39 results on '"Raccagni I"'
Search Results
2. Validation of an Engineered Cell Model for In Vitro and In Vivo HIF-1α Evaluation by Different Imaging Modalities
- Author
-
Lo Dico, A., Valtorta, S., Martelli, C., Belloli, S., Gianelli, U., Tosi, D., Bosari, S., Degrassi, A., Russo, M., Raccagni, I., Lucignani, G., Moresco, R. M., and Ottobrini, L.
- Published
- 2014
- Full Text
- View/download PDF
3. Imaging Metformin Efficacy as Add-On Therapy in Cells and Mouse Models of Human EGFR Glioblastoma
- Author
-
Valtorta, S, Lo Dico, A, Raccagni, I, Martelli, C, Pieri, V, Rainone, P, Todde, S, Zinnhardt, B, De Bernardi, E, Coliva, A, Politi, L, Viel, T, Jacobs, A, Galli, R, Ottobrini, L, Vaira, V, Moresco, R, Politi, LS, Jacobs, AH, Moresco, RM, Valtorta, S, Lo Dico, A, Raccagni, I, Martelli, C, Pieri, V, Rainone, P, Todde, S, Zinnhardt, B, De Bernardi, E, Coliva, A, Politi, L, Viel, T, Jacobs, A, Galli, R, Ottobrini, L, Vaira, V, Moresco, R, Politi, LS, Jacobs, AH, and Moresco, RM
- Abstract
Glioblastoma (GBM) is a highly aggressive tumor of the brain. Despite the efforts, response to current therapies is poor and 2-years survival rate ranging from 6-12%. Here, we evaluated the preclinical efficacy of Metformin (MET) as add-on therapy to Temozolomide (TMZ) and the ability of [F-18]FLT (activity of thymidine kinase 1 related to cell proliferation) and [F-18]VC701 (translocator protein, TSPO) Positron Emission Tomography (PET) radiotracers to predict tumor response to therapy. Indeed, TSPO is expressed on the outer mitochondrial membrane of activated microglia/macrophages, tumor cells, astrocytes and endothelial cells. TMZ-sensitive (Gli36 Delta EGFR-1 and L0627) or -resistant (Gli36 Delta EGFR-2) GBM cell lines representative of classical molecular subtype were tested in vitro and in vivo in orthotopic mouse models. Our results indicate that in vitro, MET increased the efficacy of TMZ on TMZ-sensitive and on TMZ-resistant cells by deregulating the balance between pro-survival (bcl2) and pro-apoptotic (bax/bad) Bcl-family members and promoting early apoptosis in both Gli36 Delta EGFR-1 and Gli36 Delta EGFR-2 cells. In vivo, MET add-on significantly extended the median survival of tumor-bearing mice compared to TMZ-treated ones and reduced the rate of recurrence in the TMZ-sensitive models. PET studies with the cell proliferation radiopharmaceutical [F-18]FLT performed at early time during treatment were able to distinguish responder from non-responder to TMZ but not to predict the duration of the effect. On the contrary, [F-18]VC701 uptake was reduced only in mice treated with MET plus TMZ and levels of uptake negatively correlated with animals' survival. Overall, our data showed that MET addition improved TMZ efficacy in GBM preclinical models representative of classical molecular subtype increasing survival time and reducing tumor relapsing rate. Finally, results from PET imaging suggest that the reduction of cell proliferation represents a common mecha
- Published
- 2021
4. PO-259 Inhibition of the hexosamine biosynthetic pathway by targeting PGM3 causes breast cancer growth arrest and apoptosis
- Author
-
Chiaradonna, F, Ricciardiello, F, Votta, G, Palorini, R, Raccagni, I, Brunelli, L, De Gioia, L, Pastorelli, R, Moresco, R, Ferla, B, Chiaradonna, F., Ricciardiello, F., Votta, G., Palorini, R., Raccagni, I., Brunelli, L., De Gioia, L., Pastorelli, R., Moresco, R. M., Ferla, B. La, Chiaradonna, F, Ricciardiello, F, Votta, G, Palorini, R, Raccagni, I, Brunelli, L, De Gioia, L, Pastorelli, R, Moresco, R, Ferla, B, Chiaradonna, F., Ricciardiello, F., Votta, G., Palorini, R., Raccagni, I., Brunelli, L., De Gioia, L., Pastorelli, R., Moresco, R. M., and Ferla, B. La
- Published
- 2018
5. Disruption of redox homeostasis for combinatorial drug efficacy in K-Ras tumors as revealed by metabolic connectivity profiling
- Author
-
Gaglio., D, Bonanomi, M, Valtorta, S, Bharat, R, Ripamonti, M, Conte, F, Fiscon, G, Righi, N, Napodano, E, Papa, F, Raccagni, I, J Parker, S, Cifola, I, Camboni, T, Paci, P, Colangelo, A, Vanoni, M, M Metallo, C, Moresco, R, Alberghina, L, Daniela Gaglio., Marcella Bonanomi, Silvia Valtorta, Rohit Bharat, Marilena Ripamonti, Federica Conte, Giulia Fiscon, Nicole Righi, Elisabetta Napodano, Federico Papa, Isabella Raccagni, Seth J Parker, Ingrid Cifola, Tania Camboni, Paola Paci, Anna Maria Colangelo, Marco Vanoni, Christian M Metallo, Rosa Maria Moresco, Lilia Alberghina, Gaglio., D, Bonanomi, M, Valtorta, S, Bharat, R, Ripamonti, M, Conte, F, Fiscon, G, Righi, N, Napodano, E, Papa, F, Raccagni, I, J Parker, S, Cifola, I, Camboni, T, Paci, P, Colangelo, A, Vanoni, M, M Metallo, C, Moresco, R, Alberghina, L, Daniela Gaglio., Marcella Bonanomi, Silvia Valtorta, Rohit Bharat, Marilena Ripamonti, Federica Conte, Giulia Fiscon, Nicole Righi, Elisabetta Napodano, Federico Papa, Isabella Raccagni, Seth J Parker, Ingrid Cifola, Tania Camboni, Paola Paci, Anna Maria Colangelo, Marco Vanoni, Christian M Metallo, Rosa Maria Moresco, and Lilia Alberghina
- Abstract
Background Rewiring of metabolism induced by oncogenicK-Rasin cancer cells involves both glucose and glutamine utilization sustaining enhanced, unrestricted growth. The development of effective anti-cancer treatments targeting metabolism may be facilitated by the identification and rational combinatorial targeting of metabolic pathways. Methods We performed mass spectrometric metabolomics analysis in vitro and in vivo experiments to evaluate the efficacy of drugs and identify metabolic connectivity. Results We show thatK-Ras-mutant lung and colon cancer cells exhibit a distinct metabolic rewiring, the latter being more dependent on respiration. Combined treatment with the glutaminase inhibitor CB-839 and the PI3K/aldolase inhibitor NVP-BKM120 more consistently reduces cell growth of tumor xenografts. Maximal growth inhibition correlates with the disruption of redox homeostasis, involving loss of reduced glutathione regeneration, redox cofactors, and a decreased connectivity among metabolites primarily involved in nucleic acid metabolism. Conclusions Our findings open the way to develop metabolic connectivity profiling as a tool for a selective strategy of combined drug repositioning in precision oncology.
- Published
- 2020
6. PO-259 Inhibition of the hexosamine biosynthetic pathway by targeting PGM3 causes breast cancer growth arrest and apoptosis
- Author
-
Chiaradonna, F., primary, Ricciardiello, F., additional, Votta, G., additional, Palorini, R., additional, Raccagni, I., additional, Brunelli, L., additional, De Gioia, L., additional, Pastorelli, R., additional, Moresco, R.M., additional, and Ferla, B. La, additional
- Published
- 2018
- Full Text
- View/download PDF
7. [18F]FDG and [18F]FLT PET for the evaluation of response to neo-adjuvant chemotherapy in a model of triple negative breast cancer
- Author
-
Raccagni, I, Belloli, S, Valtorta, S, Stefano, A, Presotto, L, Pascali, C, Bogni, A, Tortoreto, M, Zaffaroni, N, Daidone, M, Russo, G, Bombardieri, E, Moresco, R, Raccagni, Isabella, Belloli, Sara, Valtorta, Silvia, Stefano, Alessandro, Presotto, Luca, Pascali, Claudio, Bogni, Anna, Tortoreto, Monica, Zaffaroni, Nadia, Daidone, Maria Grazia, Russo, Giorgio, Bombardieri, Emilio, Moresco, Rosa Maria, Raccagni, I, Belloli, S, Valtorta, S, Stefano, A, Presotto, L, Pascali, C, Bogni, A, Tortoreto, M, Zaffaroni, N, Daidone, M, Russo, G, Bombardieri, E, Moresco, R, Raccagni, Isabella, Belloli, Sara, Valtorta, Silvia, Stefano, Alessandro, Presotto, Luca, Pascali, Claudio, Bogni, Anna, Tortoreto, Monica, Zaffaroni, Nadia, Daidone, Maria Grazia, Russo, Giorgio, Bombardieri, Emilio, and Moresco, Rosa Maria
- Abstract
RATIONALE: Pathological response to neo-adjuvant chemotherapy (NAC) represents a commonly used predictor of survival in triple negative breast cancer (TNBC) and the need to identify markers that predict response to NAC is constantly increasing. Aim of this study was to evaluate the potential usefulness of PET imaging with [18F]FDG and [18F]FLT for the discrimination of TNBC responders to Paclitaxel (PTX) therapy compared to the response assessed by an adapted Response Evaluation Criteria In Solid Tumors (RECIST) criteria based on tumor volume (Tumor Volume Response). METHODS: Nu/nu mice bearing TNBC lesions of different size were evaluated with [18F]FDG and [18F]FLT PET before and after PTX treatment. SUVmax, Metabolic Tumor Volume (MTV) and Total Lesion Glycolysis (TLG) and Proliferation (TLP) were assessed using a graph-based random walk algorithm. RESULTS: We found that in our TNBC model the variation of [18F]FDG and [18F]FLT SUVmax similarly defined tumor response to therapy and that SUVmax variation represented the most accurate parameter. Response evaluation using Tumor Volume Response (TVR) showed that the effectiveness of NAC with PTX was completely independent from lesions size at baseline. CONCLUSIONS: Our study provided interesting results in terms of sensitivity and specificity of PET in TNBC, revealing the similar performances of [18F]FDG and [18F]FLT in the identification of responders to Paclitaxel
- Published
- 2018
8. Inhibition of the Hexosamine Biosynthetic Pathway by targeting PGM3 causes breast cancer growth arrest and apoptosis
- Author
-
Ricciardiello, F, Votta, G, Palorini, R, Raccagni, I, Brunelli, L, Paiotta, A, Tinelli, F, D’Orazio, G, Valtorta, S, De Gioia, L, Pastorelli, R, Moresco, R, La Ferla, B, Chiaradonna, F, RICCIARDIELLO, FRANCESCA, BRUNELLI, LUCA, TINELLI, FRANCESCA, PASTORELLI, ROBERTA, Moresco, RM, Ricciardiello, F, Votta, G, Palorini, R, Raccagni, I, Brunelli, L, Paiotta, A, Tinelli, F, D’Orazio, G, Valtorta, S, De Gioia, L, Pastorelli, R, Moresco, R, La Ferla, B, Chiaradonna, F, RICCIARDIELLO, FRANCESCA, BRUNELLI, LUCA, TINELLI, FRANCESCA, PASTORELLI, ROBERTA, and Moresco, RM
- Abstract
Cancer aberrant N- and O-linked protein glycosylation, frequently resulting from an augmented flux through the Hexosamine Biosynthetic Pathway (HBP), play different roles in tumor progression. However, the low specificity and toxicity of the existing HBP inhibitors prevented their use for cancer treatment. Here we report the preclinical evaluation of FR054, a novel inhibitor of the HBP enzyme PGM3, with a remarkable anti-breast cancer effect. In fact, FR054 induces in different breast cancer cells a dramatic decrease in cell proliferation and survival. In particular, in a model of Triple Negative Breast Cancer (TNBC) cells, MDA-MB-231, we show that these effects are correlated to FR054-dependent reduction of both N- and O-glycosylation level that cause also a strong reduction of cancer cell adhesion and migration. Moreover we show that impaired survival of cancer cells upon FR054 treatment is associated with the activation of the Unfolded Protein Response (UPR) and accumulation of intracellular ROS. Finally, we show that FR054 suppresses cancer growth in MDA-MB-231 xenograft mice, supporting the advantage of targeting HBP for therapeutic purpose and encouraging further investigation about the use of this small molecule as a promising compound for breast cancer therapy.
- Published
- 2018
9. Feasibility of supervised machine learning technique for assisted diagnosis of cancer: application to PET studies in small animals
- Author
-
Gallivanone, F, Di Grigoli, G, Belloli, S, Valtorta, Silvia, Raccagni, I, MORESCO, ROSA MARIA, SALVATORE, CHRISTIAN, GILARDI, MARIA CARLA, CASTIGLIONI, ISABELLA, Gallivanone, F, Di Grigoli, G, Salvatore, C, Belloli, S, Valtorta, S, Raccagni, I, Gilardi, M, Castiglioni, I, and Moresco, R
- Subjects
machine learning - Published
- 2013
10. Metformin and temozolomide, a synergic option to overcome resistance in glioblastoma multiforme models
- Author
-
Valtorta, S, Dico, A, Raccagni, I, Gaglio, D, Belloli, S, Politi, L, Martelli, C, Diceglie, C, Bonanomi, M, Ercoli, G, Vaira, V, Ottobrini, L, Moresco, R, Valtorta, Silvia, Dico, Alessia Lo, Raccagni, Isabella, Gaglio, Daniela, Belloli, Sara, Politi, Letterio S., Martelli, Cristina, Diceglie, Cecilia, Bonanomi, Marcella, Ercoli, Giulia, Vaira, Valentina, Ottobrini, Luisa, Moresco, Rosa Maria, Valtorta, S, Dico, A, Raccagni, I, Gaglio, D, Belloli, S, Politi, L, Martelli, C, Diceglie, C, Bonanomi, M, Ercoli, G, Vaira, V, Ottobrini, L, Moresco, R, Valtorta, Silvia, Dico, Alessia Lo, Raccagni, Isabella, Gaglio, Daniela, Belloli, Sara, Politi, Letterio S., Martelli, Cristina, Diceglie, Cecilia, Bonanomi, Marcella, Ercoli, Giulia, Vaira, Valentina, Ottobrini, Luisa, and Moresco, Rosa Maria
- Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with poor survival. Cytoreduction in association with radiotherapy and temozolomide (TMZ) is the standard therapy, but response is heterogeneous and life expectancy is limited. The combined use of chemotherapeutic agents with drugs targeting cell metabolism is becoming an interesting therapeutic option for cancer treatment. Here, we found that metformin (MET) enhances TMZ effect on TMZ-sensitive cell line (U251) and overcomes TMZ-resistance in T98G GBM cell line. In particular, combined-treatment modulated apoptosis by increasing Bax/Bcl-2 ratio, and reduced Reactive Oxygen Species (ROS) production. We also observed that MET associated with TMZ was able to reduce the expression of glioma stem cells (GSC) marker CD90 particularly in T98G cells but not that of CD133. In vivo experiments showed that combined treatment with TMZ and MET significantly slowed down growth of TMZ-resistant tumors but did not affect overall survival of TMZ-sensitive tumor bearing mice. In conclusion, our results showed that metformin is able to enhance TMZ effect in TMZ-resistant cell line suggesting its potential use in TMZ refractory GBM patients. However, the lack of effect on a GBM malignancy marker like CD133 requires further evaluation since it might influence response duration.
- Published
- 2017
11. Metabolic evaluation of non-small cell lung cancer patient-derived xenograft models using 18F-FDG PET: A potential tool for early therapy response
- Author
-
Valtorta, S, Moro, M, Prisinzano, G, Bertolini, G, Tortoreto, M, Raccagni, I, Pastorino, U, Roz, L, Sozzi, G, Moresco, R, VALTORTA, SILVIA, RACCAGNI, ISABELLA, MORESCO, ROSA MARIA, Valtorta, S, Moro, M, Prisinzano, G, Bertolini, G, Tortoreto, M, Raccagni, I, Pastorino, U, Roz, L, Sozzi, G, Moresco, R, VALTORTA, SILVIA, RACCAGNI, ISABELLA, and MORESCO, ROSA MARIA
- Abstract
Lung cancer heterogeneity makes response to therapy extremely hard to predict. Patient-derived xenografts (PDXs) are a reliable preclinical model that closely recapitulates the main characteristics of the parental tumors and may represent a useful asset for testing new therapies. Here, using PET imaging, we investigated whether lung cancer PDXs reproduce the metabolic characteristics of the corresponding parental tumors. Methods: We performed longitudinal 18FFDG PET studies on 9 different PDX groups obtained by implanting primary-cancer fragments harvested from patients into mice. The SUVmax of each PDX was calculated and compared with the SUVmax of the corresponding parental tumor. Results: Tumor growth rate and uptake varied among the different PDXs and confirmed the preservation of individual characteristics. The intragroup reproducibility of PET measurements was good. Furthermore, PDXs from tumors with a higher metabolic rate displayed a rank order of uptake similar to that of the parental tumors. Conclusion: PDXs reproduced the glucose metabolism of the parental tumors and therefore represent a promising preclinical model for the early assessment of therapy efficacy.
- Published
- 2017
12. Tumour hypoxia: lessons learnt from preclinical imaging
- Author
-
Raccagni, I, Valtorta, S, Moresco, R, Belloli, S, RACCAGNI, ISABELLA, VALTORTA, SILVIA, MORESCO, ROSA MARIA, BELLOLI, SARA, Raccagni, I, Valtorta, S, Moresco, R, Belloli, S, RACCAGNI, ISABELLA, VALTORTA, SILVIA, MORESCO, ROSA MARIA, and BELLOLI, SARA
- Abstract
Purpose: In tumour, the imbalance between oxygen supply and demand leads to hypoxia, which represents a negative prognostic factor associated with aggressive tumour phenotype and therapy resistance. This review provides an overview of the use of positron emitter-labelled radiopharmaceutical used to image hypoxia in preclinical models of cancer. Methods: A critical and comprehensive PubMed search was performed identifying articles related to PET imaging for hypoxia assessment in preclinical setting from January 2007 up to January 2017. Results: We have considered and described a total of 54 original articles, exploring tumour-associated hypoxia in preclinical models. Results underlined the potential application together with the advantages and pitfalls of the use of PET in preclinical research. Multi-target imaging allowed to better define the relationship between hypoxia and other biological hallmarks of tumour; imaging of hypoxia was proved as a useful tool for lesions stratification and response prediction to radiotherapy; however, cutoff indexes were identified in few studies. Hypoxia PET showed remarkable tracer delivery limitations in the study of vascular disrupting agents but suggested the potential use of PET as a marker of response or resistance to anti-angiogenics. Finally, the effect of anaesthesia on tracer kinetics and tumour oxygenation as well as perfusion dependency in tracer uptake should be carefully evaluated to avoid artefactual results. Conclusions: Preclinical studies highlight the advantages and the limitations of the available hypoxia-radiotracers and their potential usefulness for the evaluation of treatments outcome and radiotherapy planning
- Published
- 2017
13. Leukocytes recruited by tumor-derived HMGB1 sustain peritoneal carcinomatosis
- Author
-
Cottone, L, Capobianco, A, Gualteroni, C, Monno, A, Raccagni, I, Valtorta, S, Canu, T, Tomaso, T, Lombardo, A, Esposito, A, Moresco, R, Maschio, A, Naldini, L, Rovere Querini, P, Bianchi, M, Manfredi, A, RACCAGNI, ISABELLA, Valtorta, Silvia, MORESCO, ROSA MARIA, Manfredi, A., Cottone, L, Capobianco, A, Gualteroni, C, Monno, A, Raccagni, I, Valtorta, S, Canu, T, Tomaso, T, Lombardo, A, Esposito, A, Moresco, R, Maschio, A, Naldini, L, Rovere Querini, P, Bianchi, M, Manfredi, A, RACCAGNI, ISABELLA, Valtorta, Silvia, MORESCO, ROSA MARIA, and Manfredi, A.
- Abstract
The factors that determine whether disseminated transformed cells in vivo yield neoplastic lesions have only been partially identified. We established an ad hoc model of peritoneal carcinomatosis by injecting colon carcinoma cells in mice. Tumor cells recruit inflammatory leukocytes, mostly macrophages, and generate neoplastic peritoneal lesions. Phagocyte depletion via clodronate treatment reduces neoplastic growth. Colon carcinoma cells release a prototypic damage-associated molecular pattern (DAMP)/alarmin, High Mobility Group Box1 (HMGB1), which attracts leukocytes. Exogenous HMGB1 accelerates leukocyte recruitment, macrophage infiltration, tumor growth and vascularization. Lentiviral-based HMGB1 knockdown or pharmacological interference with its extracellular impair macrophage recruitment and tumor growth. Our findings provide a preclinical proof of principle that strategies based on preventing HMGB1-driven recruitment of leukocytes could be used for treating peritoneal carcinomatosis.
- Published
- 2016
14. MONITORING PRECLINICAL MODEL OF BREAST CANCER WITH [18F]FLT AND [18F]FDG-PET
- Author
-
Belloli S, Bogni A, Valtorta S, Tortoreto M, Pascali C, Raccagni I, Zaffaroni N, Crippa F, Daidone MG, Moresco RM, and Bombardieri E
- Published
- 2013
15. Assessing cancer metabolic remodeling in animal models using PET imaging
- Author
-
Raccagni, I., Gaglio, D., Valtorta, S., Belloli, S., Di Grigoli, G., Vanoni, M., Mastroianni, F., Todde, S., Alberghina, L., and Moresco, R. M.
- Abstract
Aim: K-ras proteins have been found mutated in about 35% of human tumors and appear to be an important factor for tumorigenesis. Its expression correlates with metabolic alterations such as increased glycolysis and glutamine consumption. This ability of cancer cells to decouple glucose and glutamine uptake reprogramming their metabolism leads to a more efficient use of nutrients in order to support cell proliferation and represents an interesting target for cancer therapy. Aim of this study is to investigate the metabolic alterations occurring in k-ras transformed fibroblasts combining in vivo and in vitro studies. Materials and methods: nu/nu mice were subcutaneously implanted with 2.5 x 105 k-ras transformed murine NIH3T3 fibroblasts (oncogenic k-ras) or with 2.5 x 105 murine fibroblasts with the dominant negative mutation on GEF protein that attenuates k-ras activation reverting to the wild type phenotype (reverted). Lesions size was constantly monitored with calliper and volumes calculated as (L x l2)/2 mm3. Animals performed [18F]FDG- and [18F]FLTPET studies at several time points starting with a tumor dimension consistent with animal PET spatial resolution (approximately 2mm). Images were calibrated, corrected for isotope half-life and elaborated with PMOD software to calculate Standardized Uptake Value (SUVmax). Finally, animals were sacrificed and tumor collected for metabolomic analysis. Results: All animals injected with oncogenic k-ras fibroblasts develop in few days fast growing, aggressive and highly glycolytic tumors that appear homogeneous for both [18F]FDG and [18F]FLT SUV values. On the contrary, 40% of k-ras reverted animals develop heterogeneous tumors at later time. Among k-ras reverted animals, two distinct tumor phenotypes can be observed: small, slow growing and poor glycolitic tumors with low uptake of both tracers, and small, slow growing but highly glycolytic and proliferating tumors that present SUVmax values comparable to those of k-ras oncogenic tumors. A good correlation, even if not significant, between tumor volume and [18F]FLT SUVmax has been observed for oncogenic k-ras animals. Conclusions:PET imaging is an accurate in vivo technique able to visualize and monitor tumor development in the k-Ras fibroblasts mouse model. K-Ras transformed fibroblasts give rise to aggressive and fast-growing tumors that represent a good model to study the efficacy of cell metabolism based therapy. Finally, K-Ras reverted tumors need further investigations to understand the interaction among tumor microenvironment, metabolic alterations and genetic component that may trigger tumor development.
- Published
- 2013
16. PET imaging as a biomarker of tumor response to therapy
- Author
-
Raccagni, I, MORESCO, ROSA MARIA, RACCAGNI, ISABELLA, Raccagni, I, MORESCO, ROSA MARIA, and RACCAGNI, ISABELLA
- Abstract
Le tecniche di imaging molecolare permettono di visualizzare e caratterizzare processi biologici e rivestono un ruolo fondamentale in oncologia, consentendo di identificare marcatori per la diagnosi e la risposta al trattamento. In questo lavoro di tesi è stato valutato il ruolo della PET come possibile marcatore di risposta al trattamento in a) un modello con k-ras oncogenico e b) un modello di glioma, focalizzando l’attenzione sulle alterazioni del metabolismo e l’ipossia. L’incremento della glicolisi e del consumo di glutammina sono associati a mutazioni dell’oncogene ras in diversi tumori. Il disaccoppiamento di tali processi determina una riprogrammazione del metabolismo per supportare l’aumentata proliferazione fornendo un interessante target terapeutico. Scopo dello studio è la valutazione in vivo delle alterazioni metaboliche e della risposta alla terapia nel modello ottenuto mediante inoculo di fibroblasti con k-ras oncogenico (NIH-RAS). A tale scopo gruppi di topi sono stati monitorati longitudinalmente mediante PET-[18F]FDG e [18F]FLT per la valutazione del metabolismo glucidico e della proliferazione cellulare. I tumori sono stati sottoposti ad analisi immunoistochimiche per confermare i dati ottenuti in vivo. Nello stesso modello è stato valutato l’effetto di un inibitore dell’autofagia (Clorochina) e della glutaminasi (BPTES) singolarmente e in combinazione mediante PET-[18F]FDG e [18F]FLT. Gli animali hanno sviluppato in breve tempo tumori glicolitici e caratterizzati da un’omogenea captazione di [18F]FDG e [18F]FLT. Le immagini PET hanno mostrato un aumento della captazione di [18F]FDG nel tempo e un andamento stabile della proliferazione come mostrato dalla costante captazione di [18F]FLT. Clorochina e BPTES in combinazione hanno determinato un rallentamento della crescita tumorale rispetto ai controlli, ma non sono state osservate variazioni nella captazione di [18F]FDG e [18F]FLT. La presenza di vie alternative per la produzione di glutammato e, Molecular imaging allows the non-invasive visualization and characterization of biological processes. It can be used in oncology to identify biomarkers for the evaluation of tumor progression and response to therapy. In this thesis work, the animal PET was used as potential biomarker of tumor response to therapy focusing on altered metabolism and hypoxia in a) a model of oncogenic k-ras and b) in a model of glioma. Metabolic alterations, such as increased glycolysis and glutamine consumption, are associated with mutations in k-ras gene. The decoupling of glucose and glutamine uptake leads to a reprogramming of their metabolism to support cell proliferation representing a target for cancer therapy. The aim of this study is to investigate metabolic alterations in k-ras transformed fibroblasts (NIH-RAS) in in vivo studies and to assess response to therapy. Animals subcutaneously implanted with NIH-RAS performed [18F]FDG- and [18F]FLT-PET at several time points to evaluate glucose metabolism and cell proliferation, respectively. Tumors were collected and evaluated for different markers by immunohistochemistry (IHC) to confirm in vivo results. In the same model, the efficacy of chloroquine (autophagy blocker) and BPTES (glutaminase inhibitor) alone or in combination was monitored by [18F]FDG- and [18F]FLT-PET before and 48 hours after treatments. All animals developed fast growing and highly glycolytic tumors in few days that appear homogeneous for both [18F]FDG and [18F]FLT uptake. PET imaging showed a significant increase in [18F]FDG uptake while cell proliferation remained stable over time, as depicted by [18F]FLT uptake. IHC analyses confirmed the high aggressiveness of these cells. Chloroquine and BPTES combined treatment slowed down tumor growth only if compared to vehicle, without affecting glucose metabolism or cell proliferation. The presence of alternative pathways for glutamate production and the need of higher doses of treatments may provide explanations to t
- Published
- 2015
17. Characterization of biological features of a rat F98 GBM model: a PET-MRI study with [18F]FAZA and [18F]FDG
- Author
-
Belloli, S, Brioschi, A, Politi, L, Ronchetti, F, Calderoni, S, Raccagni, I, Pagani, A, Monterisi, C, Zenga, F, Zara, G, Fazio, F, Mauro, A, Moresco, R, RACCAGNI, ISABELLA, FAZIO, FERRUCCIO, MORESCO, ROSA MARIA, Belloli, S, Brioschi, A, Politi, L, Ronchetti, F, Calderoni, S, Raccagni, I, Pagani, A, Monterisi, C, Zenga, F, Zara, G, Fazio, F, Mauro, A, Moresco, R, RACCAGNI, ISABELLA, FAZIO, FERRUCCIO, and MORESCO, ROSA MARIA
- Abstract
The prognosis of malignant gliomas remains largely unsatisfactory for the intrinsic characteristics of the pathology and for the delayed diagnosis. Multimodal imaging based on PET and MRI may assess the dynamics of disease onset and progression allowing the validation of preclinical models of glioblastoma multiforme (GBM). The aim of this study was the characterization of a syngeneic rat model of GBM using combined in vivo imaging and immunohistochemistry.
- Published
- 2013
18. Validation of an Engineered Cell Model for In Vitro and In Vivo HIF-1α Evaluation by Different Imaging Modalities
- Author
-
Lo Dico, A., primary, Valtorta, S., additional, Martelli, C., additional, Belloli, S., additional, Gianelli, U., additional, Tosi, D., additional, Bosari, S., additional, Degrassi, A., additional, Russo, M., additional, Raccagni, I., additional, Lucignani, G., additional, Moresco, R. M., additional, and Ottobrini, L., additional
- Published
- 2013
- Full Text
- View/download PDF
19. Disruption of redox homeostasis for combinatorial drug efficacy in K-Ras tumors as revealed by metabolic connectivity profiling
- Author
-
Daniela Gaglio 1, 2, Marcella Bonanomi 2, 3, Silvia Valtorta 1, 4, Rohit Bharat 23, Marilena Ripamonti 1, Federica Conte 2, 5, Giulia Fiscon 2, Nicole Righi 2, Elisabetta Napodano 1, Federico Papa 2, Isabella Raccagni 1, 6, Seth J Parker 7, 8, Ingrid Cifola 9, Tania Camboni 9, Paola Paci 2, Anna Maria Colangelo 2, Marco Vanoni 2, Christian M Metallo 7, Rosa Maria Moresco 1, Lilia Alberghina 2, Gaglio., D, Bonanomi, M, Valtorta, S, Bharat, R, Ripamonti, M, Conte, F, Fiscon, G, Righi, N, Napodano, E, Papa, F, Raccagni, I, J Parker, S, Cifola, I, Camboni, T, Paci, P, Colangelo, A, Vanoni, M, M Metallo, C, Moresco, R, and Alberghina, L
- Subjects
Glutamine ,Metabolic rewiring, metabolic cancer therapy, metabolic signature, glycolysis, glutamine, combinatorial drugs treatment, precision oncology, metabolic connectivity ,Metabolic cancer therapy ,lcsh:RC254-282 ,Nucleic acid metabolism ,chemistry.chemical_compound ,Metabolic connectivity ,Metabolomics ,Metabolic signature ,Combinatorial drug treatment ,Glycolysis ,Metabolic rewiring ,Precision oncology ,Cancer Metabolism ,Chemistry ,Cell growth ,Glutaminase ,Research ,Metabolism ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell biology ,Psychiatry and Mental health ,Metabolic pathway ,Cancer cell ,Growth inhibition - Abstract
Abstract Background Rewiring of metabolism induced by oncogenic K-Ras in cancer cells involves both glucose and glutamine utilization sustaining enhanced, unrestricted growth. The development of effective anti-cancer treatments targeting metabolism may be facilitated by the identification and rational combinatorial targeting of metabolic pathways. Methods We performed mass spectrometric metabolomics analysis in vitro and in vivo experiments to evaluate the efficacy of drugs and identify metabolic connectivity. Results We show that K-Ras-mutant lung and colon cancer cells exhibit a distinct metabolic rewiring, the latter being more dependent on respiration. Combined treatment with the glutaminase inhibitor CB-839 and the PI3K/aldolase inhibitor NVP-BKM120 more consistently reduces cell growth of tumor xenografts. Maximal growth inhibition correlates with the disruption of redox homeostasis, involving loss of reduced glutathione regeneration, redox cofactors, and a decreased connectivity among metabolites primarily involved in nucleic acid metabolism. Conclusions Our findings open the way to develop metabolic connectivity profiling as a tool for a selective strategy of combined drug repositioning in precision oncology.
- Published
- 2020
- Full Text
- View/download PDF
20. Imaging Metformin Efficacy as Add-On Therapy in Cells and Mouse Models of Human EGFR Glioblastoma
- Author
-
Silvia Valtorta, Alessia Lo Dico, Isabella Raccagni, Cristina Martelli, Valentina Pieri, Paolo Rainone, Sergio Todde, Bastian Zinnhardt, Elisabetta De Bernardi, Angela Coliva, Letterio S. Politi, Thomas Viel, Andreas H. Jacobs, Rossella Galli, Luisa Ottobrini, Valentina Vaira, Rosa Maria Moresco, Valtorta, S, Lo Dico, A, Raccagni, I, Martelli, C, Pieri, V, Rainone, P, Todde, S, Zinnhardt, B, De Bernardi, E, Coliva, A, Politi, L, Viel, T, Jacobs, A, Galli, R, Ottobrini, L, Vaira, V, and Moresco, R
- Subjects
Cancer Research ,EGFR - epidermal growth factor receptor ,GBM - glioblastoma multiforme ,PET imaging ,TSPO ,[18F]FLT ,inflammation ,metformin ,In vivo ,medicine ,Translocator protein ,Thymidine kinase 1 ,F]FLT ,Survival rate ,RC254-282 ,Original Research ,Tumor microenvironment ,Temozolomide ,biology ,Cell growth ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Oncology ,Apoptosis ,Cancer research ,biology.protein ,[ ,business ,medicine.drug - Abstract
Glioblastoma (GBM) is a highly aggressive tumor of the brain. Despite the efforts, response to current therapies is poor and 2-years survival rate ranging from 6-12%. Here, we evaluated the preclinical efficacy of Metformin (MET) as add-on therapy to Temozolomide (TMZ) and the ability of [18F]FLT (activity of thymidine kinase 1 related to cell proliferation) and [18F]VC701 (translocator protein, TSPO) Positron Emission Tomography (PET) radiotracers to predict tumor response to therapy. Indeed, TSPO is expressed on the outer mitochondrial membrane of activated microglia/macrophages, tumor cells, astrocytes and endothelial cells. TMZ-sensitive (Gli36ΔEGFR-1 and L0627) or -resistant (Gli36ΔEGFR-2) GBM cell lines representative of classical molecular subtype were tested in vitro and in vivo in orthotopic mouse models. Our results indicate that in vitro, MET increased the efficacy of TMZ on TMZ-sensitive and on TMZ-resistant cells by deregulating the balance between pro-survival (bcl2) and pro-apoptotic (bax/bad) Bcl-family members and promoting early apoptosis in both Gli36ΔEGFR-1 and Gli36ΔEGFR-2 cells. In vivo, MET add-on significantly extended the median survival of tumor-bearing mice compared to TMZ-treated ones and reduced the rate of recurrence in the TMZ-sensitive models. PET studies with the cell proliferation radiopharmaceutical [18F]FLT performed at early time during treatment were able to distinguish responder from non-responder to TMZ but not to predict the duration of the effect. On the contrary, [18F]VC701 uptake was reduced only in mice treated with MET plus TMZ and levels of uptake negatively correlated with animals’ survival. Overall, our data showed that MET addition improved TMZ efficacy in GBM preclinical models representative of classical molecular subtype increasing survival time and reducing tumor relapsing rate. Finally, results from PET imaging suggest that the reduction of cell proliferation represents a common mechanism of TMZ and combined treatment, whereas only the last was able to reduce TSPO. This reduction was associated with the duration of treatment response. TSPO-ligand may be used as a complementary molecular imaging marker to predict tumor microenvironment related treatment effects.
- Published
- 2021
- Full Text
- View/download PDF
21. Metformin and temozolomide, a synergic option to overcome resistance in glioblastoma multiforme models
- Author
-
Sara Belloli, Isabella Raccagni, Rosa Maria Moresco, Silvia Valtorta, Giulia Ercoli, Alessia Lo Dico, Cristina Martelli, Daniela Gaglio, Cecilia Diceglie, Letterio S. Politi, Marcella Bonanomi, Valentina Vaira, Luisa Ottobrini, Valtorta, S, Dico, A, Raccagni, I, Gaglio, D, Belloli, S, Politi, L, Martelli, C, Diceglie, C, Bonanomi, M, Ercoli, G, Vaira, V, Ottobrini, L, and Moresco, R
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Brain tumor ,Glioblastoma multiforme ,03 medical and health sciences ,Glioma ,Temozolomide ,medicine ,CD90 ,business.industry ,Glioma stem cell ,medicine.disease ,Metformin ,Radiation therapy ,Metabolism ,030104 developmental biology ,Oncology ,Apoptosis ,glioma stem cells ,Cancer research ,Stem cell ,business ,Research Paper ,medicine.drug - Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with poor survival. Cytoreduction in association with radiotherapy and temozolomide (TMZ) is the standard therapy, but response is heterogeneous and life expectancy is limited. The combined use of chemotherapeutic agents with drugs targeting cell metabolism is becoming an interesting therapeutic option for cancer treatment. Here, we found that metformin (MET) enhances TMZ effect on TMZ-sensitive cell line (U251) and overcomes TMZ-resistance in T98G GBM cell line. In particular, combined-treatment modulated apoptosis by increasing Bax/Bcl-2 ratio, and reduced Reactive Oxygen Species (ROS) production. We also observed that MET associated with TMZ was able to reduce the expression of glioma stem cells (GSC) marker CD90 particularly in T98G cells but not that of CD133. In vivo experiments showed that combined treatment with TMZ and MET significantly slowed down growth of TMZ-resistant tumors but did not affect overall survival of TMZ-sensitive tumor bearing mice. In conclusion, our results showed that metformin is able to enhance TMZ effect in TMZ-resistant cell line suggesting its potential use in TMZ refractory GBM patients. However, the lack of effect on a GBM malignancy marker like CD133 requires further evaluation since it might influence response duration.
- Published
- 2017
- Full Text
- View/download PDF
22. Tumour hypoxia: lessons learnt from preclinical imaging
- Author
-
Silvia Valtorta, Isabella Raccagni, Rosa Maria Moresco, Sara Belloli, Raccagni, I, Valtorta, S, Moresco, R, and Belloli, S
- Subjects
Oncology ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,PET imaging ,Hypoxia-specific radiopharmaceutical ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Preclinical research ,Hypoxia-specific radiopharmaceuticals ,0302 clinical medicine ,Internal medicine ,medicine ,Radiology, Nuclear Medicine and imaging ,Treatment outcome ,Treatment resistance ,Hypoxia ,business.industry ,Pet imaging ,Hypoxia (medical) ,Tumour oxygenation ,Preclinical ,Radiation therapy ,030220 oncology & carcinogenesis ,Tracer uptake ,medicine.symptom ,business ,Preclinical imaging - Abstract
Purpose: In tumour, the imbalance between oxygen supply and demand leads to hypoxia, which represents a negative prognostic factor associated with aggressive tumour phenotype and therapy resistance. This review provides an overview of the use of positron emitter-labelled radiopharmaceutical used to image hypoxia in preclinical models of cancer. Methods: A critical and comprehensive PubMed search was performed identifying articles related to PET imaging for hypoxia assessment in preclinical setting from January 2007 up to January 2017. Results: We have considered and described a total of 54 original articles, exploring tumour-associated hypoxia in preclinical models. Results underlined the potential application together with the advantages and pitfalls of the use of PET in preclinical research. Multi-target imaging allowed to better define the relationship between hypoxia and other biological hallmarks of tumour; imaging of hypoxia was proved as a useful tool for lesions stratification and response prediction to radiotherapy; however, cutoff indexes were identified in few studies. Hypoxia PET showed remarkable tracer delivery limitations in the study of vascular disrupting agents but suggested the potential use of PET as a marker of response or resistance to anti-angiogenics. Finally, the effect of anaesthesia on tracer kinetics and tumour oxygenation as well as perfusion dependency in tracer uptake should be carefully evaluated to avoid artefactual results. Conclusions: Preclinical studies highlight the advantages and the limitations of the available hypoxia-radiotracers and their potential usefulness for the evaluation of treatments outcome and radiotherapy planning.
- Published
- 2017
- Full Text
- View/download PDF
23. Metabolic Evaluation of Non–Small Cell Lung Cancer Patient–Derived Xenograft Models Using 18F-FDG PET: A Potential Tool for Early Therapy Response
- Author
-
Silvia Valtorta* 1, 2, Massimo Moro* 3, Giovanna Prisinzano 1, 4, Giulia Bertolini 3, Monica Tortoreto 5, Isabella Raccagni 2, Ugo Pastorino 6, Luca Roz 3, Gabriella Sozzi +3, Rosa Maria Moresco +2, 4 *Contributed equally to this work. +Contributed equally to this work., Valtorta, S, Moro, M, Prisinzano, G, Bertolini, G, Tortoreto, M, Raccagni, I, Pastorino, U, Roz, L, Sozzi, G, and Moresco, R
- Subjects
Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Mice, Nude ,Animal Imaging ,Oncology: Lung ,PET ,[18F]FDG PET ,lung cancer ,patient-derived xenograft ,stem cells ,Mice, SCID ,18F-FDG PET, lung cancer, patient-derived xenograft, stem cells ,Early Therapy ,Sensitivity and Specificity ,18f fdg pet ,03 medical and health sciences ,18F-FDG PET ,0302 clinical medicine ,Fluorodeoxyglucose F18 ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Internal medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Therapy efficacy ,Lung cancer ,Tumor xenograft ,business.industry ,Reproducibility of Results ,Pet imaging ,medicine.disease ,Molecular Imaging ,Glucose ,Outcome and Process Assessment, Health Care ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Non small cell ,Radiopharmaceuticals ,Stem cell ,business - Abstract
PURPOSE: Lung cancer heterogeneity makes response to therapy extremely hard to predict. Patient-derived xenografts (PDXs) represent a reliable preclinical model that closely recapitulates the main characteristics of the primary tumor and could represent a useful asset to test new therapies. Here, using PET imaging, we verify how lung cancer PDXs reproduce the metabolic features of the corresponding primary tumors. METHODS: We performed longitudinal [18F]FDG-PET studies on nine different PDXs, obtained by implants of primary cancer fragments harvested from patients. Max [18F]FDG uptake values of the lesion for each group were calculated and compared to corresponding patient's uptake. RESULTS: Different PDXs showed variable tumor growth rate and [18F]FDG uptake confirming the preservation of individual characteristics. A good intra-group reproducibility of PET measurements was observed. Furthermore, the subgroup of PDXs originating from primary tumors with higher metabolic rate displayed a rank order of [18F]FDG uptake similar to that of patients' original SUV. CONCLUSION: PDXs reproduced the original glucose metabolism of primary lesions and represent therefore a promising preclinical model also for the early assessment of therapy efficacy.
- Published
- 2016
- Full Text
- View/download PDF
24. [18F]FDG and [18F]FLT PET for the evaluation of response to neo-adjuvant chemotherapy in a model of triple negative breast cancer
- Author
-
Luca Presotto, Isabella Raccagni, Giorgio Ivan Russo, Nadia Zaffaroni, Rosa Maria Moresco, Maria Grazia Daidone, Sara Belloli, Emilio Bombardieri, Anna Bogni, Claudio Pascali, Monica Tortoreto, Silvia Valtorta, Alessandro Di Stefano, Raccagni, I, Belloli, S, Valtorta, S, Stefano, A, Presotto, L, Pascali, C, Bogni, A, Tortoreto, M, Zaffaroni, N, Daidone, M, Russo, G, Bombardieri, E, and Moresco, R
- Subjects
Metabolic Processes ,Oncology ,medicine.medical_treatment ,Cancer Treatment ,lcsh:Medicine ,Triple Negative Breast Neoplasms ,Mice, SCID ,Pathology and Laboratory Medicine ,Biochemistry ,Diagnostic Radiology ,030218 nuclear medicine & medical imaging ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Breast Tumors ,Medicine and Health Sciences ,lcsh:Science ,Tomography ,Neoadjuvant therapy ,Triple-negative breast cancer ,Multidisciplinary ,TNBC, [18F]FDG, [18F]FLT, PET, treatment response ,medicine.diagnostic_test ,Pharmaceutics ,Radiology and Imaging ,Neoadjuvant Therapy ,Paclitaxel ,Positron emission tomography ,Response Evaluation Criteria in Solid Tumors ,Area Under Curve ,030220 oncology & carcinogenesis ,Female ,Anatomy ,Glycolysis ,Research Article ,Clinical Oncology ,medicine.medical_specialty ,Histology ,Imaging Techniques ,Transplantation, Heterologous ,Mice, Nude ,Neuroimaging ,Research and Analysis Methods ,Cancer Chemotherapy ,03 medical and health sciences ,Signs and Symptoms ,Text mining ,Drug Therapy ,Fluorodeoxyglucose F18 ,Diagnostic Medicine ,Cell Line, Tumor ,Internal medicine ,Breast Cancer ,medicine ,Animals ,Humans ,Chemotherapy ,business.industry ,lcsh:R ,Cancers and Neoplasms ,Biology and Life Sciences ,Antineoplastic Agents, Phytogenic ,Transplantation ,Ki-67 Antigen ,Metabolism ,ROC Curve ,chemistry ,Positron-Emission Tomography ,Lesions ,lcsh:Q ,Radiopharmaceuticals ,Clinical Medicine ,business ,Positron Emission Tomography ,Biomarkers ,Thymidine ,Neuroscience - Abstract
Rationale Pathological response to neo-adjuvant chemotherapy (NAC) represents a commonly used predictor of survival in triple negative breast cancer (TNBC) and the need to identify markers that predict response to NAC is constantly increasing. Aim of this study was to evaluate the potential usefulness of PET imaging with [18F]FDG and [18F]FLT for the discrimination of TNBC responders to Paclitaxel (PTX) therapy compared to the response assessed by an adapted Response Evaluation Criteria In Solid Tumors (RECIST) criteria based on tumor volume (Tumor Volume Response). Methods Nu/nu mice bearing TNBC lesions of different size were evaluated with [18F]FDG and [18F]FLT PET before and after PTX treatment. SUVmax, Metabolic Tumor Volume (MTV) and Total Lesion Glycolysis (TLG) and Proliferation (TLP) were assessed using a graph-based random walk algorithm. Results We found that in our TNBC model the variation of [18F]FDG and [18F]FLT SUVmax similarly defined tumor response to therapy and that SUVmax variation represented the most accurate parameter. Response evaluation using Tumor Volume Response (TVR) showed that the effectiveness of NAC with PTX was completely independent from lesions size at baseline. Conclusions Our study provided interesting results in terms of sensitivity and specificity of PET in TNBC, revealing the similar performances of [18F]FDG and [18F]FLT in the identification of responders to Paclitaxel.
- Published
- 2018
- Full Text
- View/download PDF
25. Inhibition of the Hexosamine Biosynthetic Pathway by targeting PGM3 causes breast cancer growth arrest and apoptosis
- Author
-
Giuseppina Votta, Isabella Raccagni, Ferdinando Chiaradonna, Luca De Gioia, Francesca Ricciardiello, Alice Paiotta, Silvia Valtorta, Laura Brunelli, Giuseppe D'Orazio, Rosa Maria Moresco, Barbara La Ferla, Roberta Palorini, Francesca Tinelli, Roberta Pastorelli, Ricciardiello, F, Votta, G, Palorini, R, Raccagni, I, Brunelli, L, Paiotta, A, Tinelli, F, D’Orazio, G, Valtorta, S, De Gioia, L, Pastorelli, R, Moresco, R, La Ferla, B, and Chiaradonna, F
- Subjects
0301 basic medicine ,Cancer Research ,Immunology ,Triple Negative Breast cancer, Glycosylation, Therapy, Drug Discovery ,Apoptosis ,Triple Negative Breast Neoplasms ,Hexosamine Biosynthetic Pathway ,Article ,Cell Line ,Animals ,Cell Line, Tumor ,Cell Proliferation ,Enzyme Inhibitors ,Female ,Gene Expression Regulation, Neoplastic ,Hexosamines ,Humans ,MCF-7 Cells ,Mice ,Phosphoglucomutase ,Signal Transduction ,Xenograft Model Antitumor Assays ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Breast cancer ,breast cancer ,Settore BIO/10 - Biochimica ,medicine ,lcsh:QH573-671 ,Triple-negative breast cancer ,Neoplastic ,Tumor ,lcsh:Cytology ,Chemistry ,Cancer ,Settore CHIM/06 - Chimica Organica ,Cell Biology ,medicine.disease ,BIO/10 - BIOCHIMICA ,030104 developmental biology ,Gene Expression Regulation ,Cell culture ,Tumor progression ,Cancer cell ,Cancer research ,Unfolded protein response - Abstract
Cancer aberrant N- and O-linked protein glycosylation, frequently resulting from an augmented flux through the Hexosamine Biosynthetic Pathway (HBP), play different roles in tumor progression. However, the low specificity and toxicity of the existing HBP inhibitors prevented their use for cancer treatment. Here we report the preclinical evaluation of FR054, a novel inhibitor of the HBP enzyme PGM3, with a remarkable anti-breast cancer effect. In fact, FR054 induces in different breast cancer cells a dramatic decrease in cell proliferation and survival. In particular, in a model of Triple Negative Breast Cancer (TNBC) cells, MDA-MB-231, we show that these effects are correlated to FR054-dependent reduction of both N- and O-glycosylation level that cause also a strong reduction of cancer cell adhesion and migration. Moreover we show that impaired survival of cancer cells upon FR054 treatment is associated with the activation of the Unfolded Protein Response (UPR) and accumulation of intracellular ROS. Finally, we show that FR054 suppresses cancer growth in MDA-MB-231 xenograft mice, supporting the advantage of targeting HBP for therapeutic purpose and encouraging further investigation about the use of this small molecule as a promising compound for breast cancer therapy.
- Published
- 2018
- Full Text
- View/download PDF
26. Leukocytes recruited by tumor-derived HMGB1 sustain peritoneal carcinomatosis
- Author
-
Cottone, Lucia, Capobianco, Annalisa, Gualteroni, Chiara, Monno, Antonella, Raccagni, Isabella, Valtorta, Silvia, Canu, Tamara, Di Tomaso, Tiziano, Lombardo, Angelo, Esposito, Antonio, Moresco, Rosa Maria, Del Maschio, Alessandro, Naldini, Luigi, Rovere-Querini, Patrizia, Bianchi, Marco E., Manfredi, Angelo A., Cottone, L, Capobianco, A, Gualteroni, C, Monno, A, Raccagni, I, Valtorta, S, Canu, T, Tomaso, T, Lombardo, A, Esposito, A, Moresco, R, Maschio, A, Naldini, L, Rovere Querini, P, Bianchi, M, Manfredi, A, De Tomaso, T, Lombardo, ANGELO LEONE, Esposito, Antonio, Moresco, Rm, DEL MASCHIO, Alessandro, Naldini, Luigi, ROVERE QUERINI, Patrizia, Bianchi, MARCO EMILIO, and Manfredi, ANGELO ANDREA M. A.
- Subjects
0301 basic medicine ,green fluorescent protein ,Pathology ,medicine.medical_specialty ,Murine colon adenocarcinoma cell line ,Phagocyte ,Macrophage ,Immunology ,Damage-associated molecular pattern ,chemical and pharmacologic phenomena ,High Mobility Box 1 ,macrophage ,HMGB1 ,GFP ,BoxA ,damage-associated molecular pattern ,Small hairpin RNA ,03 medical and health sciences ,0302 clinical medicine ,vascularization ,In vivo ,Leukocytes ,medicine ,Extracellular ,DAMP ,short hairpin RNA ,Immunology and Allergy ,MC-38 ,murine colon adenocarcinoma cell line ,Original Research ,Gene knockdown ,peritoneal carcinomatosi ,biology ,business.industry ,Macrophages ,Tumor-Derived ,Leukocyte ,030104 developmental biology ,medicine.anatomical_structure ,Alarmin ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,ShRNA ,business ,leukocyte ,Peritoneal carcinomatosis - Abstract
The factors that determine whether disseminated transformed cells in vivo yield neoplastic lesions have only been partially identified. We established an ad hoc model of peritoneal carcinomatosis by injecting colon carcinoma cells in mice. Tumor cells recruit inflammatory leukocytes, mostly macrophages, and generate neoplastic peritoneal lesions. Phagocyte depletion via clodronate treatment reduces neoplastic growth. Colon carcinoma cells release a prototypic damage-associated molecular pattern (DAMP)/alarmin, High Mobility Group Box1 (HMGB1), which attracts leukocytes. Exogenous HMGB1 accelerates leukocyte recruitment, macrophage infiltration, tumor growth and vascularization. Lentiviral-based HMGB1 knockdown or pharmacological interference with its extracellular impair macrophage recruitment and tumor growth. Our findings provide a preclinical proof of principle that strategies based on preventing HMGB1-driven recruitment of leukocytes could be used for treating peritoneal carcinomatosis.
- Published
- 2016
- Full Text
- View/download PDF
27. PET imaging as a biomarker of tumor response to therapy
- Author
-
RACCAGNI, ISABELLA, Raccagni, I, and MORESCO, ROSA MARIA
- Subjects
Tumor metabolism, PET imaging, glutamine, therapy monitoring, glioma, hypoxia ,BIO/11 - BIOLOGIA MOLECOLARE - Abstract
Le tecniche di imaging molecolare permettono di visualizzare e caratterizzare processi biologici e rivestono un ruolo fondamentale in oncologia, consentendo di identificare marcatori per la diagnosi e la risposta al trattamento. In questo lavoro di tesi è stato valutato il ruolo della PET come possibile marcatore di risposta al trattamento in a) un modello con k-ras oncogenico e b) un modello di glioma, focalizzando l’attenzione sulle alterazioni del metabolismo e l’ipossia. L’incremento della glicolisi e del consumo di glutammina sono associati a mutazioni dell’oncogene ras in diversi tumori. Il disaccoppiamento di tali processi determina una riprogrammazione del metabolismo per supportare l’aumentata proliferazione fornendo un interessante target terapeutico. Scopo dello studio è la valutazione in vivo delle alterazioni metaboliche e della risposta alla terapia nel modello ottenuto mediante inoculo di fibroblasti con k-ras oncogenico (NIH-RAS). A tale scopo gruppi di topi sono stati monitorati longitudinalmente mediante PET-[18F]FDG e [18F]FLT per la valutazione del metabolismo glucidico e della proliferazione cellulare. I tumori sono stati sottoposti ad analisi immunoistochimiche per confermare i dati ottenuti in vivo. Nello stesso modello è stato valutato l’effetto di un inibitore dell’autofagia (Clorochina) e della glutaminasi (BPTES) singolarmente e in combinazione mediante PET-[18F]FDG e [18F]FLT. Gli animali hanno sviluppato in breve tempo tumori glicolitici e caratterizzati da un’omogenea captazione di [18F]FDG e [18F]FLT. Le immagini PET hanno mostrato un aumento della captazione di [18F]FDG nel tempo e un andamento stabile della proliferazione come mostrato dalla costante captazione di [18F]FLT. Clorochina e BPTES in combinazione hanno determinato un rallentamento della crescita tumorale rispetto ai controlli, ma non sono state osservate variazioni nella captazione di [18F]FDG e [18F]FLT. La presenza di vie alternative per la produzione di glutammato e la necessità di dosi più elevate potrebbero spiegare l’assenza di efficacia di questi trattamenti. L’ipossia rappresenta un fenomeno sfavorevole per la progressione tumorale. L’espressione di HIF1α, principale regolatore dell’ipossia, è associata alla resistenza alla terapia in molti tumori, compreso il glioma. Per questo, una migliore comprensione della modulazione dell’attività di HIF1α nel processo di risposta alla terapia è di particolare interesse. Cellule di glioma U251-HRE-mCherry in grado di esprimere l’enzima luciferasi sotto il controllo di HRE (Hypoxia Responsive Element) e mCherry sotto controllo di un promotore costitutivo sono state utilizzate per valutare la modulazione di HIF1α in seguito a trattamento con Temozolomide (TMZ) in vitro e in vivo. La crescita tumorale è stata monitorata in vivo in animali sottoposti ad inoculo intracerebrale tramite bioluminescenza, fluorescenza, RM e PET con [18F]FAZA e [18F]FLT. In seguito, è stato valutato in vivo l’effetto di due diversi regimi di TMZ. Mediante bioluminescenza è stato possibile monitorare la crescita tumorale e identificare aree ipossiche. I dati ottenuti sono stati confermati dalle immagini di fluorescenza e PET-[18F]FAZA. Le analisi ex vivo per Ki67 hanno invece confermato i dati PET-[18F]FLT ed hanno mostrato un’elevata proliferazione cellulare. Entrambi i dosaggi di TMZ hanno determinato una diminuzione dell’attività di HIF1α a tempi precoci. Al contrario, il segnale di fluorescenza e la captazione di [18F]FLT hanno subìto una diminuzione solo a tempi più tardivi. L’attività di HIF1α può essere considerata un marcatore di risposta al TMZ e questo modello un utile strumento per la valutazione in vivo di farmaci per il trattamento del glioma. Molecular imaging allows the non-invasive visualization and characterization of biological processes. It can be used in oncology to identify biomarkers for the evaluation of tumor progression and response to therapy. In this thesis work, the animal PET was used as potential biomarker of tumor response to therapy focusing on altered metabolism and hypoxia in a) a model of oncogenic k-ras and b) in a model of glioma. Metabolic alterations, such as increased glycolysis and glutamine consumption, are associated with mutations in k-ras gene. The decoupling of glucose and glutamine uptake leads to a reprogramming of their metabolism to support cell proliferation representing a target for cancer therapy. The aim of this study is to investigate metabolic alterations in k-ras transformed fibroblasts (NIH-RAS) in in vivo studies and to assess response to therapy. Animals subcutaneously implanted with NIH-RAS performed [18F]FDG- and [18F]FLT-PET at several time points to evaluate glucose metabolism and cell proliferation, respectively. Tumors were collected and evaluated for different markers by immunohistochemistry (IHC) to confirm in vivo results. In the same model, the efficacy of chloroquine (autophagy blocker) and BPTES (glutaminase inhibitor) alone or in combination was monitored by [18F]FDG- and [18F]FLT-PET before and 48 hours after treatments. All animals developed fast growing and highly glycolytic tumors in few days that appear homogeneous for both [18F]FDG and [18F]FLT uptake. PET imaging showed a significant increase in [18F]FDG uptake while cell proliferation remained stable over time, as depicted by [18F]FLT uptake. IHC analyses confirmed the high aggressiveness of these cells. Chloroquine and BPTES combined treatment slowed down tumor growth only if compared to vehicle, without affecting glucose metabolism or cell proliferation. The presence of alternative pathways for glutamate production and the need of higher doses of treatments may provide explanations to the lack of treatments’ efficacy. Hypoxia is implicated in many aspects of tumor progression and it is involved in the intracellular stabilization of the hypoxia regulator gene HIF-1α. Since the expression of HIF-1α is associated with poor prognosis and therapy resistance in glioblastoma, a better comprehension of its involvement in tumor response to treatment can be of great interest for clinical translation. U251-HRE-mCherry cells expressing Luciferase under control of a Hypoxia Responsive Element (HRE) and mCherry under the control of a constitutive promoter have been used to assess HIF-1α modulation and cell survival after treatment, both in vitro and in vivo. In vivo analyses characterized the model obtained by stereotaxic injection of glioma U251-HRE cells in mice brain. Tumor progression was monitored comparing bioluminescence, fluorescence and PET with [18F]FAZA and [18F]FLT. Afterwards, two regimens of temozolomide (TMZ) were administered starting 21 days after cells injection. TMZ efficacy was monitored by optical and fluorescence imaging, [18F]FLT-PET and MRI. Bioluminescent signals provided information about tumor growth and hypoxia presence, confirmed by both fluorescence acquisition and [18F]FAZA PET. IHC for Ki67 confirmed data obtained by [18F]FLT-PET, showing a high rate of cell proliferation. Both TMZ regimens showed a decrease of HIF-1α-dependent Luciferase activity at early time after TMZ administration. On the contrary, mCherry fluorescence, such as [18F]FLT uptake, decreased only at the end of treatments. HIF-1α activity reduction can be considered a biomarker of tumour response to TMZ and the U251-HRE-mCherry cell model a feasible tool to evaluate HIF-1α activity and treatment effects in in vivo studies.
- Published
- 2015
28. Identification of imaging biomarkers for the assessment of tumour response to different treatments in a preclinical glioma model
- Author
-
A. Lo Dico 1, 2, C. Martelli 1, 2. S. Valtorta 3, 4, I. Raccagni 4, 5, C. Diceglie 1, 6, S. Belloli 3, U. Gianelli1 7, V. Vaira 7, 8, L. S. Politi 9, S. Bosari 1, 7, G. Lucignani 2, R.M. Moresco 4, L. Ottobrini 1, 3, Lo Dico, A, Martelli, C, Valtorta, S, Raccagni, I, Diceglie, C, Belloli, S, Gianelli, U, Vaira, V, Politi, L, Bosari, S, Lucignani, G, Moresco, R, and Ottobrini, L
- Subjects
Pathology ,medicine.medical_specialty ,Drug Evaluation, Preclinical ,Mice, Nude ,Antineoplastic Agents ,MED/50 - SCIENZE TECNICHE MEDICHE APPLICATE ,Hsp90 inhibitor ,Mice ,Transduction (genetics) ,In vivo ,Cell Line, Tumor ,Glioma ,Biomarkers, Tumor ,Temozolomide ,Animals ,Humans ,Medicine ,Radiology, Nuclear Medicine and imaging ,Luciferase ,Carbonic Anhydrases ,MED/36 - DIAGNOSTICA PER IMMAGINI E RADIOTERAPIA ,Brain Neoplasms ,business.industry ,HIF-1alpha ,Optical Imaging ,CAIX ,Biomarker ,General Medicine ,Hypoxia-Inducible Factor 1, alpha Subunit ,medicine.disease ,Dacarbazine ,Luminescent Proteins ,Cancer research ,Biomarker (medicine) ,Female ,preclinical glioma model ,business ,mCherry ,medicine.drug - Abstract
Purpose: Hypoxia-inducible factor 1α (HIF-1α) activity is one of the major players in hypoxia-mediated glioma progression and resistance to therapies, and therefore the focus of this study was the evaluation of HIF-1α modulation in relation to tumour response with the purpose of identifying imaging biomarkers able to document tumour response to treatment in a murine glioma model. Methods: U251-HRE-mCherry cells expressing Luciferase under the control of a hypoxia responsive element (HRE) and mCherry under the control of a constitutive promoter were used to assess HIF-1α activity and cell survival after treatment, both in vitro and in vivo, by optical, MRI and positron emission tomography imaging. Results: This cell model can be used to monitor HIF-1α activity after treatment with different drugs modulating transduction pathways involved in its regulation. After temozolomide (TMZ) treatment, HIF-1α activity is early reduced, preceding cell cytotoxicity. Optical imaging allowed monitoring of this process in vivo, and carbonic anhydrase IX (CAIX) expression was identified as a translatable non-invasive biomarker with potential clinical significance. A preliminary in vitro evaluation showed that reduction of HIF-1α activity after TMZ treatment was comparable to the effect of an Hsp90 inhibitor, opening the way for further elucidation of its mechanism of action. Conclusion: The results of this study suggest that the U251-HRE-mCherry cell model can be used for the monitoring of HIF-1α activity through luciferase and CAIX expression. These cells can become a useful tool for the assessment and improvement of new targeted tracers for potential theranostic procedures.
- Published
- 2015
- Full Text
- View/download PDF
29. Validation of an engineered cell model for in vitro and in vivo HIF-1α evaluation by different imaging modalities
- Author
-
Silvano Bosari, Silvia Valtorta, Isabella Raccagni, Delfina Tosi, Cristina Martelli, Luisa Ottobrini, Giovanni Lucignani, Umberto Gianelli, M. Russo, Rosa Maria Moresco, Sara Belloli, Anna Degrassi, A. Lo Dico, Lo Dico, A, Valtorta, S, Martelli, C, Belloli, S, Gianelli, U, Tosi, D, Bosari, S, Degrassi, A, Russo, M, Raccagni, I, Lucignani, G, Moresco, R, and Ottobrini, L
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Fluorescence-lifetime imaging microscopy ,Tumor model ,Biology ,Deferoxamine ,Models, Biological ,Multimodal Imaging ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Glioma ,Cell Line, Tumor ,medicine ,Bioluminescence imaging ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Luciferase ,Non-invasive molecular imaging ,Hypoxia ,Luciferases ,Cell Shape ,Glioma, Hypoxia, Non-invasive molecular imaging, Reporter gene, Optical imaging, PET, MRI, Tumor model ,030304 developmental biology ,0303 health sciences ,medicine.diagnostic_test ,Brain Neoplasms ,Optical Imaging ,Magnetic resonance imaging ,medicine.disease ,Hypoxia-Inducible Factor 1, alpha Subunit ,Immunohistochemistry ,Magnetic Resonance Imaging ,Xenograft Model Antitumor Assays ,Reporter gene ,Cell Hypoxia ,PET ,Oncology ,Positron emission tomography ,Tumor progression ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,Cancer research ,MRI ,Signal Transduction - Abstract
PURPOSE: The aim of this study was to characterize a cell-based model for the molecular study of hypoxia-inducible factor (HIF)-1? activity, in the context of hypoxia, by means of different imaging techniques. PROCEDURES: Engineered U251-HRE glioma cells were used to analyze the molecular mechanisms underlying HIF-1? activity in vitro in relation to luciferase expression. The same cells were orthotopically implanted in mice to evaluate tumor progression and hypoxia induction by bioluminescence imaging, fluorescence imaging, positron emission tomography (PET), and magnetic resonance imaging (MRI). RESULTS: In vitro analyses highlighted the relationship between HIF-1? and luciferase activity in hypoxic conditions and after pharmacological treatments in U251-HRE cells. Through in vivo studies, it was possible to assess hypoxia establishment in relation to tumor growth by optical imaging, PET and MRI. CONCLUSIONS: The findings of this study indicate that the U251-HRE orthotopic murine model can be used to reliably evaluate processes modulating HIF-1? activity, using both molecular and preclinical non-invasive imaging techniques.
- Published
- 2013
- Full Text
- View/download PDF
30. Characterization of biological features of a rat F98 GBM model: a PET-MRI study with [18F]FAZA and [18F]FDG
- Author
-
Sara Belloli a, b, c, Andrea Brioschi d, Letterio Salvatore Politi e, Francesca Ronchetti d, Sara Calderoni d, Isabella Raccagni b, Antonella Pagani e, Cristina Monterisi a, Francesco Zenga f, g, Gianpaolo Zara h, Ferruccio Fazio a, Alessandro Mauro d, f, Rosa Maria Moresco a, Belloli, S, Brioschi, A, Politi, L, Ronchetti, F, Calderoni, S, Raccagni, I, Pagani, A, Monterisi, C, Zenga, F, Zara, G, Fazio, F, Mauro, A, and Moresco, R
- Subjects
Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,GBM model, Glucose metabolism, Hypoxia, Immunohistochemistry, MRI, PET ,Fluorodeoxyglucose F18 ,In vivo ,Cell Line, Tumor ,Glioma ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Hypoxia ,Glucose metabolism ,medicine.diagnostic_test ,business.industry ,Cell Differentiation ,Magnetic resonance imaging ,medicine.disease ,Magnetic Resonance Imaging ,Survival Analysis ,Immunohistochemistry ,Rats ,PET - MRI ,Radiation therapy ,Disease Models, Animal ,PET ,Nitroimidazoles ,Tumor progression ,Positron emission tomography ,Positron-Emission Tomography ,Molecular Medicine ,GBM model ,Glioblastoma ,business ,Preclinical imaging ,MRI - Abstract
Introduction The prognosis of malignant gliomas remains largely unsatisfactory for the intrinsic characteristics of the pathology and for the delayed diagnosis. Multimodal imaging based on PET and MRI may assess the dynamics of disease onset and progression allowing the validation of preclinical models of glioblastoma multiforme (GBM). The aim of this study was the characterization of a syngeneic rat model of GBM using combined in vivo imaging and immunohistochemistry. Methods Four groups of Fischer rats were implanted in a subcortical region with increasing concentration of rat glioma F98 cells and weekly monitored with Gd-MR, [ 18 F]FDG- and [ 18 F]FAZA-PET starting one week after surgery. Different targets were evaluated on post mortem brain specimens using immunohistochemistry: VEGF, GFAP, HIF-1α, Ki-67 and nestin. Results Imaging results indicated that tumor onset but not progression was related to the number of F98 cells. Hypoxic regions identified with [ 18 F]FAZA and high-glucose metabolism regions recognized with [ 18 F]FDG were located respectively in the core and in external areas of the tumor, with partial overlap and remodeling during disease progression. Histological and immunohistochemical analysis confirmed PET/MRI results and revealed that our model resumes biological characteristics of human GBM. IHC and PET studies showed that necrotic regions, defined on the basis of [ 18 F]FDG uptake reduction, may include hypoxic clusters of vital tumor tissue identified with [ 18 F]FAZA. This last information is particularly relevant for the identification of the target volume during image- guided radiotherapy . Conclusions In conclusion, the combined use of PET and MRI allows in vivo monitoring of the biological modification of F98 lesions during tumor progression.
- Published
- 2013
- Full Text
- View/download PDF
31. Pre- and post-treatment in flexor tendon tenolysis: An observational study.
- Author
-
Seppi S, Vecchi S, Raccagni I, Novelli C, and Pajardi GE
- Subjects
- Humans, Female, Male, Retrospective Studies, Adult, Middle Aged, Pain Measurement, Visual Analog Scale, Aged, Finger Injuries surgery, Finger Injuries rehabilitation, Finger Injuries physiopathology, Tendon Injuries surgery, Tendon Injuries rehabilitation, Range of Motion, Articular physiology
- Abstract
Background: Glide deficit of the distal flexors' tendons following primary repair in zone 1-3 are very common. Adhesions of tendons have multi factorial origins and are closely related to the healing of the affected tissues. The surgical practice used to resolve these complications is tenolysis., Purpose: The purpose of this study was to identify and compare the Visual Analog Scale (VAS) relate to pain and Total Active Motion (TAM) of adult patients of both sexes undergoing tenolysis surgery. The results will then be compared to existing research to confirm their significance., Study Design: Case-series., Methods: Retrospective data for TAM and pain VAS were extracted from the medical records for 63 patients (73 fingers) who underwent flexor tenolysis between 2017 and 2019. Data were compared pre-operatively and 3 months after surgery. All patients underwent pre- and post-surgery therapy by hand therapists., Results: The sample presented very encouraging improvements, except in the VAS and active range of motion (AROM) of thumb where some patients maintained the same assessment. The fingers reported statistically significant results, whereas the thumb group did not meet significant criteria. Overall, TAM improved from 134.6° to 196.7 and VAS decreased from 2.7 to 1.2., Discussion: According to the results and the data change between pre- and post-treatment, the sample demonstrated improvements in all areas examined, reporting statistically significant results for the fingers with an improvement of TAM of 62.1° with a percentage value (%TAM) of 75.6%., Conclusions: A specific treatment for this type of surgery is required for the patients so they can return to their daily and working activities. This article can be used as a starting point for further studies., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
32. Imaging Metformin Efficacy as Add-On Therapy in Cells and Mouse Models of Human EGFR Glioblastoma.
- Author
-
Valtorta S, Lo Dico A, Raccagni I, Martelli C, Pieri V, Rainone P, Todde S, Zinnhardt B, De Bernardi E, Coliva A, Politi LS, Viel T, Jacobs AH, Galli R, Ottobrini L, Vaira V, and Moresco RM
- Abstract
Glioblastoma (GBM) is a highly aggressive tumor of the brain. Despite the efforts, response to current therapies is poor and 2-years survival rate ranging from 6-12%. Here, we evaluated the preclinical efficacy of Metformin (MET) as add-on therapy to Temozolomide (TMZ) and the ability of [
18 F]FLT (activity of thymidine kinase 1 related to cell proliferation) and [18 F]VC701 (translocator protein, TSPO) Positron Emission Tomography (PET) radiotracers to predict tumor response to therapy. Indeed, TSPO is expressed on the outer mitochondrial membrane of activated microglia/macrophages, tumor cells, astrocytes and endothelial cells. TMZ-sensitive (Gli36ΔEGFR-1 and L0627) or -resistant (Gli36ΔEGFR-2) GBM cell lines representative of classical molecular subtype were tested in vitro and in vivo in orthotopic mouse models. Our results indicate that in vitro , MET increased the efficacy of TMZ on TMZ-sensitive and on TMZ-resistant cells by deregulating the balance between pro-survival ( bcl2 ) and pro-apoptotic ( bax/bad ) Bcl-family members and promoting early apoptosis in both Gli36ΔEGFR-1 and Gli36ΔEGFR-2 cells. In vivo , MET add-on significantly extended the median survival of tumor-bearing mice compared to TMZ-treated ones and reduced the rate of recurrence in the TMZ-sensitive models. PET studies with the cell proliferation radiopharmaceutical [18 F]FLT performed at early time during treatment were able to distinguish responder from non-responder to TMZ but not to predict the duration of the effect. On the contrary, [18 F]VC701 uptake was reduced only in mice treated with MET plus TMZ and levels of uptake negatively correlated with animals' survival. Overall, our data showed that MET addition improved TMZ efficacy in GBM preclinical models representative of classical molecular subtype increasing survival time and reducing tumor relapsing rate. Finally, results from PET imaging suggest that the reduction of cell proliferation represents a common mechanism of TMZ and combined treatment, whereas only the last was able to reduce TSPO. This reduction was associated with the duration of treatment response. TSPO-ligand may be used as a complementary molecular imaging marker to predict tumor microenvironment related treatment effects., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Valtorta, Lo Dico, Raccagni, Martelli, Pieri, Rainone, Todde, Zinnhardt, De Bernardi, Coliva, Politi, Viel, Jacobs, Galli, Ottobrini, Vaira and Moresco.)- Published
- 2021
- Full Text
- View/download PDF
33. Disruption of redox homeostasis for combinatorial drug efficacy in K-Ras tumors as revealed by metabolic connectivity profiling.
- Author
-
Gaglio D, Bonanomi M, Valtorta S, Bharat R, Ripamonti M, Conte F, Fiscon G, Righi N, Napodano E, Papa F, Raccagni I, Parker SJ, Cifola I, Camboni T, Paci P, Colangelo AM, Vanoni M, Metallo CM, Moresco RM, and Alberghina L
- Abstract
Background: Rewiring of metabolism induced by oncogenic K-Ras in cancer cells involves both glucose and glutamine utilization sustaining enhanced, unrestricted growth. The development of effective anti-cancer treatments targeting metabolism may be facilitated by the identification and rational combinatorial targeting of metabolic pathways., Methods: We performed mass spectrometric metabolomics analysis in vitro and in vivo experiments to evaluate the efficacy of drugs and identify metabolic connectivity., Results: We show that K-Ras -mutant lung and colon cancer cells exhibit a distinct metabolic rewiring, the latter being more dependent on respiration. Combined treatment with the glutaminase inhibitor CB-839 and the PI3K/aldolase inhibitor NVP-BKM120 more consistently reduces cell growth of tumor xenografts. Maximal growth inhibition correlates with the disruption of redox homeostasis, involving loss of reduced glutathione regeneration, redox cofactors, and a decreased connectivity among metabolites primarily involved in nucleic acid metabolism., Conclusions: Our findings open the way to develop metabolic connectivity profiling as a tool for a selective strategy of combined drug repositioning in precision oncology., Competing Interests: Competing interestsDG, L.A., R.M.M., and SV are inventors on the patent application PCT/EP2019/079843 related to the subject matter of this study. The authors declare no other competing interests., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
34. [18F]FDG and [18F]FLT PET for the evaluation of response to neo-adjuvant chemotherapy in a model of triple negative breast cancer.
- Author
-
Raccagni I, Belloli S, Valtorta S, Stefano A, Presotto L, Pascali C, Bogni A, Tortoreto M, Zaffaroni N, Daidone MG, Russo G, Bombardieri E, and Moresco RM
- Subjects
- Animals, Antineoplastic Agents, Phytogenic therapeutic use, Area Under Curve, Cell Line, Tumor, Female, Humans, Ki-67 Antigen metabolism, Mice, Mice, Nude, Mice, SCID, Neoadjuvant Therapy, Paclitaxel therapeutic use, ROC Curve, Transplantation, Heterologous, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology, Fluorodeoxyglucose F18 chemistry, Positron-Emission Tomography, Radiopharmaceuticals chemistry, Thymidine analogs & derivatives, Triple Negative Breast Neoplasms diagnostic imaging
- Abstract
Rationale: Pathological response to neo-adjuvant chemotherapy (NAC) represents a commonly used predictor of survival in triple negative breast cancer (TNBC) and the need to identify markers that predict response to NAC is constantly increasing. Aim of this study was to evaluate the potential usefulness of PET imaging with [18F]FDG and [18F]FLT for the discrimination of TNBC responders to Paclitaxel (PTX) therapy compared to the response assessed by an adapted Response Evaluation Criteria In Solid Tumors (RECIST) criteria based on tumor volume (Tumor Volume Response)., Methods: Nu/nu mice bearing TNBC lesions of different size were evaluated with [18F]FDG and [18F]FLT PET before and after PTX treatment. SUVmax, Metabolic Tumor Volume (MTV) and Total Lesion Glycolysis (TLG) and Proliferation (TLP) were assessed using a graph-based random walk algorithm., Results: We found that in our TNBC model the variation of [18F]FDG and [18F]FLT SUVmax similarly defined tumor response to therapy and that SUVmax variation represented the most accurate parameter. Response evaluation using Tumor Volume Response (TVR) showed that the effectiveness of NAC with PTX was completely independent from lesions size at baseline., Conclusions: Our study provided interesting results in terms of sensitivity and specificity of PET in TNBC, revealing the similar performances of [18F]FDG and [18F]FLT in the identification of responders to Paclitaxel., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2018
- Full Text
- View/download PDF
35. Inhibition of the Hexosamine Biosynthetic Pathway by targeting PGM3 causes breast cancer growth arrest and apoptosis.
- Author
-
Ricciardiello F, Votta G, Palorini R, Raccagni I, Brunelli L, Paiotta A, Tinelli F, D'Orazio G, Valtorta S, De Gioia L, Pastorelli R, Moresco RM, La Ferla B, and Chiaradonna F
- Subjects
- Animals, Apoptosis drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, MCF-7 Cells, Mice, Phosphoglucomutase antagonists & inhibitors, Signal Transduction drug effects, Triple Negative Breast Neoplasms metabolism, Xenograft Model Antitumor Assays, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Hexosamines biosynthesis, Phosphoglucomutase metabolism, Triple Negative Breast Neoplasms drug therapy
- Abstract
Cancer aberrant N- and O-linked protein glycosylation, frequently resulting from an augmented flux through the Hexosamine Biosynthetic Pathway (HBP), play different roles in tumor progression. However, the low specificity and toxicity of the existing HBP inhibitors prevented their use for cancer treatment. Here we report the preclinical evaluation of FR054, a novel inhibitor of the HBP enzyme PGM3, with a remarkable anti-breast cancer effect. In fact, FR054 induces in different breast cancer cells a dramatic decrease in cell proliferation and survival. In particular, in a model of Triple Negative Breast Cancer (TNBC) cells, MDA-MB-231, we show that these effects are correlated to FR054-dependent reduction of both N- and O-glycosylation level that cause also a strong reduction of cancer cell adhesion and migration. Moreover we show that impaired survival of cancer cells upon FR054 treatment is associated with the activation of the Unfolded Protein Response (UPR) and accumulation of intracellular ROS. Finally, we show that FR054 suppresses cancer growth in MDA-MB-231 xenograft mice, supporting the advantage of targeting HBP for therapeutic purpose and encouraging further investigation about the use of this small molecule as a promising compound for breast cancer therapy.
- Published
- 2018
- Full Text
- View/download PDF
36. Metformin and temozolomide, a synergic option to overcome resistance in glioblastoma multiforme models.
- Author
-
Valtorta S, Lo Dico A, Raccagni I, Gaglio D, Belloli S, Politi LS, Martelli C, Diceglie C, Bonanomi M, Ercoli G, Vaira V, Ottobrini L, and Moresco RM
- Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with poor survival. Cytoreduction in association with radiotherapy and temozolomide (TMZ) is the standard therapy, but response is heterogeneous and life expectancy is limited. The combined use of chemotherapeutic agents with drugs targeting cell metabolism is becoming an interesting therapeutic option for cancer treatment. Here, we found that metformin (MET) enhances TMZ effect on TMZ-sensitive cell line (U251) and overcomes TMZ-resistance in T98G GBM cell line. In particular, combined-treatment modulated apoptosis by increasing Bax/Bcl-2 ratio, and reduced Reactive Oxygen Species (ROS) production. We also observed that MET associated with TMZ was able to reduce the expression of glioma stem cells (GSC) marker CD90 particularly in T98G cells but not that of CD133. In vivo experiments showed that combined treatment with TMZ and MET significantly slowed down growth of TMZ-resistant tumors but did not affect overall survival of TMZ-sensitive tumor bearing mice. In conclusion, our results showed that metformin is able to enhance TMZ effect in TMZ-resistant cell line suggesting its potential use in TMZ refractory GBM patients. However, the lack of effect on a GBM malignancy marker like CD133 requires further evaluation since it might influence response duration., Competing Interests: CONFLICTS OF INTEREST The authors declare that they have no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
37. Metabolic Evaluation of Non-Small Cell Lung Cancer Patient-Derived Xenograft Models Using 18F-FDG PET: A Potential Tool for Early Therapy Response.
- Author
-
Valtorta S, Moro M, Prisinzano G, Bertolini G, Tortoreto M, Raccagni I, Pastorino U, Roz L, Sozzi G, and Moresco RM
- Subjects
- Animals, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung therapy, Cell Line, Tumor, Female, Humans, Lung Neoplasms therapy, Male, Mice, Nude, Mice, SCID, Molecular Imaging methods, Outcome and Process Assessment, Health Care methods, Radiopharmaceuticals pharmacokinetics, Reproducibility of Results, Sensitivity and Specificity, Treatment Outcome, Tumor Cells, Cultured, Carcinoma, Non-Small-Cell Lung diagnostic imaging, Carcinoma, Non-Small-Cell Lung metabolism, Fluorodeoxyglucose F18 pharmacokinetics, Glucose metabolism, Lung Neoplasms diagnostic imaging, Lung Neoplasms metabolism
- Abstract
Lung cancer heterogeneity makes response to therapy extremely hard to predict. Patient-derived xenografts (PDXs) are a reliable preclinical model that closely recapitulates the main characteristics of the parental tumors and may represent a useful asset for testing new therapies. Here, using PET imaging, we investigated whether lung cancer PDXs reproduce the metabolic characteristics of the corresponding parental tumors., Methods: We performed longitudinal
18 F-FDG PET studies on 9 different PDX groups obtained by implanting primary-cancer fragments harvested from patients into mice. The SUVmax of each PDX was calculated and compared with the SUVmax of the corresponding parental tumor., Results: Tumor growth rate and uptake varied among the different PDXs and confirmed the preservation of individual characteristics. The intragroup reproducibility of PET measurements was good. Furthermore, PDXs from tumors with a higher metabolic rate displayed a rank order of uptake similar to that of the parental tumors., Conclusion: PDXs reproduced the glucose metabolism of the parental tumors and therefore represent a promising preclinical model for the early assessment of therapy efficacy., (© 2017 by the Society of Nuclear Medicine and Molecular Imaging.)- Published
- 2017
- Full Text
- View/download PDF
38. The 18-kDa mitochondrial translocator protein in gliomas: from the bench to bedside.
- Author
-
Janczar K, Su Z, Raccagni I, Anfosso A, Kelly C, Durrenberger PF, Gerhard A, and Roncaroli F
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms metabolism, Glioma drug therapy, Glioma metabolism, Humans, Molecular Targeted Therapy, Positron-Emission Tomography, Precision Medicine, Prognosis, Up-Regulation, Brain Neoplasms pathology, Glioma pathology, Receptors, GABA metabolism
- Abstract
The 18-kDa mitochondrial translocator protein (TSPO) is known to be highly expressed in several types of cancer, including gliomas, whereas expression in normal brain is low. TSPO functions in glioma are still incompletely understood. The TSPO can be quantified pre-operatively with molecular imaging making it an ideal candidate for personalized treatment of patient with glioma. Studies have proposed to exploit the TSPO as a transporter of chemotherapics to selectively target tumour cells in the brain. Our studies proved that positron emission tomography (PET)-imaging can contribute to predict progression of patients with glioma and that molecular imaging with TSPO-specific ligands is suitable to stratify patients in view of TSPO-targeted treatment. Finally, we proved that TSPO in gliomas is predominantly expressed by tumour cells., (© 2015 Authors; published by Portland Press Limited.)
- Published
- 2015
- Full Text
- View/download PDF
39. Characterization of biological features of a rat F98 GBM model: a PET-MRI study with [18F]FAZA and [18F]FDG.
- Author
-
Belloli S, Brioschi A, Politi LS, Ronchetti F, Calderoni S, Raccagni I, Pagani A, Monterisi C, Zenga F, Zara G, Fazio F, Mauro A, and Moresco RM
- Subjects
- Animals, Cell Differentiation, Cell Line, Tumor, Disease Models, Animal, Glioblastoma metabolism, Glioblastoma pathology, Humans, Male, Rats, Survival Analysis, Fluorodeoxyglucose F18, Glioblastoma diagnostic imaging, Magnetic Resonance Imaging, Nitroimidazoles, Positron-Emission Tomography
- Abstract
Introduction: The prognosis of malignant gliomas remains largely unsatisfactory for the intrinsic characteristics of the pathology and for the delayed diagnosis. Multimodal imaging based on PET and MRI may assess the dynamics of disease onset and progression allowing the validation of preclinical models of glioblastoma multiforme (GBM). The aim of this study was the characterization of a syngeneic rat model of GBM using combined in vivo imaging and immunohistochemistry., Methods: Four groups of Fischer rats were implanted in a subcortical region with increasing concentration of rat glioma F98 cells and weekly monitored with Gd-MR, [(18)F]FDG- and [(18)F]FAZA-PET starting one week after surgery. Different targets were evaluated on post mortem brain specimens using immunohistochemistry: VEGF, GFAP, HIF-1α, Ki-67 and nestin., Results: Imaging results indicated that tumor onset but not progression was related to the number of F98 cells. Hypoxic regions identified with [(18)F]FAZA and high-glucose metabolism regions recognized with [(18)F]FDG were located respectively in the core and in external areas of the tumor, with partial overlap and remodeling during disease progression. Histological and immunohistochemical analysis confirmed PET/MRI results and revealed that our model resumes biological characteristics of human GBM. IHC and PET studies showed that necrotic regions, defined on the basis of [(18)F]FDG uptake reduction, may include hypoxic clusters of vital tumor tissue identified with [(18)F]FAZA. This last information is particularly relevant for the identification of the target volume during image-guided radiotherapy., Conclusions: In conclusion, the combined use of PET and MRI allows in vivo monitoring of the biological modification of F98 lesions during tumor progression., (Crown Copyright © 2013. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.