Dev, H.
Chiang, T.-.
Lescale, C.
de Krijger, I.
Martin, A.G.
Pilger, D.
Coates, J.
Sczaniecka-Clift, M.
Wei, W.
Ostermaier, M.
Herzog, M.
Lam, J.
Shea, A.
Demir, M.
Wu, Q.
Yang, F.
Fu, B.
Lai, Z.
Balmus, G.
Belotserkovskaya, R.
Serra, V.
O'Connor, M.J.
Bruna, A.
Beli, P.
Pellegrini, L.
Caldas, C.
Deriano, L.
Jacobs, J.J.
Galanty, Y.
Jackson, S.P.
(2018). Shieldin complex promotes DNA end-joining and counters homologous recombination in BRCA1-null cells. Nat cell biol,
Vol.20
(8),
pp. 954-965.
show abstract
full text
BRCA1 deficiencies cause breast, ovarian, prostate and other cancers, and render tumours hypersensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. To understand the resistance mechanisms, we conducted whole-genome CRISPR-Cas9 synthetic-viability/resistance screens in BRCA1-deficient breast cancer cells treated with PARP inhibitors. We identified two previously uncharacterized proteins, C20orf196 and FAM35A, whose inactivation confers strong PARP-inhibitor resistance. Mechanistically, we show that C20orf196 and FAM35A form a complex, 'Shieldin' (SHLD1/2), with FAM35A interacting with single-stranded DNA through its C-terminal oligonucleotide/oligosaccharide-binding fold region. We establish that Shieldin acts as the downstream effector of 53BP1/RIF1/MAD2L2 to promote DNA double-strand break (DSB) end-joining by restricting DSB resection and to counteract homologous recombination by antagonizing BRCA2/RAD51 loading in BRCA1-deficient cells. Notably, Shieldin inactivation further sensitizes BRCA1-deficient cells to cisplatin, suggesting how defining the SHLD1/2 status of BRCA1-deficient tumours might aid patient stratification and yield new treatment opportunities. Highlighting this potential, we document reduced SHLD1/2 expression in human breast cancers displaying intrinsic or acquired PARP-inhibitor resistance..
Tacconi, E.M.
Lai, X.
Folio, C.
Porru, M.
Zonderland, G.
Badie, S.
Michl, J.
Sechi, I.
Rogier, M.
Matía García, V.
Batra, A.S.
Rueda, O.M.
Bouwman, P.
Jonkers, J.
Ryan, A.
Reina-San-Martin, B.
Hui, J.
Tang, N.
Bruna, A.
Biroccio, A.
Tarsounas, M.
(2017). BRCA1 and BRCA2 tumor suppressors protect against endogenous acetaldehyde toxicity. Embo mol med,
Vol.9
(10),
pp. 1398-1414.
show abstract
full text
Maintenance of genome integrity requires the functional interplay between Fanconi anemia (FA) and homologous recombination (HR) repair pathways. Endogenous acetaldehyde, a product of cellular metabolism, is a potent source of DNA damage, particularly toxic to cells and mice lacking the FA protein FANCD2. Here, we investigate whether HR-compromised cells are sensitive to acetaldehyde, similarly to FANCD2-deficient cells. We demonstrate that inactivation of HR factors BRCA1, BRCA2, or RAD51 hypersensitizes cells to acetaldehyde treatment, in spite of the FA pathway being functional. Aldehyde dehydrogenases (ALDHs) play key roles in endogenous acetaldehyde detoxification, and their chemical inhibition leads to cellular acetaldehyde accumulation. We find that disulfiram (Antabuse), an ALDH2 inhibitor in widespread clinical use for the treatment of alcoholism, selectively eliminates BRCA1/2-deficient cells. Consistently, Aldh2 gene inactivation suppresses proliferation of HR-deficient mouse embryonic fibroblasts (MEFs) and human fibroblasts. Hypersensitivity of cells lacking BRCA2 to acetaldehyde stems from accumulation of toxic replication-associated DNA damage, leading to checkpoint activation, G2/M arrest, and cell death. Acetaldehyde-arrested replication forks require BRCA2 and FANCD2 for protection against MRE11-dependent degradation. Importantly, acetaldehyde specifically inhibits in vivo the growth of BRCA1/2-deficient tumors and ex vivo in patient-derived tumor xenograft cells (PDTCs), including those that are resistant to poly (ADP-ribose) polymerase (PARP) inhibitors. The work presented here therefore identifies acetaldehyde metabolism as a potential therapeutic target for the selective elimination of BRCA1/2-deficient cells and tumors..
Cassidy, J.W.
Batra, A.S.
Greenwood, W.
Bruna, A.
(2016). Patient-derived tumour xenografts for breast cancer drug discovery. Endocr relat cancer,
Vol.23
(12),
pp. T259-T270.
show abstract
full text
Despite remarkable advances in our understanding of the drivers of human malignancies, new targeted therapies often fail to show sufficient efficacy in clinical trials. Indeed, the cost of bringing a new agent to market has risen substantially in the last several decades, in part fuelled by extensive reliance on preclinical models that fail to accurately reflect tumour heterogeneity. To halt unsustainable rates of attrition in the drug discovery process, we must develop a new generation of preclinical models capable of reflecting the heterogeneity of varying degrees of complexity found in human cancers. Patient-derived tumour xenograft (PDTX) models prevail as arguably the most powerful in this regard because they capture cancer's heterogeneous nature. Herein, we review current breast cancer models and their use in the drug discovery process, before discussing best practices for developing a highly annotated cohort of PDTX models. We describe the importance of extensive multidimensional molecular and functional characterisation of models and combination drug-drug screens to identify complex biomarkers of drug resistance and response. We reflect on our own experiences and propose the use of a cost-effective intermediate pharmacogenomic platform (the PDTX-PDTC platform) for breast cancer drug and biomarker discovery. We discuss the limitations and unanswered questions of PDTX models; yet, still strongly envision that their use in basic and translational research will dramatically change our understanding of breast cancer biology and how to more effectively treat it..
Bruna, A.
Rueda, O.M.
Greenwood, W.
Batra, A.S.
Callari, M.
Batra, R.N.
Pogrebniak, K.
Sandoval, J.
Cassidy, J.W.
Tufegdzic-Vidakovic, A.
Sammut, S.-.
Jones, L.
Provenzano, E.
Baird, R.
Eirew, P.
Hadfield, J.
Eldridge, M.
McLaren-Douglas, A.
Barthorpe, A.
Lightfoot, H.
O'Connor, M.J.
Gray, J.
Cortes, J.
Baselga, J.
Marangoni, E.
Welm, A.L.
Aparicio, S.
Serra, V.
Garnett, M.J.
Caldas, C.
(2016). A Biobank of Breast Cancer Explants with Preserved Intra-tumor Heterogeneity to Screen Anticancer Compounds. Cell,
Vol.167
(1),
pp. 260-274.e22.
show abstract
full text
The inter- and intra-tumor heterogeneity of breast cancer needs to be adequately captured in pre-clinical models. We have created a large collection of breast cancer patient-derived tumor xenografts (PDTXs), in which the morphological and molecular characteristics of the originating tumor are preserved through passaging in the mouse. An integrated platform combining in vivo maintenance of these PDTXs along with short-term cultures of PDTX-derived tumor cells (PDTCs) was optimized. Remarkably, the intra-tumor genomic clonal architecture present in the originating breast cancers was mostly preserved upon serial passaging in xenografts and in short-term cultured PDTCs. We assessed drug responses in PDTCs on a high-throughput platform and validated several ex vivo responses in vivo. The biobank represents a powerful resource for pre-clinical breast cancer pharmacogenomic studies (http://caldaslab.cruk.cam.ac.uk/bcape), including identification of biomarkers of response or resistance..
Bruna, A.
Rueda, O.M.
Caldas, C.
(2016). Modeling Breast Cancer Intertumor and Intratumor Heterogeneity Using Xenografts. Cold spring harb symp quant biol,
Vol.81,
pp. 227-230.
show abstract
Breast cancer is a heterogeneous disease that can be stratified in at least 10 different subtypes. We present here a platform for derivation of preclinical models based on patient-derived tumor xenografts (PDTXs) that represent these subgroups. These models preserve the transcriptome, methylome, copy-number, and mutational landscape features of the tumor of origin through different passaging. Furthermore, the intratumoral composition of these models is formed by communities of clones very similar to the ones present in the originating tumor. Finally, we show that short-term cultures of cells from these models (PDTX-derived tumor cells, PDTCs) also preserve the molecular features of the tumor and can be used for high-throughput drug testing of single compounds or combinations, with high reproducibility and clinical predictive power..
Cassidy, J.W.
Caldas, C.
Bruna, A.
(2015). Maintaining Tumor Heterogeneity in Patient-Derived Tumor Xenografts. Cancer res,
Vol.75
(15),
pp. 2963-2968.
show abstract
Preclinical models often fail to capture the diverse heterogeneity of human malignancies and as such lack clinical predictive power. Patient-derived tumor xenografts (PDX) have emerged as a powerful technology: capable of retaining the molecular heterogeneity of their originating sample. However, heterogeneity within a tumor is governed by both cell-autonomous (e.g., genetic and epigenetic heterogeneity) and non-cell-autonomous (e.g., stromal heterogeneity) drivers. Although PDXs can largely recapitulate the polygenomic architecture of human tumors, they do not fully account for heterogeneity in the tumor microenvironment. Hence, these models have substantial utility in basic and translational research in cancer biology; however, study of stromal or immune drivers of malignant progression may be limited. Similarly, PDX models offer the ability to conduct patient-specific in vivo and ex vivo drug screens, but stromal contributions to treatment responses may be under-represented. This review discusses the sources and consequences of intratumor heterogeneity and how these are recapitulated in the PDX model. Limitations of the current generation of PDXs are discussed and strategies to improve several aspects of the model with respect to preserving heterogeneity are proposed..
Eirew, P.
Steif, A.
Khattra, J.
Ha, G.
Yap, D.
Farahani, H.
Gelmon, K.
Chia, S.
Mar, C.
Wan, A.
Laks, E.
Biele, J.
Shumansky, K.
Rosner, J.
McPherson, A.
Nielsen, C.
Roth, A.J.
Lefebvre, C.
Bashashati, A.
de Souza, C.
Siu, C.
Aniba, R.
Brimhall, J.
Oloumi, A.
Osako, T.
Bruna, A.
Sandoval, J.L.
Algara, T.
Greenwood, W.
Leung, K.
Cheng, H.
Xue, H.
Wang, Y.
Lin, D.
Mungall, A.J.
Moore, R.
Zhao, Y.
Lorette, J.
Nguyen, L.
Huntsman, D.
Eaves, C.J.
Hansen, C.
Marra, M.A.
Caldas, C.
Shah, S.P.
Aparicio, S.
(2015). Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution. Nature,
Vol.518
(7539),
pp. 422-426.
show abstract
full text
Human cancers, including breast cancers, comprise clones differing in mutation content. Clones evolve dynamically in space and time following principles of Darwinian evolution, underpinning important emergent features such as drug resistance and metastasis. Human breast cancer xenoengraftment is used as a means of capturing and studying tumour biology, and breast tumour xenografts are generally assumed to be reasonable models of the originating tumours. However, the consequences and reproducibility of engraftment and propagation on the genomic clonal architecture of tumours have not been systematically examined at single-cell resolution. Here we show, using deep-genome and single-cell sequencing methods, the clonal dynamics of initial engraftment and subsequent serial propagation of primary and metastatic human breast cancers in immunodeficient mice. In all 15 cases examined, clonal selection on engraftment was observed in both primary and metastatic breast tumours, varying in degree from extreme selective engraftment of minor (<5% of starting population) clones to moderate, polyclonal engraftment. Furthermore, ongoing clonal dynamics during serial passaging is a feature of tumours experiencing modest initial selection. Through single-cell sequencing, we show that major mutation clusters estimated from tumour population sequencing relate predictably to the most abundant clonal genotypes, even in clonally complex and rapidly evolving cases. Finally, we show that similar clonal expansion patterns can emerge in independent grafts of the same starting tumour population, indicating that genomic aberrations can be reproducible determinants of evolutionary trajectories. Our results show that measurement of genomically defined clonal population dynamics will be highly informative for functional studies using patient-derived breast cancer xenoengraftment..
Tufegdzic Vidakovic, A.
Rueda, O.M.
Vervoort, S.J.
Sati Batra, A.
Goldgraben, M.A.
Uribe-Lewis, S.
Greenwood, W.
Coffer, P.J.
Bruna, A.
Caldas, C.
(2015). Context-Specific Effects of TGF-β/SMAD3 in Cancer Are Modulated by the Epigenome. Cell rep,
Vol.13
(11),
pp. 2480-2490.
show abstract
full text
The transforming growth factor beta (TGF-β) signaling pathway exerts opposing effects on cancer cells, acting as either a tumor promoter or a tumor suppressor. Here, we show that these opposing effects are a result of the synergy between SMAD3, a downstream effector of TGF-β signaling, and the distinct epigenomes of breast-tumor-initiating cells (BTICs). These effects of TGF-β are associated with distinct gene expression programs, but genomic SMAD3 binding patterns are highly similar in the BTIC-promoting and BTIC-suppressing contexts. Our data show cell-type-specific patterns of DNA and histone modifications provide a modulatory layer by determining accessibility of genes to regulation by TGF-β/SMAD3. LBH, one such context-specific target gene, is regulated according to its DNA methylation status and is crucial for TGF-β-dependent promotion of BTICs. Overall, these results reveal that the epigenome plays a central and previously overlooked role in shaping the context-specific effects of TGF-β in cancer..
Bruna, A.
Greenwood, W.
Le Quesne, J.
Teschendorff, A.
Miranda-Saavedra, D.
Rueda, O.M.
Sandoval, J.L.
Vidakovic, A.T.
Saadi, A.
Pharoah, P.
Stingl, J.
Caldas, C.
(2012). TGFβ induces the formation of tumour-initiating cells in claudinlow breast cancer. Nat commun,
Vol.3,
p. 1055.
show abstract
The role of transforming growth factor-beta (TGFβ) in the progression of different molecular subtypes of breast cancer has not been clarified. Here we show that TGFβ increases breast tumour-initiating cell (BTIC) numbers but only in claudin(low) breast cancer cell lines by orchestrating a specific gene signature enriched in stem cell processes that predicts worse clinical outcome in breast cancer patients. NEDD9, a member of the Cas family of integrin scaffold proteins, is necessary to mediate these TGFβ-specific effects through a positive feedback loop that integrates TGFβ/Smad and Rho-actin-SRF-dependent signals. In normal human mammary epithelium, TGFβ induces progenitor activity only in the basal/stem cell compartment, where claudin(low) cancers are presumed to arise. These data show opposing responses to TGFβ in both breast malignant cell subtypes and normal mammary epithelial cell subpopulations and suggest therapeutic strategies for a subset of human breast cancers..
Bruna, A.
Darken, R.S.
Rojo, F.
Ocaña, A.
Peñuelas, S.
Arias, A.
Paris, R.
Tortosa, A.
Mora, J.
Baselga, J.
Seoane, J.
(2007). High TGFbeta-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer cell,
Vol.11
(2),
pp. 147-160.
show abstract
TGFbeta acts as a tumor suppressor in normal epithelial cells and early-stage tumors and becomes an oncogenic factor in advanced tumors. The molecular mechanisms involved in the malignant function of TGFbeta are not fully elucidated. We demonstrate that high TGFbeta-Smad activity is present in aggressive, highly proliferative gliomas and confers poor prognosis in patients with glioma. We discern the mechanisms and molecular determinants of the TGFbeta oncogenic response with a transcriptomic approach and by analyzing primary cultured patient-derived gliomas and human glioma biopsies. The TGFbeta-Smad pathway promotes proliferation through the induction of PDGF-B in gliomas with an unmethylated PDGF-B gene. The epigenetic regulation of the PDGF-B gene dictates whether TGFbeta acts as an oncogenic factor inducing PDGF-B and proliferation in human glioma..