predicting a strategy to treat patients with genetic aberrations in these genes particularly relevant for EIF5A2 amplified tumours.