Network


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

Hotspot


Dive into the research topics where Mateus Crespo is active.

Publication


Featured researches published by Mateus Crespo.


European Urology | 2014

Visceral disease in castration-resistant prostate cancer

Carmel Pezaro; Aurelius Omlin; David Lorente; Daniel Nava Rodrigues; Roberta Ferraldeschi; Diletta Bianchini; Deborah Mukherji; Ruth Riisnaes; Amelia Altavilla; Mateus Crespo; Nina Tunariu; Johann S. de Bono; Gerhardt Attard

Metastatic involvement of the viscera in men with advanced castration-resistant prostate cancer (CRPC) has been poorly characterised to date. In 359 CRPC patients treated between June 2003 and December 2011, the frequency of radiologically detected visceral metastases before death was 32%. Of the 92 patients with computed tomography performed within 3 mo of death, 49% had visceral metastases. Visceral metastases most commonly involved the liver (20%) and lung (13%). Median survival from diagnosis of visceral disease was 7.1 mo (95% confidence interval, 5.9-8.3). Survival was affected by the degree of bone involvement at detection of visceral disease, varying from 6.1 mo in men with more than six bone metastases to 18.2 mo in men with no bone metastases (p=0.001). Heterogeneity was noted in clinical phenotypes and prostate-specific antigen trends at development of visceral metastases. Visceral metastases are now more commonly detected in men with CRPC, likely due to the introduction of novel survival-prolonging treatments.


British Journal of Cancer | 2013

First-in-human Phase I study of EZN-4176, a locked nucleic acid antisense oligonucleotide to exon 4 of the androgen receptor mRNA in patients with castration-resistant prostate cancer.

Diletta Bianchini; Aurelius Omlin; Carmel Pezaro; David Lorente; Roberta Ferraldeschi; Deborah Mukherji; Mateus Crespo; Ines Figueiredo; Susana Miranda; Ruth Riisnaes; A. Zivi; A Buchbinder; Dana E. Rathkopf; Gerhardt Attard; Howard I. Scher; J. S. De Bono; Daniel C. Danila

Background:Prostate cancer remains dependent of androgen receptor (AR) signalling, even after emergence of castration resistance. EZN-4176 is a third-generation antisense oligonucleotide that binds to the hinge region (exon 4) of AR mRNA resulting in full-length AR mRNA degradation and decreased AR protein expression. This Phase I study aimed to evaluate EZN-4176 in men with castration-resistant prostate cancer (CRPC).Methods:Patients with progressing CRPC were eligible; prior abiraterone and enzalutamide treatment were allowed. EZN-4176 was administered as a weekly (QW) 1-h intravenous infusion. The starting dose was 0.5 mg kg−1 with a 4-week dose-limiting toxicity (DLT) period and a 3+3 modified Fibonacci dose escalation design. After determination of the DLT for weekly administration, an every 2 weeks schedule was initiated.Results:A total of 22 patients were treated with EZN-4176. At 10 mg kg−1 QW, two DLTs were observed due to grade 3–4 ALT or AST elevation. No confirmed biochemical or soft tissue responses were observed. Of eight patients with ⩾5 circulating tumour cells at baseline, a conversion to <5 was observed in three (38%) patients. The most common EZN-4176-related toxicities (all grades) were fatigue (59%), reversible abnormalities in liver function tests ALT (41%) and AST (41%) and infusion-related reactions including chills (36%) and pyrexia (14%).Conclusion:Activity of EZN-4176 at the doses and schedules explored was minimal. The highest dose of 10 mg kg−1 QW was associated with significant but reversible transaminase elevation.


Molecular Cancer Therapeutics | 2012

The Association of PI3 Kinase Signaling and Chemoresistance in Advanced Ovarian Cancer

Craig P. Carden; Aengus Stewart; Parames Thavasu; Emma Kipps; Lorna Pope; Mateus Crespo; Susana Miranda; Gerhardt Attard; Michelle D. Garrett; Paul A. Clarke; Paul Workman; J. S. De Bono; Martin Gore; Stan B. Kaye; Udai Banerji

Evidence that the phosphoinositide 3-kinase (PI3K) pathway is deregulated in ovarian cancer is largely based on the analysis of surgical specimens sampled at diagnosis and may not reflect the biology of advanced ovarian cancer. We aimed to investigate PI3K signaling in cancer cells isolated from patients with advanced ovarian cancer. Ascites samples were analyzed from 88 patients, of whom 61 received further treatment. Cancer cells were immunomagnetically separated from ascites, and the signaling output of the PI3K pathway was studied by quantifying p-AKT, p-p70S6K, and p-GSK3β by ELISA. Relevant oncogenes, such as PIK3CA and AKT, were sequenced by PCR-amplified mass spectroscopy detection methods. In addition, PIK3CA and AKT2 amplifications and PTEN deletions were analyzed by FISH. p-p70S6K levels were significantly higher in cells from 37 of 61 patients who did not respond to subsequent chemotherapy (0.7184 vs. 0.3496; P = 0.0100), and this difference was greater in patients who had not received previous chemotherapy. PIK3CA and AKT mutations were present in 5% and 0% of samples, respectively. Amplification of PIK3CA and AKT2 and deletion of PTEN was seen in 10%, 10%, and 27% of samples, respectively. Mutations of PIK3CA and amplification of PIK3CA/AKT2 or deletion of PTEN did not correlate with levels of p-AKT, p-p70S6K, and p-GSK3β. In patients with advanced ovarian cancer, there is an association between levels of p-p70S6K and response to subsequent chemotherapy. There is no clear evidence that this is driven specifically by PIK3CA or AKT mutations or by amplifications or deletion of PTEN. Mol Cancer Ther; 11(7); 1609–17. ©2012 AACR.


British Journal of Cancer | 2015

Androgen receptor expression in Circulating Tumor Cells from castration-resistant prostate cancer patients with novel endocrine agents

Mateus Crespo; G. van Dalum; Roberta Ferraldeschi; Zafeiris Zafeiriou; Spyridon Sideris; David Lorente; Diletta Bianchini; Daniel Nava Rodrigues; Ruth Riisnaes; Susana Miranda; Ines Figueiredo; Penny Flohr; Karolina Nowakowska; J. S. De Bono; Leonardus Wendelinus Mathias Marie Terstappen; G. Attard

Background:Abiraterone and enzalutamide are novel endocrine treatments that abrogate androgen receptor (AR) signalling in castration-resistant prostate cancer (CRPC). Here, we developed a circulating tumour cells (CTCs)-based assay to evaluate AR expression in real-time in CRPC and investigated nuclear AR expression in CTCs in patients treated with enzalutamide and abiraterone.Methods:CTCs were captured and characterised using the CellSearch system. An automated algorithm to identify CTCs and quantify AR expression was employed. The primary aim was to evaluate the association between CTC AR expression and prior treatment with abiraterone or enzalutamide.Results:AR expression in CTCs was evaluated in 94 samples from 48 metastatic CRPC patients. We observed large intra-patient heterogeneity of AR expression in CTCs. Prior exposure to abiraterone or enzalutamide was not associated with a change in CTCs AR expression (median intensity and distribution of AR-positive classes). In support of this, we also confirmed maintained nuclear AR expression in tissue samples collected after progression on abiraterone. AR staining also identified additional AR-positive CD45-negative circulating cells that were CK-negative/weak and therefore missed using standard protocols. The number of these events correlated with traditional CTCs and was associated with worse outcome on univariate analysis.Conclusions:We developed a non-invasive method to monitor AR nuclear expression in CTCs. Our studies confirm nuclear AR expression in CRPC patients progressing on novel endocrine treatments. Owing to the significant heterogeneity of AR expression in CTCs, studies in larger cohorts of patients are required to identify associations with outcome.


Journal of the National Cancer Institute | 2014

Preclinical Evaluation of Imaging Biomarkers for Prostate Cancer Bone Metastasis and Response to Cabozantinib

Timothy J. Graham; Gary Box; Nina Tunariu; Mateus Crespo; Terry J. Spinks; Susana Miranda; Gerhardt Attard; Johann S. de Bono; Suzanne A. Eccles; Faith E. Davies; Simon P. Robinson

BACKGROUND Prostate cancer is incurable once it has metastasized to the bone. Appropriate preclinical models are lacking. The therapeutic efficacy of the multikinase inhibitor cabozantinib was assessed in an orthotopic xenograft model of castration-resistant prostate cancer (CRPC) bone metastasis using noninvasive, multimodality functional imaging. METHODS NOD/SCID mice were injected intratibially with luciferase-expressing ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) rearranged VCaP human prostate carcinoma cells. The response of VCaP xenografts (n = 7 per group) to cabozantinib was investigated using bioluminescence imaging and anatomical and diffusion weighted magnetic resonance imaging. This enabled quantitation of tumor volume and apparent diffusion coefficient (ADC). Bone uptake of technetium-methylene diphosphonate ((99m)Tc-MDP) was assessed by single-photon emission computed tomography. Ex vivo micro computed tomography was used to quantify bone volume and correlated with appropriate histopathology. Statistical significance was determined using the two-sided Mann-Whitney test or Wilcoxon signed rank test. RESULTS VCaP xenografts were predominantly osteosclerotic with some osteolytic activity. Fluorescent in situ hybridization analysis confirmed retention of ERG oncogene rearrangements. Cabozantinib induced a statistically significant 52% reduction in tumor luminance (P = .02) and stasis in tumor volume after 15 days of treatment. Tumor ADC statistically significantly increased with cabozantinib and was associated with extensive necrosis (after 10 days, mean tumor ADC ± SD = 556±43×10(-6) mm(2)/s vs pretreatment ADC = 485±43×10(-6) mm(2)/s; P = .02 ). Tumor-associated uptake of (99m)Tc-MDP was statistically significantly reduced after 3 days of treatment (P = .02), sustained over 15 days treatment, and associated with a statistically significant (P = .048) reduction in bone growth on the tibial cortex, yet a highly statistically significant (P = .001) increase in trabecular bone volume. CONCLUSIONS The intratibial VCaP model faithfully emulates clinical disease. Cabozantinib exerts potent effects on both tumor and tumor-induced bone matrix remodeling, and quantitation of ADC provides a clinically translatable imaging biomarker for early, sensitive assessment of treatment response in CRPC bone metastasis.


European Urology | 2016

Decline in Circulating Tumor Cell Count and Treatment Outcome in Advanced Prostate Cancer

David Lorente; David Olmos; Joaquin Mateo; Diletta Bianchini; George Seed; Martin Fleisher; Daniel C. Danila; Penny Flohr; Mateus Crespo; Ines Figueiredo; Susana Miranda; Kurt Baeten; Arturo Molina; Thian Kheoh; Robert McCormack; Leon W.M.M. Terstappen; Howard I. Scher; Johann S. de Bono

Background Treatment response biomarkers are urgently needed for castration-resistant prostate cancer (CRPC). Baseline and post-treatment circulating tumor cell (CTC) counts of ≥5 cells/7.5 ml are associated with poor CRPC outcome. Objective To determine the value of a ≥30% CTC decline as a treatment response indicator. Design, setting, and participants We identified patients with a baseline CTC count ≥5 cells/7.5 ml and evaluable post-treatment CTC counts in two prospective trials. Intervention Patients were treated in the COU-AA-301 (abiraterone after chemotherapy) and IMMC-38 (chemotherapy) trials. Outcome measures and statistical analysis The association between a ≥30% CTC decline after treatment and survival was evaluated using univariable and multivariable Cox regression models at three landmark time points (4, 8, and 12 wk). Model performance was evaluated by calculating the area under the receiver operating characteristic curve (AUC) and c-indices. Results Overall 486 patients (122 in IMMC-38 and 364 in COU-AA-301) had a CTC count ≥5 cells/7.5 ml at baseline, with 440, 380, and 351 patients evaluable at 4, 8, and 12 wk, respectively. A 30% CTC decline was associated with increased survival at 4 wk (hazard ratio [HR] 0.45, 95% confidence interval [CI] 0.36–0.56; p < 0.001), 8 wk (HR 0.41, 95% CI 0.33–0.53; p < 0.001), and 12 wk (HR 0.39, 95% CI 0.3–0.5; p < 0.001) in univariable and multivariable analyses. Stable CTC count (<30% fall or <30% increase) was not associated with a survival benefit when compared with increased CTC count. The association between a 30% CTC decline after treatment and survival was independent of baseline CTC count. CTC declines significantly improved the AUC at all time-points. Finally, in the COU-AA-301 trial, patients with CTC ≥5 cells/7.5 ml and a 30% CTC decline had similar overall survival in both arms. Conclusions A 30% CTC decline after treatment from an initial count ≥5 cells/7.5 ml is independently associated with CRPC overall survival following abiraterone and chemotherapy, improving the performance of a multivariable model as early as 4 wk after treatment. This potential surrogate must now be prospectively evaluated. Patient summary Circulating tumor cells (CTCs) are cancer cells that can be detected in the blood of prostate cancer patients. We analyzed changes in CTCs after treatment with abiraterone and chemotherapy in two large clinical trials, and found that patients who have a decline in CTC count have a better survival outcome.


Cancer Research | 2016

Second-Generation HSP90 Inhibitor Onalespib Blocks mRNA Splicing of Androgen Receptor Variant 7 in Prostate Cancer Cells

Roberta Ferraldeschi; Jonathan C. Welti; Marissa V. Powers; Wei Yuan; Tomoko Smyth; George Seed; Ruth Riisnaes; Somaieh Hedayat; Hannah Wang; Mateus Crespo; Daniel Nava Rodrigues; Ines Figueiredo; Susana Miranda; Suzanne Carreira; John Lyons; Swee Y. Sharp; Stephen R. Plymate; Gerhardt Attard; Nicola G. Wallis; Paul Workman; Johann S. de Bono

Resistance to available hormone therapies in prostate cancer has been associated with alternative splicing of androgen receptor (AR) and specifically, the expression of truncated and constitutively active AR variant 7 (AR-V7). The transcriptional activity of steroid receptors, including AR, is dependent on interactions with the HSP90 chaperone machinery, but it is unclear whether HSP90 modulates the activity or expression of AR variants. Here, we investigated the effects of HSP90 inhibition on AR-V7 in prostate cancer cell lines endogenously expressing this variant. We demonstrate that AR-V7 and full-length AR (AR-FL) were depleted upon inhibition of HSP90. However, the mechanisms underlying AR-V7 depletion differed from those for AR-FL. Whereas HSP90 inhibition destabilized AR-FL and induced its proteasomal degradation, AR-V7 protein exhibited higher stability than AR-FL and did not require HSP90 chaperone activity. Instead, HSP90 inhibition resulted in the reduction of AR-V7 mRNA levels but did not affect total AR transcript levels, indicating that HSP90 inhibition disrupted AR-V7 splicing. Bioinformatic analyses of transcriptome-wide RNA sequencing data confirmed that the second-generation HSP90 inhibitor onalespib altered the splicing of at least 557 genes in prostate cancer cells, including AR. These findings indicate that the effects of HSP90 inhibition on mRNA splicing may prove beneficial in prostate cancers expressing AR-V7, supporting further clinical investigation of HSP90 inhibitors in malignancies no longer responsive to androgen deprivation. Cancer Res; 76(9); 2731-42. ©2016 AACR.


BJUI | 2017

Phenotypic diversity of circulating tumour cells in patients with metastatic castration-resistant prostate cancer

Andrew S. McDaniel; Roberta Ferraldeschi; Rachel Krupa; Mark Landers; Ryon Graf; Jessica Louw; Adam Jendrisak; Natalee Bales; Dena Marrinucci; Zafeiris Zafeiriou; Penelope Flohr; Spyridon Sideris; Mateus Crespo; Ines Figueiredo; Joaquin Mateo; Johann S. de Bono; Ryan Dittamore; Scott A. Tomlins; Gerhardt Attard

To use a non‐biased assay for circulating tumour cells (CTCs) in patients with prostate cancer (PCa) in order to identify non‐traditional CTC phenotypes potentially excluded by conventional detection methods that are reliant on antigen‐ and/or size‐based enrichment.


Annals of Oncology | 2017

CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error-prone double-strand break repair

T. R Shenoy; G Boysen; M. Y Wang; Q. Z Xu; Weilong Guo; Fong Ming Koh; C Wang; L. Z Zhang; Ying Wang; Gil; Sara Aziz; Rossitza Christova; Daniel Nava Rodrigues; Mateus Crespo; Pasquale Rescigno; Nina Tunariu; Ruth Riisnaes; Zafeiris Zafeiriou; Penny Flohr; Wei Yuan; Eleanor Knight; Amanda Swain; Miguel Ramalho-Santos; D. Y Xu; J. S. De Bono; Hong Wu

Background Deletion of the chromatin remodeler chromodomain helicase DNA-binding protein 1 (CHD1) is a common genomic alteration found in human prostate cancers (PCas). CHD1 loss represents a distinct PCa subtype characterized by SPOP mutation and higher genomic instability. However, the role of CHD1 in PCa development in vivo and its clinical utility remain unclear. Patients and methods To study the role of CHD1 in PCa development and its loss in clinical management, we generated a genetically engineered mouse model with prostate-specific deletion of murine Chd1 as well as isogenic CHD1 wild-type and homozygous deleted human benign and PCa lines. We also developed patient-derived organoid cultures and screened patients with metastatic PCa for CHD1 loss. Results We demonstrate that CHD1 loss sensitizes cells to DNA damage and causes a synthetic lethal response to DNA damaging therapy in vitro, in vivo, ex vivo, in patient-derived organoid cultures and in a patient with metastatic PCa. Mechanistically, CHD1 regulates 53BP1 stability and CHD1 loss leads to decreased error-free homologous recombination (HR) repair, which is compensated by increased error-prone non-homologous end joining (NHEJ) repair for DNA double-strand break (DSB) repair. Conclusions Our study provides the first in vivo and in patient evidence supporting the role of CHD1 in DSB repair and in response to DNA damaging therapy. We uncover mechanistic insights that CHD1 modulates the choice between HR and NHEJ DSB repair and suggest that CHD1 loss may contribute to the genomic instability seen in this subset of PCas.


Cancer Research | 2017

Disrupting Androgen Receptor Signaling Induces Snail-Mediated Epithelial–Mesenchymal Plasticity in Prostate Cancer

Lu Miao; Lin Yang; Rui Li; Daniel Nava Rodrigues; Mateus Crespo; Jer Tsong Hsieh; Wayne D. Tilley; Johann S. de Bono; Luke A. Selth; Ganesh V. Raj

Epithelial-to-mesenchymal plasticity (EMP) has been linked to metastasis, stemness, and drug resistance. In prostate cancer, EMP has been associated with both suppression and activation of the androgen receptor (AR) signaling. Here we investigated the effect of the potent AR antagonist enzalutamide on EMP in multiple preclinical models of prostate cancer and patient tissues. Enzalutamide treatment significantly enhanced the expression of EMP drivers (ZEB1, ZEB2, Snail, Twist, and FOXC2) and mesenchymal markers (N-cadherin, fibronectin, and vimentin) in prostate cancer cells, enhanced prostate cancer cell migration, and induced prostate cancer transformation to a spindle, fibroblast-like morphology. Enzalutamide-induced EMP required concomitant suppression of AR signaling and activation of the EMP-promoting transcription factor Snail, as evidenced by both knockdown and overexpression studies. Supporting these findings, AR signaling and Snail expression were inversely correlated in C4-2 xenografts, patient-derived castration-resistant metastases, and clinical samples. For the first time, we elucidate a mechanism explaining the inverse relationship between AR and Snail. Specifically, we found that AR directly repressed SNAI1 gene expression by binding to specific AR-responsive elements within the SNAI1 promoter. Collectively, our findings demonstrate that de-repression of Snail and induction of EMP is an adaptive response to enzalutamide with implications for therapy resistance. Cancer Res; 77(11); 3101-12. ©2017 AACR.

Collaboration


Dive into the Mateus Crespo's collaboration.

Top Co-Authors

Avatar

Johann S. de Bono

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Susana Miranda

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Ines Figueiredo

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Ruth Riisnaes

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Diletta Bianchini

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Joaquin Mateo

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Wei Yuan

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Adam Sharp

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

George Seed

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge