22 results on '"Paolo Chetta"'
Search Results
2. Supplementary Fig. S1-S4, Table S1 from Molecular Characterization of Prostate Cancer with Associated Gleason Score Using Mass Spectrometry Imaging
- Author
-
Nathalie Y.R. Agar, Fiona M. Fennessy, Clare M. Tempany, Massimo Loda, Jeffrey N. Agar, Sankha S. Basu, Sudeepa Syamala, Begona G.C. Lopez, Paolo Chetta, Giorgia Zadra, and Elizabeth C. Randall
- Abstract
S1. Segmentation map of a subset of lower Gleason tissue specimens, demonstrates regional differences between tumor and normal tissue, and between the two different Gleason grades S2. Example MALDI ion images of 2 long chain acylcarnitine species S3. Annotated H&E and serial frozen sections of human prostate tissue S4. Scores plot from PLSDA multivariate analysis of LESA MS data from tumor and normal prostate tissue Table 1. Pathological description of specimens used for MALDI MSI, cases were staged according to AJCC TNM 8th edition
- Published
- 2023
- Full Text
- View/download PDF
3. Supplementary Material and Methods from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
Supplementary Material and Methods
- Published
- 2023
- Full Text
- View/download PDF
4. Data from Molecular Characterization of Prostate Cancer with Associated Gleason Score Using Mass Spectrometry Imaging
- Author
-
Nathalie Y.R. Agar, Fiona M. Fennessy, Clare M. Tempany, Massimo Loda, Jeffrey N. Agar, Sankha S. Basu, Sudeepa Syamala, Begona G.C. Lopez, Paolo Chetta, Giorgia Zadra, and Elizabeth C. Randall
- Abstract
Diagnosis of prostate cancer is based on histologic evaluation of tumor architecture using a system known as the “Gleason score.” This diagnostic paradigm, while the standard of care, is time-consuming, shows intraobserver variability, and provides no information about the altered metabolic pathways, which result in altered tissue architecture. Characterization of the molecular composition of prostate cancer and how it changes with respect to the Gleason score (GS) could enable a more objective and faster diagnosis. It may also aid in our understanding of disease onset and progression. In this work, we present mass spectrometry imaging for identification and mapping of lipids and metabolites in prostate tissue from patients with known prostate cancer with GS from 6 to 9. A gradient of changes in the intensity of various lipids was observed, which correlated with increasing GS. Interestingly, these changes were identified in both regions of high tumor cell density, and in regions of tissue that appeared histologically benign, possibly suggestive of precancerous metabolomic changes. A total of 31 lipids, including several phosphatidylcholines, phosphatidic acids, phosphatidylserines, phosphatidylinositols, and cardiolipins were detected with higher intensity in GS (4+3) compared with GS (3+4), suggesting they may be markers of prostate cancer aggression. Results obtained through mass spectrometry imaging studies were subsequently correlated with a fast, ambient mass spectrometry method for potential use as a clinical tool to support image-guided prostate biopsy.Implications:In this study, we suggest that metabolomic differences between prostate cancers with different Gleason scores can be detected by mass spectrometry imaging.
- Published
- 2023
- Full Text
- View/download PDF
5. Figure S1 from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
TRAILR2 and CDH17 RNA expression in related normal tissues.
- Published
- 2023
- Full Text
- View/download PDF
6. Figure S4 from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
IHC for cleaved caspase-8 and cleaved caspase-3 24h after the second administration of BI 905711 in GP2d xenograft tumors.
- Published
- 2023
- Full Text
- View/download PDF
7. Table S1, S2, S3 and S4 from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
S1: Evaluation of CDH17 and TRAILR2 IHC staining in hepatic metastases of colorectal cancer -S2:BI 905711 kinetic and steady-state measurements -S3: CDH17 and TRAILR2 mean fluorescence intensity, antibody binding capacity and BI 905711 / TAS266' minPOC in 24 CRC cell lines -S4: Intergroup comparison of gross pathology observations in Cynomolgus Monkey small intestine, large intestine and pancreas
- Published
- 2023
- Full Text
- View/download PDF
8. Data from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
Activation of TRAILR2 has emerged as an important therapeutic concept in cancer treatment. TRAILR2 agonistic molecules have only had limited clinical success, to date, due either to lack of efficacy or hepatotoxicity. BI 905711 is a novel tetravalent bispecific antibody targeting both TRAILR2 and CDH17 and represents a novel liver-sparing TRAILR2 agonist specifically designed to overcome the disadvantages of previous strategies. Here, we show that BI 905711 effectively triggered apoptosis in a broad panel of CDH17-positive colorectal cancer tumor cells in vitro. Efficient induction of apoptosis was dependent on the presence of CDH17, as exemplified by the greater than 1,000-fold drop in potency in CDH17-negative cells. BI 905711 demonstrated single-agent tumor regressions in CDH17-positive colorectal cancer xenografts, an effect that was further enhanced upon combination with irinotecan. Antitumor efficacy correlated with induction of caspase activation, as measured in both the tumor and plasma. Effective tumor growth inhibition was further demonstrated across a series of different colorectal cancer PDX models. BI 905711 induced apoptosis in both a cis (same cell) as well as trans (adjacent cell) fashion, translating into significant antitumor activity even in xenograft models with heterogeneous CDH17 expression. In summary, we demonstrate that BI 905711 has potent and selective antitumor activity in CDH17-positive colorectal cancer models both in vitro and in vivo. The high prevalence of over 95% CDH17-positive tumors in patients with colorectal cancer, the molecule preclinical efficacy together with its potential for a favorable safety profile, support the ongoing BI 905711 phase I trial in colorectal cancer and additional CDH17-positive cancer types (NCT04137289).
- Published
- 2023
- Full Text
- View/download PDF
9. Figure S5 from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
Impact of CDH17 targeting on bi-specific molecules in vivo efficacy.
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S2 from Selective Tumor Cell Apoptosis and Tumor Regression in CDH17-Positive Colorectal Cancer Models using BI 905711, a Novel Liver-Sparing TRAILR2 Agonist
- Author
-
Klaus Peter Kuenkele, Mark Pearson, Norbert Kraut, Frank Hilberg, Iñigo Tirapu, Maria Antonietta Impagnatiello, Craig Giragossian, Joerg Rinnenthal, Rachel Kroe-Barrett, Philip N. Gorman, Darrin Dutcher, Jason Ho, Catarina Pinto, Paolo Chetta, Andreas Wernitznig, Cordula Weishaeupl, Shirley Wang, and Juan Manuel García-Martínez
- Abstract
Effect of BI 905711-mediated tumor cell death on dendritic cells.
- Published
- 2023
- Full Text
- View/download PDF
11. Precision RNAi using synthetic shRNAmir target sites
- Author
-
Thomas Hoffmann, Alexandra Hörmann, Maja Corcokovic, Jakub Zmajkovic, Matthias Hinterndorfer, Jasko Salkanovic, Fiona Spreitzer, Anna Köferle, Katrin Gitschtaler, Alexandra Popa, Sarah Oberndorfer, Florian Andersch, Markus Schäfer, Michaela Fellner, Nicole Budano, Jan G. Ruppert, Paolo Chetta, Melanie Wurm, Johannes Zuber, and Ralph A. Neumüller
- Abstract
Loss-of-function genetic tools are widely applied for validating therapeutic targets, but their utility remains limited by incomplete on- and uncontrolled off-target effects. We describe artificial RNA interference (ARTi) based on synthetic, ultra-potent, off-target-free shRNAs that enable efficient, inducible, and reversible suppression of any gene upon introduction of a synthetic target sequence into non-coding transcript regions. ARTi establishes a scalable loss-of-function tool with full control over on- and off-target effects.
- Published
- 2023
- Full Text
- View/download PDF
12. ELOVL5 Is a Critical and Targetable Fatty Acid Elongase in Prostate Cancer
- Author
-
Wayne D. Tilley, Andrew M. Scott, Irene Zinonos, Paolo Chetta, Ryan Carelli, Zeyad D. Nassar, Jonas Dehairs, Joanna L. Gillis, Massimo Loda, Nicolas H. Voelcker, Andreas Evdokiou, Johannes V. Swinnen, Etienne Waelkens, Luke A. Selth, Vasilios Panagopoulos, Natalie K. Ryan, Chui Yan Mah, Ian G. Mills, Elizabeth D. Williams, Deanna C. Miller, Julia S. Scott, Katarzyna Bloch, Margaret M. Centenera, Lisa M. Butler, Emma Evergren, Rita Derua, Giorgia Zadra, Terence Tieu, Ingrid Burvenich, Clyde Bango, Adrienne R. Hanson, Jelle Machiels, and Anna Cifuentes-Rius
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Fatty Acid Elongases ,Mice, SCID ,Gene Expression Regulation, Enzymologic ,Metastasis ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Cell Movement ,Mice, Inbred NOD ,Prostate ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,RNA, Small Interfering ,Cell Proliferation ,chemistry.chemical_classification ,Science & Technology ,Prostatic Neoplasms ,Fatty acid ,Lipid metabolism ,Lipidome ,Lipid Metabolism ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,Receptors, Androgen ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Cancer research ,Fatty acid elongation ,Life Sciences & Biomedicine - Abstract
The androgen receptor (AR) is the key oncogenic driver of prostate cancer, and despite implementation of novel AR targeting therapies, outcomes for metastatic disease remain dismal. There is an urgent need to better understand androgen-regulated cellular processes to more effectively target the AR dependence of prostate cancer cells through new therapeutic vulnerabilities. Transcriptomic studies have consistently identified lipid metabolism as a hallmark of enhanced AR signaling in prostate cancer, yet the relationship between AR and the lipidome remains undefined. Using mass spectrometry–based lipidomics, this study reveals increased fatty acyl chain length in phospholipids from prostate cancer cells and patient-derived explants as one of the most striking androgen-regulated changes to lipid metabolism. Potent and direct AR-mediated induction of ELOVL fatty acid elongase 5 (ELOVL5), an enzyme that catalyzes fatty acid elongation, was demonstrated in prostate cancer cells, xenografts, and clinical tumors. Assessment of mRNA and protein in large-scale data sets revealed ELOVL5 as the predominant ELOVL expressed and upregulated in prostate cancer compared with nonmalignant prostate. ELOVL5 depletion markedly altered mitochondrial morphology and function, leading to excess generation of reactive oxygen species and resulting in suppression of prostate cancer cell proliferation, 3D growth, and in vivo tumor growth and metastasis. Supplementation with the monounsaturated fatty acid cis-vaccenic acid, a direct product of ELOVL5 elongation, reversed the oxidative stress and associated cell proliferation and migration effects of ELOVL5 knockdown. Collectively, these results identify lipid elongation as a protumorigenic metabolic pathway in prostate cancer that is androgen-regulated, critical for metastasis, and targetable via ELOVL5. Significance: This study identifies phospholipid elongation as a new metabolic target of androgen action that is critical for prostate tumor metastasis.
- Published
- 2021
- Full Text
- View/download PDF
13. Ductal adenocarcinoma of the prostate: A systematic review and meta‐analysis of incidence, presentation, prognosis, and management
- Author
-
Ian G. Mills, Alastair D. Lamb, Richard J. Bryant, Massimo Loda, Eli Harriss, Paolo Chetta, Niall M. Corcoran, Lucy Davies, Freddie C. Hamdy, Clare Verrill, Yiannis Philippou, Altan Omer, Christopher M. Hovens, Declan G. Murphy, Nithesh Ranasinha, Timothy Rajakumar, Ken Chow, and Andrew Erickson
- Subjects
Biochemical recurrence ,Oncology ,medicine.medical_specialty ,recurrence ,business.industry ,Prostatectomy ,medicine.medical_treatment ,Acinar adenocarcinoma ,ductal carcinoma ,General Medicine ,prostate cancer ,medicine.disease ,survival ,Cystoprostatectomy ,Prostate-specific antigen ,Prostate cancer ,Prostate Ductal Adenocarcinoma ,Internal medicine ,medicine ,Adenocarcinoma ,acinar carcinoma ,business - Abstract
Context Ductal adenocarcinoma (DAC) is relatively rare, but is nonetheless the second most common subtype of prostate cancer. First described in 1967, opinion is still divided regarding its biology, prognosis, and outcome. Objectives To systematically interrogate the literature to clarify the epidemiology, diagnosis, management, progression, and survival statistics of DAC. Materials and methods We conducted a literature search of five medical databases from inception to May 04 2020 according to PRISMA criteria using search terms “prostate ductal adenocarcinoma” OR “endometriod adenocarcinoma of prostate” and variations of each. Results Some 114 studies were eligible for inclusion, presenting 2 907 170 prostate cancer cases, of which 5911 were DAC. [Correction added on 16 January 2021 after the first online publication: the preceding statement has been corrected in this current version.] DAC accounts for 0.17% of prostate cancer on meta‐analysis (range 0.0837%‐13.4%). The majority of DAC cases were admixed with predominant acinar adenocarcinoma (AAC). Median Prostate Specific Antigen at diagnosis ranged from 4.2 to 9.6 ng/mL in the case series. DAC was more likely to present as T3 (RR1.71; 95%CI 1.53‐1.91) and T4 (RR7.56; 95%CI 5.19‐11.01) stages, with far higher likelihood of metastatic disease (RR4.62; 95%CI 3.84‐5.56; all P‐values < .0001), compared to AAC. Common first treatments included surgery (radical prostatectomy (RP) or cystoprostatectomy for select cases) or radiotherapy (RT) for localized disease, and hormonal or chemo‐therapy for metastatic disease. Few studies compared RP and RT modalities, and those that did present mixed findings, although cancer‐specific survival rates seem worse after RP. Biochemical recurrence rates were increased with DAC compared to AAC. Additionally, DAC metastasized to unusual sites, including penile and peritoneal metastases. Where compared, all studies reported worse survival for DAC compared to AAC. Conclusion When drawing conclusions about DAC it is important to note the heterogenous nature of the data. DAC is often diagnosed incidentally post‐treatment, perhaps due to lack of a single, universally applied histopathological definition. As such, DAC is likely underreported in clinical practice and the literature. Poorer prognosis and outcomes for DAC compared to AAC merit further research into genetic composition, evolution, diagnosis, and treatment of this surprisingly common prostate cancer sub‐type. Patient summary Ductal prostate cancer is a rare but important form of prostate cancer. This review demonstrates that it tends to be more serious at detection and more likely to spread to unusual parts of the body. Overall survival is worse with this type of prostate cancer and urologists need to be aware of the presence of ductal prostate cancer to alter management decisions and follow‐up.
- Published
- 2021
- Full Text
- View/download PDF
14. A selective and orally bioavailable VHL-recruiting PROTAC achieves SMARCA2 degradation in vivo
- Author
-
Christiane Kofink, Nicole Trainor, Barbara Mair, Simon Wöhrle, Melanie Wurm, Nikolai Mischerikow, Michael J. Roy, Gerd Bader, Peter Greb, Géraldine Garavel, Emelyne Diers, Ross McLennan, Claire Whitworth, Vesna Vetma, Klaus Rumpel, Maximilian Scharnweber, Julian E. Fuchs, Thomas Gerstberger, Yunhai Cui, Gabriela Gremel, Paolo Chetta, Stefan Hopf, Nicole Budano, Joerg Rinnenthal, Gerhard Gmaschitz, Moriz Mayer, Manfred Koegl, Alessio Ciulli, Harald Weinstabl, and William Farnaby
- Subjects
Adenosine Triphosphatases ,Multidisciplinary ,Von Hippel-Lindau Tumor Suppressor Protein ,Ubiquitin-Protein Ligases ,Proteolysis ,DNA Helicases ,General Physics and Astronomy ,Humans ,Nuclear Proteins ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology ,Transcription Factors - Abstract
Targeted protein degradation offers an alternative modality to classical inhibition and holds the promise of addressing previously undruggable targets to provide novel therapeutic options for patients. Heterobifunctional molecules co-recruit a target protein and an E3 ligase, resulting in ubiquitylation and proteosome-dependent degradation of the target. In the clinic, the oral route of administration is the option of choice but has only been achieved so far by CRBN- recruiting bifunctional degrader molecules. We aimed to achieve orally bioavailable molecules that selectively degrade the BAF Chromatin Remodelling complex ATPase SMARCA2 over its closely related paralogue SMARCA4, to allow in vivo evaluation of the synthetic lethality concept of SMARCA2 dependency in SMARCA4-deficient cancers. Here we outline structure- and property-guided approaches that led to orally bioavailable VHL-recruiting degraders. Our tool compound, ACBI2, shows selective degradation of SMARCA2 over SMARCA4 in ex vivo human whole blood assays and in vivo efficacy in SMARCA4-deficient cancer models. This study demonstrates the feasibility for broadening the E3 ligase and physicochemical space that can be utilised for achieving oral efficacy with bifunctional molecules.
- Published
- 2022
15. Patient-derived head and neck tumor slice cultures: a versatile tool to study oncolytic virus action
- Author
-
Annette Runge, Melissa Mayr, Theresa Schwaiger, Susanne Sprung, Paolo Chetta, Timo Gottfried, Jozsef Dudas, Maria C. Greier, Marlies C. Glatz, Johannes Haybaeck, Knut Elbers, Herbert Riechelmann, Patrik Erlmann, and Monika Petersson
- Subjects
Mice ,Oncolytic Viruses ,Multidisciplinary ,Head and Neck Neoplasms ,Squamous Cell Carcinoma of Head and Neck ,Animals ,Humans ,Immunotherapy ,Ecosystem - Abstract
Head and neck cancer etiology and architecture is quite diverse and complex, impeding the prediction whether a patient could respond to a particular cancer immunotherapy or combination treatment. A concomitantly arising caveat is obviously the translation from pre-clinical, cell based in vitro systems as well as syngeneic murine tumor models towards the heterogeneous architecture of the human tumor ecosystems. To bridge this gap, we have established and employed a patient-derived HNSCC (head and neck squamous cell carcinoma) slice culturing system to assess immunomodulatory effects as well as permissivity and oncolytic virus (OV) action. The heterogeneous contexture of the human tumor ecosystem including tumor cells, cancer-associated fibroblasts and immune cells was preserved in our HNSCC slice culturing approach. Importantly, the immune cell compartment remained to be functional and cytotoxic T-cells could be activated by immunostimulatory antibodies. In addition, we uncovered that a high proportion of the patient-derived HNSCC slice cultures were susceptible to the OV VSV-GP. More specifically, VSV-GP infects a broad spectrum of tumor-associated lineages including epithelial and stromal cells and can induce apoptosis. In sum, this human tumor ex vivo platform might complement pre-clinical studies to eventually propel cancer immune-related drug discovery and ease the translation to the clinics.
- Published
- 2021
16. Molecular Characterization of Prostate Cancer with Associated Gleason Score Using Mass Spectrometry Imaging
- Author
-
Sankha S. Basu, Clare M. Tempany, Paolo Chetta, Massimo Loda, Nathalie Y. R. Agar, Sudeepa Syamala, Begoña Gimenez-Cassina Lopez, Jeffrey N. Agar, Elizabeth C. Randall, Giorgia Zadra, and Fiona M. Fennessy
- Subjects
Image-Guided Biopsy ,Male ,0301 basic medicine ,Cancer Research ,Prostate biopsy ,medicine.medical_treatment ,Mass spectrometry ,Article ,Mass Spectrometry ,Mass spectrometry imaging ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Metabolomics ,Prostate ,Biomarkers, Tumor ,medicine ,Humans ,Gleason scores ,Molecular Biology ,Prostatectomy ,medicine.diagnostic_test ,business.industry ,Prostatic Neoplasms ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Lipidomics ,Disease Progression ,Cancer research ,Neoplasm Grading ,business - Abstract
Diagnosis of prostate cancer is based on histologic evaluation of tumor architecture using a system known as the “Gleason score.” This diagnostic paradigm, while the standard of care, is time-consuming, shows intraobserver variability, and provides no information about the altered metabolic pathways, which result in altered tissue architecture. Characterization of the molecular composition of prostate cancer and how it changes with respect to the Gleason score (GS) could enable a more objective and faster diagnosis. It may also aid in our understanding of disease onset and progression. In this work, we present mass spectrometry imaging for identification and mapping of lipids and metabolites in prostate tissue from patients with known prostate cancer with GS from 6 to 9. A gradient of changes in the intensity of various lipids was observed, which correlated with increasing GS. Interestingly, these changes were identified in both regions of high tumor cell density, and in regions of tissue that appeared histologically benign, possibly suggestive of precancerous metabolomic changes. A total of 31 lipids, including several phosphatidylcholines, phosphatidic acids, phosphatidylserines, phosphatidylinositols, and cardiolipins were detected with higher intensity in GS (4+3) compared with GS (3+4), suggesting they may be markers of prostate cancer aggression. Results obtained through mass spectrometry imaging studies were subsequently correlated with a fast, ambient mass spectrometry method for potential use as a clinical tool to support image-guided prostate biopsy. Implications: In this study, we suggest that metabolomic differences between prostate cancers with different Gleason scores can be detected by mass spectrometry imaging.
- Published
- 2019
- Full Text
- View/download PDF
17. Discovery of potent and selective HER2 inhibitors with efficacy against HER2 exon 20 insertion-driven tumors, which preserve wild-type EGFR signaling
- Author
-
Birgit Wilding, Dirk Scharn, Dietrich Böse, Anke Baum, Valeria Santoro, Paolo Chetta, Renate Schnitzer, Dana A. Botesteanu, Christoph Reiser, Stefan Kornigg, Petr Knesl, Alexandra Hörmann, Anna Köferle, Maja Corcokovic, Simone Lieb, Guido Scholz, Jens Bruchhaus, Markus Spina, Josef Balla, Biljana Peric-Simov, Jasmin Zimmer, Sophie Mitzner, Thomas N. Fett, Alexandra Beran, Lyne Lamarre, Thomas Gerstberger, Daniel Gerlach, Markus Bauer, Andreas Bergner, Andreas Schlattl, Gerd Bader, Matthias Treu, Harald Engelhardt, Stephan Zahn, Julian E. Fuchs, Johannes Zuber, Peter Ettmayer, Mark Pearson, Mark Petronczki, Norbert Kraut, Darryl B. McConnell, Flavio Solca, and Ralph A. Neumüller
- Subjects
ErbB Receptors ,Cancer Research ,Lung Neoplasms ,Oncology ,Receptor, ErbB-2 ,Carcinoma, Non-Small-Cell Lung ,Humans ,Exons ,Genes, erbB-2 ,Protein Kinase Inhibitors - Abstract
Oncogenic alterations in human epidermal growth factor receptor 2 (HER2) occur in approximately 2% of patients with non-small cell lung cancer and predominantly affect the tyrosine kinase domain and cluster in exon 20 of the ERBB2 gene. Most clinical-grade tyrosine kinase inhibitors are limited by either insufficient selectivity against wild-type (WT) epidermal growth factor receptor (EGFR), which is a major cause of dose-limiting toxicity or by potency against HER2 exon 20 mutant variants. Here we report the discovery of covalent tyrosine kinase inhibitors that potently inhibit HER2 exon 20 mutants while sparing WT EGFR, which reduce tumor cell survival and proliferation in vitro and result in regressions in preclinical xenograft models of HER2 exon 20 mutant non-small cell lung cancer, concomitant with inhibition of downstream HER2 signaling. Our results suggest that HER2 exon 20 insertion-driven tumors can be effectively treated by a potent and highly selective HER2 inhibitor while sparing WT EGFR, paving the way for clinical translation.
- Published
- 2021
18. Metabolic reprogramming as an emerging mechanism of resistance to endocrine therapies in prostate cancer
- Author
-
Giorgia Zadra and Paolo Chetta
- Subjects
Prostate cancer ,Metabolomics ,Castration Resistance ,Mechanism (biology) ,business.industry ,Metabolic imaging ,Metabolic reprogramming ,Cancer research ,medicine ,Endocrine system ,medicine.disease ,business - Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death in the US. Androgen receptor (AR) signaling is the driver of both PCa development and progression and, thus, the major target of current in-use therapies. However, despite the survival benefit of second-generation inhibitors of AR signaling in the metastatic setting, resistance mechanisms inevitably occur. Thus, novel strategies are required to circumvent resistance occurrence and thereby to improve PCa survival. Among the key cellular processes that are regulated by androgens, metabolic reprogramming stands out because of its intricate links with cancer cell biology. In this review, we discuss how cancer metabolism and lipid metabolism in particular are regulated by androgens and contribute to the acquisition of resistance to endocrine therapy. We describe the interplay between genetic alterations, metabolic vulnerabilities and castration resistance. Since PCa cells adapt their metabolism to excess nutrient supply to promote cancer progression, we review our current knowledge on the association between diet/obesity and resistance to anti-androgen therapies. We briefly describe the metabolic symbiosis between PCa cells and tumor microenvironment and how this crosstalk might contribute to PCa progression. We discuss how tackling PCa metabolic vulnerabilities represents a potential approach of synthetic lethality to endocrine therapies. Finally, we describe how the continuous advances in analytical technologies and metabolic imaging have led to the identification of potential new prognostic and predictive biomarkers, and non-invasive approaches to monitor therapy response.
- Published
- 2020
19. MP67-19 DUCTAL ADENOCARCINOMA OF THE PROSTATE: A SYSTEMATIC REVIEW AND META-ANALYSIS OF INCIDENCE, PRESENTATION AND MANAGEMENT
- Author
-
Alastair D. Lamb, Massimo Loda, Nithesh Ranasinha, Niall M. Corcoran, Declan G. Murphy, Christopher M. Hovens, Freddie C. Hamdy, Andrew Erickson, Altan Omer, Clare Verrill, Lucy Davies, Eli Harriss, Paolo Chetta, Ian G. Mills, Ken Chow, and Richard J. Bryant
- Subjects
Oncology ,medicine.medical_specialty ,medicine.anatomical_structure ,Prostate ,business.industry ,Urology ,Internal medicine ,Meta-analysis ,Incidence (epidemiology) ,medicine ,Ductal adenocarcinoma ,Presentation (obstetrics) ,business - Published
- 2020
- Full Text
- View/download PDF
20. Abstract 985: BI 905711 selectively induces apoptosis and anti-tumor response in TRAILR2/CDH17- expressing pancreatic cancer models
- Author
-
Norbert Schweifer, Mikhila Mahendra, Paolo Chetta, Juan Manuel Garcia-Martinez, Timothy P. Heffernan, Annette Machado, Christopher P. Vellano, Christopher A. Bristow, Joseph R. Marszalek, Ningping Feng, Alessandro Carugo, Frank Hilberg, Justin K. Huang, Jing Han, Poojabahen Gandhi, Benjamin J. Bivona, Joseph R. Daniele, and Robert A. Mullinax
- Subjects
Cancer Research ,biology ,business.industry ,Cancer ,medicine.disease ,Oncology ,Apoptosis ,Pancreatic cancer ,Gene expression ,medicine ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,Receptor ,business ,Survival rate ,Caspase - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal adult cancers with an average 5-year survival rate of less than 10% due in part to the limited number of effective therapies. Activation of TRAILR2 (Tumor necrosis factor (TNF)-Related Apoptosis-Inducing Ligand Receptor 2) has emerged as an important therapeutic concept in cancer treatment. Traditional TRAILR2 agonists have had limited clinical success due to lack of efficacy or, importantly, severe hepatotoxicity. Here we present anti-tumor activity in preclinical PDAC models for BI 905711, a first-in-class tetravalent bispecific antibody specifically designed to overcome the disadvantages of previous strategies targeting TRAILR2. BI 905711 serves as a uniquely specific, and liver-sparing therapeutic by targeting tumors that co-express TRAILR2 and another cell surface protein CDH17, which has ~40% prevalence in PDAC and is not expressed in normal liver. Working from a large cohort of molecularly characterized PDAC PDX models, we provide the first preclinical evidence of BI 905711 exhibiting robust anti-tumor activity in difficult to treat PDAC PDX models. Anti-tumor efficacy in responding models correlated with strong induction of Caspases 3/7 and 8 activation in tumors 24 hours after a single dose of BI 905711, and was associated with the presence and expression levels of TRAILR2 and CDH17 proteins. Evaluation of models with differential TRAILR2 and CDH17 expression profiles helped define the expression threshold for each target that is associated with response, upon which clinical assay development is in process for future patient stratification. Additionally, response was also associated with PDAC molecular subtypes utilizing a novel proprietary gene co-expression network developed from a curated cohort of PDAC PDX tumors. Responders to BI 905711 were identified primarily within the classical and quasi-basal/hybrid subtypes when TRAILR2 was adequately co-expressed. This correlates with an enrichment pattern of CDH17 gene expression that is mostly within the classical gene cluster and strongly anti-correlated with basal-like cluster enrichment. Robust preclinical anti-tumor activity of BI 905711 in TRAILR2 and CDH17-expressing PDAC PDX models, along with this antibody's potential for a favorable safety profile, has justified the enrollment of pancreatic cancer patients in the ongoing BI 905711 FIH Phase I clinical trial (NCT04137289). Citation Format: Jing Han, Annette A. Machado, Mikhila Mahendra, Joseph R. Daniele, Christopher A. Bristow, Justin Kwang-Lay Huang, Alessandro Carugo, Robert A. Mullinax, Benjamin J. Bivona, Ningping Feng, Poojabahen Gandhi, Norbert Schweifer, Paolo Maria Chetta, Juan Manuel Garcia-Martinez, Frank Hilberg, Christopher P. Vellano, Timothy P. Heffernan, Joseph R. Marszalek. BI 905711 selectively induces apoptosis and anti-tumor response in TRAILR2/CDH17- expressing pancreatic cancer models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 985.
- Published
- 2021
- Full Text
- View/download PDF
21. Abstract 4522: Potent induction of a tumor-specific immune response by a cyclic dinucleotide STING agonist
- Author
-
Achim Grube, Martin Fleck, Vittoria Zinzalla, Norbert Kraut, Georg Rast, Maria Antonietta Impagnatiello, Sebastian Carotta, Gabriela Gremel, Esther Schmidt, Paolo Chetta, Jonathon Sedgwick, Thorsten Oost, Sophia Blake, Thorsten Laux, Thomas Zichner, Marco H. Hofmann, Tom Bretschneider, Ute Klinkhardt, Kirsten Arndt-Schmitz, Herbert Nar, and Otmar Schaaf
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,medicine.medical_treatment ,medicine.disease ,Primary tumor ,eye diseases ,03 medical and health sciences ,Sting ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Cytokine ,Oncology ,In vivo ,Interferon ,Cancer research ,Medicine ,Cytokine secretion ,business ,Ex vivo ,030215 immunology ,medicine.drug - Abstract
Introduction: Stimulator of interferon genes (STING) is activated by cyclic dinucleotides that are generated by cyclic GMP-AMP synthase in the presence of cytosolic DNA, resulting in a type 1 interferon response and the secretion of pro-inflammatory cytokines. BI-STING mimics the natural ligand of STING and hijacks the STING signaling pathway to trigger a potent anti-tumor immune response. Here we report the pre-clinical characterization of BI-STING in human cells lines in vitro, in human ex vivo systems and in vivo mouse models. Methods: Activity and specificity of BI-STING were tested in vitro in the human monocytic leukemia cell line THP1. Target engagement was confirmed using ex vivo studies in human whole blood and in human precision cut colorectal cancer slices. For in vivo studies, BI-STING was administered intratumorally (i.tu.) in a range of subcutaneous, syngeneic murine tumor models. The modulation of cytokine secretion over time was recorded and the induction of a tumor-specific immune response demonstrated. Results: Using THP1 cells, activation of down-stream signaling events by BI-STING was seen specifically in STING wild type, but not STING knock out cells. Ex vivo treatment of human whole blood and human precision cut colorectal cancer slices with BI-STING resulted in the significant induction of cytokine secretion (including IFNβ) and in an array of transcriptional changes associated with the activation of STING signaling. Treatment of tumor-bearing mice with a single dose of BI-STING i.tu. led to a transient increase of cytokine levels in tumor and plasma. In all tested models, i.tu. administration of BI-STING resulted in dose-dependent local tumor control. Importantly, animals whose primary tumor was cured by BI-STING treatment did not develop tumors when re-challenged with the same tumor cell line and an abscopal delay in tumor growth was seen for non-injected (or anenestic) lesions in mice carrying bilateral tumors. Abscopal tumor control was further improved when combined with anti-PD-1. Finally, ELISpot analysis resulted in a significantly increased number of immunospots in splenocytes from BI-STING treated animals as compared to vehicle control mice, confirming the induction of a tumor-specific immune response. Conclusions: BI-STING is a potent and specific inducer of the STING signaling pathway. In addition to local tumor control, i.tu. treatment with BI-STING results in a systemic anti-tumor immune response in mice, which was enhanced upon anti-PD-1 combination. A clinical trial to evaluate i.tu. administration of BI-STING alone and in combination with anti-PD-1 in patients with advanced solid tumors is ongoing. Citation Format: Gabriela Gremel, Maria A. Impagnatiello, Sebastian Carotta, Otmar Schaaf, Paolo M. Chetta, Thorsten Oost, Thomas Zichner, Marco Hofmann, Sophia Blake, Tom Bretschneider, Martin Fleck, Achim Grube, Herbert Nar, Georg Rast, Esther Schmidt, Ute Klinkhardt, Kirsten Arndt-Schmitz, Thorsten Laux, Vittoria Zinzalla, Jonathon Sedgwick, Norbert Kraut. Potent induction of a tumor-specific immune response by a cyclic dinucleotide STING agonist [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4522.
- Published
- 2020
- Full Text
- View/download PDF
22. Abstract 2748: The role of TMPRSS2_ERG fusions in modulating tumor microenvironment in prostate cancer
- Author
-
Caroline F. Ribeiro, Basudev Chowdhury, Giuseppe Nicolò Fanelli, Zhe Li, Sudeepa Syamala, Rory Kirchner, Giorgia Zadra, Paolo Chetta, Massimo Loda, and Hubert Pakula
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,ETS transcription factor family ,Wnt signaling pathway ,Biology ,medicine.disease ,TMPRSS2 ,WNT6 ,Prostate cancer ,Oncology ,WNT2 ,Cancer research ,medicine - Abstract
The control region of TMPRSS2 fuses with ETS transcription factor family members, particularly ERG. This occurs in ~20-50% of human prostate cancers depending on the ethnicity. TMPRSS2-ERG (T2E) fusions, however, do not display a notable phenotype in genetically engineered mouse models (GEMM) recapitulating the fusion expression and T2E also has no prognostic significance in humans, despite its high incidence. Wnt signaling has been implicated in prostate cancer. Here, we report a discovery of a role for T2E fusion that impacts the prostate stroma in pre-clinical models. The composition of stromal cells within the microenvironment in T2E mice was studied by single cell RNAseq analysis. We identified stromal cell clusters in T2E mice that differed from wild type mice in the expression of Pdgfrβ, Pdgfrα and Col1a1. Interestingly, these clusters showed an increase of expression of Wnt receptors such as Lgr5 and Fzd7 as well as Wnt ligands such as Sfrp2, Wnt2 and Wnt6. T2E GEMMs also exhibited upregulation of the Wnt-secretion regulator porcupine (PORCN-Protein-serine O-palmitoleoyltransferase) in the stroma adjacent to the T2E-expression prostate epithelial cells. Furthermore, lineage-tracing with the Wnt reporter and epithelial stem cell marker Lgr5 showed that Wnt-active cells were increased in prostatic intraepithelial neoplasia (PIN) lesions in T2E;Pten+/- mice. Strikingly, Lgr5+ cells were also found in the prostate stroma surrounding these PINs. Since, TMPRSS2/ERG fusions represent an early event in prostate tumorigenesis, here we provide a mechanism whereby induction of Wnt signaling in the stroma by T2E-expressing prostate epithelial cells increases the stem cell compartments in both epithelial and stromal cells. These data suggest that aberrant ERG-expressing prostate epithelial cells activate upregulation of Wnt signaling in stromal cells, which provides a possible route for enhancing prostate carcinogenesis. Citation Format: Hubert Pakula, Caroline F. Ribeiro, Giuseppe N. Fanelli, Rory Kirchner, Sudeepa Syamala, Basudev Chowdhury, Giorgia Zadra, Paolo Chetta, Zhe Li, Massimo Loda. The role of TMPRSS2_ERG fusions in modulating tumor microenvironment in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 2748.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.