38 results on '"Saturno G"'
Search Results
2. The paradox-breaking panRAF plus SRC family kinase inhibitor, CCT3833, is effective in mutant KRAS-driven cancers
- Author
-
Saturno, G., Lopes, F., Niculescu-Duvaz, I., Niculescu-Duvaz, D., Zambon, A., Davies, L., Johnson, L., Preece, N., Lee, R., Viros, A., Holovanchuk, D., Pedersen, M., McLeary, R., Lorigan, P., Dhomen, N., Fisher, C., Banerji, U., Dean, E., Krebs, M.G., Gore, M., Larkin, J., Marais, R., and Springer, C.
- Published
- 2021
- Full Text
- View/download PDF
3. P-141 Independent external pre-clinical validation of iDAScore v1.0 in 1232 PGT-A cycles with 3604 biopsied blastocysts and 808 euploid transfers.
- Author
-
Casciani, V, primary, Cimadomo, D, additional, Chiappetta, V, additional, Innocenti, F, additional, Saturno, G, additional, Albricci, L, additional, Maggiulli, R, additional, Coticchio, G, additional, Ahlström, A, additional, Berntsen, J, additional, Larman, M, additional, Borini, A, additional, Vaiarelli, A, additional, Ubaldi, F M, additional, and Rienzi, L, additional
- Published
- 2023
- Full Text
- View/download PDF
4. P-297 Comprehensive artificial intelligence-powered investigation of blastocyst expansion dynamics: associations with competence
- Author
-
Trio, S, primary, Innocenti, F, additional, Saturno, G, additional, Taggi, M, additional, Hickman, C, additional, Kantor, B, additional, Taub, D, additional, Ben-Meir, A, additional, Har-Vardi, I, additional, Marconetto, A, additional, Vitiello, C, additional, Alfano, S, additional, Ubaldi, F M, additional, Rienzi, L, additional, and Cimadomo, D, additional
- Published
- 2023
- Full Text
- View/download PDF
5. O-119 Time-lapse comprehensive portrait of cytoplasmic strings appearing during blastulation: prevalence and implications for embryo assessment.
- Author
-
Marconetto, A, primary, Innocenti, F, additional, Saturno, G, additional, Taggi, M, additional, Chiappetta, V, additional, Albricci, L, additional, Maggiulli, R, additional, Soscia, D, additional, Casciani, V, additional, De Falco, F, additional, Coticchio, G, additional, Ahlström, A, additional, Ubaldi, F M, additional, Rienzi, L, additional, and Cimadomo, D, additional
- Published
- 2023
- Full Text
- View/download PDF
6. Whole-genome sequencing reveals complex mechanisms of intrinsic resistance to BRAF inhibition
- Author
-
Turajlic, S., Furney, S.J., Stamp, G., Rana, S., Ricken, G., Oduko, Y., Saturno, G., Springer, C., Hayes, A., Gore, M., Larkin, J., and Marais, R.
- Published
- 2014
- Full Text
- View/download PDF
7. Selective modulation of ER-β by estradiol and xenoestrogens in human breast cancer cell lines
- Author
-
Cappelletti, V., Saturno, G., Miodini, P., Körner, W., and Daidone, M. G.
- Published
- 2003
- Full Text
- View/download PDF
8. Erratum: Paradox-Breaking RAF Inhibitors that Also Target SRC Are Effective in Drug-Resistant BRAF Mutant Melanoma (Cancer Cell (2015) 27(1) (85–96) (S1535610814004577) (10.1016/j.ccell.2014.11.006))
- Author
-
Girotti, M. R., Lopes, F., Preece, N., Niculescu-Duvaz, D., Zambon, A., Davies, L., Whittaker, S., Saturno, G., Viros, A., Pedersen, M., Suijkerbuijk, B. M. J. M., Menard, D., Mcleary, R., Johnson, L., Fish, L., Ejiama, S., Sanchez-Laorden, B., Hohloch, J., Carragher, N., Macleod, K., Ashton, G., Marusiak, A. A., Fusi, A., Brognard, J., Frame, M., Lorigan, P., Marais, R., and Springer, C.
- Published
- 2017
9. Gene-specific interactions between ultraviolet radiation and melanoma
- Author
-
Viriós, A., primary, Pedersen, M., additional, Furney, S.J., additional, Girotti, M.R., additional, Hogan, K., additional, Saturno, G., additional, Galvani, E., additional, Sanchez-Laorden, B., additional, Ng, C., additional, Reis-Filho, J.S., additional, Lorigan, P., additional, Cook, M., additional, and Marais, R., additional
- Published
- 2016
- Full Text
- View/download PDF
10. Lysyl oxidase regulates EGFR signalling through the extracellular matrix
- Author
-
Tang, H., primary, Leung, L., additional, Saturno, G., additional, Viros, A., additional, Di Leva, G., additional, Morrison, E., additional, Smith, D., additional, Johnson, L., additional, Dhomen, N., additional, Springer, C., additional, and Marais, R., additional
- Published
- 2016
- Full Text
- View/download PDF
11. Therapeutic efficacy of the paradox-breaking panRAF and SRC drug CCT3833/BAL3833 in KRAS-driven cancer models
- Author
-
Saturno, G., primary, Lopes, F., additional, Girotti, M.R., additional, Niculescu-Duvaz, I., additional, Niculescu-Duvaz, D., additional, Zambon, A., additional, Davies, L., additional, Johnson, L., additional, Preece, N., additional, Viros, A., additional, Pedersen, M., additional, McLeary, R., additional, Knight, R., additional, Lee, R., additional, Holovanchuk, D., additional, Fusi, A., additional, Lorigan, P., additional, Dhomen, N., additional, Marais, R., additional, and Springer, C., additional
- Published
- 2016
- Full Text
- View/download PDF
12. Application of sequencing, liquid biopsies and patient-derived xenografts for personalized medicine in melanoma
- Author
-
Girotti, M.R., primary, Gremel, G., additional, Lee, R., additional, Galvani, E., additional, Rothwell, D., additional, Mandal, A.K., additional, Lim, K.H.J., additional, Saturno, G., additional, Furney, S.J., additional, Baenke, F., additional, Pedersen, M., additional, Smith, M., additional, Fusi, A., additional, Dhomen, N., additional, Brady, G., additional, Lorigan, P., additional, Dive, C., additional, and Marais, R., additional
- Published
- 2016
- Full Text
- View/download PDF
13. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy
- Author
-
Miller, BW, Morton, JP, Pinese, M, Saturno, G, Jamieson, NB, McGhee, E, Timpson, P, Leach, J, McGarry, L, Shanks, E, Bailey, P, Chang, D, Oien, K, Karim, S, Au, A, Steele, C, Carter, CR, McKay, C, Anderson, K, Evans, TRJ, Marais, R, Springer, C, Biankin, A, Erler, JT, Sansom, OJ, Miller, BW, Morton, JP, Pinese, M, Saturno, G, Jamieson, NB, McGhee, E, Timpson, P, Leach, J, McGarry, L, Shanks, E, Bailey, P, Chang, D, Oien, K, Karim, S, Au, A, Steele, C, Carter, CR, McKay, C, Anderson, K, Evans, TRJ, Marais, R, Springer, C, Biankin, A, Erler, JT, and Sansom, OJ
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality. Despite significant advances made in the treatment of other cancers, current chemotherapies offer little survival benefit in this disease. Pancreaticoduodenectomy offers patients the possibility of a cure, but most will die of recurrent or metastatic disease. Hence, preventing metastatic disease in these patients would be of significant benefit. Using principal component analysis (PCA), we identified a LOX/hypoxia signature associated with poor patient survival in resectable patients. We found that LOX expression is upregulated in metastatic tumors from Pdx1-Cre Kras(G12D/+) Trp53(R172H/+) (KPC) mice and that inhibition of LOX in these mice suppressed metastasis. Mechanistically, LOX inhibition suppressed both migration and invasion of KPC cells. LOX inhibition also synergized with gemcitabine to kill tumors and significantly prolonged tumor-free survival in KPC mice with early-stage tumors. This was associated with stromal alterations, including increased vasculature and decreased fibrillar collagen, and increased infiltration of macrophages and neutrophils into tumors. Therefore, LOX inhibition is able to reverse many of the features that make PDAC inherently refractory to conventional therapies and targeting LOX could improve outcome in surgically resectable disease.
- Published
- 2015
14. 862 - Therapeutic efficacy of the paradox-breaking panRAF and SRC drug CCT3833/BAL3833 in KRAS-driven cancer models
- Author
-
Saturno, G., Lopes, F., Girotti, M.R., Niculescu-Duvaz, I., Niculescu-Duvaz, D., Zambon, A., Davies, L., Johnson, L., Preece, N., Viros, A., Pedersen, M., McLeary, R., Knight, R., Lee, R., Holovanchuk, D., Fusi, A., Lorigan, P., Dhomen, N., Marais, R., and Springer, C.
- Published
- 2016
- Full Text
- View/download PDF
15. 836 - Application of sequencing, liquid biopsies and patient-derived xenografts for personalized medicine in melanoma
- Author
-
Girotti, M.R., Gremel, G., Lee, R., Galvani, E., Rothwell, D., Mandal, A.K., Lim, K.H.J., Saturno, G., Furney, S.J., Baenke, F., Pedersen, M., Smith, M., Fusi, A., Dhomen, N., Brady, G., Lorigan, P., Dive, C., and Marais, R.
- Published
- 2016
- Full Text
- View/download PDF
16. 297 - Lysyl oxidase regulates EGFR signalling through the extracellular matrix
- Author
-
Tang, H., Leung, L., Saturno, G., Viros, A., Di Leva, G., Morrison, E., Smith, D., Johnson, L., Dhomen, N., Springer, C., and Marais, R.
- Published
- 2016
- Full Text
- View/download PDF
17. 289 - Gene-specific interactions between ultraviolet radiation and melanoma
- Author
-
Viriós, A., Pedersen, M., Furney, S.J., Girotti, M.R., Hogan, K., Saturno, G., Galvani, E., Sanchez-Laorden, B., Ng, C., Reis-Filho, J.S., Lorigan, P., Cook, M., and Marais, R.
- Published
- 2016
- Full Text
- View/download PDF
18. Modulation of cardiac contractility by antagonism of pleckstrin-homology (PH) domain and akt-1 silencing
- Author
-
Chisci, R., primary, Rocchetti, M., additional, Menduni, F., additional, Mostacciuolo, G., additional, Cipolla, L., additional, Saturno, G., additional, Castoldi, R., additional, Venturi, M., additional, and Zaza, A., additional
- Published
- 2008
- Full Text
- View/download PDF
19. Selective modulation of ER-? by estradiol and xenoestrogens in human breast cancer cell lines
- Author
-
Cappelletti, V., primary, Saturno, G., additional, Miodini, P., additional, K�rner, W., additional, and Daidone, M. G., additional
- Published
- 2003
- Full Text
- View/download PDF
20. Application of sequencing, liquid biopsies and patient-derived xenografts for personalized medicine in melanoma
- Author
-
Girotti MR, Gremel G, Lee R, Galvani E, Rothwell D, Viros A, Amit Kumar Mandal, Kh, Lim, Saturno G, Sj, Furney, Baenke F, Pedersen M, Rogan J, Swan J, Smith M, Fusi A, Oudit D, Dhomen N, Brady G, and Lorigan P
21. Cytoplasmic strings in human blastocysts: hypotheses of their role and implications for embryo selection.
- Author
-
Marconetto A, Innocenti F, Saturno G, Taggi M, Chiappetta V, Trio S, De Falco F, Albricci L, Coticchio G, Ahlström A, Fiorentino G, Maggiulli R, Vaiarelli A, Zuccotti M, Rienzi L, and Cimadomo D
- Subjects
- Humans, Female, Retrospective Studies, Pregnancy, Adult, Cytoplasm metabolism, Aneuploidy, Vitrification, Embryo Transfer methods, Fertilization in Vitro, Embryo Implantation physiology, Blastocyst, Embryo Culture Techniques, Embryonic Development physiology
- Abstract
Study Question: What are the implications of the presence cytoplasmic strings (Cyt-S) and their quantity and dynamics for the pre-implantation development of human blastocysts?, Summary Answer: Cyt-S are common in human embryos and are associated with faster blastocyst development, larger expansion, and better morphological quality., What Is Known Already: Cyt-S are dynamic cellular projections connecting inner cell mass and trophectoderm (TE) cells, that can be observed during blastocyst expansion. Their prevalence in human embryos has been estimated to be between 44% and 93%. Data relevant to their clinical implications and role in development are lacking, limited, or controversial., Study Design, Size, Duration: Retrospective study conducted at a single IVF center between May 2013 and November 2014 and involving 124 pre-implantation genetic testing for aneuploidy cycles in a time-lapse incubator with ≥1 blastocyst biopsied and vitrified (N = 370 embryos assessed). These cycles resulted in 87 vitrified-warmed single-euploid blastocyst transfers., Participants/materials, Setting, Methods: ICSI, continuous blastocyst culture (Days 5-7), TE biopsy of fully expanded blastocysts without Day 3 zona pellucida drilling, qPCR to assess uniform full-chromosome aneuploidies, and vitrification were all performed. Only vitrified-warmed euploid single-embryo-transfers were conducted. Blastocyst morphological quality was defined according to Gardner's criteria. The AI-based software CHLOE™ (Fairtility) automatically registered timings from time of starting blastulation (tSB) to biopsy (t-biopsy, i.e. blastocyst full-expansion) as hours-post-insemination (hpi), embryo area (including zona pellucida in µm2), and spontaneous blastocyst collapses. One senior embryologist manually annotated Cyt-S presence, quantity, timings, and type (thick cell-to-cell connections and/or threads). All significant associations were confirmed through regression analyses. All couples', cycles', and embryos' main features were also tested for associations with Cyt-S presence, quantity, and dynamics., Main Results and the Role of Chance: About 94.3% of the patients (N = 117/124) had ≥1 embryo with Cyt-S. Out of a total of 370 blastocysts, 55 degenerated between blastulation and full-expansion (N = 55/370, 14.9%). The degeneration rate among embryos with ≥1 Cyt-S was 10.8% (N = 33/304), significantly lower than that of embryos without Cyt-S (33.3%, N = 22/66, P < 0.01). Of the remaining 315 viable blastocysts analyzed, 86% (N = 271/315; P < 0.01) had ≥1 Cyt-S, on average 3.5 ± 2.1 per embryo ranging 1-13. The first Cyt-S per viable embryo appeared at 115.3 ± 12.5 hpi (85.7-157.7), corresponding to 10.5 ± 5.8 h (0.5-31) after tSB. Overall, we analyzed 937 Cyt-S showing a mean duration of 3.8 ± 2.7 h (0.3-20.9). Cyt-S were mostly threads (N = 508/937, 54.2%) or thick cell-to-cell connections becoming threads (N = 382/937, 40.8%) than thick bridges (N = 47/937, 5.0%). The presence and quantity of Cyt-S were significantly associated with developmentally faster (on average 6-12 h faster) and more expanded (on average 2700 µm2-larger blastocyst's area at t-biopsy) embryos. Also, the presence and duration of Cyt-S were associated with better morphology. Lastly, while euploidy rates were comparable between blastocysts with and without Cyt-S, all euploid blastocysts transferred from the latter group failed to implant (N = 10)., Limitations, Reasons for Caution: Cyt-S presence and dynamics were assessed manually on seven focal planes from video frames recorded every 15 min. The patients included were mostly of advanced maternal age. Only associations could be reported, but no causations/consequences. Lastly, larger datasets are required to better assess Cyt-S associations with clinical outcomes., Wider Implications of the Findings: Cyt-S are common during human blastocyst expansion, suggesting their physiological implication in this process. Their presence, quantity and dynamics mirror embryo viability, and morphological quality, yet their role is still unknown. Future basic science studies are encouraged to finally describe Cyt-S molecular nature and biophysical properties, and Artificial Intelligence tools should aid these studies by incorporating Cyt-S assessment., Study Funding/competing Interest(s): None., Trial Registration Number: N/A., (© The Author(s) 2024. Published by Oxford University Press on behalf of European Society of Human Reproduction and Embryology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
22. Quantitative Standardized Expansion Assay: An Artificial Intelligence-Powered Morphometric Description of Blastocyst Expansion and Zona Thinning Dynamics.
- Author
-
Cimadomo D, Trio S, Canosi T, Innocenti F, Saturno G, Taggi M, Soscia DM, Albricci L, Kantor B, Dvorkin M, Svensson A, Huang T, Vaiarelli A, Gennarelli G, and Rienzi L
- Abstract
Artificial intelligence applied to time-lapse microscopy may revolutionize embryo selection in IVF by automating data collection and standardizing the assessments. In this context, blastocyst expansion dynamics, although being associated with reproductive fitness, have been poorly studied. This retrospective study (N = 2184 blastocysts from 786 cycles) exploited both technologies to picture the association between embryo and inner-cell-mass (ICM) area in µm
2 , the ICM/Trophectoderm ratio, and the zona pellucida thickness in µm (zp-T) at sequential blastocyst expansion stages, with (i) euploidy and (ii) live-birth per transfer (N = 548 transfers). A quantitative-standardized-expansion-assay (qSEA) was also set-up; a novel approach involving automatic annotations of all expansion metrics every 30 min across 5 h following blastulation. Multivariate regressions and ROC curve analyses were conducted. Aneuploid blastocysts were slower, expanded less and showed thicker zp. The qSEA outlined faster and more consistent zp thinning processes among euploid blastocysts, being more or as effective as the embryologists in ranking euploid embryo as top-quality of their cohorts in 69% of the cases. The qSEA also outlined faster and more consistent blastocyst expansion and zp thinning dynamics among euploid implanted versus not implanted blastocysts, disagreeing with embryologists' priority choice in about 50% of the cases. In conclusion, qSEA is a promising objective, quantitative, and user-friendly strategy to predict embryo competence that now deserves prospective validations.- Published
- 2024
- Full Text
- View/download PDF
23. Discovery and optimization of 4-pyrazolyl-2-aminopyrimidine derivatives as potent spleen tyrosine kinase (SYK) inhibitors.
- Author
-
Cervi G, D'Alessio R, Bindi S, Buffa L, Burocchi A, Canevari G, Modugno M, Motto I, Saturno G, and Orsini P
- Subjects
- Humans, Syk Kinase metabolism, Intracellular Signaling Peptides and Proteins metabolism, Signal Transduction, Phosphorylation, Protein Kinase Inhibitors therapeutic use, Protein-Tyrosine Kinases, Hematologic Neoplasms drug therapy, Pyrimidines
- Abstract
Spleen tyrosine kinase (Syk) is a key signal transduction mediator of the B cell receptor (BCR) signaling pathway. Abnormal BCR signaling plays a key role in initiation and development of B-cell-derived hematological malignancies, therefore, Syk represents a potential target for inhibiting the BCR signaling resulting in a therapeutic effect in these cancers. Herein, we describe a novel series of SYK inhibitors with 4-(3'-pyrazolyl)-2-amino-pyrimidine scaffold. Extensive study of structure-activity relationships led to the identification of 1 (NMS-0963), a highly potent Syk inhibitor (IC
50 = 3 nM) endowed with high selectivity within a panel of tested kinases and high antiproliferative activity in SYK-dependent BaF3-TEL/SYK cells and in other BCR-dependent hematological tumor cell lines. Additionally, 1 effectively inhibited Syk phosphorylation and downstream signaling mediators of the BCR in treated cells. In in vivo pharmacokinetics studies, 1, displayed good pharmacokinetics properties, with linear exposure with dose and excellent oral bioavailability. These findings suggest that 1 is a promising new Syk inhibitor for treating BCR-dependent hematological cancers., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Masson SAS. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
24. How should the best human embryo in vitro be? Current and future challenges for embryo selection.
- Author
-
Cimadomo D, Innocenti F, Taggi M, Saturno G, Campitiello MR, Guido M, Vaiarelli A, Ubaldi FM, and Rienzi L
- Subjects
- Humans, Genetic Testing, Fertilization in Vitro, Aneuploidy, Artificial Intelligence, Blastocyst pathology
- Abstract
In-vitro fertilization (IVF) aims at overcoming the causes of infertility and lead to a healthy live birth. To maximize IVF efficiency, it is critical to identify and transfer the most competent embryo within a cohort produced by a couple during a cycle. Conventional static embryo morphological assessment involves sequential observations under a light microscope at specific timepoints. The introduction of time-lapse technology enhanced morphological evaluation via the continuous monitoring of embryo preimplantation in vitro development, thereby unveiling features otherwise undetectable via multiple static assessments. Although an association exists, blastocyst morphology poorly predicts chromosomal competence. In fact, the only reliable approach currently available to diagnose the embryonic karyotype is trophectoderm biopsy and comprehensive chromosome testing to assess non-mosaic aneuploidies, namely preimplantation genetic testing for aneuploidies (PGT-A). Lately, the focus is shifting towards the fine-tuning of non-invasive technologies, such as "omic" analyses of waste products of IVF (e.g., spent culture media) and/or artificial intelligence-powered morphologic/morphodynamic evaluations. This review summarizes the main tools currently available to assess (or predict) embryo developmental, chromosomal, and reproductive competence, their strengths, the limitations, and the most probable future challenges.
- Published
- 2024
- Full Text
- View/download PDF
25. Towards Automation in IVF: Pre-Clinical Validation of a Deep Learning-Based Embryo Grading System during PGT-A Cycles.
- Author
-
Cimadomo D, Chiappetta V, Innocenti F, Saturno G, Taggi M, Marconetto A, Casciani V, Albricci L, Maggiulli R, Coticchio G, Ahlström A, Berntsen J, Larman M, Borini A, Vaiarelli A, Ubaldi FM, and Rienzi L
- Abstract
Preimplantation genetic testing for aneuploidies (PGT-A) is arguably the most effective embryo selection strategy. Nevertheless, it requires greater workload, costs, and expertise. Therefore, a quest towards user-friendly, non-invasive strategies is ongoing. Although insufficient to replace PGT-A, embryo morphological evaluation is significantly associated with embryonic competence, but scarcely reproducible. Recently, artificial intelligence-powered analyses have been proposed to objectify and automate image evaluations. iDAScore v1.0 is a deep-learning model based on a 3D convolutional neural network trained on time-lapse videos from implanted and non-implanted blastocysts. It is a decision support system for ranking blastocysts without manual input. This retrospective, pre-clinical, external validation included 3604 blastocysts and 808 euploid transfers from 1232 cycles. All blastocysts were retrospectively assessed through the iDAScore v1.0; therefore, it did not influence embryologists' decision-making process. iDAScore v1.0 was significantly associated with embryo morphology and competence, although AUCs for euploidy and live-birth prediction were 0.60 and 0.66, respectively, which is rather comparable to embryologists' performance. Nevertheless, iDAScore v1.0 is objective and reproducible, while embryologists' evaluations are not. In a retrospective simulation, iDAScore v1.0 would have ranked euploid blastocysts as top quality in 63% of cases with one or more euploid and aneuploid blastocysts, and it would have questioned embryologists' ranking in 48% of cases with two or more euploid blastocysts and one or more live birth. Therefore, iDAScore v1.0 may objectify embryologists' evaluations, but randomized controlled trials are required to assess its clinical value.
- Published
- 2023
- Full Text
- View/download PDF
26. Author Correction: Lysyl oxidase drives tumour progression by trapping EGF receptors at the cell surface.
- Author
-
Tang H, Leung L, Saturno G, Viros A, Smith D, Leva GD, Morrison E, Niculescu-Duvaz D, Lopes F, Johnson L, Dhomen N, Springer C, and Marais R
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
27. Anti-metastatic Inhibitors of Lysyl Oxidase (LOX): Design and Structure-Activity Relationships.
- Author
-
Leung L, Niculescu-Duvaz D, Smithen D, Lopes F, Callens C, McLeary R, Saturno G, Davies L, Aljarah M, Brown M, Johnson L, Zambon A, Chambers T, Ménard D, Bayliss N, Knight R, Fish L, Lawrence R, Challinor M, Tang H, Marais R, and Springer C
- Subjects
- Administration, Oral, Animals, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Biological Availability, Cell Line, Tumor, Enzyme Inhibitors pharmacokinetics, Enzyme Inhibitors therapeutic use, Humans, Mice, Neoplasm Metastasis drug therapy, Structure-Activity Relationship, Thiophenes chemistry, Thiophenes pharmacokinetics, Thiophenes pharmacology, Thiophenes therapeutic use, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Drug Design, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Protein-Lysine 6-Oxidase antagonists & inhibitors
- Abstract
Lysyl oxidase (LOX) is a secreted copper-dependent amine oxidase that cross-links collagens and elastin in the extracellular matrix and is a critical mediator of tumor growth and metastatic spread. LOX is a target for cancer therapy, and thus the search for therapeutic agents against LOX has been widely sought. We report herein the medicinal chemistry discovery of a series of LOX inhibitors bearing an aminomethylenethiophene (AMT) scaffold. High-throughput screening provided the initial hits. Structure-activity relationship (SAR) studies led to the discovery of AMT inhibitors with sub-micromolar half-maximal inhibitory concentrations (IC
50 ) in a LOX enzyme activity assay. Further SAR optimization yielded the orally bioavailable LOX inhibitor CCT365623 with good anti-LOX potency, selectivity, pharmacokinetic properties, as well as anti-metastatic efficacy.- Published
- 2019
- Full Text
- View/download PDF
28. Lysyl oxidase drives tumour progression by trapping EGF receptors at the cell surface.
- Author
-
Tang H, Leung L, Saturno G, Viros A, Smith D, Di Leva G, Morrison E, Niculescu-Duvaz D, Lopes F, Johnson L, Dhomen N, Springer C, and Marais R
- Subjects
- Aminopropionitrile chemistry, Aminopropionitrile pharmacology, Animals, Biosensing Techniques, Cell Line, Tumor, Cell Membrane drug effects, Cell Proliferation drug effects, Dogs, Enzyme Activation, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Epidermal Growth Factor pharmacology, High-Temperature Requirement A Serine Peptidase 1 metabolism, Humans, Matrilin Proteins metabolism, Mice, Models, Biological, Neoplasm Metastasis, Protein-Lysine 6-Oxidase antagonists & inhibitors, Rats, Signal Transduction drug effects, Transforming Growth Factor beta1 metabolism, Cell Membrane metabolism, Disease Progression, ErbB Receptors metabolism, Neoplasms metabolism, Neoplasms pathology, Protein-Lysine 6-Oxidase metabolism
- Abstract
Lysyl oxidase (LOX) remodels the tumour microenvironment by cross-linking the extracellular matrix. LOX overexpression is associated with poor cancer outcomes. Here, we find that LOX regulates the epidermal growth factor receptor (EGFR) to drive tumour progression. We show that LOX regulates EGFR by suppressing TGFβ1 signalling through the secreted protease HTRA1. This increases the expression of Matrilin2 (MATN2), an EGF-like domain-containing protein that traps EGFR at the cell surface to facilitate its activation by EGF. We describe a pharmacological inhibitor of LOX, CCT365623, which disrupts EGFR cell surface retention and delays the growth of primary and metastatic tumour cells in vivo. Thus, we show that LOX regulates EGFR cell surface retention to drive tumour progression, and we validate the therapeutic potential of inhibiting this pathway with the small molecule inhibitor CCT365623.
- Published
- 2017
- Full Text
- View/download PDF
29. Paradox-Breaking RAF Inhibitors that Also Target SRC Are Effective in Drug-Resistant BRAF Mutant Melanoma.
- Author
-
Girotti MR, Lopes F, Preece N, Niculescu-Duvaz D, Zambon A, Davies L, Whittaker S, Saturno G, Viros A, Pedersen M, Suijkerbuijk BMJM, Menard D, McLeary R, Johnson L, Fish L, Ejiama S, Sanchez-Laorden B, Hohloch J, Carragher N, Macleod K, Ashton G, Marusiak AA, Fusi A, Brognard J, Frame M, Lorigan P, Marais R, and Springer C
- Published
- 2017
- Full Text
- View/download PDF
30. Application of Sequencing, Liquid Biopsies, and Patient-Derived Xenografts for Personalized Medicine in Melanoma.
- Author
-
Girotti MR, Gremel G, Lee R, Galvani E, Rothwell D, Viros A, Mandal AK, Lim KH, Saturno G, Furney SJ, Baenke F, Pedersen M, Rogan J, Swan J, Smith M, Fusi A, Oudit D, Dhomen N, Brady G, Lorigan P, Dive C, and Marais R
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Biopsy, Cluster Analysis, Disease Management, Disease Progression, Drug Resistance, Neoplasm, Gene Expression Profiling, High-Throughput Nucleotide Sequencing, Humans, Molecular Targeted Therapy, Mutation, Neoplasm Staging, Reproducibility of Results, Treatment Outcome, Xenograft Model Antitumor Assays, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Melanoma diagnosis, Melanoma drug therapy, Precision Medicine
- Abstract
Unlabelled: Targeted therapies and immunotherapies have transformed melanoma care, extending median survival from ∼9 to over 25 months, but nevertheless most patients still die of their disease. The aim of precision medicine is to tailor care for individual patients and improve outcomes. To this end, we developed protocols to facilitate individualized treatment decisions for patients with advanced melanoma, analyzing 364 samples from 214 patients. Whole exome sequencing (WES) and targeted sequencing of circulating tumor DNA (ctDNA) allowed us to monitor responses to therapy and to identify and then follow mechanisms of resistance. WES of tumors revealed potential hypothesis-driven therapeutic strategies for BRAF wild-type and inhibitor-resistant BRAF-mutant tumors, which were then validated in patient-derived xenografts (PDX). We also developed circulating tumor cell-derived xenografts (CDX) as an alternative to PDXs when tumors were inaccessible or difficult to biopsy. Thus, we describe a powerful technology platform for precision medicine in patients with melanoma., Significance: Although recent developments have revolutionized melanoma care, most patients still die of their disease. To improve melanoma outcomes further, we developed a powerful precision medicine platform to monitor patient responses and to identify and validate hypothesis-driven therapies for patients who do not respond, or who develop resistance to current treatments., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
31. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy.
- Author
-
Miller BW, Morton JP, Pinese M, Saturno G, Jamieson NB, McGhee E, Timpson P, Leach J, McGarry L, Shanks E, Bailey P, Chang D, Oien K, Karim S, Au A, Steele C, Carter CR, McKay C, Anderson K, Evans TR, Marais R, Springer C, Biankin A, Erler JT, and Sansom OJ
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Deoxycytidine analogs & derivatives, Deoxycytidine therapeutic use, Humans, Hypoxia, Mice, Neoplasm Metastasis prevention & control, Protein-Lysine 6-Oxidase antagonists & inhibitors, Treatment Outcome, Gemcitabine, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology, Protein-Lysine 6-Oxidase metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality. Despite significant advances made in the treatment of other cancers, current chemotherapies offer little survival benefit in this disease. Pancreaticoduodenectomy offers patients the possibility of a cure, but most will die of recurrent or metastatic disease. Hence, preventing metastatic disease in these patients would be of significant benefit. Using principal component analysis (PCA), we identified a LOX/hypoxia signature associated with poor patient survival in resectable patients. We found that LOX expression is upregulated in metastatic tumors from Pdx1-Cre Kras(G12D/+) Trp53(R172H/+) (KPC) mice and that inhibition of LOX in these mice suppressed metastasis. Mechanistically, LOX inhibition suppressed both migration and invasion of KPC cells. LOX inhibition also synergized with gemcitabine to kill tumors and significantly prolonged tumor-free survival in KPC mice with early-stage tumors. This was associated with stromal alterations, including increased vasculature and decreased fibrillar collagen, and increased infiltration of macrophages and neutrophils into tumors. Therefore, LOX inhibition is able to reverse many of the features that make PDAC inherently refractory to conventional therapies and targeting LOX could improve outcome in surgically resectable disease., (© 2015 Cancer Research UK Beatson Institute. Published under the terms of the CC BY 4.0 license.)
- Published
- 2015
- Full Text
- View/download PDF
32. Paradox-breaking RAF inhibitors that also target SRC are effective in drug-resistant BRAF mutant melanoma.
- Author
-
Girotti MR, Lopes F, Preece N, Niculescu-Duvaz D, Zambon A, Davies L, Whittaker S, Saturno G, Viros A, Pedersen M, Suijkerbuijk BM, Menard D, McLeary R, Johnson L, Fish L, Ejiama S, Sanchez-Laorden B, Hohloch J, Carragher N, Macleod K, Ashton G, Marusiak AA, Fusi A, Brognard J, Frame M, Lorigan P, Marais R, and Springer C
- Subjects
- Animals, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Melanoma pathology, Melanoma, Experimental, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Melanoma drug therapy, Phenylurea Compounds pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Pyrazines pharmacology, src-Family Kinases antagonists & inhibitors
- Abstract
BRAF and MEK inhibitors are effective in BRAF mutant melanoma, but most patients eventually relapse with acquired resistance, and others present intrinsic resistance to these drugs. Resistance is often mediated by pathway reactivation through receptor tyrosine kinase (RTK)/SRC-family kinase (SFK) signaling or mutant NRAS, which drive paradoxical reactivation of the pathway. We describe pan-RAF inhibitors (CCT196969, CCT241161) that also inhibit SFKs. These compounds do not drive paradoxical pathway activation and inhibit MEK/ERK in BRAF and NRAS mutant melanoma. They inhibit melanoma cells and patient-derived xenografts that are resistant to BRAF and BRAF/MEK inhibitors. Thus, paradox-breaking pan-RAF inhibitors that also inhibit SFKs could provide first-line treatment for BRAF and NRAS mutant melanomas and second-line treatment for patients who develop resistance., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. No longer an untreatable disease: how targeted and immunotherapies have changed the management of melanoma patients.
- Author
-
Girotti MR, Saturno G, Lorigan P, and Marais R
- Subjects
- Animals, Antineoplastic Agents pharmacology, Humans, Melanoma immunology, Melanoma metabolism, Melanoma pathology, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Proto-Oncogene Proteins B-raf metabolism, Signal Transduction drug effects, Skin drug effects, Skin immunology, Skin metabolism, Skin pathology, Skin Neoplasms immunology, Skin Neoplasms metabolism, Skin Neoplasms pathology, Antineoplastic Agents therapeutic use, Immunotherapy methods, Melanoma therapy, Molecular Targeted Therapy methods, Skin Neoplasms therapy
- Abstract
The discovery that BRAF is a driver oncogene in cancer, and complementary improvements in our understanding of the immune system have resulted in new targeted and immune-therapies for metastatic melanoma. Targeted therapies achieve impressive clinical results in carefully selected patients but the development of resistance seems inevitable in most cases. Conversely, immune-checkpoints inhibitors can achieve long-term remission and cures, but in a smaller proportion of patients, and biomarkers to predict which patients will respond are not available. Nevertheless, melanoma has led the evolution of cancer treatment from relatively nonspecific cytotoxic agents to highly selective therapies and here we review the lessons from this paradigm shift in treatment and the opportunities for further improvements in outcomes for melanoma patients., (Copyright © 2014 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
34. BRAF inhibitors induce metastasis in RAS mutant or inhibitor-resistant melanoma cells by reactivating MEK and ERK signaling.
- Author
-
Sanchez-Laorden B, Viros A, Girotti MR, Pedersen M, Saturno G, Zambon A, Niculescu-Duvaz D, Turajlic S, Hayes A, Gore M, Larkin J, Lorigan P, Cook M, Springer C, and Marais R
- Subjects
- Animals, Blotting, Western, Cell Shape physiology, Dimethyl Sulfoxide, Drug Resistance, Neoplasm, Enzyme-Linked Immunosorbent Assay, Humans, Indoles pharmacology, Interleukin-8 metabolism, Mice, Mice, Nude, Neoplasm Invasiveness genetics, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins p21(ras) genetics, Statistics, Nonparametric, Sulfonamides pharmacology, MAP Kinase Signaling System physiology, Melanoma physiopathology, Neoplasm Invasiveness physiopathology, Neoplasm Metastasis physiopathology, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Skin Neoplasms physiopathology
- Abstract
Melanoma is a highly metastatic and lethal form of skin cancer. The protein kinase BRAF is mutated in about 40% of melanomas, and BRAF inhibitors improve progression-free and overall survival in these patients. However, after a relatively short period of disease control, most patients develop resistance because of reactivation of the RAF-ERK (extracellular signal-regulated kinase) pathway, mediated in many cases by mutations in RAS. We found that BRAF inhibition induces invasion and metastasis in RAS mutant melanoma cells through a mechanism mediated by the reactivation of the MEK (mitogen-activated protein kinase kinase)-ERK pathway, increased expression and secretion of interleukin 8, and induction of protease-dependent invasion. These events were accompanied by a cell morphology switch from predominantly rounded to predominantly elongated cells. We also observed similar responses in BRAF inhibitor-resistant melanoma cells. These data show that BRAF inhibitors can induce melanoma cell invasion and metastasis in tumors that develop resistance to these drugs.
- Published
- 2014
- Full Text
- View/download PDF
35. Combining trail with PI3 kinase or HSP90 inhibitors enhances apoptosis in colorectal cancer cells via suppression of survival signaling.
- Author
-
Saturno G, Valenti M, De Haven Brandon A, Thomas GV, Eccles S, Clarke PA, and Workman P
- Subjects
- Animals, Apoptosis drug effects, Benzoquinones administration & dosage, Cell Line, Tumor, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Drug Screening Assays, Antitumor, Furans administration & dosage, Furans pharmacology, HCT116 Cells, HSP90 Heat-Shock Proteins metabolism, HT29 Cells, Humans, Lactams, Macrocyclic administration & dosage, Mice, Mice, Nude, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Pyridines administration & dosage, Pyridines pharmacology, Pyrimidines administration & dosage, Pyrimidines pharmacology, Signal Transduction drug effects, TNF-Related Apoptosis-Inducing Ligand administration & dosage, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzoquinones pharmacology, Colorectal Neoplasms drug therapy, HSP90 Heat-Shock Proteins antagonists & inhibitors, Lactams, Macrocyclic pharmacology, Phosphoinositide-3 Kinase Inhibitors, TNF-Related Apoptosis-Inducing Ligand pharmacology
- Abstract
TRAIL has been shown to induce apoptosis in cancer cells, but in some cases they fail to respond to this ligand. We explored the ability of representative phosphatidylinositol-3-kinase (PI3 Kinase)/mTOR and HSP90 inhibitors to overcome TRAIL resistance by increasing apoptosis in colorectal cancer models. We determined the sensitivity of 27 human colorectal cancer and 2 non-transformed colon epithelial cell lines to TRAIL treatment. A subset of the cancer cell lines with a range of responses to TRAIL was selected from the panel for treatment with TRAIL combined with the PI3 Kinase/mTOR inhibitor PI-103 or the HSP90 inhibitor 17-AAG (tanespimycin). Two TRAIL-resistant cell lines were selected for in vivo combination studies with TRAIL and 17-AAG. We found that 13 colorectal cancer cell lines and the 2 non-transformed colon epithelial cell lines were resistant to TRAIL. We demonstrated that co-treatment of TRAIL and PI-103 or 17-AAG was synergistic or additive and significantly enhanced apoptosis in colorectal cancer cells. This was associated with decreased expression or activity of survival protein biomarkers such as ERBB2, AKT, IKKα and XIAP. In contrast, the effect of the combination treatments in non-transformed colon cells was minimal. We show here for the first time that co-treatment in vivo with TRAIL and 17-AAG in two TRAIL-resistant human colorectal cancer xenograft models resulted in significantly greater tumor growth inhibition compared to single treatments. We propose that combining TRAIL with PI3 Kinase/mTOR or HSP90 inhibitors has therapeutic potential in the treatment of TRAIL-resistant colorectal cancers.
- Published
- 2013
- Full Text
- View/download PDF
36. Expression of serine/threonine protein-kinases and related factors in normal monkey and human retinas: the mechanistic understanding of a CDK2 inhibitor induced retinal toxicity.
- Author
-
Saturno G, Pesenti M, Cavazzoli C, Rossi A, Giusti AM, Gierke B, Pawlak M, and Venturi M
- Subjects
- Animals, Female, Fluorescent Antibody Technique, Glycogen Synthase Kinase 3 analysis, Glycogen Synthase Kinase 3 beta, Humans, Immunohistochemistry, Macaca fascicularis, Male, Microdissection, Phosphorylation, Polymerase Chain Reaction, Retina pathology, tau Proteins analysis, Cyclin-Dependent Kinase 2 antagonists & inhibitors, Protein Kinase Inhibitors toxicity, Protein Serine-Threonine Kinases analysis, Retina drug effects, Retina enzymology
- Abstract
Protein-kinase inhibitors are among the most advanced compounds in development using the new drug discovery paradigm of developing small-molecule drugs against specific molecular targets in cancer. After treatment with a cyclin dependent kinase CDK2 inhibitor in monkey, histopathological analysis of the eye showed specific cellular damage in the photoreceptor layer. Since this CDK2 inhibitor showed activity also on other CDKs, in order to investigate the mechanism of toxicity of this compound, we isolated cones and rods from the retina of normal monkey and humans by Laser Capture Microdissection. Using Real-Time PCR we first measured the expression of cyclin dependent protein-kinases (CDK)1, 2, 4, 5, Glycogen synthase kinase 3beta (GSK3beta) and microtubule associated protein TAU. We additionally verified the presence of these proteins in monkey eye sections by immuno-histochemistry and immunofluorescence analysis and afterwards quantified GSK3beta, phospho-GSK3beta and TAU by Reverse Phase Protein Microarrays. With this work we demonstrate how complementary gene expression and protein-based technologies constitute a powerful tool for the understanding of the molecular mechanism of a CDK2 inhibitor induced toxicity. Moreover, this investigative approach is helpful to better understand and characterize the mechanism of species-specific toxicities and further support a rational, molecular mechanism-based safety assessment in humans.
- Published
- 2007
- Full Text
- View/download PDF
37. An in vivo and in vitro comparison of CYP gene induction in mice using liver slices and quantitative RT-PCR.
- Author
-
Martignoni M, de Kanter R, Grossi P, Saturno G, Barbaria E, and Monshouwer M
- Subjects
- Animals, Cytochrome P-450 Enzyme System genetics, Dexamethasone pharmacology, Gene Expression Regulation, Enzymologic drug effects, In Vitro Techniques, Male, Mice, Mice, Inbred Strains, Phenobarbital pharmacology, RNA, Messenger metabolism, Transcriptional Activation, beta-Naphthoflavone pharmacology, Biological Assay, Cytochrome P-450 Enzyme System metabolism, Liver enzymology
- Abstract
The scope of this study was to compare in vitro and in vivo cytochrome P450 (CYP) gene induction in mice, using liver slices as an in vitro model. We have chosen to study mice to be able to better interpret CYP induction during long-term safety studies in this species. Mouse liver slices were incubated with beta-naphthoflavone (betaNF), phenobarbital (PB) or dexamethasone (DEX) for 24 h. In addition, in an in vivo study, mice were treated with the same compounds for three days. The mRNA expression of cyp1a1, cyp1a2, cyp2b10 and cyp3a11, which are important for drug metabolism and inducible by xenobiotics, were investigated in vivo and in vitro by real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). Both in mouse liver slices and in vivo, betaNF was found to be a potent inducer of cyp1a1 and to a lesser extent of cyp1a2. All three compounds induced cyp2b10 mRNA levels, while the cyp3a11 mRNA level was induced only by DEX. Overall, these data demonstrated a good predictive in vitro-in vivo correlation of CYP induction.
- Published
- 2006
- Full Text
- View/download PDF
38. An in vivo and in vitro comparison of CYP induction in rat liver and intestine using slices and quantitative RT-PCR.
- Author
-
Martignoni M, de Kanter R, Grossi P, Mahnke A, Saturno G, and Monshouwer M
- Subjects
- Animals, Base Sequence, Cytochrome P-450 Enzyme System genetics, DNA Primers, Dexamethasone pharmacology, Enzyme Induction, In Vitro Techniques, Intestines enzymology, Liver enzymology, Male, Phenobarbital pharmacology, RNA, Messenger genetics, Rats, Rats, Wistar, beta-Naphthoflavone pharmacology, Cytochrome P-450 Enzyme System biosynthesis, Intestines drug effects, Liver drug effects, Reverse Transcriptase Polymerase Chain Reaction methods
- Abstract
Xenobiotics, including drugs, can influence cytochrome P450 (CYP) activity by upregulating the transcription of CYP genes. To minimize potential drug interactions, it is important to ascertain whether a compound will be an inducer of CYP enzymes early in the development of new therapeutic agents. In vivo and in vitro studies are reported that demonstrate the use of liver and intestinal slices as an in vitro model to predict potential CYP induction in vivo. Rat liver slices and intestinal slices were incubated, for 24 h and 6 h, respectively, with beta-naphthoflavone (betaNF), phenobarbital (PB) or dexamethasone (DEX). In an in vivo study, rats were treated with the same compounds for 3 days. In vivo and in vitro CYP mRNA levels were measured by using real-time quantitative reverse transcription-polymerase chain reaction (RT-PCR). In addition, CYP enzyme activities were determined in rat liver slices after 48 h incubation. In both rat liver and intestinal slices, betaNF significantly induced CYP1A1, CYP1A2 and CYP2B1 mRNA levels. PB significantly induced CYP2B1. In liver slices a minor induction of CYP1A1 and CYP3A1 by PB was observed, whereas DEX significantly induced CYP3A1, CYP2B1 and CYP1A2 mRNA levels. The induction profiles (qualitative and quantitative) observed in vivo and in vitro are quite similar. All together, these data demonstrate that liver and intestinal slices are a useful and predictive tool to study CYP induction.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.