42 results on '"Lawlor, R.T."'
Search Results
2. 1150P Search for biomarkers to personalize treatment with streptozotocin plus 5-fluorouracil or everolimus in patients with advanced pancreatic neuroendocrine tumors: The randomized phase III SEQTOR trial (GETNE-1206)
- Author
-
Salazar Soler, R., Scarpa, A., Lawlor, R.T., Sadanandam, A., Tafuto, S., Krogh, M., Teule, A., Garcia-Carbonero, R., Klumpen, H.J., Cremer, B., Sevilla, I., Eriksson, B.K., Mitkina Tabaksblat, E., Metges, J-P., Reed, N.S., Schrader, J., Bozzarelli, S., Venerito, M., Navarro, V., and Capdevila Castillon, J.
- Published
- 2024
- Full Text
- View/download PDF
3. Refining targeted therapeutic approaches in pancreatic cancer: from histology and molecular pathology to the clinic
- Author
-
Mattiolo, P., Kryklyva, V., Brosens, L.A.A., Mafficini, A., Lawlor, R.T., Milella, M., Scarpa, A., Corbo, V., Luchini, C., Mattiolo, P., Kryklyva, V., Brosens, L.A.A., Mafficini, A., Lawlor, R.T., Milella, M., Scarpa, A., Corbo, V., and Luchini, C.
- Abstract
Item does not contain fulltext
- Published
- 2022
4. Juvenile polyposis diagnosed with an integrated histological, immunohistochemical and molecular approach identifying new SMAD4 pathogenic variants
- Author
-
Mafficini, A., Brosens, L.A.A., Piredda, M.L., Conti, C., Mattiolo, P., Turri, G., Mastrosimini, M.G., Cingarlini, S., Crinò, S.F., Fassan, M., Piccoli, P., Simbolo, M., Nottegar, A., Lawlor, R.T., Guglielmi, A., Scarpa, A., Pedrazzani, C., Luchini, C., Mafficini, A., Brosens, L.A.A., Piredda, M.L., Conti, C., Mattiolo, P., Turri, G., Mastrosimini, M.G., Cingarlini, S., Crinò, S.F., Fassan, M., Piccoli, P., Simbolo, M., Nottegar, A., Lawlor, R.T., Guglielmi, A., Scarpa, A., Pedrazzani, C., and Luchini, C.
- Abstract
Item does not contain fulltext, Juvenile polyposis (JP) is a rare familial syndrome characterized by the development of numerous hamartomatous polyps of the gastrointestinal tract and by an increased risk of developing gastrointestinal cancers. It follows a pattern of autosomal dominant inheritance and is associated with germline variants of SMAD4 or BMPR1A genes. Differential diagnosis may be difficult based on histology alone, due to morphological similarities to other familial syndromes. Here we report a case of familial JP diagnosed in a 50-years woman with a familial history positive for gastrointestinal cancers and other tumor types. The patient presented with severe iron deficiency anemia and showed numerous polyps in the stomach and jejunum according to endoscopy and imaging. She underwent an intra-gastric laparoscopic removal of the major gastric polyp, followed by jejunal exploration and resection of a segment with multiple neoformations. Histological examination revealed the presence of hamartomatous polyposis. Gastric and intestinal samples were analyzed with next-generation sequencing. Molecular analysis showed that the patient harbored a germline splicing site variant of SMAD4, c.1139 + 3A > G, which was complemented by different somatic variants of the same gene in the different polyps. Immunohistochemistry for SMAD4 confirmed loss of protein expression in the polyps, with regular expression in normal cells. cDNA sequencing further confirmed the findings. We thus definitively diagnosed the woman as having JP thanks to an integrated approach based on histology, immunohistochemistry and molecular analysis. The identified variants, all previously reported as variants of unknown significance, were classified as pathogenic as they complemented each other leading to SMAD4 loss.
- Published
- 2022
5. Association of genetic variants affecting microRNAs and pancreatic cancer risk
- Author
-
Lu, Y., Corradi, C., Gentiluomo, M., Theodoropoulos, G.E., Roth, S., Maiello, E., Morelli, L., L.Archibugi, Izbicki, J.R., Sarlós, P., Kiudelis, V., Oliverius, M., López de Maturana, E., Aoki, M.N., Vashist, Y., van Eijck, C.H.J., Gazouli, M., Talar-Wojnarowska, R., Mambrini, A., Pezzilli, R., Bueno-de-Mesquita, B., Hegyi, P., Souček, P., Neoptolemos, J., Di Franco, G., Sperti, C., Kauffmann, E.F., Hlaváč, V., Uzunoğlu, F.G., Ermini, S., Małecka-Panas, E., Lucchesi, M., Vanella, G., Dijk, F., Mohelníková-Duchoňová, B., Bambi, F., Petrone, M.C., Jamroziak, K., Guo, F., Kolarova, K., Capretti, G., Milanetto, A.C., Ginocchi, L., Loveček, M., Puzzono, M., van Laarhoven, H.W.M., Carrara, S., Ivanauskas, A., Papiris, K., Basso, D., Arcidiacono, P.G., Izbéki, F., Chammas, R., Vodicka, P., Hackert, T., Pasquali, C., Piredda, M.L., Costello-Goldring, E., Cavestro, G.M., Szentesi, A., Tavano, F., Włodarczyk, B., Brenner, H., Kreivenaite, E., Gao, X., Bunduc, S., Schneider, M.A., Latiano, A., Gioffreda, D., Testoni, S.G.G., Malats, N., Kupcinskas, J., Lawlor, R.T., Capurso, G., Campa, D., and Canzian, F.
- Published
- 2021
- Full Text
- View/download PDF
6. Combinatorial Effect of Magnetic Field and Radiotherapy in Pancreatic Ductal Adenocarcinoma Organoids
- Author
-
Nicosia, L., primary, Andreani, S., additional, Ruggieri, R., additional, Rusev, B., additional, Lawlor, R.T., additional, Pea, A., additional, Agolli, L., additional, Scarpa, A., additional, Corbo, V., additional, D'Agosto, S., additional, and Alongi, F., additional
- Published
- 2021
- Full Text
- View/download PDF
7. A functional role for splicing factor EIF4A3 dysregulation in pancreatic ductal adenocarcinoma
- Author
-
Blazquez-Encinas, R., primary, Alors-Pérez, E., additional, Mafficini, A., additional, Moreno-Montilla, T., additional, Viyuela-García, C., additional, Abollo-Jiménez, F., additional, Sánchez-Hidalgo, J.M., additional, Sánchez-Frías, M.E., additional, García-Vioque, V., additional, Luchini, C., additional, Pea, A., additional, Ventura, S., additional, Gahete, M.D., additional, Arjona, Á., additional, Lawlor, R.T., additional, Luque, R.M., additional, Scarpa, A., additional, Ibáñez-Costa, A., additional, and Castaño, J.P., additional
- Published
- 2021
- Full Text
- View/download PDF
8. Modelling evolution of PDAC towards aggressive phenotypes using the organoid culture system
- Author
-
Malinova, A., primary, Franceschini, A., additional, Andreani, S., additional, D'agosto, S., additional, Delfino, P., additional, Sereni, E., additional, Pea, A., additional, Salvia, R., additional, Lawlor, R.T., additional, Rusev, B., additional, Scarpa, A., additional, Fiorini, E., additional, and Corbo, V., additional
- Published
- 2021
- Full Text
- View/download PDF
9. Leukocyte-DNA methylome explains a large proportion of the risk variability of pancreatic cancer
- Author
-
López De Maturana, E., primary, Alonso, L., additional, Carrato, A., additional, Iglesias, M., additional, Molero, X., additional, Hidalgo, M., additional, Perea, J., additional, Barberá, V., additional, Farré, A., additional, Tardón, A., additional, Dominguez-Muñoz, E., additional, Muñoz-Bellvís, L., additional, Crnogorac-Jurcevic, T., additional, Greenhalf, W., additional, Gress, T., additional, Löhr, M., additional, Lawlor, R.T., additional, Michalski, C., additional, Rorke, M.O., additional, Sharp, L., additional, Real, F.X., additional, and Malats, N., additional
- Published
- 2021
- Full Text
- View/download PDF
10. Splicing machinery dysregulation and its relationship with PDAC aggressiveness
- Author
-
Perez, E. Alors, primary, Blázquez-Encinas, R., additional, Mafficini, A., additional, Montilla, T. Moreno, additional, Viyuela-Garcia, C., additional, Jiménez-Vacas, J.M., additional, Sánchez-Hidalgo, J.M., additional, Berbel, I., additional, Marín-Sanz, J.A., additional, Sánchez-Fría, M.E., additional, Luchini, C., additional, Pea, A., additional, Ventura, S., additional, Gahete, M.D., additional, Arjona-Sánchez, Á., additional, Lawlor, R.T., additional, Luque, R.M., additional, Scarpa, A., additional, Ibáñez-Costa, A., additional, and Castaño, J.P., additional
- Published
- 2021
- Full Text
- View/download PDF
11. Genome-wide scan of long noncoding RNA single nucleotide polymorphisms and pancreatic cancer susceptibility
- Author
-
Corradi, C. Gentiluomo, M. Gajdán, L. Cavestro, G.M. Kreivenaite, E. Di Franco, G. Sperti, C. Giaccherini, M. Petrone, M.C. Tavano, F. Gioffreda, D. Morelli, L. Soucek, P. Andriulli, A. Izbicki, J.R. Napoli, N. Małecka-Panas, E. Hegyi, P. Neoptolemos, J.P. Landi, S. Vashist, Y. Pasquali, C. Lu, Y. Cervena, K. Theodoropoulos, G.E. Moz, S. Capurso, G. Strobel, O. Carrara, S. Hackert, T. Hlavac, V. Archibugi, L. Oliverius, M. Vanella, G. Vodicka, P. Arcidiacono, P.G. Pezzilli, R. Milanetto, A.C. Lawlor, R.T. Ivanauskas, A. Szentesi, A. Kupcinskas, J. Testoni, S.G.G. Lovecek, M. Nentwich, M. Gazouli, M. Luchini, C. Zuppardo, R.A. Darvasi, E. Brenner, H. Gheorghe, C. Jamroziak, K. Canzian, F. Campa, D.
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is projected to become the second cancer-related cause of death by 2030. Identifying novel risk factors, including genetic risk loci, could be instrumental in risk stratification and implementation of prevention strategies. Long noncoding RNAs (lncRNAs) are involved in regulation of key biological processes, and the possible role of their genetic variability has been unexplored so far. Combining genome wide association studies and functional data, we investigated the genetic variability in all lncRNAs. We analyzed 9893 PDAC cases and 9969 controls and identified a genome-wide significant association between the rs7046076 SNP and risk of developing PDAC (P = 9.73 × 10−9). This SNP is located in the NONHSAG053086.2 (lnc-SMC2-1) gene and the risk allele is predicted to disrupt the binding of the lncRNA with the micro-RNA (miRNA) hsa-mir-1256 that regulates several genes involved in cell cycle, such as CDKN2B. The CDKN2B region is pleiotropic and its genetic variants have been associated with several human diseases, possibly though an imperfect interaction between lncRNA and miRNA. We present a novel PDAC risk locus, supported by a genome-wide statistical significance and a plausible biological mechanism. © 2021 UICC
- Published
- 2021
12. Epithelial-mesenchymal transition in undifferentiated carcinoma of the pancreas with and without osteoclast-like giant cells
- Author
-
Mattiolo, P., Fiadone, G., Paolino, G., Chatterjee, D., Bernasconi, R., Piccoli, P., Parolini, C., Aidi, M. El, Sperandio, N., Malleo, G., Salvia, R., Brosens, L.A.A., Wood, L.D., Scarpa, A., Lawlor, R.T., Luchini, C., Mattiolo, P., Fiadone, G., Paolino, G., Chatterjee, D., Bernasconi, R., Piccoli, P., Parolini, C., Aidi, M. El, Sperandio, N., Malleo, G., Salvia, R., Brosens, L.A.A., Wood, L.D., Scarpa, A., Lawlor, R.T., and Luchini, C.
- Abstract
Item does not contain fulltext, Undifferentiated carcinoma (UC) and undifferentiated carcinoma with osteoclast-like giant cells (UCOGC) are peculiar variants of pancreatic ductal adenocarcinoma (PDAC), characterized by hypercellularity and absence of glandular patterns. The inflammatory microenvironment is peculiar in UCOGC, since it is dominated by macrophages and osteoclast-like giant cells. However, from a molecular point of view, both UC and UCOGC are very similar to conventional PDAC, sharing alterations of the most common genetic drivers. Clinically, UC usually show a worse prognosis, whereas UCOGC may show a better prognosis if it is not associated with a PDAC component. To highlight potential biological differences between these entities, we investigated the role of the epithelial to mesenchymal transition (EMT) in UC and UCOGC. Specifically, we analyzed the immunohistochemical expression of three well-known EMT markers, namely Twist1, Snai2, and E-cadherin, in 16 cases of UCOGC and 10 cases of UC. We found that EMT is more frequently activated in UC (10/10 cases) than in UCOGC (8/16 cases; p = 0.05). Furthermore, in UCOGC, EMT was activated with a higher frequency in cases with an associated PDAC component. Snai2 was the most frequently and strongly expressed marker in both tumor types (10/10 UC, 8/16 UCOGC), and its expression was higher in UC than in UCOGC (mean immunohistochemical score: 4.8 in UC vs. 2.1 in UCOGC, p < 0.01). Our results shed new light on the biology of UC and UCOGC: EMT appeared as a more important process in UC, and Snai2 emerged as a central EMT effector in this setting.
- Published
- 2021
13. Genomic characterization of hepatoid tumors: context matters
- Author
-
Lawlor, R.T., Mafficini, Andrea, Sciammarella, Concetta, Cantu, Cinzia, Rusev, Borislav, Piredda, M.L., Brosens, L.A.A., Scarpa, Aldo, Luchini, Claudio, Lawlor, R.T., Mafficini, Andrea, Sciammarella, Concetta, Cantu, Cinzia, Rusev, Borislav, Piredda, M.L., Brosens, L.A.A., Scarpa, Aldo, and Luchini, Claudio
- Abstract
Item does not contain fulltext
- Published
- 2021
14. Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Pancreatic Cancer: Systematic Review and Still-Open Questions
- Author
-
Lawlor, R.T., Mattiolo, P., Mafficini, A., Hong, S.M., Piredda, M.L., Taormina, S.V., Malleo, G., Marchegiani, G., Pea, A., Salvia, R., Kryklyva, V., Shin, J.I., Brosens, L.A.A., Milella, M., Scarpa, A., Luchini, C., Lawlor, R.T., Mattiolo, P., Mafficini, A., Hong, S.M., Piredda, M.L., Taormina, S.V., Malleo, G., Marchegiani, G., Pea, A., Salvia, R., Kryklyva, V., Shin, J.I., Brosens, L.A.A., Milella, M., Scarpa, A., and Luchini, C.
- Abstract
Contains fulltext : 238941.pdf (Publisher’s version ) (Open Access), Tumor mutational burden (TMB) is a numeric index that expresses the number of mutations per megabase (muts/Mb) harbored by tumor cells in a neoplasm. TMB can be determined using different approaches based on next-generation sequencing. In the case of high values, it indicates a potential response to immunotherapy. In this systematic review, we assessed the potential predictive role of high-TMB in pancreatic ductal adenocarcinoma (PDAC), as well as the histo-molecular features of high-TMB PDAC. High-TMB appeared as a rare but not-negligible molecular feature in PDAC, being present in about 1.1% of cases. This genetic condition was closely associated with mucinous/colloid and medullary histology (p < 0.01). PDAC with high-TMB frequently harbored other actionable alterations, with microsatellite instability/defective mismatch repair as the most common. Immunotherapy has shown promising results in high-TMB PDAC, but the sample size of high-TMB PDAC treated so far is quite small. This study highlights interesting peculiarities of PDAC harboring high-TMB and may represent a reliable starting point for the assessment of TMB in the clinical management of patients affected by pancreatic cancer.
- Published
- 2021
15. Association of Genetic Variants Affecting microRNAs and Pancreatic Cancer Risk
- Author
-
Sub ISEP overig, IRAS OH Epidemiology Chemical Agents, dIRAS RA-2, Lu, Y., Corradi, C., Gentiluomo, M., López de Maturana, E., Theodoropoulos, G.E., Roth, S., Maiello, E., Morelli, L., Archibugi, L., Izbicki, J.R., Sarlós, P., Kiudelis, V., Oliverius, M., Aoki, M.N., Vashist, Y., van Eijck, C.H.J., Gazouli, M., Talar-Wojnarowska, R., Mambrini, A., Pezzilli, R., Bueno-de-Mesquita, B., Hegyi, P., Souček, P., Neoptolemos, J.P., Di Franco, G., Sperti, C., Kauffmann, E.F., Hlaváč, V., Uzunoğlu, F.G., Ermini, S., Małecka-Panas, E., Lucchesi, M., Vanella, G., Dijk, F., Mohelníková-Duchoňová, B., Bambi, F., Petrone, M.C., Jamroziak, K., Guo, F., Kolarova, K., Capretti, G., Milanetto, A.C., Ginocchi, L., Loveček, M., Puzzono, M., van Laarhoven, H.W.M., Carrara, S., Ivanauskas, A., Papiris, K., Basso, D., Arcidiacono, P.G., Izbéki, F., Chammas, R., Vodicka, P., Hackert, T., Pasquali, C., Piredda, M.L., Costello-Goldring, E., Cavestro, G.M., Szentesi, A., Tavano, F., Włodarczyk, B., Brenner, H., Kreivenaite, E., Gao, X., Bunduc, S., Vermeulen, R.C.H., Schneider, M.A., Latiano, A., Gioffreda, D., Testoni, S.G.G., Kupcinskas, J., Lawlor, R.T., Capurso, G., Malats, N., Campa, D., Canzian, F., Sub ISEP overig, IRAS OH Epidemiology Chemical Agents, dIRAS RA-2, Lu, Y., Corradi, C., Gentiluomo, M., López de Maturana, E., Theodoropoulos, G.E., Roth, S., Maiello, E., Morelli, L., Archibugi, L., Izbicki, J.R., Sarlós, P., Kiudelis, V., Oliverius, M., Aoki, M.N., Vashist, Y., van Eijck, C.H.J., Gazouli, M., Talar-Wojnarowska, R., Mambrini, A., Pezzilli, R., Bueno-de-Mesquita, B., Hegyi, P., Souček, P., Neoptolemos, J.P., Di Franco, G., Sperti, C., Kauffmann, E.F., Hlaváč, V., Uzunoğlu, F.G., Ermini, S., Małecka-Panas, E., Lucchesi, M., Vanella, G., Dijk, F., Mohelníková-Duchoňová, B., Bambi, F., Petrone, M.C., Jamroziak, K., Guo, F., Kolarova, K., Capretti, G., Milanetto, A.C., Ginocchi, L., Loveček, M., Puzzono, M., van Laarhoven, H.W.M., Carrara, S., Ivanauskas, A., Papiris, K., Basso, D., Arcidiacono, P.G., Izbéki, F., Chammas, R., Vodicka, P., Hackert, T., Pasquali, C., Piredda, M.L., Costello-Goldring, E., Cavestro, G.M., Szentesi, A., Tavano, F., Włodarczyk, B., Brenner, H., Kreivenaite, E., Gao, X., Bunduc, S., Vermeulen, R.C.H., Schneider, M.A., Latiano, A., Gioffreda, D., Testoni, S.G.G., Kupcinskas, J., Lawlor, R.T., Capurso, G., Malats, N., Campa, D., and Canzian, F.
- Published
- 2021
16. Association of Genetic Variants Affecting microRNAs and Pancreatic Cancer Risk
- Author
-
Sub IER overig, IRAS OH Epidemiology Chemical Agents, dIRAS RA-2, Lu, Y., Corradi, C., Gentiluomo, M., López de Maturana, E., Theodoropoulos, G.E., Roth, S., Maiello, E., Morelli, L., Archibugi, L., Izbicki, J.R., Sarlós, P., Kiudelis, V., Oliverius, M., Aoki, M.N., Vashist, Y., van Eijck, C.H.J., Gazouli, M., Talar-Wojnarowska, R., Mambrini, A., Pezzilli, R., Bueno-de-Mesquita, B., Hegyi, P., Souček, P., Neoptolemos, J.P., Di Franco, G., Sperti, C., Kauffmann, E.F., Hlaváč, V., Uzunoğlu, F.G., Ermini, S., Małecka-Panas, E., Lucchesi, M., Vanella, G., Dijk, F., Mohelníková-Duchoňová, B., Bambi, F., Petrone, M.C., Jamroziak, K., Guo, F., Kolarova, K., Capretti, G., Milanetto, A.C., Ginocchi, L., Loveček, M., Puzzono, M., van Laarhoven, H.W.M., Carrara, S., Ivanauskas, A., Papiris, K., Basso, D., Arcidiacono, P.G., Izbéki, F., Chammas, R., Vodicka, P., Hackert, T., Pasquali, C., Piredda, M.L., Costello-Goldring, E., Cavestro, G.M., Szentesi, A., Tavano, F., Włodarczyk, B., Brenner, H., Kreivenaite, E., Gao, X., Bunduc, S., Vermeulen, R.C.H., Schneider, M.A., Latiano, A., Gioffreda, D., Testoni, S.G.G., Kupcinskas, J., Lawlor, R.T., Capurso, G., Malats, N., Campa, D., Canzian, F., Sub IER overig, IRAS OH Epidemiology Chemical Agents, dIRAS RA-2, Lu, Y., Corradi, C., Gentiluomo, M., López de Maturana, E., Theodoropoulos, G.E., Roth, S., Maiello, E., Morelli, L., Archibugi, L., Izbicki, J.R., Sarlós, P., Kiudelis, V., Oliverius, M., Aoki, M.N., Vashist, Y., van Eijck, C.H.J., Gazouli, M., Talar-Wojnarowska, R., Mambrini, A., Pezzilli, R., Bueno-de-Mesquita, B., Hegyi, P., Souček, P., Neoptolemos, J.P., Di Franco, G., Sperti, C., Kauffmann, E.F., Hlaváč, V., Uzunoğlu, F.G., Ermini, S., Małecka-Panas, E., Lucchesi, M., Vanella, G., Dijk, F., Mohelníková-Duchoňová, B., Bambi, F., Petrone, M.C., Jamroziak, K., Guo, F., Kolarova, K., Capretti, G., Milanetto, A.C., Ginocchi, L., Loveček, M., Puzzono, M., van Laarhoven, H.W.M., Carrara, S., Ivanauskas, A., Papiris, K., Basso, D., Arcidiacono, P.G., Izbéki, F., Chammas, R., Vodicka, P., Hackert, T., Pasquali, C., Piredda, M.L., Costello-Goldring, E., Cavestro, G.M., Szentesi, A., Tavano, F., Włodarczyk, B., Brenner, H., Kreivenaite, E., Gao, X., Bunduc, S., Vermeulen, R.C.H., Schneider, M.A., Latiano, A., Gioffreda, D., Testoni, S.G.G., Kupcinskas, J., Lawlor, R.T., Capurso, G., Malats, N., Campa, D., and Canzian, F.
- Published
- 2021
17. Bioengineered 3D models of human pancreatic cancer recapitulate in vivo tumour biology
- Author
-
Erkan, Murat Mert (ORCID 0000-0002-2753-0234 & YÖK ID 214689), de la Pena, Osuna D.; Trabulo, S.M.D.; Collin, E.; Liu, Y.; Sharma, S.; Tatari, M., Behrens, D.; Lawlor, R.T.; Scarpa, A.; Heeschen, C.; Mata, A.; Loessner, D., Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), School of Medicine, Erkan, Murat Mert (ORCID 0000-0002-2753-0234 & YÖK ID 214689), de la Pena, Osuna D.; Trabulo, S.M.D.; Collin, E.; Liu, Y.; Sharma, S.; Tatari, M., Behrens, D.; Lawlor, R.T.; Scarpa, A.; Heeschen, C.; Mata, A.; Loessner, D., Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), and School of Medicine
- Abstract
Patient-derived in vivo models of human cancer have become a reality, yet their turnaround time is inadequate for clinical applications. Therefore, tailored ex vivo models that faithfully recapitulate in vivo tumour biology are urgently needed. These may especially benefit the management of pancreatic ductal adenocarcinoma (PDAC), where therapy failure has been ascribed to its high cancer stem cell (CSC) content and high density of stromal cells and extracellular matrix (ECM). To date, these features are only partially reproduced ex vivo using organoid and sphere cultures. We have now developed a more comprehensive and highly tuneable ex vivo model of PDAC based on the 3D co-assembly of peptide amphiphiles (PAs) with custom ECM components (PA-ECM). These cultures maintain patient-specific transcriptional profiles and exhibit CSC functionality, including strong in vivo tumourigenicity. User-defined modification of the system enables control over niche-dependent phenotypes such as epithelial-to-mesenchymal transition and matrix deposition. Indeed, proteomic analysis of these cultures reveals improved matrisome recapitulation compared to organoids. Most importantly, patient-specific in vivo drug responses are better reproduced in self-assembled cultures than in other models. These findings support the use of tuneable self-assembling platforms in cancer research and pave the way for future precision medicine approaches., Biotechnology and Biological Sciences Research Council; LIDo Grant; Medical Research Council; UK Regenerative Medicine Platform Acellular/Smart Materials-3D Architecture; Fondazione Italiana Malattie Pancreas; Italian Ministry of Health; Fondazione Cariverona: Oncology Biobank Project “Antonio Schiavi”; European Union (EU); Horizon 2020; European Community Seventh Framework Programme; FP7/2007-2013; CAM-PaC Consortium; BIOMORPH; Marie Curie Integration Grant; STROFUNSCAFF; ERC Starting Grant; Associazione Italiana Ricerca Cancro; Barts Cancer Institute Catalyst; IMPETUS Awards
- Published
- 2021
18. Genome-wide association study identifies an early onset pancreatic cancer risk locus
- Author
-
Campa, D. Gentiluomo, M. Obazee, O. Ballerini, A. Vodickova, L. Hegyi, P. Soucek, P. Brenner, H. Milanetto, A.C. Landi, S. Gao, X. Bozzato, D. Capurso, G. Tavano, F. Vashist, Y. Hackert, T. Bambi, F. Bursi, S. Oliverius, M. Gioffreda, D. Schöttker, B. Ivanauskas, A. Mohelnikova-Duchonova, B. Darvasi, E. Pezzilli, R. Małecka-Panas, E. Strobel, O. Gazouli, M. Katzke, V. Szentesi, A. Cavestro, G.M. Farkas, G., Jr. Izbicki, J.R. Moz, S. Archibugi, L. Hlavac, V. Vincze, Á. Talar-Wojnarowska, R. Rusev, B. Kupcinskas, J. Greenhalf, B. Dijk, F. Giese, N. Boggi, U. Andriulli, A. Busch, O.R. Vanella, G. Vodicka, P. Nentwich, M. Lawlor, R.T. Theodoropoulos, G.E. Jamroziak, K. Zuppardo, R.A. Moletta, L. Ginocchi, L. Kaaks, R. Neoptolemos, J.P. Lucchesi, M. Canzian, F.
- Abstract
Early onset pancreatic cancer (EOPC) is a rare disease with a very high mortality rate. Almost nothing is known on the genetic susceptibility of EOPC, therefore, we performed a genome-wide association study (GWAS) to identify novel genetic variants specific for patients diagnosed with pancreatic ductal adenocarcinoma (PDAC) at younger ages. In the first phase, conducted on 821 cases with age of onset ≤60 years, of whom 198 with age of onset ≤50, and 3227 controls from PanScan I-II, we observed four SNPs (rs7155613, rs2328991, rs4891017 and rs12610094) showing an association with EOPC risk (P < 1 × 10−4). We replicated these SNPs in the PANcreatic Disease ReseArch (PANDoRA) consortium and used additional in silico data from PanScan III and PanC4. Among these four variants rs2328991 was significant in an independent set of 855 cases with age of onset ≤60 years, of whom 265 with age of onset ≤50, and 4142 controls from the PANDoRA consortium while in the in silico data, we observed no statistically significant association. However, the resulting meta-analysis supported the association (P = 1.15 × 10−4). In conclusion, we propose a novel variant rs2328991 to be involved in EOPC risk. Even though it was not possible to find a mechanistic link between the variant and the function, the association is supported by a solid statistical significance obtained in the largest study on EOPC genetics present so far in the literature. © 2020 UICC
- Published
- 2020
19. 30P Assessing the potential role of RICTOR expression as predictive factor of response to PI3K/mTOR pathway inhibitors in preclinical models of squamous cell lung cancer
- Author
-
Gkountakos, A., primary, Simbolo, M., additional, Vicentini, C., additional, Lawlor, R.T., additional, Veghini, L., additional, Pilotto, S., additional, Ludovini, V., additional, Chiari, R., additional, Novello, S., additional, Tortora, G., additional, Milella, M., additional, Scarpa, A., additional, Corbo, V., additional, and Bria, E., additional
- Published
- 2020
- Full Text
- View/download PDF
20. Germline BRCA2 K3326X and CHEK2 I157T mutations increase risk for sporadic pancreatic ductal adenocarcinoma
- Author
-
Obazee, O. Archibugi, L. Andriulli, A. Soucek, P. Małecka-Panas, E. Ivanauskas, A. Johnson, T. Gazouli, M. Pausch, T. Lawlor, R.T. Cavestro, G.M. Milanetto, A.C. Di Leo, M. Pasquali, C. Hegyi, P. Szentesi, A. Radu, C.E. Gheorghe, C. Theodoropoulos, G.E. Bergmann, F. Brenner, H. Vodickova, L. Katzke, V. Campa, D. Strobel, O. Kaiser, J. Pezzilli, R. Federici, F. Mohelnikova-Duchonova, B. Boggi, U. Lemstrova, R. Johansen, J.S. Bojesen, S.E. Chen, I. Jensen, B.V. Capurso, G. Pazienza, V. Dervenis, C. Sperti, C. Mambrini, A. Hackert, T. Kaaks, R. Basso, D. Talar-Wojnarowska, R. Maiello, E. Izbicki, J.R. Cuk, K. Saum, K.U. Cantore, M. Kupcinskas, J. Palmieri, O. Delle Fave, G. Landi, S. Salvia, R. Fogar, P. Vashist, Y.K. Scarpa, A. Vodicka, P. Tjaden, C. Iskierka-Jazdzewska, E. Canzian, F.
- Abstract
Rare truncating BRCA2 K3326X (rs11571833) and pathogenic CHEK2 I157T (rs17879961) variants have previously been implicated in familial pancreatic ductal adenocarcinoma (PDAC), but not in sporadic cases. The effect of both mutations in important DNA repair genes on sporadic PDAC risk may shed light on the genetic architecture of this disease. Both mutations were genotyped in germline DNA from 2,935 sporadic PDAC cases and 5,626 control subjects within the PANcreatic Disease ReseArch (PANDoRA) consortium. Risk estimates were evaluated using multivariate unconditional logistic regression with adjustment for possible confounders such as sex, age and country of origin. Statistical analyses were two-sided with p values
- Published
- 2019
21. Corrigendum: Common genetic variants associated with pancreatic adenocarcinoma may also modify risk of pancreatic neuroendocrine neoplasms [Carcinogenesis, 39, 3, (2018), (360-367)] DOI: 10.1093/carcin/bgx150
- Author
-
Obazee, O. Capurso, G. Tavano, F. Archibugi, L. De Bonis, A. Greenhalf, W. Key, T. Pasquali, C. Milanetto, A.C. Hackert, T. Fogar, P. Lico, V. Dervenis, C. Lawlor, R.T. Landoni, L. Gazouli, M. Zambon, C.F. Funel, N. Strobel, O. Jamroziak, K. Cantu, C. Malecka-Panas, E. Landi, S. Neoptolemos, J.P. Basso, D. Talar-Wojnarowska, R. Rinzivillo, M. Andriulli, A. Canzian, F. Campa, D.
- Subjects
GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,GeneralLiterature_MISCELLANEOUS - Abstract
In the originally published version of this article, the institutional affiliations of some authors were listed incorrectly. The correct affiliations are as listed above. This has been corrected online. The authors wish to apologize for this error. © The Author(s) 2018. Published by Oxford University Press. All rights reserved.
- Published
- 2018
22. Genome-wide meta-analysis identifies five new susceptibility loci for pancreatic cancer
- Author
-
Klein, A.P. Wolpin, B.M. Risch, H.A. Stolzenberg-Solomon, R.Z. Mocci, E. Zhang, M. Canzian, F. Childs, E.J. Hoskins, J.W. Jermusyk, A. Zhong, J. Chen, F. Albanes, D. Andreotti, G. Arslan, A.A. Babic, A. Bamlet, W.R. Beane-Freeman, L. Berndt, S.I. Blackford, A. Borges, M. Borgida, A. Bracci, P.M. Brais, L. Brennan, P. Brenner, H. Bueno-De-Mesquita, B. Buring, J. Campa, D. Capurso, G. Cavestro, G.M. Chaffee, K.G. Chung, C.C. Cleary, S. Cotterchio, M. Dijk, F. Duell, E.J. Foretova, L. Fuchs, C. Funel, N. Gallinger, S. Gaziano, J.M.M. Gazouli, M. Giles, G.G. Giovannucci, E. Goggins, M. Goodman, G.E. Goodman, P.J. Hackert, T. Haiman, C. Hartge, P. Hasan, M. Hegyi, P. Helzlsouer, K.J. Herman, J. Holcatova, I. Holly, E.A. Hoover, R. Hung, R.J. Jacobs, E.J. Jamroziak, K. Janout, V. Kaaks, R. Khaw, K.-T. Klein, E.A. Kogevinas, M. Kooperberg, C. Kulke, M.H. Kupcinskas, J. Kurtz, R.J. Laheru, D. Landi, S. Lawlor, R.T. Lee, I.-M. Lemarchand, L. Lu, L. Malats, N. Mambrini, A. Mannisto, S. Milne, R.L. Mohelníková-Duchoňová, B. Neale, R.E. Neoptolemos, J.P. Oberg, A.L. Olson, S.H. Orlow, I. Pasquali, C. Patel, A.V. Peters, U. Pezzilli, R. Porta, M. Real, F.X. Rothman, N. Scelo, G. Sesso, H.D. Severi, G. Shu, X.-O. Silverman, D. Smith, J.P. Soucek, P. Sund, M. Talar-Wojnarowska, R. Tavano, F. Thornquist, M.D. Tobias, G.S. Van Den Eeden, S.K. Vashist, Y. Visvanathan, K. Vodicka, P. Wactawski-Wende, J. Wang, Z. Wentzensen, N. White, E. Yu, H. Yu, K. Zeleniuch-Jacquotte, A. Zheng, W. Kraft, P. Li, D. Chanock, S. Obazee, O. Petersen, G.M. Amundadottir, L.T.
- Abstract
In 2020, 146,063 deaths due to pancreatic cancer are estimated to occur in Europe and the United States combined. To identify common susceptibility alleles, we performed the largest pancreatic cancer GWAS to date, including 9040 patients and 12,496 controls of European ancestry from the Pancreatic Cancer Cohort Consortium (PanScan) and the Pancreatic Cancer Case-Control Consortium (PanC4). Here, we find significant evidence of a novel association at rs78417682 (7p12/TNS3, P = 4.35 × 10-8). Replication of 10 promising signals in up to 2737 patients and 4752 controls from the PANcreatic Disease ReseArch (PANDoRA) consortium yields new genome-wide significant loci: Rs13303010 at 1p36.33 (NOC2L, P = 8.36 × 10-14), rs2941471 at 8q21.11 (HNF4G, P = 6.60 × 10-10), rs4795218 at 17q12 (HNF1B, P = 1.32 × 10-8), and rs1517037 at 18q21.32 (GRP, P = 3.28 × 10-8). rs78417682 is not statistically significantly associated with pancreatic cancer in PANDoRA. Expression quantitative trait locus analysis in three independent pancreatic data sets provides molecular support of NOC2L as a pancreatic cancer susceptibility gene. © 2018 The Author(s).
- Published
- 2018
23. Common genetic variants associated with pancreatic adenocarcinoma may also modify risk of pancreatic neuroendocrine neoplasms
- Author
-
Obazee, O. Capurso, G. Tavano, F. Archibugi, L. Bonis, A.D. Greenhalf, W. Key, T. Pasquali, C. Milanetto, A.C. Hackert, T. Fogar, P. Liço, V. Dervenis, C. Lawlor, R.T. Landoni, L. Gazouli, M. Zambon, C.F. Funel, N. Strobel, O. Jamroziak, K. Cantù, C. Malecka-Panas, E. Landi, S. Neoptolemos, J.P. Basso, D. Talar-Wojnarowska, R. Rinzivillo, M. Andriulli, A. Canzian, F. Campa, D.
- Abstract
Pancreatic neuroendocrine neoplasms (pNEN) account for less than 5% of all pancreatic neoplasms and genetic association studies on susceptibility to the disease are limited. We sought to identify possible overlap of genetic susceptibility loci between pancreatic ductal adenocarcinoma (PDAC) and pNEN; therefore, PDAC susceptibility variants (n = 23) from Caucasian genome-wide association studies (GWAS) were genotyped in 369 pNEN cases and 3277 controls from the PANcreatic Disease ReseArch (PANDoRA) consortium to evaluate the odds associated with pNEN risk, disease onset and tumor characteristics. Main effect analyses showed four PDAC susceptibility variants-rs9854771, rs1561927, rs9543325 and rs10919791 to be associated with pNEN risk. Subsequently, only associations with rs9543325, rs10919791 and rs1561927 were noteworthy with false positive report probability (FPRP) tests. Stratified analyses considering age at onset (50-year threshold), showed rs2736098, rs16986825 and rs9854771 to be associated with risk of developing pNEN at a younger age. Stratified analyses also showed some single nucleotide polymorphisms to be associated with different degrees of tumor grade, metastatic potential and functionality. Our results identify known GWAS PDAC susceptibility loci, which may also be involved in sporadic pNEN etiology and suggest that some genetic mechanisms governing pathogenesis of these two entities may be similar, with few of these loci being more influential in younger cases or tumor subtypes. © The Author(s) 2018. Published by Oxford University Press. All rights reserved.
- Published
- 2018
24. Validation of a tumour mutational burden workflow on routine histological samples of colorectal cancer and assessment of a cohort with synchronous hepatic metastases
- Author
-
Mafficini, A., primary, Cantù, C., additional, Antonello, D., additional, Simbolo, M., additional, Piredda, L., additional, Lever, V., additional, Rusev, B., additional, Pecori, S., additional, Milella, M., additional, Lawlor, R.T., additional, and Scarpa, A., additional
- Published
- 2019
- Full Text
- View/download PDF
25. Identification of recessively inherited genetic variants for pancreatic cancer risk
- Author
-
Lu, Y., Gentiluomo, M., Macauda, A., Gioffreda, D., Gazouli, M., Petrone, M.C., Kelemen, D., L.Ginocchi, Morelli, L., Papiris, K., Greenhalf, W., Izbicki, J.R., Kiudelis, V., Mohelníková-Duchoňová, B., Bueno-de-Mesquita, B., Vodicka, P., Brenner, H., Diener, M.K., Pezzilli, R., Ivanauskas, A., Salvia, R., Szentesi, A., Aoki, M.N., Németh, B.C., Sperti, C., Jamroziak, K., Chammas, R., Oliverius, M., Archibugi, L., Ermini, S., Novák, J., Kupcinskas, J., Strouhal, O., Souček, P., Cavestro, G.M., Milanetto, A.C., Vanella, G., Neoptolemos, J.P., Theodoropoulos, G.E., van Laarhoven, H.W.M., Mambrini, A., Moz, S., Kala, Z., Loveček, M., Basso, D., Uzunoglu, F.G., Hackert, T., Testoni, S.G.G., Hlavac, V., Andriulli, A., Lucchesi, M., Tavano, F., Carrara, S., Hegyi, P., Arcidiacono, P.G., Busch, O.R., Lawlor, R.T., Puzzono, M., Boggi, U., Guo, F., Małecka-Panas, E., Capurso, G., Landi, S., R.Talar-Wojnarowska, Strobel, O., Gao, X., Vashist, Y., Campa, D., and Canzian, F.
- Published
- 2021
- Full Text
- View/download PDF
26. Association of variants of genes involved in mitochondrial metabolism with pancreatic cancer risk
- Author
-
Peduzzi, G., Gentiluomo, M., Tavano, F., Arcidiacono, P.G., Ermini, S., Boggi, U., Cavestro, G.M., Capurso, G., Morelli, L., Milanetto, A.C., Pezzilli, R., Lawlor, R.T., Carrara, S., Lovecek, M., Souček, P., Vodičková, Guo, F., Hackert, T., Uzunoğlu, F.G., Gazouli, M., Párniczky, A., Kupcinskas, J., Bijlsm, M.F., Bueno-de-Mesquita, B., Vermeulen, R.C.H., van Eijck, C.H.J., Jamroziak, K., Talar-Wojnarowska, R., Greenhalf, W., Gioffreda, D., Petrone, M.C., Landi, S., Archibugi, L., Puzzono, M., Funel, N., Sperti, C., Piredda, M.L., Mohelnikova-Duchonova, B., Hlaváč, V., Gao, X., Schneider, M., Izbicki, J.R., Theodoropoulos, G., Bunduc, S., Kreivenaite, E., Busch, O.R., Małecka-Panas, E., Costello, E., Perri, F., Testoni, S.G.G., Vanella, G., Pasquali, C., Oliverius, M., H.Brenner, Loos, M., Götz, M., Georgiou, K., Erőss, B., Maiello, E., Szentes i, A., Bazzocchi, F., Basso, D., Neoptolemos, J.P., Hegyi, P., Kiudelis, V., Canzian, F., and Campa, D.
- Published
- 2021
- Full Text
- View/download PDF
27. Common germline variants within the CDKN2A/2B region affect risk of pancreatic neuroendocrine tumors
- Author
-
Campa, D. Capurso, G. Pastore, M. Talar-Wojnarowska, R. Milanetto, A.C. Landoni, L. Maiello, E. Lawlor, R.T. Malecka-Panas, E. Funel, N. Gazouli, M. De Bonis, A. Klüter, H. Rinzivillo, M. Delle Fave, G. Hackert, T. Landi, S. Bugert, P. Bambi, F. Archibugi, L. Scarpa, A. Katzke, V. Dervenis, C. Liço, V. Furlanello, S. Strobel, O. Tavano, F. Basso, D. Kaaks, R. Pasquali, C. Gentiluomo, M. Rizzato, C. Canzian, F.
- Abstract
Pancreatic neuroendocrine tumors (PNETs) are heterogeneous neoplasms which represent only 2% of all pancreatic neoplasms by incidence, but 10% by prevalence. Genetic risk factors could have an important role in the disease aetiology, however only a small number of case control studies have been performed yet. To further our knowledge, we genotyped 13 SNPs belonging to the pleiotropic CDKN2A/B gene region in 320 PNET cases and 4436 controls, the largest study on the disease so far. We observed a statistically significant association between the homozygotes for the minor allele of the rs2518719 SNP and an increased risk of developing PNET (ORhom = 2.08, 95% CI 1.05-4.11, p = 0.035). This SNP is in linkage disequilibrium with another polymorphic variant associated with increased risk of several cancer types. In silico analysis suggested that the SNP could alter the sequence recognized by the Neuron-Restrictive Silencer Factor (NRSF), whose deregulation has been associated with the development of several tumors. The mechanistic link between the allele and the disease has not been completely clarified yet but the epidemiologic evidences that link the DNA region to increased cancer risk are convincing. In conclusion, our results suggest rs2518719 as a pleiotropic CDKN2A variant associated with the risk of developing PNETs. © 2016 The Author(s).
- Published
- 2016
28. Undiscovered immune heterogeneity in pancreatic adenocarcinoma (PDAC)
- Author
-
Ragulan, C., primary, Poudel, P., additional, Young, K., additional, Nyamundanda, G., additional, Lawlor, R.T., additional, Scarpa, A., additional, and Sadanandam, A., additional
- Published
- 2017
- Full Text
- View/download PDF
29. Immune landscape of pancreatic neuroendocrine tumours (PanNETs)
- Author
-
Young, K., primary, Ragulan, C., additional, Nyamundanda, G., additional, Patil, Y., additional, Lawlor, R.T., additional, Cunningham, D., additional, Starling, N., additional, Scarpa, A., additional, and Sadanandam, A., additional
- Published
- 2017
- Full Text
- View/download PDF
30. A systems approach identifies time-dependent associations of multimorbidities with pancreatic cancer risk
- Author
-
Gomez-Rubio, P., primary, Rosato, V., additional, Márquez, M., additional, Bosetti, C., additional, Molina-Montes, E., additional, Rava, M., additional, Piñero, J., additional, Michalski, C.W., additional, Farré, A., additional, Molero, X., additional, Löhr, M., additional, Ilzarbe, L., additional, Perea, J., additional, Greenhalf, W., additional, O’Rorke, M., additional, Tardón, A., additional, Gress, T., additional, Barberá, V.M., additional, Crnogorac-Jurcevic, T., additional, Muñoz-Bellvís, L., additional, Domínguez-Muñoz, E., additional, Gutiérrez-Sacristán, A., additional, Balsells, J., additional, Costello, E., additional, Guillén-Ponce, C., additional, Huang, J., additional, Iglesias, M., additional, Kleeff, J., additional, Kong, B., additional, Mora, J., additional, Murray, L., additional, O’Driscoll, D., additional, Peláez, P., additional, Poves, I., additional, Lawlor, R.T., additional, Carrato, A., additional, Hidalgo, M., additional, Scarpa, A., additional, Sharp, L., additional, Furlong, L.I., additional, Real, F.X., additional, La Vecchia, C., additional, and Malats, N., additional
- Published
- 2017
- Full Text
- View/download PDF
31. 1405P - Validation of a tumour mutational burden workflow on routine histological samples of colorectal cancer and assessment of a cohort with synchronous hepatic metastases
- Author
-
Mafficini, A., Cantù, C., Antonello, D., Simbolo, M., Piredda, L., Lever, V., Rusev, B., Pecori, S., Milella, M., Lawlor, R.T., and Scarpa, A.
- Published
- 2019
- Full Text
- View/download PDF
32. Detection of EGFR alterations in circulating tumor DNA of non-small cell lung cancer by digital PCR and Next Generation Sequencing
- Author
-
Malpeli, G., primary, Simbolo, M., additional, Pilotto, S., additional, Lawlor, R.T., additional, Peretti, U., additional, Bonizzato, G., additional, Kinspergher, S., additional, Ferrara, R., additional, Cantu', C., additional, Caccese, M., additional, Tortora, G., additional, Bria, E., additional, and Scarpa, A., additional
- Published
- 2015
- Full Text
- View/download PDF
33. 1177P - Undiscovered immune heterogeneity in pancreatic adenocarcinoma (PDAC)
- Author
-
Ragulan, C., Poudel, P., Young, K., Nyamundanda, G., Lawlor, R.T., Scarpa, A., and Sadanandam, A.
- Published
- 2017
- Full Text
- View/download PDF
34. 428O - Immune landscape of pancreatic neuroendocrine tumours (PanNETs)
- Author
-
Young, K., Ragulan, C., Nyamundanda, G., Patil, Y., Lawlor, R.T., Cunningham, D., Starling, N., Scarpa, A., and Sadanandam, A.
- Published
- 2017
- Full Text
- View/download PDF
35. 980: Identification of potential biomarkers in pancreatic ductal adenocarcinoma associated to tumor–stroma interaction
- Author
-
Resovi, A., primary, Taraboletti, G., additional, Scarpa, A., additional, Lawlor, R.T., additional, Giavazzi, R., additional, and Belotti, D., additional
- Published
- 2014
- Full Text
- View/download PDF
36. Bioengineered 3D models of human pancreatic cancer recapitulate in vivo tumour biology
- Author
-
Estelle Collin, Ying Liu, Sara Trabulo, Rita T. Lawlor, Diana Behrens, David Osuna de la Peña, Daniela Loessner, Marianthi Tatari, Christopher Heeschen, Aldo Scarpa, Shreya Sharma, Mert Erkan, Alvaro Mata, Erkan, Murat Mert (ORCID 0000-0002-2753-0234 & YÖK ID 214689), de la Pena, Osuna D., Trabulo, S.M.D., Collin, E., Liu, Y., Sharma, S., Tatari, M., Behrens, D., Lawlor, R.T., Scarpa, A., Heeschen, C., Mata, A., Loessner, D., Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), and School of Medicine
- Subjects
Cancer microenvironment ,EXPRESSION ,STEM-CELL, STELLATE CELLS, BIOMATERIALS, VITRO, EXPRESSION, PHENOTYPES, HYDROGELS, PLATFORM, COLLAGEN, PROMOTE ,STELLATE CELLS ,Stromal cell ,Science ,HYDROGELS ,Cell Culture Techniques ,General Physics and Astronomy ,Bioengineering ,PHENOTYPES ,Biology ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Article ,Extracellular matrix ,In vivo ,Cancer stem cell ,Pancreatic cancer ,medicine ,Tumor Cells, Cultured ,Humans ,PROMOTE ,VITRO ,Cancer models ,BIOMATERIALS ,Science and technology ,Multidisciplinary ,Cancer stem cells ,Gene Expression Profiling ,Reproducibility of Results ,PLATFORM ,General Chemistry ,medicine.disease ,Precision medicine ,Peptide ,Cell culture techniques ,Gene expression profiling ,Gene expression regulation ,Neoplastic stem cells ,Pancreatic neoplasms ,Reproducibility of results ,Stromal cells ,COLLAGEN ,STEM-CELL ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Cancer research ,Neoplastic Stem Cells ,Stem cell ,Stromal Cells ,Ex vivo ,Carcinoma, Pancreatic Ductal - Abstract
Patient-derived in vivo models of human cancer have become a reality, yet their turnaround time is inadequate for clinical applications. Therefore, tailored ex vivo models that faithfully recapitulate in vivo tumour biology are urgently needed. These may especially benefit the management of pancreatic ductal adenocarcinoma (PDAC), where therapy failure has been ascribed to its high cancer stem cell (CSC) content and high density of stromal cells and extracellular matrix (ECM). To date, these features are only partially reproduced ex vivo using organoid and sphere cultures. We have now developed a more comprehensive and highly tuneable ex vivo model of PDAC based on the 3D co-assembly of peptide amphiphiles (PAs) with custom ECM components (PA-ECM). These cultures maintain patient-specific transcriptional profiles and exhibit CSC functionality, including strong in vivo tumourigenicity. User-defined modification of the system enables control over niche-dependent phenotypes such as epithelial-to-mesenchymal transition and matrix deposition. Indeed, proteomic analysis of these cultures reveals improved matrisome recapitulation compared to organoids. Most importantly, patient-specific in vivo drug responses are better reproduced in self-assembled cultures than in other models. These findings support the use of tuneable self-assembling platforms in cancer research and pave the way for future precision medicine approaches., Biotechnology and Biological Sciences Research Council; LIDo Grant; Medical Research Council; UK Regenerative Medicine Platform Acellular/Smart Materials-3D Architecture; Fondazione Italiana Malattie Pancreas; Italian Ministry of Health; Fondazione Cariverona: Oncology Biobank Project “Antonio Schiavi”; European Union (EU); Horizon 2020; European Community Seventh Framework Programme; FP7/2007-2013; CAM-PaC Consortium; BIOMORPH; Marie Curie Integration Grant; STROFUNSCAFF; ERC Starting Grant; Associazione Italiana Ricerca Cancro; Barts Cancer Institute Catalyst; IMPETUS Awards
- Published
- 2021
37. The Italian Rare Pancreatic Exocrine Cancer Initiative
- Author
-
Felice Giuliante, Enrico Vasile, Oronzo Brunetti, Chiara Alessandra Cella, Rossana Berardi, Stefano Cascinu, Anita Mangia, Sara Lonardi, Mario Scartozzi, Rita T. Lawlor, Daniele Santini, Fernando De Vita, Michele Milella, Claudio Luchini, Giuseppe Aprile, Antonella Argentiero, Stefania Tommasi, Andrea Casadei Gardini, Sandro Barni, Aldo Scarpa, Nicola Silvestris, Giovanni Brandi, Francesco Di Costanzo, Sara Delfanti, Claudio Doglioni, Vincenzo Mazzaferro, Ivana Cataldo, Evaristo Maiello, Paolo Marchetti, Brunetti, O., Luchini, C., Argentiero, A., Tommasi, S., Mangia, A., Aprile, G., Marchetti, P., Vasile, E., Casadei Gardini, A., Scartozzi, M., Barni, S., Delfanti, S., De Vita, F., Di Costanzo, F., Milella, M., Cella, C. A., Berardi, R., Cataldo, I., Santini, D., Doglioni, C., Maiello, E., Lawlor, R. T., Mazzaferro, V., Lonardi, S., Giuliante, F., Brandi, G., Scarpa, A., Cascinu, S., Silvestris, N., Brunetti O., Luchini C., Argentiero A., Tommasi S., Mangia A., Aprile G., Marchetti P., Vasile E., Casadei Gardini A., Scartozzi M., Barni S., Delfanti S., De Vita F., Di Costanzo F., Milella M., Cella C.A., Berardi R., Cataldo I., Santini D., Doglioni C., Maiello E., Lawlor R.T., Mazzaferro V., Lonardi S., Giuliante F., Brandi G., Scarpa A., Cascinu S., and Silvestris N.
- Subjects
Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Pancreatic ductal adenocarcinoma ,Adenosquamous carcinoma ,pancreatic cancer ,Acinar Cell ,Carcinoma, Adenosquamou ,chemotherapy ,Gastroenterology ,Carcinoma, Adenosquamous ,Adenosquamous ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Pancreatic cancer ,Carcinosarcoma ,medicine ,Humans ,Anaplastic carcinoma ,Retrospective Studies ,Rare tumors ,Carcinoma, Acinar Cell ,business.industry ,Rare tumor ,Carcinoma ,Pancreatic Neoplasm ,General Medicine ,medicine.disease ,Immunohistochemistry ,Cystic Neoplasm ,Pancreatic Neoplasms ,030104 developmental biology ,Italy ,Oncology ,Pancreatic exocrine cancer ,Medullary carcinoma ,Pancreatic Ductal ,030220 oncology & carcinogenesis ,biomolecular characterization ,Female ,Carcinoma, Pancreatic Ductal ,business ,Human - Abstract
Introduction:Exocrine pancreatic cancers include common type pancreatic ductal adenocarcinoma and cystic neoplasms, which account for 85% and 10% of cases, respectively. The remaining 5% are rare histotypes, comprising adenosquamous carcinoma, acinar cell carcinoma, signet ring cell carcinoma, medullary carcinoma, pancreatoblastoma, hepatoid carcinoma, undifferentiated carcinoma and its variant with osteoclast-like giant cells, solid pseudopapillary carcinoma, and carcinosarcoma. Due to their low incidence, little knowledge is available on their clinical and molecular features as well as on treatment choices. The national initiative presented here aims at the molecular characterization of series of rare histotypes for which therapeutic and follow-up data are available.Methods:A nationwide Italian Rare Pancreatic Cancer (IRaPaCa) task force whose first initiative is a multicentric retrospective study involving 21 Italian cancer centers to retrieve histologic material and clinical and treatment data of at least 100 patients with rare exocrine pancreatic cancers has been created. After histologic revision by a panel of expert pathologists, DNA and RNA from paraffin tissues will be investigated by next-generation sequencing using molecular pathway–oriented and immune-oriented mutational and expression profiling panels constructed availing of the information from the International Cancer Genome Consortium. Bioinformatic analysis of data will drive validation studies by immunohistochemistry and in situ hybridization, as well as nanostring assays.Conclusions:We expect to gather novel data on rare pancreatic cancer types that will be useful to inform the design of therapeutic choices.
- Published
- 2019
38. Perineural Invasion is a Strong Prognostic Moderator in Ampulla of Vater Carcinoma
- Author
-
Rita T. Lawlor, Laura D. Wood, Alessia Nottegar, Andrea Mafficini, Aldo Scarpa, Camilla Pilati, Claudio Luchini, Michele Simbolo, Vincenzo Corbo, Liang Cheng, Shinichi Yachida, Nicola Veronese, Brendon Stubbs, Giulio Riva, Aldo Mombello, Roberto Salvia, Luchini, C., Veronese, N., Nottegar, A., Riva, G., Pilati, C., Mafficini, A., Stubbs, B., Simbolo, M., Mombello, A., Corbo, V., Cheng, L., Yachida, S., Wood, L.D., Lawlor, R.T., Salvia, R., and Scarpa, A.
- Subjects
Ampulla of Vater ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,PNI ,Perineural invasion ,Perineum ,Gastroenterology ,03 medical and health sciences ,Broad spectrum ,0302 clinical medicine ,Endocrinology ,papilla, PNI, perineural, Vater ,Risk Factors ,Internal medicine ,Internal Medicine ,medicine ,Carcinoma ,Humans ,Neoplasm Invasiveness ,Vater ,papilla ,Hepatology ,business.industry ,Hazard ratio ,Prognosis ,medicine.disease ,Confidence interval ,Pancreatic Neoplasms ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Relative risk ,Meta-analysis ,perineural ,030211 gastroenterology & hepatology ,Neoplasm Recurrence, Local ,business ,Carcinoma, Pancreatic Ductal - Abstract
Objective Ampulla of Vater carcinoma (AVC) has a broad spectrum of different prognoses. As such, new moderators of survival are urgently needed. We aimed at clarifying the prognostic role of perineural invasion in AVC. Methods Using PubMed and SCOPUS databases, we conducted the first systematic review and meta-analysis on this topic. Results Analyzing 29 articles for a total of 2379 patients, we found that the presence of perineural invasion increased the risk of all-cause mortality more than 2 times (relative risk [RR], 2.07; 95% confidence interval [CI], 1.78-2.42 [P < 0.0001]; hazard ratio [HR], 2.72; 95% CI, 1.86-3.97 [P < 0.0001]), of cancer-specific mortality more than 6 times (RR, 6.12; 95% CI, 3.25-11.54 [P < 0.0001]; HR, 6.59; 95% CI, 2.29-3.49 [P < 0.0001]), and of recurrence more than 2 times (RR, 2.63; 95% CI, 1.89-3.67 [P < 0.0001]; HR, 2.54; 95% CI, 1.24-5.21 [P = 0.01]). Conclusions Perineural invasion is strongly associated with a poorer prognosis in AVC, influencing both survival and risk of recurrence. It should be reported in the final pathology report and should be taken into account by future oncologic staging systems, identifying a group of AVC with a more malignant biological behavior. © Wolters Kluwer Health, Inc. All rights reserved.
- Published
- 2019
39. Alternative lengthening of telomeres (ALT) influences survival in soft tissue sarcomas: a systematic review with meta-analysis
- Author
-
Antonio Pea, Alessia Nottegar, Nicola Veronese, Camilla Pilati, Lee Smith, Liang Cheng, Jacopo Demurtas, Rita T. Lawlor, Matteo Fassan, Claudio Luchini, Lawlor, R.T., Veronese, N., Pea, A., Nottegar, A., Smith, L., Pilati, C., Demurtas, J., Fassan, M., Cheng, L., and Luchini, C.
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Multivariate analysis ,Mesenchymal ,Survival ,ALT ,lcsh:RC254-282 ,digestive system ,Risk Assessment ,not known ,03 medical and health sciences ,0302 clinical medicine ,Surgical oncology ,Internal medicine ,Genetics ,medicine ,Humans ,Clinical significance ,ATR ,ATRX ,Prognosis ,Sarcoma ,Proportional Hazards Models ,business.industry ,Hazard ratio ,Soft tissue ,Telomere Homeostasis ,Middle Aged ,Telomere ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Survival Analysis ,digestive system diseases ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Relative risk ,Meta-analysis ,business ,Research Article - Abstract
Background Alternative lengthening of telomeres (ALT) is a telomerase-independent mechanism used by a broad range of neoplasms to maintain telomere length, permitting uncontrolled replication during their progression. ALT has been described in different types of sarcoma, but a comprehensive analysis of its clinical significance is still lacking. Therefore, we provide here the first meta-analysis on this topic. Methods We searched SCOPUS and PubMed through July 2018 to identify all studies that investigated the prognostic role of ALT in sarcomas. We considered the risk of death (risk ratio, RR) calculated as the number of death vs. total participants during follow-up in ALT+ versus ALT- patients as the primary outcome. The secondary outcome was the hazard ratio (HR), adjusted for the maximum number of covariates available, using ALT- patients as reference. Results Eight articles comprising a total of 551 patients with sarcomas (226 ALT+ and 325 ALT-) were selected. The ALT+ group showed a higher mitotic count and a higher tumor grade compared with the ALT- group (p
- Published
- 2019
40. Telomere length and health outcomes: An umbrella review of systematic reviews and meta-analyses of observational studies
- Author
-
Lin Yang, Guillermo F. López-Sánchez, Peter Willeit, Pinar Soysal, James Johnstone, Nicola Veronese, Joseph Firth, Lee Smith, Mark Hamer, Ai Koyanagi, Alessia Nottegar, Rita T. Lawlor, Justin D. Roberts, Adam D. Abbs, Brendon Stubbs, Claudio Luchini, Mike Loosemore, Thomas Waldhoer, Jacopo Demurtas, SOYSAL, PINAR, Smith, L., Luchini, C., Demurtas, J., Soysal, P., Stubbs, B., Hamer, M., Nottegar, A., Lawlor, R.T., Lopez-Sanchez, G.F., Firth, J., Koyanagi, A., Roberts, J., Willeit, P., Waldhoer, T., Loosemore, M., Abbs, A.D., Johnstone, J., Yang, L., and Veronese, N.
- Subjects
0301 basic medicine ,Aging ,Population ,Disease ,Biochemistry ,03 medical and health sciences ,Umbrella review ,0302 clinical medicine ,Alzheimer Disease ,Stomach Neoplasms ,Diabetes Mellitus ,Medicine ,Humans ,education ,Observational studies ,Molecular Biology ,education.field_of_study ,Telomere length ,business.industry ,Incidence (epidemiology) ,Incidence ,Evidence-based medicine ,Telomere ,Confidence interval ,Observational Studies as Topic ,030104 developmental biology ,Systematic review ,Treatment Outcome ,Neurology ,Relative risk ,Case-Control Studies ,Observational study ,business ,030217 neurology & neurosurgery ,Biotechnology ,Demography - Abstract
The aim of the present study was to map and grade evidence for the relationships between telomere length with a diverse range of health outcomes, using an umbrella review of systematic reviews with meta-analyses. We searched for meta-analyses of observational studies reporting on the association of telomere length with any health outcome (clinical disease outcomes and intermediate traits). For each association, random-effects summary effect size, 95% confidence interval (CI), and 95% prediction interval were calculated. To evaluate the credibility of the identified evidence, we assessed also heterogeneity, evidence for small-study effect and evidence for excess significance bias. Twenty-one relevant meta-analyses were identified reporting on 50 different outcomes and including a total of 326 observational studies. The level of evidence was high only for the association of short telomeres with higher risk of gastric cancer in the general population (relative risk, RR=1.95, 95%CI: 1.68-2.26), and moderate for the association of shorter telomeres with diabetes or with Alzheimer’s disease, even if limited to meta-analyses of case-control studies. There was weak evidence for twenty outcomes and not significant association for 27 health outcomes. The present umbrella review demonstrates that shorter telomere length may have an important role in incidence gastric cancer and, probably, diabetes and Alzheimer’s disease. At the same time, conversely to general assumptions, it does not find strong evidence supporting the notion that shorter telomere length plays an important role in many health outcomes that have been studied thus far.
- Published
- 2019
41. PD-1, PD-L1, and CD163 in pancreatic undifferentiated carcinoma with osteoclast-like giant cells: A expression patterns and clinical implications
- Author
-
Claudio Luchini, Alessia Nottegar, Antonio Pea, Jérôme Cros, Claudia Parolini, Lodewijk A.A. Brosens, Mattia Barbareschi, Vincenzo Corbo, Rita T. Lawlor, Camilla Pilati, Paola Piccoli, Laura D. Wood, Matteo Fassan, Paola Capelli, Aldo Scarpa, Peter Chianchiano, Giulio Riva, Nicola Sperandio, G. Johan A. Offerhaus, Giuseppe Malleo, Nicola Veronese, Andrea Mafficini, Michaël Noë, Liang Cheng, Borislav Rusev, Luchini, C., Cros, J., Pea, A., Pilati, C., Veronese, N., Rusev, B., Capelli, P., Mafficini, A., Nottegar, A., Brosens, L.A.A., Noë, M., Offerhaus, G.J.A., Chianchiano, P., Riva, G., Piccoli, P., Parolini, C., Malleo, G., Lawlor, R.T., Corbo, V., Sperandio, N., Barbareschi, M., Fassan, M., Cheng, L., Wood, L.D., and Scarpa, A.
- Subjects
Male ,0301 basic medicine ,Indiana ,Programmed Cell Death 1 Receptor ,Osteoclast ,PDAC ,Pancreatic Cancer ,Tumor-Associated Macrophages ,UCOGC ,Osteoclasts ,Giant Cells ,B7-H1 Antigen ,0302 clinical medicine ,Tumours of the digestive tract Radboud Institute for Molecular Life Sciences [Radboudumc 14] ,Lymphocytes ,Aged, 80 and over ,biology ,Tumor-associated macrophages ,Cell Differentiation ,Middle Aged ,Pancreatic cancer ,2734 ,Immunohistochemistry ,Europe ,Phenotype ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Antibody ,Carcinoma, Pancreatic Ductal ,Adult ,Antigens, Differentiation, Myelomonocytic ,Receptors, Cell Surface ,Pathology and Forensic Medicine ,03 medical and health sciences ,Immune system ,All institutes and research themes of the Radboud University Medical Center ,Antigens, CD ,PD-L1 ,Biomarkers, Tumor ,medicine ,Humans ,Histiocyte ,Aged ,Neoplasm Staging ,Histiocytes ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,Giant cell ,Cancer research ,biology.protein ,CD163 - Abstract
Undifferentiated carcinoma with osteoclast-like giant cells (UCOGC), a variant of pancreatic ductal adenocarcinoma (PDAC), has a striking genetic similarity to PDAC but a significantly improved overall survival. We hypothesize that this difference could be due to the immune response to the tumor, and as such, we investigated the expression of PD-1, PD-L1, and CD163 in a series of UCOGC. To this aim, 27 pancreatic UCOGCs (11 pure and 16 PDAC-associated), 5 extrapancreatic tumors with osteoclast-like giant cells and 10 pancreatic anaplastic carcinomas were immunostained using antibodies against PD-1, PD-L1, and CD163. In pancreatic UCOGCs, PD-L1 was expressed in neoplastic cells of 17 (63%) of 27 cases, more often in cases with an associated PDAC (P =.04). Expression of PD-L1 was associated with poor prognosis, confirmed by multivariate analysis: patients with PD-L1–positive UCOGCs had a risk of all-cause mortality that was 3 times higher than did patients with PD-L1–negative UCOGCs (hazard ratio, 3.397; 95% confidence interval, 1.023-18.375; P =.034). PD-L1 expression on tumor cells was also associated with aberrant P53 expression (P =.035). PD-1 was expressed on rare lymphocytes in 12 UCOGCs (44.4%), mainly located at the tumor periphery. CD163 was expressed on histiocytes, with a diffuse and strong staining pattern in all UCOGCs. Extrapancreatic tumors with osteoclast-like giant cells showed very similar staining patterns for the same proteins. Anaplastic carcinomas have some similarities to UCOGCs, but PD-L1 has no prognostic roles. Our results may have important implications for immunotherapeutic strategies in UCOGCs; these tumors may also represent a model for future therapeutic approaches against PDAC. © 2018 Elsevier Inc.
- Published
- 2018
42. Prognostic Role of High-Grade Tumor Budding in Pancreatic Ductal Adenocarcinoma: A Systematic Review and Meta-Analysis with a Focus on Epithelial to Mesenchymal Transition
- Author
-
Nicola Silvestris, Alessia Nottegar, Nicola Veronese, Giuseppe Malleo, Rita T. Lawlor, Roberto Salvia, Lee Smith, Aldo Scarpa, Jacopo Demurtas, Claudio Luchini, Liang Cheng, Laura D. Wood, Lawlor, R.T., Veronese, N., Nottegar, A., Malleo, G., Smith, L., Demurtas, J., Cheng, L., Wood, L.D., Silvestris, N., Salvia, R., Scarpa, A., and Luchini, C.
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pancreatic ductal adenocarcinoma ,pancreatic cancer ,High grade tumor ,Review ,EMT ,budding ,buds ,epithelial to mesenchymal transition ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Tumor budding ,Pancreatic cancer ,Internal medicine ,Medicine ,Clinical significance ,Epithelial–mesenchymal transition ,business.industry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,bud ,3. Good health ,030104 developmental biology ,Increased risk ,030220 oncology & carcinogenesis ,Meta-analysis ,business - Abstract
This study aims at clarifying the prognostic role of high-grade tumor budding (TB) in pancreatic ductal adenocarcinoma (PDAC) with the first systematic review and meta-analysis on this topic. Furthermore, we analyzed with a systematic review the relationship between TB and a recently suggested TB-associated mechanism: the epithelial to mesenchymal transition (EMT). Analyzing a total of 613 patients, 251 of them (40.9%) with high grade-TB, we found an increased risk of all-cause mortality (RR, 1.46; 95% CI, 1.13-1.88, p = 0.004; HR, 2.65; 95% CI, 1.79-3.91; p < 0.0001) and of recurrence (RR, 1.61; 95% CI, 1.05-2.47, p = 0.03) for PDAC patients with high-grade TB. Moreover, we found that EMT is a central process in determining the presence of TB in PDAC. Thanks to this meta-analysis, we demonstrate the potential clinical significance of high-grade TB for prognostic stratification of PDAC. TB also shows a clear association with the process of EMT. Based on the results of the present study, TB should be conveyed in pathology reports and taken into account by future oncologic staging systems. © 2019 by the authors. Licensee MDPI, Basel, Switzerland.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.