177 results on '"Grazia Graziani"'
Search Results
2. Combination Treatment of Resistant Acute Promyelocytic Leukemia Cells with Arsenic Trioxide and Anti-Apoptotic Gene Inhibitors
- Author
-
Manuela Giansanti, Tiziana Ottone, Serena Travaglini, Maria Teresa Voso, Grazia Graziani, and Isabella Faraoni
- Subjects
APL ,arsenic resistance ,apoptosis ,BCL2 ,venetoclax ,BIRC3 ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Background: Arsenic trioxide (ATO) is an anticancer agent for treating acute promyelocytic leukemia (APL). However, 5–10% of patients fail to respond, developing relapsed/refractory disease. The aim of this study was to identify potential new therapeutic approaches for ATO-unresponsive APL by targeting the anti-apoptotic genes that contribute to drug resistance. Methods: RNA expression of dysregulated genes involved in the apoptotic pathway was analyzed by comparing ATO-resistant APL cell clones generated in our lab with the corresponding sensitive clones, at basal levels and after 48 h of treatment with ATO. Results: ATO-resistant APL cells showed upregulation of APAF1, BCL2, BIRC3, and NOL3 genes, while CD70 and IL10 genes were downregulated, compared to ATO-sensitive cells. Treatment with ATO strongly increased the expression of the anti-apoptotic genes BIRC3, NOL3, and BCL2A1 and significantly downregulated BCL2 in ATO-sensitive clones. Although all these genes can be relevant to ATO-resistance, we selected BCL2 and BIRC3 as druggable targets. A direct correlation between BCL2 expression and the sensitivity to the BCL2 inhibitor venetoclax was observed, indicating BCL2 as predictive biomarker of the response. Moreover, the combination of venetoclax with ATO exerted synergistic cytotoxic effects, thus reverting the resistance to ATO. APL treatment with SMAC mimetics such as LCL161 and xevinapant (inhibitors of BIRC3) was not as effective as the BCL2 inhibitor as a monotherapy but exerted synergistic effects in combination with ATO in cells with low BIRC expression. Conclusions: This study demonstrates the therapeutic potential of venetoclax in combination with ATO in vitro and strongly encourages further investigation of relapsed/refractory APL with high BCL2 expression.
- Published
- 2024
- Full Text
- View/download PDF
3. The VEGFs/VEGFRs system in Alzheimer’s and Parkinson’s diseases: Pathophysiological roles and therapeutic implications
- Author
-
Claudia Ceci, Pedro Miguel Lacal, Maria Luisa Barbaccia, Nicola Biagio Mercuri, Grazia Graziani, and Ada Ledonne
- Subjects
Parkinson’s disease ,Alzheimer’s disease ,VEGF-A ,VEGFR-1 ,VEGFR-2 ,Angiogenesis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The vascular endothelial growth factors (VEGFs) and their cognate receptors (VEGFRs), besides their well-known involvement in physiological angiogenesis/lymphangiogenesis and in diseases associated to pathological vessel formation, play multifaceted functions in the central nervous system (CNS). In addition to shaping brain development, by controlling cerebral vasculogenesis and regulating neurogenesis as well as astrocyte differentiation, the VEGFs/VEGFRs axis exerts essential functions in the adult brain both in physiological and pathological contexts. In this article, after describing the physiological VEGFs/VEGFRs functions in the CNS, we focus on the VEGFs/VEGFRs involvement in neurodegenerative diseases by reviewing the current literature on the rather complex VEGFs/VEGFRs contribution to the pathogenic mechanisms of Alzheimer’s (AD) and Parkinson’s (PD) diseases. Thereafter, based on the outcome of VEGFs/VEGFRs targeting in animal models of AD and PD, we discuss the factual relevance of pharmacological VEGFs/VEGFRs modulation as a novel and potential disease-modifying approach for these neurodegenerative pathologies. Specific VEGFRs targeting, aimed at selective VEGFR-1 inhibition, while preserving VEGFR-2 signal transduction, appears as a promising strategy to hit the molecular mechanisms underlying AD pathology. Moreover, therapeutic VEGFs-based approaches can be proposed for PD treatment, with the aim of fine-tuning their brain levels to amplify neurotrophic/neuroprotective effects while limiting an excessive impact on vascular permeability.
- Published
- 2024
- Full Text
- View/download PDF
4. A novel and atypical NF-KB pro-inflammatory program regulated by a CamKII-proteasome axis is involved in the early activation of Muller glia by high glucose
- Author
-
Diego Sbardella, Grazia Raffaella Tundo, Alice Mecchia, Camilla Palumbo, Maria Grazia Atzori, Lauretta Levati, Alessandra Boccaccini, Anna Maria Caccuri, Paolo Cascio, Pedro Miguel Lacal, Grazia Graziani, Monica Varano, Massimiliano Coletta, and Mariacristina Parravano
- Subjects
Diabetic retinopathy ,Muller glia ,NF-kB pathway ,Proteasome ,CamKII ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Key Points High glucose quickly induces an atypical NF-kB pro-inflammatory program. CamKII phosphorylation of Rpt6 subunit of the proteasome stimulates IkBα turnover and p65-p50 release. Inhibition of either CamkII or proteasome blocks this pathway.
- Published
- 2022
- Full Text
- View/download PDF
5. Targeting of PDGF-C/NRP-1 autocrine loop as a new strategy for counteracting the invasiveness of melanoma resistant to braf inhibitors
- Author
-
Federica Ruffini, Claudia Ceci, Maria Grazia Atzori, Simona Caporali, Lauretta Levati, Laura Bonmassar, Gian Carlo Antonini Cappellini, Stefania D’Atri, Grazia Graziani, and Pedro Miguel Lacal
- Subjects
Melanoma ,BRAF inhibitors ,Neuropilin-1 ,PDGF-C ,Dabrafenib ,Extracellular matrix invasion ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Melanoma resistance to BRAF inhibitors (BRAFi) is often accompanied by a switch from a proliferative to an invasive phenotype. Therefore, the identification of signaling molecules involved in the development of metastatic properties by resistant melanoma cells is of primary importance. We have previously demonstrated that activation of neuropilin-1 (NRP-1) by platelet-derived growth factor (PDGF)-C confers melanoma cells with an invasive behavior similar to that of BRAFi resistant tumors. Aims of the present study were to evaluate the role of PDGF-C/NRP-1 autocrine loop in the acquisition of an invasive and BRAFi-resistant phenotype by melanoma cells and the effect of its inhibition on drug resistance and extracellular matrix (ECM) invasion. Furthermore, we investigated whether PDGF-C serum levels were differentially modulated by drug treatment in metastatic melanoma patients responsive or refractory to BRAFi as single agents or in combination with MEK inhibitors (MEKi). The results indicated that human melanoma cells resistant to BRAFi express higher levels of PDGF-C and NRP-1 as compared to their susceptible counterparts. Overexpression occurs early during development of drug resistance and contributes to the invasive properties of resistant cells. Accordingly, silencing of NRP-1 or PDGF-C reduces tumor cell invasiveness. Analysis of PDGF-C in the serum collected from patients treated with BRAFi or BRAFi+MEKi, showed that in responders PDGF-C levels decrease after treatment and raise again at tumor progression. Conversely, in non-responders treatment does not affect PDGF-C serum levels. Thus, blockade of NRP-1 activation by PDGF-C might represent a new therapeutic approach to counteract the invasiveness of BRAFi-resistant melanoma.
- Published
- 2023
- Full Text
- View/download PDF
6. The Insulin-Degrading Enzyme from Structure to Allosteric Modulation: New Perspectives for Drug Design
- Author
-
Grazia Raffaella Tundo, Giuseppe Grasso, Marco Persico, Oleh Tkachuk, Francesco Bellia, Alessio Bocedi, Stefano Marini, Mariacristina Parravano, Grazia Graziani, Caterina Fattorusso, and Diego Sbardella
- Subjects
insulin-degrading enzyme ,Alzheimer’s disease ,diabetes ,proteasome ,insulin ,allostery ,Microbiology ,QR1-502 - Abstract
The insulin-degrading enzyme (IDE) is a Zn2+ peptidase originally discovered as the main enzyme involved in the degradation of insulin and other amyloidogenic peptides, such as the β-amyloid (Aβ) peptide. Therefore, a role for the IDE in the cure of diabetes and Alzheimer’s disease (AD) has been long envisaged. Anyway, its role in degrading amyloidogenic proteins remains not clearly defined and, more recently, novel non-proteolytic functions of the IDE have been proposed. From a structural point of view, the IDE presents an atypical clamshell structure, underscoring unique enigmatic enzymological properties. A better understanding of the structure–function relationship may contribute to solving some existing paradoxes of IDE biology and, in light of its multifunctional activity, might lead to novel therapeutic approaches.
- Published
- 2023
- Full Text
- View/download PDF
7. VEGF-A/VEGFR-1 signalling and chemotherapy-induced neuropathic pain: therapeutic potential of a novel anti-VEGFR-1 monoclonal antibody
- Author
-
Laura Micheli, Carmen Parisio, Elena Lucarini, Alessia Vona, Alessandra Toti, Alessandra Pacini, Tommaso Mello, Serena Boccella, Flavia Ricciardi, Sabatino Maione, Grazia Graziani, Pedro Miguel Lacal, Paola Failli, Carla Ghelardini, and Lorenzo Di Cesare Mannelli
- Subjects
VEGF-A ,VEGFR-1 ,Neuropathy biomarker ,Astrocytes ,D16F7 mAb ,Oxaliplatin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Neuropathic pain is a clinically relevant adverse effect of several anticancer drugs that markedly impairs patients’ quality of life and frequently leads to dose reduction or therapy discontinuation. The poor knowledge about the mechanisms involved in neuropathy development and pain chronicization, and the lack of effective therapies, make treatment of chemotherapy-induced neuropathic pain an unmet medical need. In this context, the vascular endothelial growth factor A (VEGF-A) has emerged as a candidate neuropathy hallmark and its decrease has been related to pain relief. In the present study, we have investigated the role of VEGF-A and its receptors, VEGFR-1 and VEGFR-2, in pain signalling and in chemotherapy-induced neuropathy establishment as well as the therapeutic potential of receptor blockade in the management of pain. Methods Behavioural and electrophysiological analyses were performed in an in vivo murine model, by using selective receptor agonists, blocking monoclonal antibodies or siRNA-mediated silencing of VEGF-A and VEGFRs. Expression of VEGF-A and VEGFR-1 in astrocytes and neurons was detected by immunofluorescence staining and confocal microscopy analysis. Results In mice, the intrathecal infusion of VEGF-A (VEGF165 isoforms) induced a dose-dependent noxious hypersensitivity and this effect was mediated by VEGFR-1. Consistently, electrophysiological studies indicated that VEGF-A strongly stimulated the spinal nociceptive neurons activity through VEGFR-1. In the dorsal horn of the spinal cord of animals affected by oxaliplatin-induced neuropathy, VEGF-A expression was increased in astrocytes while VEGFR-1 was mainly detected in neurons, suggesting a VEGF-A/VEGFR-1-mediated astrocyte-neuron cross-talk in neuropathic pain pathophysiology. Accordingly, the selective knockdown of astrocytic VEGF-A by intraspinal injection of shRNAmir blocked the development of oxaliplatin-induced neuropathic hyperalgesia and allodynia. Interestingly, both intrathecal and systemic administration of the novel anti-VEGFR-1 monoclonal antibody D16F7, endowed with anti-angiogenic and antitumor properties, reverted oxaliplatin-induced neuropathic pain. Besides, D16F7 effectively relieved hypersensitivity induced by other neurotoxic chemotherapeutic agents, such as paclitaxel and vincristine. Conclusions These data strongly support the role of the VEGF-A/VEGFR-1 system in mediating chemotherapy-induced neuropathic pain at the central nervous system level. Thus, treatment with the anti-VEGFR-1 mAb D16F7, besides exerting antitumor activity, might result in the additional advantage of attenuating neuropathic pain when combined with neurotoxic anticancer agents.
- Published
- 2021
- Full Text
- View/download PDF
8. Insulin-Degrading Enzyme Is a Non Proteasomal Target of Carfilzomib and Affects the 20S Proteasome Inhibition by the Drug
- Author
-
Grazia Raffaella Tundo, Diego Sbardella, Francesco Oddone, Giuseppe Grasso, Stefano Marini, Maria Grazia Atzori, Anna Maria Santoro, Danilo Milardi, Francesco Bellia, Gabriele Macari, Grazia Graziani, Fabio Polticelli, Paolo Cascio, Mariacristina Parravano, and Massimo Coletta
- Subjects
proteasome ,carfilzomib ,insulin-degrading enzyme ,cancer ,neurodegeneration ,Microbiology ,QR1-502 - Abstract
Carfilzomib is a last generation proteasome inhibitor (PI) with proven clinical efficacy in the treatment of relapsed/refractory multiple myeloma. This drug is considered to be extremely specific in inhibiting the chymotrypsin-like activity of the 20S proteasome, encoded by the β5 subunit, overcoming some bortezomib limitations, the first PI approved for multiple myeloma therapy which is however burdened by a significant toxicity profile, due also to its off-target effects. Here, molecular approaches coupled with molecular docking studies have been used to unveil that the Insulin-Degrading Enzyme, a ubiquitous and highly conserved Zn2+ peptidase, often found to associate with proteasome in cell-based models, is targeted by carfilzomib in vitro. The drug behaves as a modulator of IDE activity, displaying an inhibitory effect over 10-fold lower than for the 20S. Notably, the interaction of IDE with the 20S enhances in vitro the inhibitory power of carfilzomib on proteasome, so that the IDE-20S complex is an even better target of carfilzomib than the 20S alone. Furthermore, IDE gene silencing after delivery of antisense oligonucleotides (siRNA) significantly reduced carfilzomib cytotoxicity in rMC1 cells, a validated model of Muller glia, suggesting that, in cells, the inhibitory activity of this drug on cell proliferation is somewhat linked to IDE and, possibly, also to its interaction with proteasome.
- Published
- 2022
- Full Text
- View/download PDF
9. The anti-vascular endothelial growth factor receptor-1 monoclonal antibody D16F7 inhibits invasiveness of human glioblastoma and glioblastoma stem cells
- Author
-
Maria Grazia Atzori, Lucio Tentori, Federica Ruffini, Claudia Ceci, Lucia Lisi, Elena Bonanno, Manuel Scimeca, Eskil Eskilsson, Thomas Daubon, Hrvoje Miletic, Lucia Ricci Vitiani, Roberto Pallini, Pierluigi Navarra, Rolf Bjerkvig, Stefania D’Atri, Pedro Miguel Lacal, and Grazia Graziani
- Subjects
VEGFR-1 ,PlGF ,VEGF-A ,Glioblastoma ,Angiogenesis ,Molecular marker ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Glioblastoma (GBM) is a highly migratory, invasive, and angiogenic brain tumor. Like vascular endothelial growth factor-A (VEGF-A), placental growth factor (PlGF) promotes GBM angiogenesis. VEGF-A is a ligand for both VEGF receptor-1 (VEGFR-1) and VEGFR-2, while PlGF interacts exclusively with VEGFR-1. We recently generated the novel anti-VEGFR-1 monoclonal antibody (mAb) D16F7 that diminishes VEGFR-1 homodimerization/activation without affecting VEGF-A and PlGF binding. Methods In the present study, we evaluated the expression of VEGFR-1 in human GBM tissue samples (n = 42) by immunohistochemistry, in cell lines (n = 6) and GBM stem cells (GSCs) (n = 18) by qRT-PCR and/or western blot analysis. In VEGFR-1 positive GBM or GSCs we also analyzed the ability of D16F7 to inhibit GBM invasiveness in response to VEGF-A and PlGF. Results Most of GBM specimens stained positively for VEGFR-1 and all but one GBM cell lines expressed VEGFR-1. On the other hand, in GSCs the expression of the receptor was heterogeneous. D16F7 reduced migration and invasion of VEGFR-1 positive GBM cell lines and patient-derived GSCs in response to VEGF-A and PlGF. Interestingly, this effect was also observed in VEGFR-1 positive GSCs transfected to over-express wild-type EGFR (EGFRwt+) or mutant EGFR (ligand binding domain-deficient EGFRvIII+). Furthermore, D16F7 suppressed intracellular signal transduction in VEGFR-1 over-expressing GBM cells by reducing receptor auto-phosphorylation at tyrosine 1213 and downstream Erk1/2 activation induced by receptor ligands. Conclusion The results from this study suggest that VEGFR-1 is a relevant target for GBM therapy and that D16F7-derived humanized mAbs warrant further investigation.
- Published
- 2017
- Full Text
- View/download PDF
10. Saffron and Its Major Ingredients’ Effect on Colon Cancer Cells with Mismatch Repair Deficiency and Microsatellite Instability
- Author
-
Amr Amin, Aaminah Farrukh, Chandraprabha Murali, Akbar Soleimani, Françoise Praz, Grazia Graziani, Hassan Brim, and Hassan Ashktorab
- Subjects
colorectal cancer ,saffron ,safranal ,crocin ,HCT116 ,MLH1 ,Organic chemistry ,QD241-441 - Abstract
Background: Colorectal cancer (CRC) is one of the most common cancers worldwide. One of its subtypes is associated with defective mismatch repair (dMMR) genes. Saffron has many potentially protective roles against colon malignancy. However, these roles in the context of dMMR tumors have not been explored. In this study, we aimed to investigate the effects of saffron and its constituents in CRC cell lines with dMMR. Methods: Saffron crude extracts and specific compounds (safranal and crocin) were used in the human colorectal cancer cell lines HCT116, HCT116+3 (inserted MLH1), HCT116+5 (inserted MSH3), and HCT116+3+5 (inserted MLH1 and MSH3). CDC25b, p-H2AX, TPDP1, and GAPDH were analyzed by Western blot. Proliferation and cytotoxicity were analyzed by MTT. The scratch wound assay was also performed. Results: Saffron crude extracts restricted (up to 70%) the proliferation in colon cells with deficient MMR (HCT116) compared to proficient MMR. The wound healing assay indicates that deficient MMR cells are doing better (up to 90%) than proficient MMR cells when treated with saffron. CDC25b and TDP1 downregulated (up to 20-fold) in proficient MMR cells compared to deficient MMR cells, while p.H2AX was significantly upregulated in both cell types, particularly at >10 mg/mL saffron in a concentration-dependent manner. The reduction in cellular proliferation was accompanied with upregulation of caspase 3 and 7. The major active saffron compounds, safranal and crocin reproduced most of the saffron crude extracts’ effects. Conclusions: Saffron’s anti-proliferative effect is significant in cells with deficient MMR. This novel effect may have therapeutic implications and benefits for MSI CRC patients who are generally not recommended for the 5-fluorouracil-based treatment.
- Published
- 2021
- Full Text
- View/download PDF
11. PARP-1 modulates amyloid beta peptide-induced neuronal damage.
- Author
-
Sara Martire, Andrea Fuso, Dante Rotili, Italo Tempera, Cesare Giordano, Ivana De Zottis, Alessia Muzi, Patrizia Vernole, Grazia Graziani, Emanuela Lococo, Martina Faraldi, Bruno Maras, Sigfrido Scarpa, Luciana Mosca, and Maria d'Erme
- Subjects
Medicine ,Science - Abstract
Amyloid beta peptide (Aβ) causes neurodegeneration by several mechanisms including oxidative stress, which is known to induce DNA damage with the consequent activation of poly (ADP-ribose) polymerase (PARP-1). To elucidate the role of PARP-1 in the neurodegenerative process, SH-SY5Y neuroblastoma cells were treated with Aβ25-35 fragment in the presence or absence of MC2050, a new PARP-1 inhibitor. Aβ25-35 induces an enhancement of PARP activity which is prevented by cell pre-treatment with MC2050. These data were confirmed by measuring PARP-1 activity in CHO cells transfected with amylod precursor protein and in vivo in brains specimens of TgCRND8 transgenic mice overproducing the amyloid peptide. Following Aβ25-35 exposure a significant increase in intracellular ROS was observed. These data were supported by the finding that Aβ25-35 induces DNA damage which in turn activates PARP-1. Challenge with Aβ25-35 is also able to activate NF-kB via PARP-1, as demonstrated by NF-kB impairment upon MC2050 treatment. Moreover, Aβ25-35 via PARP-1 induces a significant increase in the p53 protein level and a parallel decrease in the anti-apoptotic Bcl-2 protein. These overall data support the hypothesis of PARP-1 involvment in cellular responses induced by Aβ and hence a possible rationale for the implication of PARP-1 in neurodegeneration is discussed.
- Published
- 2013
- Full Text
- View/download PDF
12. Reduced proficiency in homologous recombination underlies the high sensitivity of embryonal carcinoma testicular germ cell tumors to Cisplatin and poly (adp-ribose) polymerase inhibition.
- Author
-
Francesca Cavallo, Grazia Graziani, Cristina Antinozzi, Darren R Feldman, Jane Houldsworth, George J Bosl, Raju S K Chaganti, Mary Ellen Moynahan, Maria Jasin, and Marco Barchi
- Subjects
Medicine ,Science - Abstract
Testicular Germ Cell Tumors (TGCT) and patient-derived cell lines are extremely sensitive to cisplatin and other interstrand cross-link (ICL) inducing agents. Nevertheless, a subset of TGCTs are either innately resistant or acquire resistance to cisplatin during treatment. Understanding the mechanisms underlying TGCT sensitivity/resistance to cisplatin as well as the identification of novel strategies to target cisplatin-resistant TGCTs have major clinical implications. Herein, we have examined the proficiency of five embryonal carcinoma (EC) cell lines to repair cisplatin-induced ICLs. Using γH2AX staining as a marker of double strand break formation, we found that EC cell lines were either incapable of or had a reduced ability to repair ICL-induced damage. The defect correlated with reduced Homologous Recombination (HR) repair, as demonstrated by the reduction of RAD51 foci formation and by direct evaluation of HR efficiency using a GFP-reporter substrate. HR-defective tumors cells are known to be sensitive to the treatment with poly(ADP-ribose) polymerase (PARP) inhibitor. In line with this observation, we found that EC cell lines were also sensitive to PARP inhibitor monotherapy. The magnitude of sensitivity correlated with HR-repair reduced proficiency and with the expression levels and activity of PARP1 protein. In addition, we found that PARP inhibition strongly enhanced the response of the most resistant EC cells to cisplatin, by reducing their ability to overcome the damage. These results point to a reduced proficiency of HR repair as a source of sensitivity of ECs to ICL-inducing agents and PARP inhibitor monotherapy, and suggest that pharmacological inhibition of PARP can be exploited to target the stem cell component of the TGCTs (namely ECs) and to enhance the sensitivity of cisplatin-resistant TGCTs to standard treatments.
- Published
- 2012
- Full Text
- View/download PDF
13. Exogenous Control of the Expression of Group I CD1 Molecules Competent for Presentation of Microbial Nonpeptide Antigens to Human T Lymphocytes
- Author
-
Angelo Aquino, Grazia Graziani, Ornella Franzese, Salvatore P. Prete, Enzo Bonmassar, Laura Bonmassar, and Stefania D'Atri
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
Group I CD1 (CD1a, CD1b, and CD1c) glycoproteins expressed on immature and mature dendritic cells present nonpeptide antigens (i.e., lipid or glycolipid molecules mainly of microbial origin) to T cells. Cytotoxic CD1-restricted T lymphocytes recognizing mycobacterial lipid antigens were found in tuberculosis patients. However, thanks to a complex interplay between mycobacteria and CD1 system, M. tuberculosis possesses a successful tactic based, at least in part, on CD1 downregulation to evade CD1-dependent immunity. On the ground of these findings, it is reasonable to hypothesize that modulation of CD1 protein expression by chemical, biological, or infectious agents could influence host's immune reactivity against M. tuberculosis-associated lipids, possibly affecting antitubercular resistance. This scenario prompted us to perform a detailed analysis of the literature concerning the effect of external agents on Group I CD1 expression in order to obtain valuable information on the possible strategies to be adopted for driving properly CD1-dependent immune functions in human pathology and in particular, in human tuberculosis.
- Published
- 2011
- Full Text
- View/download PDF
14. Physical Exercise and the Hallmarks of Breast Cancer: A Narrative Review
- Author
-
Celia García-Chico, Susana López-Ortiz, Saúl Peñín-Grandes, José Pinto-Fraga, Pedro L. Valenzuela, Enzo Emanuele, Claudia Ceci, Grazia Graziani, Carmen Fiuza-Luces, Simone Lista, Alejandro Lucia, and Alejandro Santos-Lozano
- Subjects
Cancer Research ,breast cancer ,training ,mammary cancer ,Oncology ,Settore BIO/14 ,biomarkers ,physical activity - Abstract
Growing evidence suggests that, among the different molecular/cellular pathophysiological mechanisms associated with cancer, there are 14 hallmarks that play a major role, including: (i) sustaining proliferative signaling, (ii) evading growth suppressors, (iii) activating invasion and metastasis, (iv) enabling replicative immortality, (v) inducing angiogenesis, (vi) resisting cell death, (vii) reprogramming energy metabolism, (viii) evading immune destruction, (ix) genome instability and mutations, (x) tumor-promoting inflammation, (xi) unlocking phenotypic plasticity, (xii) nonmutational epigenetic reprogramming, (xiii) polymorphic microbiomes, and (xiv) senescent cells. These hallmarks are also associated with the development of breast cancer, which represents the most prevalent tumor type in the world. The present narrative review aims to describe, for the first time, the effects of physical activity/exercise on these hallmarks. In summary, an active lifestyle, and particularly regular physical exercise, provides beneficial effects on all major hallmarks associated with breast cancer, and might therefore help to counteract the progression of the disease or its associated burden.
- Published
- 2022
15. The Anti-Vascular Endothelial Growth Factor Receptor 1 (VEGFR-1) D16F7 Monoclonal Antibody Inhibits Melanoma Adhesion to Soluble VEGFR-1 and Tissue Invasion in Response to Placenta Growth Factor
- Author
-
Maria Grazia Atzori, Claudia Ceci, Federica Ruffini, Manuel Scimeca, Rosella Cicconi, Maurizio Mattei, Pedro Miguel Lacal, and Grazia Graziani
- Subjects
Cancer Research ,PlGF ,VEGF-A ,VEGFR-1 ,melanoma ,metastasis ,Oncology ,Settore BIO/14 - Abstract
Placenta growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family involved in tumor-associated angiogenesis and melanoma invasion of the extra-cellular matrix (ECM) through activation of membrane VEGF receptor 1 (VEGFR-1). A soluble VEGFR-1 (sVEGFR-1) form is released in the ECM, where it sequesters proangiogenic factors and stimulates endothelial or tumor cell adhesion and chemotaxis through interaction with α5β1 integrin. The anti-VEGFR-1 monoclonal antibody (D16F7 mAb) inhibits VEGF-A or PlGF-mediated signal transduction without affecting ligand interaction, thus preserving sVEGFR-1 decoy function. The aim of this study was to investigate whether D16F7 mAb hampers melanoma spread by in vitro analysis of cell adhesion to sVEGFR-1, ECM invasion, transmigration through an endothelial cell monolayer and in vivo evaluation of tumor infiltrative potential in a syngeneic murine model. Results indicate that D16F7 mAb significantly inhibits melanoma adhesion to sVEGFR-1 and ECM invasion, as well as transmigration in response to PlGF. Moreover, treatment of melanoma-bearing mice with the anti-VEGFR-1 mAb not only inhibits tumor growth but also induces a significant reduction in bone infiltration associated with a decrease in PlGF-positive melanoma cells. Furthermore, D16F7 mAb reduces PlGF production by melanoma cells. Therefore, blockade of PLGF/VEGFR-1 signaling represents a suitable strategy to counteract the metastatic potential of melanoma.
- Published
- 2022
- Full Text
- View/download PDF
16. Role of PARP Inhibitors in Cancer Immunotherapy: Potential Friends to Immune Activating Molecules and Foes to Immune Checkpoints
- Author
-
Ornella Franzese and Grazia Graziani
- Subjects
PD-L1 ,Cancer Research ,PARP inhibitor ,Oncology ,BRCA ,PD-1 ,Settore BIO/14 ,cancer ,immune checkpoint inhibitor ,CTLA-4 ,immunotherapy ,DNA damage response ,combination therapy - Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) induce cytotoxic effects as single agents in tumors characterized by defective repair of DNA double-strand breaks deriving from BRCA1/2 mutations or other abnormalities in genes associated with homologous recombination. Preclinical studies have shown that PARPi-induced DNA damage may affect the tumor immune microenvironment and immune-mediated anti-tumor response through several mechanisms. In particular, increased DNA damage has been shown to induce the activation of type I interferon pathway and up-regulation of PD-L1 expression in cancer cells, which can both enhance sensitivity to Immune Checkpoint Inhibitors (ICIs). Despite the recent approval of ICIs for a number of advanced cancer types based on their ability to reinvigorate T-cell-mediated antitumor immune responses, a consistent percentage of treated patients fail to respond, strongly encouraging the identification of combination therapies to overcome resistance. In the present review, we analyzed both established and unexplored mechanisms that may be elicited by PARPi, supporting immune reactivation and their potential synergism with currently used ICIs. This analysis may indicate novel and possibly patient-specific immune features that might represent new pharmacological targets of PARPi, potentially leading to the identification of predictive biomarkers of response to their combination with ICIs.
- Published
- 2022
17. Poly(ADP-Ribose) Polymerase Inhibitors for Arsenic Trioxide–Resistant Acute Promyelocytic Leukemia: Synergistic In Vitro Antitumor Effects with Hypomethylating Agents or High-Dose Vitamin C
- Author
-
Grazia Graziani, Maria Domenica Divona, Maria Teresa Voso, Antonio De Gabrieli, Fabio Ciccarone, Manuela Giansanti, Isabella Faraoni, Terry Karimi, Tiziana Ottone, and S.P. Prete
- Subjects
0301 basic medicine ,Pharmacology ,Acute promyelocytic leukemia ,Veliparib ,Azacitidine ,Settore BIO/14 ,Decitabine ,Poly(ADP-ribose) Polymerase Inhibitors ,medicine.disease ,Poly (ADP-Ribose) Polymerase Inhibitor ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Arsenic Trioxide ,chemistry ,medicine ,Cancer research ,Phthalazines ,Molecular Medicine ,Arsenic trioxide ,Rucaparib ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Arsenic trioxide (ATO) is an anticancer agent used for the treatment ofacute promyelocytic leukemia (APL). However, 5%–10% of patients fail to respond or experience disease relapse. Based on poly(ADP-ribose) polymerase (PARP) 1 involvement in the processing of DNA demethylation, here we have tested the in vitro susceptibility of ATO-resistant clones (derived from the human APL cell line NB4) to PARP inhibitors (PARPi) in combination with hypomethylating agents (azacitidine and decitabine) or high-dose vitamin C (ascorbate), which induces 5-hydroxymethylcytosine (5hmC)-mediated DNA demethylation. ATO-sensitive and -resistant APL cell clones were generated and initially analyzed for their susceptibility to five clinically used PARPi (olaparib, niraparib, rucaparib, veliparib, and talazoparib). The obtained PARPi IC50 values were far below (olaparib and niraparib), within the range (talazoparib), or above (rucaparib and veliparib) the Cmax reported in patients, likely as a result of differences in the mechanisms of their cytotoxic activity. ATO-resistant APL cells were also susceptible to clinically relevant concentrations of azacitidine and decitabine and to high-dose ascorbate. Interestingly, the combination of these agents with olaparib, niraparib, or talazoparib resulted in synergistic antitumor activity. In combination with ascorbate, PARPi increased the ascorbate-mediated induction of 5hmC, which likely resulted in stalled DNA repair and cytotoxicity. Talazoparib was the most effective PARPi in synergizing with ascorbate, in accordance with its marked ability to trap PARP1 at damaged DNA. These findings suggest that ATO and PARPi have nonoverlapping resistance mechanisms and support further investigation on PARPi combination with hypomethylating agents or high-dose ascorbate for relapsed/ATO-refractory APL, especially in frail patients. SIGNIFICANCE STATEMENT This study found that poly(ADP-ribose) inhibitors (PARPi) show activity as single agents against human acute promyelocytic leukemia cells resistant to arsenic trioxide at clinically relevant concentrations. Furthermore, PARPi enhance the in vitro efficacy of azacitidine, decitabine, and high-dose vitamin C, all agents that alter DNA methylation. In combination with vitamin C, PARPi increase the levels of 5-hydroxymethylcytosine, likely as a result of altered processing of the oxidized intermediates associated with DNA demethylation.
- Published
- 2021
- Full Text
- View/download PDF
18. Monoclonal Antibodies to CTLA-4 with Focus on Ipilimumab
- Author
-
Grazia, Graziani, Lucia, Lisi, Lucio, Tentori, and Pierluigi, Navarra
- Subjects
Antibodies, Monoclonal ,Humans ,CTLA-4 Antigen ,Neoplasm Recurrence, Local ,Ipilimumab ,Melanoma - Abstract
The immune checkpoint cytotoxic T lymphocyte-associated antigen 4 (CTLA-4 or CD152) is a negative regulator of T-cell-mediated immune responses which plays a critical role in suppressing autoimmunity and maintaining immune homeostasis. Because of its inhibitory activity on T cells, CTLA-4 has been investigated as a drug target to induce immunostimulation, blocking the interaction with its ligands. The antitumor effects mediated by CTLA-4 blockade have been attributed to a sustained active immune response against cancer cells, due to the release of a brake on T cell activation. Ipilimumab (Yervoy, Bristol-Myers Squibb) is a fully human anti-CTLA-4 IgG1κ monoclonal antibody (mAb) that represents the first immune checkpoint inhibitor approved as monotherapy by FDA and EMA in 2011 for the treatment of unresectable/metastatic melanoma. In 2015, FDA also granted approval to ipilimumab monotherapy as adjuvant treatment of stage III melanoma to reduce the risk of tumour recurrence. The subsequent approved indications of ipilimumab for metastatic melanoma, regardless of BRAF mutational status, and other advanced/metastatic solid tumours always involve its use in association with the anti-programmed cell death protein 1 (PD-1) mAb nivolumab. Currently, ipilimumab is evaluated in ongoing clinical trials for refractory/advanced solid tumours mainly in combination with additional immunostimulating agents.
- Published
- 2022
19. Monoclonal Antibodies to CTLA-4 with Focus on Ipilimumab
- Author
-
Grazia Graziani, Lucia Lisi, Lucio Tentori, and Pierluigi Navarra
- Published
- 2022
- Full Text
- View/download PDF
20. Clinical experience with CTLA-4 blockade for cancer immunotherapy: From the monospecific monoclonal antibody ipilimumab to probodies and bispecific molecules targeting the tumor microenvironment
- Author
-
Maria Martire, Lucia Lisi, Grazia Graziani, Pierluigi Navarra, and Pedro Miguel Lacal
- Subjects
PD-L1 ,CTLA-4 ,Immune checkpoint inhibitors ,Immunotherapy ,Ipilimumab ,Melanoma ,Monoclonal antibodies ,PD-1 ,Settore BIO/14 - FARMACOLOGIA ,medicine.drug_class ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Antineoplastic Agents ,Monoclonal antibody ,Antibodies ,Antineoplastic Agents, Immunological ,Cancer immunotherapy ,Neoplasms ,Antibodies, Bispecific ,medicine ,Tumor Microenvironment ,Animals ,Humans ,CTLA-4 Antigen ,Pharmacology ,Tumor microenvironment ,business.industry ,Settore BIO/14 ,Immune checkpoint ,Immunological ,Cancer research ,Bispecific ,Nivolumab ,business ,medicine.drug - Abstract
The immune checkpoint cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is an inhibitory regulator of T-cell mediated responses that has been investigated as target of monoclonal antibodies (mAbs) for cancer immunotherapy. The anti-CTLA-4 mAb ipilimumab represents the first immune checkpoint inhibitor that significantly improved overall survival in patients with unresectable/metastatic melanoma. The subsequent approved indications (often in the first-line setting) for melanoma and other advanced/metastatic solid tumors always require ipilimumab combination with nivolumab, an anti-programmed cell death protein 1 (PD-1) mAb. However, the improved clinical efficacy of the mAb combination is associated with increased immune-related adverse events, which might require treatment discontinuation even in responding patients. This drawback is expected to be overcome by the recent development of anti-CTLA-4 probodies proteolitycally activated in the tumor microenvironment and bispecific molecules targeting both CTLA-4 and PD-1, whose co-expression is characteristic of tumor-infiltrating T cells. These molecules would preferentially stimulate immune responses against the tumor, reducing toxicity toward normal tissues.
- Published
- 2022
21. Targeting immunoproteasome in neurodegeneration: A glance to the future
- Author
-
Grazia R. Tundo, Paolo Cascio, Danilo Milardi, Anna Maria Santoro, Grazia Graziani, Pedro Miguel Lacal, Alessio Bocedi, Francesco Oddone, Mariacristina Parravano, Andrea Coletta, Massimo Coletta, and Diego Sbardella
- Subjects
Pharmacology ,Immunoproteasome ,Neurodegeneration ,PA28 ,Retina ,Pharmacology (medical) - Abstract
The immunoproteasome is a specialized form of proteasome equipped with modified catalytic subunits that was initially discovered to play a pivotal role in MHC class I antigen processing and immune system modulation. However, over the last years, this proteolytic complex has been uncovered to serve additional functions unrelated to antigen presentation. Accordingly, it has been proposed that immunoproteasome synergizes with canonical proteasome in different cell types of the nervous system, regulating neurotransmission, metabolic pathways and adaptation of the cells to redox or inflammatory insults. Hence, studying the alterations of immunoproteasome expression and activity is gaining research interest to define the dynamics of neuroinflammation as well as the early and late molecular events that are likely involved in the pathogenesis of a variety of neurological disorders. Furthermore, these novel functions foster the perspective of immunoproteasome as a potential therapeutic target for neurodegeneration. In this review, we provide a brain and retina-wide overview, trying to correlate present knowledge on structure-function relationships of immunoproteasome with the variety of observed neuro-modulatory functions.
- Published
- 2023
- Full Text
- View/download PDF
22. A Novel and Atypical Nf-Kb Proinflammatory Program Regulated by A Camkii-Proteasome Axis Is Involved in the Early Activation of Muller Glia by High Glucose
- Author
-
Diego Ph.D. Diego Sba, Grazia Raffaella Tundo, Alice Mecchia, Camilla Palumbo, Maria Grazia Atzori, Lauretta Levati, Anna Maria Caccuri, Paolo Cascio, Pedro Lacal, Grazia Graziani, Monica Varano, Massimiliano Coletta, and Maria Cristina Parravano
- Abstract
BackgroundDiabetic retinopathy (DR) is a microvascular complication of diabetes with a heavy impact on the life-quality of subjects and with a dramatic burden for health and economic systems on a global scale.Although the pathogenesis of DR is largely unknown, several preclinical data have pointed out to a main role of Muller glia, a cell type which spans across the retina layers providing nourishment and support for Retina Ganglion Cells (RGCs), in sensing glycemia and in acquiring a proinflammatory polarization in response to this insult.ResultsBy using a validated experimental model of DR in vitro, the rMC1 cells challenged with high glucose, we uncovered the induction of an early (within minutes) and atypical NF-kB signalling pathway regulated by a CamKII-proteasome axis. Phosphorylation of proteasome subunit Rpt6 (at serine 120) by CamKII stimulated the accelerated turnover of IkBα (i.e., the natural inhibitor of p65-50 transcription factor), regardless of the phosphorylation at serine 32 which labels canonical NF-kB signalling. This event allowed the p65-p50 heterodimer to migrate into the nucleus and to induce the selective transcription of IL-8, Il-1β and MCP-1. Pharmacological inhibition of CamKII or proteasome stopped this proinflammatory program, whereas introduction of a Rpt6 phospho-dead mutant (Rpt6-S120A) stimulated a paradoxical effect on NF-kB probably through the activation of a compensatory mechanism which may involve phosphorylation of 20S α4 subunit.ConclusionsThis study introduces a novel pathway of MG activation by high glucose and casts some light on the biological relevance of proteasome post-translational modifications in modulating pathways regulated through targeted proteolysis.
- Published
- 2021
- Full Text
- View/download PDF
23. A novel and atypical NF-KB pro-inflammatory program regulated by a CamKII-proteasome axis is involved in the early activation of Muller glia by high glucose
- Author
-
Diego Sbardella, Grazia Raffaella Tundo, Alice Mecchia, Camilla Palumbo, Maria Grazia Atzori, Lauretta Levati, Alessandra Boccaccini, Anna Maria Caccuri, Paolo Cascio, Pedro Miguel Lacal, Grazia Graziani, Monica Varano, Massimiliano Coletta, and Mariacristina Parravano
- Subjects
CamKII ,Muller glia ,Proteasome ,Diabetic retinopathy ,Settore BIO/10 ,NF-kB pathway ,General Biochemistry, Genetics and Molecular Biology - Abstract
Background Diabetic retinopathy (DR) is a microvascular complication of diabetes with a heavy impact on the quality of life of subjects and with a dramatic burden for health and economic systems on a global scale. Although the pathogenesis of DR is largely unknown, several preclinical data have pointed out to a main role of Muller glia (MG), a cell type which spans across the retina layers providing nourishment and support for Retina Ganglion Cells (RGCs), in sensing hyper-glycemia and in acquiring a pro-inflammatory polarization in response to this insult. Results By using a validated experimental model of DR in vitro, rMC1 cells challenged with high glucose, we uncovered the induction of an early (within minutes) and atypical Nuclear Factor-kB (NF-kB) signalling pathway regulated by a calcium-dependent calmodulin kinase II (CamKII)-proteasome axis. Phosphorylation of proteasome subunit Rpt6 (at Serine 120) by CamKII stimulated the accelerated turnover of IkBα (i.e., the natural inhibitor of p65-50 transcription factor), regardless of the phosphorylation at Serine 32 which labels canonical NF-kB signalling. This event allowed the p65-p50 heterodimer to migrate into the nucleus and to induce transcription of IL-8, Il-1β and MCP-1. Pharmacological inhibition of CamKII as well as proteasome inhibition stopped this pro-inflammatory program, whereas introduction of a Rpt6 phospho-dead mutant (Rpt6-S120A) stimulated a paradoxical effect on NF-kB probably through the activation of a compensatory mechanism which may involve phosphorylation of 20S α4 subunit. Conclusions This study introduces a novel pathway of MG activation by high glucose and casts some light on the biological relevance of proteasome post-translational modifications in modulating pathways regulated through targeted proteolysis.
- Published
- 2021
24. TRF1 poly(ADP-ribosyl)ation by PARP1 allows proper telomere replication through helicase recruitment in non-ALT cells
- Author
-
A. Dello Stritto, Antonella Sgura, Francesco Berardinelli, Erica Salvati, Luca Pompili, E. Vertecchi, Antonio Antoccia, E. Petti, Grazia Graziani, C. D'Angelo, Carmen Maresca, and Annamaria Biroccio
- Subjects
PARP1 ,biology ,Chemistry ,DNA damage ,Poly ADP ribose polymerase ,biology.protein ,DNA replication ,Helicase ,Shelterin ,Chromatin ,Telomere ,Cell biology - Abstract
Telomeres are nucleoprotein structures at eukaryotic chromosome termini. Their stability is preserved by a six-protein complex named shelterin. Among these, TRF1 binds telomere duplex and assists DNA replication with mechanisms only partly clarified. Poly (ADP-ribose) polymerase 1 (PARP1) is a chromatin associated enzyme which adds poly (ADP-ribose) polymers (PARs) to acceptor proteins by covalent hetero-modification. Here we found that TRF1 is covalently PARylated by PARP1 during DNA synthesis. PARP1 downregulation perturbs bromodeoxyuridine incorporation at telomeres in S-phase, triggering replication-dependent DNA damage and telomere fragility. PARylated TRF1 recruits WRN and BLM helicases in S-phase in a PARP1-dependent manner, probably through non-covalent PAR binding to solve secondary structures during telomere replication. ALT telomeres are less affected by PARP1 downregulation and are less sensitive to PARP inhibitors. This work unveils an unprecedented role for PARP1 as a “surveillant” of telomere replication, in absence of exogenous DNA insults, which orchestrates protein dynamics at proceeding replication fork.
- Published
- 2021
- Full Text
- View/download PDF
25. On the Horizon: Targeting Next-Generation Immune Checkpoints for Cancer Treatment
- Author
-
Grazia Graziani, Grazia R. Tundo, Diego Sbardella, Stefano Marini, and Pedro Miguel Lacal
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Pembrolizumab ,Bioinformatics ,0302 clinical medicine ,Non-small cell lung cancer ,Cancer immunotherapy ,Immunologic ,Neoplasms ,Monoclonal ,Receptors ,Drug Discovery ,Atezolizumab ,Metastatic melanoma ,Nivolumab ,Antibodies, Monoclonal ,Antigens, CD ,CTLA-4 Antigen ,Clinical Trials as Topic ,Hepatitis A Virus Cellular Receptor 2 ,Humans ,Receptors, Immunologic ,Tumor Microenvironment ,Pharmacology (medical) ,Settore BIO/14 ,General Medicine ,Lymphocyte Activation Gene 3 Protein ,CD ,Infectious Diseases ,Oncology ,030220 oncology & carcinogenesis ,medicine.drug_class ,030106 microbiology ,Monoclonal antibody ,Antibodies ,03 medical and health sciences ,Immune system ,TIGIT ,medicine ,Antigens ,Pharmacology ,business.industry ,Cancer ,medicine.disease ,business - Abstract
Background: Immune checkpoints are critical regulatory pathways of the immune system which finely tune the response to biological threats. Among them, the CD-28/CTLA-4 and PD-1/PD-L1 axes play a key role in tumour immune escape and are well-established targets of cancer immunotherapy. Summary: The clinical experience accumulated to date provides unequivocal evidence that anti-CTLA-4, PD-1, or PD-L1 monoclonal antibodies, used as monotherapy or in combination regimes, are effective in a variety of advanced/metastatic types of cancer, with improved clinical outcomes compared to conventional chemotherapy. However, the therapeutic success is currently restricted to a limited subset of patients and reliable predictive biomarkers are still lacking. Key Message: The identification and characterization of additional co-inhibitory pathways as novel pharmacological targets to improve the clinical response in refractory patients has led to the development of different immune checkpoint inhibitors, the activities of which are currently under investigation. In this review, we discuss recent literature data concerning the mechanisms of action of next-generation monoclonal antibodies targeting LAG-3, TIM-3, and TIGIT co-inhibitory molecules that are being explored in clinical trials, as single agents or in combination with other immune-stimulating agents.
- Published
- 2019
- Full Text
- View/download PDF
26. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential
- Author
-
Claudia Ceci, Pedro Miguel Lacal, and Grazia Graziani
- Subjects
Pharmacology ,Immunoconjugates ,Neoplasms ,Settore BIO/14 ,Tumor Microenvironment ,Antibodies, Monoclonal ,Humans ,Monoclonal antibodies ,Antineoplastic Agents ,Pharmacology (medical) ,Immunotherapy ,Cancer - Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
- Published
- 2022
- Full Text
- View/download PDF
27. hTERT Transduction Extends the Lifespan of Primary Pediatric Low-Grade Glioma Cells While Preserving the Biological Response to NGF
- Author
-
Angela Maria Di Francesco, Riccardo Riccardi, Gabriella Cusano, Antonio Ruggiero, Lauretta Levati, Ornella Franzese, Grazia Graziani, and Daniela Meco
- Subjects
0301 basic medicine ,Cancer Research ,Telomerase ,senescence ,Cellular differentiation ,Cell Culture Techniques ,Tropomyosin receptor kinase A ,Pathology and Forensic Medicine ,Transduction ,03 medical and health sciences ,0302 clinical medicine ,Genetic ,Transduction, Genetic ,Glioma ,Nerve Growth Factor ,Tumor Cells, Cultured ,medicine ,Humans ,Telomerase reverse transcriptase ,neoplasms ,Original Research ,low-grade glioma ,Cultured ,biology ,Brain Neoplasms ,NGF (nerve growth factor) ,Settore BIO/14 ,differentiation ,General Medicine ,medicine.disease ,Tumor Cells ,Society Journal Archive ,030104 developmental biology ,Nerve growth factor ,nervous system ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,Oncology ,Cell culture ,biology.protein ,Cancer research ,hTERT ,030217 neurology & neurosurgery ,Neurotrophin - Abstract
The neurotrophin nerve growth factor (NGF) modulates the growth of human gliomas and is able to induce cell differentiation through the engagement of tropomyosin receptor kinase A (TrkA) receptor, although the role played in controlling glioma survival has proved controversial. Unfortunately, the slow growth rate of low-grade gliomas (LGG) has made it difficult to investigate NGF effects on these tumors in preclinical models. In fact, patient-derived low-grade human astrocytoma cells duplicate only a limited number of times in culture before undergoing senescence. Nevertheless, replicative senescence can be counteracted by overexpression of hTERT, the catalytic subunit of telomerase, which potentially increases the proliferative potential of human cells without inducing cancer-associated changes. We have extended, by hTERT transduction, the proliferative in vitro potential of a human LGG cell line derived from a pediatric pilocytic astrocytoma (PA) surgical sample. Remarkably, the hTERT-transduced LGG cells showed a behavior similar to that of the parental line in terms of biological responses to NGF treatment, including molecular events associated with induction of NGF-related differentiation. Therefore, transduction of LGG cells with hTERT can provide a valid approach to increase the in vitro life-span of patient-derived astrocytoma primary cultures, characterized by a finite proliferative potential.
- Published
- 2021
- Full Text
- View/download PDF
28. VEGF-A/VEGFR-1: a painful astrocyte-mediated signaling blocked by the anti-VEGFR-1 mAb D16F7
- Author
-
Elena Lucarini, Carmen Parisio, Carla Ghelardini, Alessandra Pacini, Serena Boccella, Alessia Vona, Alessandra Toti, L. Di Cesare Mannelli, Grazia Graziani, Paola Failli, Tommaso Mello, Flavia Ricciardi, Sabatino Maione, Pedro Miguel Lacal, and Laura Micheli
- Subjects
Gene knockdown ,education.field_of_study ,business.industry ,Population ,Neurotoxicity ,Context (language use) ,Pharmacology ,medicine.disease ,Vascular endothelial growth factor A ,medicine.anatomical_structure ,Mediator ,Neuropathic pain ,medicine ,business ,education ,Astrocyte - Abstract
Chemotherapy-induced neuropathic pain is a clinically relevant adverse effect of several anticancer drugs leading to dose reduction or therapy discontinuation. The lack of knowledge about the mechanisms of neuropathy development and pain chronicization makes chemotherapy-induced neuropathic pain treatment an unmet medical need. In this context, the vascular endothelial growth factor A (VEGF-A) has emerged as a neurotoxicity biomarker in a model of chemotherapy-induced neuropathy, and its decrease has been related to pain relief. Aim of this study was to clarify the VEGF-A-dependent pain signaling in the CNS for individuating new targeted therapeutic approaches. In mice, the intrathecal infusion of VEGF-A induced a dose-dependent noxious hypersensitivity mediated by the VEGF receptor 1 (VEGFR-1) as demonstrated by pharmacological and genetic tools. In electrophysiological study, VEGF-A stimulated the spinal nociceptive neurons activity through VEGFR-1. In the dorsal horn of the spinal cord, VEGF-A increased in astrocytes of animals affected by neuropathy suggesting this cell population as a source of the potent pain mediator. Accordingly, the selective knockdown of astrocytic VEGF-A, by shRNAmir, blocked the development of oxaliplatin-induced neuropathic pain. Besides, the anti-VEGFR-1 mAb D16F7 (previously described as anticancer) effectively relieved neuropathic pain induced by chemotherapeutic agents. In conclusion, astrocyte-released VEGF-A is a new player in the complex neuron-glia network that oversees physiological and pathological pain and D16F7 mAb rises as a potent pain killer strategy.
- Published
- 2021
- Full Text
- View/download PDF
29. At the cutting edge against cancer: A perspective on immunoproteasome and immune checkpoints modulation as a potential therapeutic intervention
- Author
-
Grazia Graziani, Alexey A. Belogurov, Francesco Oddone, Grazia R. Tundo, Diego Sbardella, Anna Kudriaeva, Stefano Marini, and Pedro Miguel Lacal
- Subjects
Cancer Research ,medicine.medical_treatment ,proteasome inhibitors ,Computational biology ,Review ,Major histocompatibility complex ,immunoproteasome ,Immune system ,Antigen ,Medicine ,RC254-282 ,biology ,Antigen processing ,business.industry ,Settore BIO/14 ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,immune checkpoints ,medicine.disease ,ubiquitin–proteasome system ,Oncology ,Proteasome ,Cancer cell ,biology.protein ,immunotherapy ,ubiquitin-proteasome system ,business - Abstract
Simple Summary Immunoproteasome plays a key role in the generation of antigenic peptides. Immune checkpoints therapy is a front-line treatment of advanced/metastatic tumors, and to improve its efficacy, a broader knowledge of the dynamics of antigen repertoire processing by cancer cells is mandatory. The scope of this review is to offer a picture of the role of immunoproteasome in antigen presentation to fuel the hypothesis of novel therapeutic interventions based on the modulation of this proteolytic complex and immune checkpoints. Abstract Immunoproteasome is a noncanonical form of proteasome with enzymological properties optimized for the generation of antigenic peptides presented in complex with class I MHC molecules. This enzymatic property makes the modulation of its activity a promising area of research. Nevertheless, immunotherapy has emerged as a front-line treatment of advanced/metastatic tumors providing outstanding improvement of life expectancy, even though not all patients achieve a long-lasting clinical benefit. To enhance the efficacy of the currently available immunotherapies and enable the development of new strategies, a broader knowledge of the dynamics of antigen repertoire processing by cancer cells is needed. Therefore, a better understanding of the role of immunoproteasome in antigen processing and of the therapeutic implication of its modulation is mandatory. Studies on the potential crosstalk between proteasome modulators and immune checkpoint inhibitors could provide novel perspectives and an unexplored treatment option for a variety of cancers.
- Published
- 2021
30. The Effects of Association of Topical Polydatin Improves the Preemptive Systemic Treatment on EGFR Inhibitors Cutaneous Adverse Reactions
- Author
-
Pietro Sollena, Maria Concetta Romano, Ketty Peris, Elena Campione, Deborah Coletta, Luca Bianchi, Mario Roselli, Mauro Bavetta, Grazia Graziani, Vincenzo Formica, and Dionisio Silvaggio
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Epidermal Growth Factor Receptor Inhibitors ,papulopustular rash ,cutaneous adverse events ,lcsh:Medicine ,Topical Product ,Article ,03 medical and health sciences ,Settore MED/35 ,0302 clinical medicine ,Papulopustular ,Quality of life ,Statistical significance ,polydatin ,Medicine ,Papulopustular rash ,EGFR inhibitors ,business.industry ,lcsh:R ,Cancer ,General Medicine ,medicine.disease ,Dermatology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Settore MED/35 - MALATTIE CUTANEE E VENEREE ,business - Abstract
Epidermal Growth Factor Receptor inhibitors (EGFRi) are approved as therapeutic options in several solid tumors. Cutaneous papulopustular eruption is the most frequent cutaneous adverse-event (AE), usually treated with emollient or corticosteroids according to toxicity grade. Our study evaluated the efficacy and safety of a topical product containing polydatin, a glycosylated polyphenol, natural precursor of resveratrol showing anti-inflammatory and anti-oxidative activities, for the prevention and treatment of skin papulopustular rash in EGFRi-treated patients. Forty oncologic patients treated with EGFRi were enrolled in two groups: group-A, 20 patients with papulopustular AE, and group-B, 20 patients without cutaneous manifestations. The study consisted of twice-daily application of polydatin cream 1.5% (group-A) and 0.8% (group-B) for 6 months. In group-A patients, we observed at week 4 a remarkable improvement of skin manifestation and quality of life evaluated with National-Cancer-Institute-Common-Terminology-Criteria for Adverse-Events (NCI-CTCAE), Dermatology-Life-Quality-Index (DLQI) score and Visual-Analogue-Scale (VAS) pruritus, with a statistical significance of p <, 0.05. None of the patients of group-B developed skin AEs to EGFRi. No cutaneous AEs related to the polydatin product were reported in both groups. Polydatin can be a good topical aid for the prevention and management of papulopustular rash in cancer patients receiving EGFRi, also capable of improving cancer patients&rsquo, quality of life.
- Published
- 2021
31. Targeting Tumor-Associated Macrophages to Increase the Efficacy of Immune Checkpoint Inhibitors: A Glimpse into Novel Therapeutic Approaches for Metastatic Melanoma
- Author
-
Claudia Ceci, Maria Grazia Atzori, Grazia Graziani, and Pedro Miguel Lacal
- Subjects
0301 basic medicine ,PD-L1 ,Cancer Research ,medicine.medical_treatment ,Review ,lcsh:RC254-282 ,VEGFR-1 ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,stomatognathic system ,PD-1 ,medicine ,melanoma ,metastasis ,immune checkpoint ,biology ,CTLA-4 ,immune escape ,macrophages ,business.industry ,Melanoma ,Settore BIO/14 ,Cancer ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,bacteria ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Simple Summary Stimulation of the host immune responses, through the use of biotech drugs that remove a brake on the immune system (immune checkpoint inhibitors), is a current widely used strategy to treat a variety of advanced-stage tumors with impressive outcomes also in patients refractory to standard chemotherapy. However, as in the case of metastatic melanoma, many patients fail to achieve a long-lasting clinical benefit. The aim of this article is to provide an overview of the current scientific evidence concerning the role played by cells of the tumor micro-environment, and in particular tumor-associated M2 macrophages, on the innate or acquired resistance of melanoma to immune checkpoint inhibitors. A special focus will be given to potential therapeutic interventions capable of counteracting tumor ability to evade the control of the immune system in order to enhance the efficacy of immune checkpoint inhibitors. Abstract Immune checkpoint inhibitors (ICIs) represent a promising therapeutic intervention for a variety of advanced/metastatic solid tumors, including melanoma, but in a large number of cases, patients fail to establish a sustained anti-tumor immunity and to achieve a long-lasting clinical benefit. Cells of the tumor micro-environment such as tumor-associated M2 macrophages (M2-TAMs) have been reported to limit the efficacy of immunotherapy, promoting tumor immune evasion and progression. Thus, strategies targeting M2-TAMs have been suggested to synergize with immune checkpoint blockade. This review recapitulates the molecular mechanisms by which M2-TAMs promote cancer immune evasion, with focus on the potential cross-talk between pharmacological interventions targeting M2-TAMs and ICIs for melanoma treatment.
- Published
- 2020
32. PDIA3 Expression in Glioblastoma Modulates Macrophage/Microglia Pro-Tumor Activation
- Author
-
Gabriella Maria Pia Ciotti, Grazia Graziani, Fabio Altieri, Lucia Lisi, Pierluigi Navarra, Marta Chiavari, Francesco Canonico, and Pedro Miguel Lacal
- Subjects
Male ,microglia ,PDIA3 ,lcsh:Chemistry ,STAT3 ,Cohort Studies ,glioma ,Tumor Microenvironment ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,Aged, 80 and over ,Microglia ,Brain Neoplasms ,Settore BIO/14 ,General Medicine ,Middle Aged ,Computer Science Applications ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Disease Progression ,Immunohistochemistry ,Female ,Adult ,Settore BIO/14 - FARMACOLOGIA ,IL6 ,punicalagin ,Protein Disulfide-Isomerases ,Biology ,Catalysis ,Article ,Gene Expression Regulation, Enzymologic ,Inorganic Chemistry ,Glioma ,Parenchyma ,medicine ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,Aged ,Innate immune system ,Endoplasmic reticulum ,Macrophages ,Organic Chemistry ,Macrophage Activation ,medicine.disease ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer research ,Glioblastoma - Abstract
The glioblastoma (GB) microenvironment includes cells of the innate immune system identified as glioma-associated microglia/macrophages (GAMs) that are still poorly characterized. A potential role on the mechanisms regulating GAM activity might be played by the endoplasmic reticulum protein ERp57/PDIA3 (protein disulfide-isomerase A3), the modulation of which has been reported in a variety of cancers. Moreover, by using The Cancer Genome Atlas database, we found that overexpression of PDIA3 correlated with about 55% reduction of overall survival of glioma patients. Therefore, we analyzed the expression of ERp57/PDIA3 using specimens obtained after surgery from 18 GB patients. Immunohistochemical analysis of tumor samples revealed ERp57/PDIA3 expression in GB cells as well as in GAMs. The ERp57/PDIA3 levels were higher in GAMs than in the microglia present in the surrounding parenchyma. Therefore, we studied the role of PDIA3 modulation in microglia&ndash, glioma interaction, based on the ability of conditioned media collected from human GB cells to induce the activation of microglial cells. The results indicated that reduced PDIA3 expression/activity in GB cells significantly limited the microglia pro-tumor polarization towards the M2 phenotype and the production of pro-inflammatory factors. Our data support a role of PDIA3 expression in GB-mediated protumor activation of microglia.
- Published
- 2020
33. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges
- Author
-
Roberto Purrello, Diego Sbardella, Grazia R. Tundo, Giuseppe Grasso, Grazia Graziani, Massimo Coletta, Andrea Coletta, Danilo Milardi, Pedro Miguel Lacal, Anna Maria Santoro, Stefano Marini, and Francesco Oddone
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,SARS-Cov-2 ,Druggability ,Drug Resistance ,Muscle Proteins ,E2F4 Transcription Factor ,Proteasome inhibitors ,Bioinformatics ,Cancer ,Neurodegeneration ,Proteasome ,Article ,Ixazomib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,medicine ,Humans ,Pharmacology (medical) ,Settore BIO/10 ,Pharmacology ,Bortezomib ,Ubiquitin ,Settore BIO/14 ,NF-kappa B ,Neurodegenerative Diseases ,Lipid Droplets ,medicine.disease ,Carfilzomib ,Cyclin-Dependent Kinases ,030104 developmental biology ,Proteostasis ,Drug development ,chemistry ,030220 oncology & carcinogenesis ,Tumor Suppressor Protein p53 ,Holoenzymes ,medicine.drug ,Molecular Chaperones - Abstract
Ubiquitin Proteasome System ( UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs. (C) 2020 Elsevier Inc. All rights reserved.
- Published
- 2020
- Full Text
- View/download PDF
34. Vascular endothelial growth factor receptor 1 in glioblastoma‑associated microglia/macrophages
- Author
-
Marta Chiavari, Lucia Lisi, Pedro Miguel Lacal, Gabriella Maria Pia Ciotti, Grazia Graziani, Federica Ruffini, and Pierluigi Navarra
- Subjects
Adult ,Male ,Vascular Endothelial Growth Factor A ,Angiogenesis ,Glioblastoma ,Macrophages ,Melanoma ,Microglia ,VEGF-A ,VEGFR-1 ,0301 basic medicine ,Cancer Research ,Settore BIO/14 - FARMACOLOGIA ,Bevacizumab ,Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Humans ,Receptor ,Aged ,Cell chemotaxis ,Vascular Endothelial Growth Factor Receptor-1 ,Oncogene ,Brain Neoplasms ,Settore BIO/14 ,Membrane Proteins ,General Medicine ,Middle Aged ,Survival Analysis ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunohistochemistry ,Female ,medicine.drug - Abstract
The anti‑vascular endothelial growth factor‑A (VEGF‑A) monoclonal antibody (mAb) bevacizumab is an FDA‑approved monotherapy for the treatment of recurrent glioblastoma (GB), a highly angiogenic and infiltrative tumour. However, bevacizumab does not increase overall survival and blockade of VEGF‑A/VEGF receptor (VEGFR)‑2 signal transduction is associated with severe adverse effects due to inhibition of physiological angiogenesis. Conversely, VEGFR‑1 does not play a relevant role in physiological angiogenesis in the adult. VEGFR‑1 is activated by both VEGF‑A and placenta growth factor (PlGF), a protein involved in tumour growth and progression. In previous studies, it was demonstrated that inhibition of VEGFR‑1 using a specific mAb developed in our laboratories reduced angiogenesis and GB cell chemotaxis and increased the survival of tumour‑bearing mice. Failure of treatments directed toward the VEGF‑A/VEGFR‑2 axis could in part be due to inefficient targeting of the tumour microenvironment. In the present study, VEGFR‑1 expression was investigated in GB‑associated microglia/macrophages (GAMs) by analysing surgical specimens collected from 42 patients with GB. Data obtained from The Cancer Genome Atlas (TCGA) database revealed that upregulation of the VEGFR‑1 ligands VEGF‑A and PlGF was associated with a significant reduction in overall survival for patients with GB, highlighting the potential relevance of this receptor in the aggressiveness of GB. Immunohistochemical analysis indicated that VEGFR‑1 is expressed not only in GB tissue but also in GAMs. Furthermore, the percentage of VEGFR‑1‑positive GAMs was significantly higher in the tumour region compared with that noted in the surrounding parenchyma. Thus, VEGFR‑1 represents a potential therapeutic target for the treatment of GB, being present not only in GB and endothelial cells, but also in GAMs that are involved in tumour progression.
- Published
- 2020
- Full Text
- View/download PDF
35. Role of VEGFs/VEGFR-1 Signaling and its Inhibition in Modulating Tumor Invasion: Experimental Evidence in Different Metastatic Cancer Models
- Author
-
Grazia Graziani, Claudia Ceci, Pedro Miguel Lacal, and Maria Grazia Atzori
- Subjects
Vascular Endothelial Growth Factor A ,Angiogenesis ,Review ,Biology ,Catalysis ,VEGFR-1 ,VEGF-A ,Metastasis ,Inorganic Chemistry ,Extracellular matrix ,lcsh:Chemistry ,chemistry.chemical_compound ,angiogenesis ,Cell surface receptor ,Neoplasms ,medicine ,melanoma ,Animals ,Humans ,cancer ,metastasis ,Neoplasm Invasiveness ,Physical and Theoretical Chemistry ,Progenitor cell ,Neoplasm Metastasis ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,Cell chemotaxis ,Vascular Endothelial Growth Factor Receptor-1 ,integumentary system ,Organic Chemistry ,Settore BIO/14 ,immune escape ,Cancer ,Flt-1 ,Immune escape ,Melanoma ,PlGF ,General Medicine ,medicine.disease ,Computer Science Applications ,Vascular endothelial growth factor ,chemistry ,lcsh:Biology (General) ,lcsh:QD1-999 ,embryonic structures ,Cancer research ,cardiovascular system ,Signal Transduction - Abstract
The vascular endothelial growth factor (VEGF) family members, VEGF-A, placenta growth factor (PlGF), and to a lesser extent VEGF-B, play an essential role in tumor-associated angiogenesis, tissue infiltration, and metastasis formation. Although VEGF-A can activate both VEGFR-1 and VEGFR-2 membrane receptors, PlGF and VEGF-B exclusively interact with VEGFR-1. Differently from VEGFR-2, which is involved both in physiological and pathological angiogenesis, in the adult VEGFR-1 is required only for pathological angiogenesis. Besides this role in tumor endothelium, ligand-mediated stimulation of VEGFR-1 expressed in tumor cells may directly induce cell chemotaxis and extracellular matrix invasion. Furthermore, VEGFR-1 activation in myeloid progenitors and tumor-associated macrophages favors cancer immune escape through the release of immunosuppressive cytokines. These properties have prompted a number of preclinical and clinical studies to analyze VEGFR-1 involvement in the metastatic process. The aim of the present review is to highlight the contribution of VEGFs/VEGFR-1 signaling in the progression of different tumor types and to provide an overview of the therapeutic approaches targeting VEGFR-1 currently under investigation.
- Published
- 2020
36. Defective proteasome biogenesis into skin fibroblasts isolated from Rett syndrome subjects with MeCP2 non-sense mutations
- Author
-
Diego, Sbardella, Grazia, Raffaella, Tundo, Vincenzo, Cunsolo, Giuseppe, Grasso, Raffaella, Cascella, Valerio, Caputo, Anna Maria, Santoro, Danilo, Mailardi, Pecorelli, Alessandra, Chiara, Ciaccio, Donato, Di Pierro, Silvia, Leoncini, Luisa, Campagnolo, Virginia, Pironi, Francesco, Oddone, Priscilla, Manni, Salvatore, Foti, Emiliano, Giardina, Claudio, De Felice, Joussef, Hayekk, Paolo, Curatolo, Cinzia, Galasso, Valacchi, Giuseppe, Massimiliano, Coletta, Grazia, Graziani, and Stefano, Marini
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Proteasome Endopeptidase Complex ,Rett syndrome,Skin primary fibroblasts, Proteasome,α-Ring, PAC1, PAC2 ,Methyl-CpG-Binding Protein 2 ,Primary Cell Culture ,Rett syndrome ,Protein degradation ,Biology ,NO ,MECP2 ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,medicine ,PAC1 ,PAC2 ,Proteasome ,Skin primary fibroblasts ,α-Ring ,Humans ,Epigenetics ,Settore BIO/10 ,Molecular Biology ,?-Ring ,Skin ,Regulation of gene expression ,Dual Specificity Phosphatase 2 ,Genetic Diseases, X-Linked ,Fibroblasts ,medicine.disease ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,Codon, Nonsense ,Neurodevelopmental Disorders ,Proteolysis ,biology.protein ,Molecular Medicine ,030217 neurology & neurosurgery ,Biogenesis - Abstract
Rett Syndrome (RTT) is a rare X-linked neurodevelopmental disorder which affects about 1: 10000 live births. In >95% of subjects RTT is caused by a mutation in Methyl-CpG binding protein-2 (MECP2) gene, which encodes for a transcription regulator with pleiotropic genetic/epigenetic activities. The molecular mechanisms underscoring the phenotypic alteration of RTT are largely unknown and this has impaired the development of therapeutic approaches to alleviate signs and symptoms during disease progression. A defective proteasome biogenesis into two skin primary fibroblasts isolated from RTT subjects harbouring non-sense (early-truncating) MeCP2 mutations (i.e., R190fs and R255X) is herewith reported. Proteasome is the proteolytic machinery of Ubiquitin Proteasome System (UPS), a pathway of overwhelming relevance for post-mitotic cells metabolism. Molecular, transcription and proteomic analyses indicate that MeCP2 mutations down-regulate the expression of one proteasome subunit, ?7, and of two chaperones, PAC1 and PAC2, which bind each other in the earliest step of proteasome biogenesis. Furthermore, this molecular alteration recapitulates in neuron-like SH-SY5Y cells upon silencing of MeCP2 expression, envisaging a general significance of this transcription regulator in proteasome biogenesis.
- Published
- 2020
- Full Text
- View/download PDF
37. Targeting the vascular endothelial growth factor receptor-1 by the monoclonal antibody D16F7 to increase the activity of immune checkpoint inhibitors against cutaneous melanoma
- Author
-
Rosella Cicconi, Grazia Graziani, Manuel Scimeca, Roberta Bernardini, Pedro Miguel Lacal, Lucio Tentori, Elena Bonanno, Maria Grazia Atzori, Federica Ruffini, Stefania D'Atri, and Maurizio Mattei
- Subjects
0301 basic medicine ,Male ,Skin Neoplasms ,Angiogenesis ,medicine.medical_treatment ,Melanoma, Experimental ,Tregs ,CD8-Positive T-Lymphocytes ,VEGFR-1 ,03 medical and health sciences ,Mice ,Immune checkpoint inhibitors ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,Tumor-Associated Macrophages ,medicine ,Tumor Microenvironment ,Animals ,Humans ,M2 macrophages ,Vasculogenic mimicry ,Melanoma ,Pharmacology ,Tumor microenvironment ,Vascular Endothelial Growth Factor Receptor-1 ,Chemistry ,Monocyte ,Settore BIO/14 ,Antibodies, Monoclonal ,Immunotherapy ,Macrophage Activation ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Tumor progression ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Cancer research ,Monoclonal antibodies - Abstract
The vascular endothelial growth factor receptor-1 (VEGFR-1) is a membrane receptor for VEGF-A, placenta growth factor (PlGF) and VEGF-B that plays a crucial role in melanoma invasiveness, vasculogenic mimicry and tumor-associated angiogenesis. Furthermore, activation of VEGFR-1 is involved in the mobilization of myeloid progenitors from the bone marrow that infiltrate the tumor. Myeloid-derived suppressor cells and tumor-associated macrophages have been involved in tumor progression and resistance to cancer treatment with immune checkpoint inhibitors (ICIs). We have recently demonstrated that the anti-VEGFR-1 monoclonal antibody (mAb) D16F7 developed in our laboratories is able to inhibit melanoma growth in preclinical in vivo models and to reduce monocyte/macrophage progenitor mobilization and tumor infiltration by myeloid cells. Aim of the study was to investigate whether the anti-VEGFR-1 mAb D16F7 affects the activity of protumoral M2 macrophages in vitro in response to PlGF and inhibits the recruitment of these cells to the melanoma site in vivo. Finally, we tested whether, through its multi-targeted action, D16F7 mAb might increase the efficacy of ICIs against melanoma. The results indicated that VEGFR-1 expression is up-regulated in human activated M2 macrophages compared to activated M1 cells and exposure to the D16F7 mAb decreases in vitro chemotaxis of activated M2 macrophages. In vivo treatment with the anti-VEGFR-1 mAb D16F7 of B6D2F1 mice injected with syngeneic B16F10 melanoma cells resulted in tumor growth inhibition associated with the modification of tumor microenvironment that involves a decrease of melanoma infiltration by M2 macrophages and PD-1+ and FoxP3+ cells. These alterations result in increased M1/M2 and CD8+/FoxP3+ ratios, which favor an antitumor and immunostimulating milieu. Accordingly, D16F7 mAb increased the antitumor activity of the ICIs anti-CTLA-4 and anti-PD-1 mAbs. Overall, these data reinforce the role of VEGFR-1-mediated-signalling as a valid target for reducing tumor infiltration by protumoral macrophages and for improving the efficacy of immunotherapy with ICIs.
- Published
- 2020
38. Strategies to improve ellagic acid bioavailability: from natural or semisynthetic derivatives to nanotechnological approaches based on innovative carriers
- Author
-
Isabella Faraoni, Grazia Graziani, Ilaria Cacciotti, and Claudia Ceci
- Subjects
Materials science ,Antioxidant ,Dried fruit ,health promotion ,Chemical structure ,medicine.medical_treatment ,Anti-Inflammatory Agents ,Administration, Oral ,Biological Availability ,Bioengineering ,Antineoplastic Agents ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Antioxidants ,chemistry.chemical_compound ,Pharmacokinetics ,ellagic acid ,medicine ,melanoma ,Animals ,Humans ,Nanotechnology ,cancer ,General Materials Science ,Food science ,Electrical and Electronic Engineering ,Drug Carriers ,Mechanical Engineering ,polymeric nano ,Settore BIO/14 ,General Chemistry ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,Bioavailability ,chemistry ,Mechanics of Materials ,Polyphenol ,Fruit ,Drug delivery ,drug delivery ,and microspheres ,0210 nano-technology ,bioavailability ,Ellagic acid - Abstract
Ellagic acid (EA) is a polyphenolic compound whose dietary consumption is mainly associated with the intake of red fruits, including pomegranates, strawberries, blackberries, blackcurrants, raspberries, grapes or dried fruits, like walnuts and almonds. A number of studies indicate that EA exerts health-beneficial effects against several chronic pathologies associated with oxidative damage, including different kinds of cancer, cardiovascular and neurodegenerative diseases. Furthermore, EA possesses wound-healing properties, antibacterial and antiviral effects, and acts as a systemic antioxidant. However, clinical applications of this polyphenol have been hampered and prevented by its poor water solubility (9.7 ± 3.2 μg ml-1 in water) and pharmacokinetic profile (limited absorption rate and plasma half-life
- Published
- 2020
39. DNA inhibitors for the treatment of brain tumors
- Author
-
Grazia Graziani, Marta Chiavari, Pierluigi Navarra, Gabriella Maria Pia Ciotti, Lucia Lisi, and Pedro Miguel Lacal
- Subjects
Drug ,Antimetabolites, Antineoplastic ,Settore BIO/14 - FARMACOLOGIA ,Topoisomerase Inhibitors ,DNA damage ,medicine.medical_treatment ,media_common.quotation_subject ,Central nervous system ,DNA inhibitors ,Glioblastoma ,anti-VEGF therapy ,brain tumors ,chemotherapy ,clinical trials ,immune checkpoint inhibitors ,monoclonal antibodies ,temozolomide ,Antineoplastic Agents ,Toxicology ,Bioinformatics ,Tumor Microenvironment ,medicine ,Animals ,Humans ,media_common ,Pharmacology ,Chemotherapy ,Tumor microenvironment ,Temozolomide ,Brain Neoplasms ,business.industry ,Settore BIO/14 ,DNA ,General Medicine ,Clinical trial ,medicine.anatomical_structure ,Cancer cell ,business ,medicine.drug - Abstract
Introduction: The worldwide incidence of central nervous system (CNS) primary tumors is increasing. Most of the chemotherapeutic agents used for treating these cancer types induce DNA damage, and their activity is affected by the functional status of repair systems involved in the detection or correction of DNA lesions. Unfortunately, treatment of malignant high-grade tumors is still an unmet medical need.Areas covered: We summarize the action mechanisms of the main DNA inhibitors used for the treatment of brain tumors. In addition, studies on new agents or drug combinations investigated for this indication are reviewed, focusing our attention on clinical trials that in the last 3 years have been completed, terminated or are still recruiting patients.Expert opinion: Much still needs to be done to render aggressive CNS tumors curable or at least to transform them from lethal to chronic diseases, as it is possible for other cancer types. Drugs with improved penetration in the CNS, toxicity profile, and activity against primary and recurrent tumors are eagerly needed. Targeted agents with innovative mechanisms of action and ability to harness the cells of the tumor microenvironment against cancer cells represent a promising approach for improving the clinical outcome of CNS tumors.
- Published
- 2020
40. Approaching coronavirus disease 2019: Mechanisms of action of repurposed drugs with potential activity against SARS-CoV-2
- Author
-
Pedro Miguel Lacal, Grazia Graziani, Maria Luisa Barbaccia, and Lucia Lisi
- Subjects
MSCs, mesenchymal stem cells ,CoV, coronavirus ,Biochemistry ,AAK1, AP2-associated kinase 1 ,0302 clinical medicine ,HBV, hepatitis B ,Medicine ,NF-kB, nuclear factor-kB ,CAR, chimeric antigen receptors ,media_common ,Coronavirus ,Settore BIO/14 ,RBD, receptor-binding domain ,3CLpro, chymotrypsin‐like protease ,MERS-CoV, Middle East respiratory syndrome coronavirus ,CT, computed tomography ,STAT, signal transducer and activator of transcription ,TNFα, tumor necrosis factor α ,Outcome and Process Assessment, Health Care ,Cardiovascular Diseases ,sHLH, secondary hemophagocytic lymphohistiocytosis ,030220 oncology & carcinogenesis ,S1P, sphingosine 1-phosphate ,Cytokines ,G-CSF, granulocyte-colony stimulating factor ,PDE-5, phosphodiesterase-5 ,Drug ,S, spike ,medicine.medical_specialty ,2019-nCoV, 2019 novel coronavirus ,media_common.quotation_subject ,Pneumonia, Viral ,E, envelope ,COVID-19, Corona Virus Disease 2019 ,ACE2, angiotensin-converting enzyme 2 ,Article ,Betacoronavirus ,03 medical and health sciences ,Viral entry ,VEGF-A, vascular endothelial growth factor-A ,CAS, Chemical Abstracts Service ,Humans ,SARS, severe acute respiratory syndrome ,DAMPs, danger-associated molecular patterns ,mAb, monoclonal antibody ,PLpro, papain‐like protease ,Intensive care medicine ,ARDS, acute respiratory distress syndrome ,Pharmacology ,IRF3, interferon regulatory factor 3 ,Pneumonia ,medicine.disease ,IL, interleukin ,Health Care ,Clinical trial ,030104 developmental biology ,nsps, nonstructural proteins ,JAK, Janus kinases ,0301 basic medicine ,HCV, hepatitis C ,tPa, tissue-type plasminogen activator ,ADE, antibody-dependent enhancement, ALI, acute lung injury ,medicine.disease_cause ,Cytokine storm ,ORF, open reading frames ,TMPRSS2, transmembrane protease serine 2 protease ,PRRs, pattern recognition receptors ,Pandemic ,Viral ,MRSA, methicillin-resistant Staphylococcus Aureus ,Clinical Trials as Topic ,biology ,ICU, intensive care unit ,Drug repositioning ,MIP1α, macrophage inflammatory protein ,Coronavirus Infections ,TLR, Toll-like receptor ,Settore BIO/14 - FARMACOLOGIA ,Outcome and Process Assessment ,ERGIC, endoplasmic reticulum-Golgi apparatus intermediate compartment ,M, membrane ,Animals ,UFH, unfractionated heparin ,Pandemics ,ComputingMethodologies_COMPUTERGRAPHICS ,business.industry ,SARS-CoV-2 ,VIP, vasoactive intestinal polypeptide ,COVID-19 ,MCP, monocyte chemoattractant protein ,biology.organism_classification ,LMWH, low molecular weight heparin ,COVID-19 Drug Treatment ,GAK, cyclin G–associated kinase ,Mpro, main protease ,Antiviral agents ,HScore, hemophagocytosis score ,business ,IP-10, interferon-γ-inducible protein 10 ,N, nucleocapsid - Abstract
Graphical abstract, On March 11, 2020, the World Health Organization (WHO) declared the severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2) a global pandemic. As of July 2020, SARS-CoV-2 has infected more than 14 million people and provoked more than 590,000 deaths, worldwide. From the beginning, a variety of pharmacological treatments has been empirically used to cope with the life-threatening complications associated with Corona Virus Disease 2019 (COVID-19). Thus far, only a couple of them and not consistently across reports have been shown to further decrease mortality, respect to what can be achieved with supportive care. In most cases, and due to the urgency imposed by the number and severity of the patients’ clinical conditions, the choice of treatment has been limited to repurposed drugs, approved for other indications, or investigational agents used for other viral infections often rendered available on a compassionate-use basis. The rationale for drug selection was mainly, though not exclusively, based either i) on the activity against other coronaviruses or RNA viruses in order to potentially hamper viral entry and replication in the epithelial cells of the airways, and/or ii) on the ability to modulate the excessive inflammatory reaction deriving from dysregulated host immune responses against the SARS-CoV-2. In several months, an exceptionally large number of clinical trials have been designed to evaluate the safety and efficacy of anti-COVID-19 therapies in different clinical settings (treatment or pre- and post-exposure prophylaxis) and levels of disease severity, but only few of them have been completed so far. This review focuses on the molecular mechanisms of action that have provided the scientific rationale for the empirical use and evaluation in clinical trials of structurally different and often functionally unrelated drugs during the SARS-CoV-2 pandemic.
- Published
- 2020
41. Therapeutic implication of vascular endothelial growth factor receptor-1 (VEGFR-1) targeting in cancer cells and tumor microenvironment by competitive and non-competitive inhibitors
- Author
-
Pedro Miguel Lacal and Grazia Graziani
- Subjects
0301 basic medicine ,Angiogenesis ,Axitinib: (PubMED CID: 6450551) ,Bevacizumab: (PubMED CID: 24801580) ,Cabozantinib: (PubMED CID: 25102847) ,Immune checkpoint inhibitors ,Lenvatinib: (PubMED CID: 9823820) ,Melanoma ,Nintedanib: (PubMED CID: 9809715) ,Non-small cell lung cancer ,Pazopanib: (PubMED CID: 10113978) ,Regorafenib: (PubMED CID: 11167602) ,Sorafenib: (PubMED CID: 216239) ,Sunitinib: (PubMed CID: 5329102) ,Tumor-associated ,Vandetanib: (PubMed CID: 3081361) ,Vascular endothelial growth factor receptor-1 ,macrophages ,medicine.medical_treatment ,Antineoplastic Agents ,Receptor tyrosine kinase ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Myeloid Cells ,Vasculogenic mimicry ,Pharmacology ,Tumor microenvironment ,Vascular Endothelial Growth Factor Receptor-1 ,integumentary system ,biology ,Chemistry ,Growth factor ,Settore BIO/14 ,Vascular endothelial growth factor ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,embryonic structures ,cardiovascular system ,biology.protein ,Cancer research ,Tyrosine kinase - Abstract
The vascular endothelial growth factor receptor-1 (VEGFR-1) is a tyrosine kinase receptor for VEGF-A, VEGF-B, and placental growth factor (PlGF) ligands that is expressed in endothelial, myelomonocytic and tumor cells. VEGF-B and PlGF exclusively bind to VEGFR-1, whereas VEGF-A also binds to VEGFR-2. At variance with VEGFR-2, VEGFR-1 does not play a relevant role in physiological angiogenesis in the adult, while it is important in tumor-associated angiogenesis. VEGFR-1 and PlGF are expressed in a variety of tumors, promote invasiveness and contribute to resistance to anti-VEGF-A therapy. The currently approved antiangiogenic therapies for the treatment of a variety of solid tumors hamper VEGF-A signaling mediated by both VEGFR-2 and VEGFR-1 [i.e., the monoclonal antibody (mAb) anti-VEGF-A bevacizumab, the chimeric molecule aflibercept and several small molecule tyrosine kinase inhibitors] or exclusively by VEGFR-2 (i.e., the mAb anti-VEGFR-2 ramucirumab). However, molecules that interfere with VEGF-A/VEGFR-2 signaling determine severe adverse effects due to inhibition of physiological angiogenesis and their efficacy is hampered by tumor infiltration of protumoral myeloid cells. Blockade of VEGFR-1 may exert anti-tumor activity by multiple mechanisms: a) inhibition of tumor-associated angiogenesis; b) reduction of myeloid progenitor mobilization and tumor infiltration by VEGFR-1 expressing M2 macrophages, which contribute to tumor progression and spreading; c) inhibition of invasiveness, vasculogenic mimicry and survival of VEGFR-1 positive tumor cells. As a consequence of these properties, molecules targeting VEGFR-1 are expected to produce less adverse effects and to counteract resistance towards anti-VEGF-A therapies. More interestingly, selective VEGFR-1 inhibition might enhance the efficacy of immunotherapy with immune checkpoint inhibitors. In this review, we will examine the experimental evidence available so far that supports targeting VEGFR-1 signal transduction pathway for cancer treatment by competitive inhibitors that prevent growth factor interaction with the receptor and non-competitive inhibitors that hamper receptor activation without affecting ligand binding.
- Published
- 2018
- Full Text
- View/download PDF
42. Modulation of GDF11 expression and synaptic plasticity by age and training
- Author
-
Paola Grimaldi, Grazia Graziani, Giovanna D'Arcangelo, Emanuela De Domenico, Lucio Tentori, Mattia Palmieri, Isabella Faraoni, and Virginia Tancredi
- Subjects
0301 basic medicine ,medicine.medical_specialty ,hippocampus ,Population ,Physical exercise ,Biology ,Stimulus (physiology) ,Settore BIO/09 ,GDF11 ,Gerotarget ,exercise ,long-term potentiation ,skeletal muscle ,03 medical and health sciences ,Research Paper: Gerotarget (Focus on Aging) ,0302 clinical medicine ,Internal medicine ,medicine ,Hippocampus (mythology) ,education ,education.field_of_study ,Settore BIO/14 ,Skeletal muscle ,Long-term potentiation ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Synaptic plasticity ,Immunology ,030217 neurology & neurosurgery - Abstract
The Growth Differentiation Factor 11 (GDF11) has been controversially involved in the aging/rejuvenation process. To clarify whether GDF11 is differently expressed during aging, we have evaluated GDF11 levels in skeletal muscles and hippocampi of young and old mice, sedentary or subjected to a 12-weeks triweekly training protocol. The results of real-time PCR and Western blot analyses indicate that skeletal muscles of sedentary old mice express higher levels of GDF11 compared to young animals (p < 0.05). Conversely, in hippocampi no significant differences of GDF11 expression are detected. Analysis of long-term potentiation, a synaptic plasticity phenomenon, reveals that population spikes in response to a tetanic stimulus are significantly higher in sedentary young mice than in old animals (p < 0.01). Training induces a significant improvement of long-term potentiation in both young and old animals (p < 0.05), an increase (p < 0.05) of skeletal muscle GDF11 levels in young mice and a reduction of GDF11 expression in hippocampi of old mice (p < 0.05). Overall, data suggest that GDF11 can be considered an aging biomarker for skeletal muscles. Moreover, physical exercise has a positive impact on long-term potentiation in both young and old mice, while it has variable effects on GDF11 expression depending on age and on the tissue analyzed.
- Published
- 2017
- Full Text
- View/download PDF
43. Role of VEGFR-1 in melanoma acquired resistance to the BRAF inhibitor vemurafenib
- Author
-
Claudia Ceci, Maria Luisa Barbaccia, Pedro Miguel Lacal, Maria Grazia Atzori, Stefania D'Atri, Mauro Trapani, Grazia Graziani, Lucio Tentori, and Federica Ruffini
- Subjects
0301 basic medicine ,Placental growth factor ,Vascular Endothelial Growth Factor A ,VEGF‐A ,Skin Neoplasms ,Angiogenesis ,Receptor tyrosine kinase ,VEGF-A ,chemistry.chemical_compound ,angiogenesis ,0302 clinical medicine ,Vemurafenib ,biology ,Melanoma ,Settore BIO/14 ,BRAF inhibitors ,Transfection ,Extracellular Matrix ,Vascular endothelial growth factor ,Phenotype ,030220 oncology & carcinogenesis ,embryonic structures ,Molecular Medicine ,Original Article ,medicine.drug ,Proto-Oncogene Proteins B-raf ,VEGFR‐1 ,VEGFR-1 ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,melanoma ,Gene silencing ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Protein Kinase Inhibitors ,Cell Proliferation ,Placenta Growth Factor ,Vascular Endothelial Growth Factor Receptor-1 ,business.industry ,Cell Biology ,Original Articles ,medicine.disease ,030104 developmental biology ,chemistry ,vemurafenib ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,business - Abstract
The vascular endothelial growth factor receptor‐1 (VEGFR‐1) is a tyrosine kinase receptor frequently expressed in melanoma. Its activation by VEGF‐A or placental growth factor (PlGF) promotes tumour cell survival, migration and invasiveness. Moreover, VEGFR‐1 stimulation contributes to pathological angiogenesis and induces recruitment of tumour‐associated macrophages. Since melanoma acquired resistance to BRAF inhibitors (BRAFi) has been associated with activation of pro‐angiogenic pathways, we have investigated VEGFR‐1 involvement in vemurafenib resistance. Results indicate that human melanoma cells rendered resistant to vemurafenib secrete greater amounts of VEGF‐A and express higher VEGFR‐1 levels compared with their BRAFi‐sensitive counterparts. Transient VEGFR‐1 silencing in susceptible melanoma cells delays resistance development, whereas in resistant cells it increases sensitivity to the BRAFi. Consistently, enforced VEGFR‐1 expression, by stable gene transfection in receptor‐negative melanoma cells, markedly reduces sensitivity to vemurafenib. Moreover, melanoma cells expressing VEGFR‐1 are more invasive than VEGFR‐1 deficient cells and receptor blockade by a specific monoclonal antibody (D16F7 mAb) reduces extracellular matrix invasion triggered by VEGF‐A and PlGF. These data suggest that VEGFR‐1 up‐regulation might contribute to melanoma progression and spreading after acquisition of a drug‐resistant phenotype. Thus, VEGFR‐1 inhibition with D16F7 mAb might be a suitable adjunct therapy for VEGFR‐1 positive tumours with acquired resistance to vemurafenib.
- Published
- 2019
44. Targeting ADP-ribosylation by PARP inhibitors in acute myeloid leukaemia and related disorders
- Author
-
Francesco Lo-Coco, Isabella Faraoni, Manuela Giansanti, Maria Teresa Voso, and Grazia Graziani
- Subjects
0301 basic medicine ,Genome instability ,Myeloid ,Synthetic lethality ,DNA Repair ,DNA repair ,Poly ADP ribose polymerase ,AML ,DNA damage repair ,Myeloproliferative neoplasms ,PARP inhibitors ,Gene mutation ,Biology ,Poly(ADP-ribose) Polymerase Inhibitors ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,ADP-Ribosylation ,Drug Delivery Systems ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Pharmacology ,Clinical Trials as Topic ,Settore BIO/14 ,Settore MED/15 ,Fusion protein ,Leukemia, Myeloid, Acute ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Poly(ADP-ribose) Polymerases - Abstract
Acute myeloid leukaemia (AML) is a highly heterogeneous disease characterized by uncontrolled proliferation, block in myeloid differentiation and recurrent genetic abnormalities. In the search of new effective therapies, identification of synthetic lethal partners of AML genetic alterations might represent a suitable approach to tailor patient treatment. Genetic mutations directly affecting DNA repair genes are not commonly present in AML. Nevertheless, several studies indicate that AML cells show high levels of DNA lesions and genomic instability. Leukaemia-driving oncogenes (e.g., RUNX1-RUNXT1, PML-RARA, TCF3-HLF, IDH1/2, TET2) or treatment with targeted agents directed against aberrant kinases (e.g., JAK1/2 and FLT3 inhibitors) have been associated with reduced DNA repair gene expression/activity that would render leukaemia blasts selectively sensitive to synthetic lethality induced by poly(ADP-ribose) polymerase inhibitors (PARPi). Thus, specific oncogenic chimeric proteins or gene mutations, rare or typically distinctive of certain leukaemia subtypes, may allow tagging cancer cells for destruction by PARPi. In this review, we will discuss the rationale for using PARPi in AML subtypes characterized by a specific genetic background and summarize the preclinical and clinical evidence reported so far on their activity when used as single agents or in combination with classical cytotoxic chemotherapy or with agents targeting AML-associated mutated proteins.
- Published
- 2019
45. Effects of Glutathione Transferase-Targeting Nitrobenzoxadiazole Compounds in Relation to PD-L1 Status in Human Melanoma Cells
- Author
-
Camilla Palumbo, Lucio Tentori, Dante Rotili, Grazia Graziani, Chiara Fulci, Francesca Sciarretta, and Anna Maria Caccuri
- Subjects
0301 basic medicine ,Proto-Oncogene Proteins B-raf ,PD-L1 ,tumor ,030106 microbiology ,Population ,Cell ,Sulforhodamine B ,NBDHEX ,B7-H1 Antigen ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Western blot ,Cell Line, Tumor ,Drug Discovery ,medicine ,Autophagy ,Cytotoxic T cell ,Humans ,Pharmacology (medical) ,Settore BIO/10 ,education ,Vemurafenib ,Melanoma ,Nitrobenzenes ,Chloroquine ,Glutathione Transferase ,Pharmacology ,education.field_of_study ,Oxadiazoles ,medicine.diagnostic_test ,Chemistry ,autophagy ,chloroquine ,melanoma ,vemurafenib ,B7-H1 antigen ,cell line, tumor ,gene expression regulation ,glutathione transferase ,humans ,nitrobenzenes ,oxadiazoles ,proto-oncogene proteins B-raf ,General Medicine ,cell line ,medicine.disease ,Infectious Diseases ,medicine.anatomical_structure ,Oncology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Cancer research ,medicine.drug - Abstract
Background: PD-L1 is a membrane protein with inhibitory effects on immune responses, whose expression has been correlated with high aggressiveness and the propensity of melanoma to metastasize. The nitrobenzoxadiazole (NBD) NBDHEX and its analog MC3181 are endowed with strong antitumor activity towards melanoma and a significant ability to reduce its adhesion and invasiveness. Therefore, we investigated whether PD-L1 status could affect cell sensitivity to the cytotoxic effects of NBDs. We then evaluated the effects of NBDHEX on PD-L1 expression and autophagy in melanoma cells. We used the BRAF-mutated A375 melanoma cell line and an A375 variant population enriched for PD-L1+ cells as a model. The cytotoxic effects of NBDs were evaluated in comparison to those of the BRAF inhibitor vemurafenib and the autophagy inhibitor chloroquine. Methods: The effect of NBDHEX on autophagy was determined by measuring LC3-II and p62 protein levels by Western blot. The cytotoxic activity of the compounds was evaluated by sulforhodamine B assay. PD-L1 expression and plasma membrane localization were analyzed by FACS and Western blot analysis. Results: NBDHEX behaves as a late-autophagy inhibitor in A375 melanoma cells, as previously found in other tumor cell lines. NBDHEX and MC3181 showed strong and comparable cytotoxic activity in both parental and PD-L1+ A375 cells, with IC50 values in the sub-micromolar range. Conversely, cells sorted for high PD-L1 expression had lower sensitivity to both the BRAF inhibitor vemurafenib and the autophagy inhibitor chloroquine. NBDHEX treatment did not change the total expression and cell surface localization of PD-L1 in both parental and PD-L1+ A375 cells. Conclusions: Our data suggest that NBDs may represent a promising treatment strategy for melanoma with elevated PD-L1 expression.
- Published
- 2019
46. Antitumor activity of a novel anti-vascular endothelial growth factor receptor-1 monoclonal antibody that does not interfere with ligand binding
- Author
-
Pedro Miguel Lacal, Stefania D'Atri, Veronica Morea, Cristina Maria Failla, Elena Bonanno, Manuel Scimeca, Grazia Graziani, Federica Ruffini, Maria Grazia Atzori, Annalisa Susanna Dorio, and Lucio Tentori
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Angiogenesis ,Melanoma, Experimental ,Angiogenesis Inhibitors ,Ligands ,Monocytes ,Mice ,angiogenesis ,chemistry.chemical_compound ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Cell Movement ,Medicine ,Phosphorylation ,Receptor ,Neovascularization, Pathologic ,Melanoma ,Settore BIO/14 ,monocyte/macrophage ,Vascular endothelial growth factor ,Endothelial stem cell ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Female ,Tyrosine kinase ,Protein Binding ,Research Paper ,VEGFR-1 ,03 medical and health sciences ,Cell Line, Tumor ,Human Umbilical Vein Endothelial Cells ,melanoma ,Animals ,Humans ,PlGF ,Vasculogenic mimicry ,Vascular Endothelial Growth Factor Receptor-1 ,business.industry ,Macrophages ,Monocyte ,Membrane Proteins ,Hematopoietic Stem Cells ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Immunology ,Cancer research ,Protein Multimerization ,business - Abstract
Vascular endothelial growth factor receptor-1 (VEGFR-1) is a tyrosine kinase transmembrane receptor that has also a soluble isoform containing most of the extracellular ligand binding domain (sVEGFR-1). VEGF-A binds to both VEGFR-2 and VEGFR-1, whereas placenta growth factor (PlGF) interacts exclusively with VEGFR-1. In this study we generated an anti-VEGFR-1 mAb (D16F7) by immunizing BALB/C mice with a peptide that we had previously reported to inhibit angiogenesis and endothelial cell migration induced by PlGF. D16F7 did not affect binding of VEGF-A or PlGF to VEGFR-1, thus allowing sVEGFR-1 to act as decoy receptor for these growth factors, but it hampered receptor homodimerization and activation. D16F7 inhibited both the chemotactic response of human endothelial, myelomonocytic and melanoma cells to VEGFR-1 ligands and vasculogenic mimicry by tumor cells. Moreover, D16F7 exerted in vivo antiangiogenic effects in a matrigel plug assay. Importantly, D16F7 inhibited tumor growth and was well tolerated by B6D2F1 mice injected with syngeneic B16F10 melanoma cells. The antitumor effect was associated with melanoma cell apoptosis, vascular abnormalities and decrease of both monocyte/macrophage infiltration and myeloid progenitor mobilization. For all the above, D16F7 may be exploited in the therapy of metastatic melanoma and other tumors or pathological conditions involving VEGFR-1 activation.
- Published
- 2016
- Full Text
- View/download PDF
47. Placenta growth factor and neuropilin-1 collaborate in promoting melanoma aggressiveness
- Author
-
Cristina Fortes, Alessandro Scoppola, Paolo Marchetti, Elena Pagani, Pedro Miguel Lacal, Gian Carlo Antonini Cappellini, Stefania D'Atri, Federica Ruffini, and Grazia Graziani
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Drug Resistance ,Aged ,Bevacizumab ,Cell Line, Tumor ,Cell Movement ,Humans ,Melanoma ,Middle Aged ,Neoplasm Metastasis ,Neuropilin-1 ,Placenta Growth Factor ,Vascular Endothelial Growth Factor Receptor-1 ,Young Adult ,Drug Resistance, Neoplasm ,Biology ,Receptor tyrosine kinase ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neuropilin 1 ,medicine ,Vasculogenic mimicry ,Tumor ,Oncogene ,Settore BIO/14 ,Cell cycle ,medicine.disease ,Molecular medicine ,Vascular endothelial growth factor ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,biology.protein ,Neoplasm - Abstract
The placenta growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family, which shares with VEGF-A the tyrosine kinase receptor VEGFR-1 and the co-receptor neuropilin-1 (NRP-1). In melanoma models, PlGF enhances tumour growth and neovessel formation, whereas NRP-1 promotes the metastatic process. Increased secretion of PlGF and expression of NRP-1 have also been involved in intrinsic or acquired resistance to anti‑angiogenic therapies. In this study we investigated whether PlGF and NRP-1 cooperate in promoting melanoma aggressiveness independently of VEGFR-1. For this purpose, the melanoma cell clones M14-N, expressing NRP-1 and lacking VEGFR-1, and M14-C, devoid of both receptors, were used. M14-N cells are characterized by an invasive phenotype and vasculogenic mimicry, whereas M14-C cells possess a negligible invasive capacity. The results indicated that M14-N cells secrete higher levels of PlGF than M14-C cells and that PlGF is involved in the invasion of the extracellular matrix (ECM) and vasculogenic mimicry of M14-N cells. In fact, neutralizing antibodies against PlGF reverted ECM invasion in response to PlGF and markedly reduced the formation of tubule-like structures. Moreover, M14-N cells migrated in response to PlGF and chemotaxis was specifically abrogated by anti-NRP-1 antibodies, demonstrating that PlGF directly activates NRP-1 in the absence of VEGFR-1. We also compared the levels of PlGF in the plasma of patients affected by metastatic melanoma with those of healthy donors and evaluated whether PlGF levels could be affected by a bevacizumab-containing chemotherapy regimen. Melanoma patients showed a 20-fold increase in plasma PlGF and the bevacizumab-containing regimen induced a reduction of VEGF-A and in a further increase of PlGF. In conclusion, our studies suggest that the activation of NRP-1 by PlGF directly contributes to melanoma aggressiveness and represents a potential compensatory pro-angiogenic mechanism that may contribute to the resistance to therapies targeting VEGF-A.
- Published
- 2016
- Full Text
- View/download PDF
48. Experimental Evidence of the Antitumor, Antimetastatic and Antiangiogenic Activity of Ellagic Acid
- Author
-
Roberto Miano, Grazia Graziani, Pedro Miguel Lacal, Maria Gabriella De Martino, Lucio Tentori, and Claudia Ceci
- Subjects
0301 basic medicine ,Antioxidant ,Dried fruit ,Angiogenesis ,medicine.medical_treatment ,Antineoplastic Agents ,lcsh:TX341-641 ,Angiogenesis Inhibitors ,colorectal cancer ,Review ,03 medical and health sciences ,chemistry.chemical_compound ,angiogenesis ,0302 clinical medicine ,breast cancer ,Ellagic Acid ,In vivo ,polyphenolic compounds ,Phytogenic ,medicine ,melanoma ,bladder cancer ,metastases ,non-small cell lung cancer (NSCLC) ,ovarian cancer ,prostate cancer ,Animals ,Antineoplastic Agents, Phytogenic ,Nutrition and Dietetics ,Chemistry ,Settore BIO/14 ,medicine.disease ,In vitro ,Settore MED/24 ,030104 developmental biology ,Polyphenol ,030220 oncology & carcinogenesis ,Cancer research ,Ovarian cancer ,lcsh:Nutrition. Foods and food supply ,Food Science ,Ellagic acid - Abstract
Ellagic acid (EA) is a naturally occurring polyphenolic compound endowed with strong antioxidant and anticancer properties that is present in high quantity in a variety of berries, pomegranates, and dried fruits. The antitumor activity of EA has been mostly attributed to direct antiproliferative and apoptotic effects. Moreover, EA can inhibit tumour cell migration, extra-cellular matrix invasion and angiogenesis, all processes that are crucial for tumour infiltrative behaviour and the metastatic process. In addition, EA may increase tumour sensitivity to chemotherapy and radiotherapy. The aim of this review is to summarize the in vitro and in vivo experimental evidence supporting the anticancer activity of pure EA, its metabolites, and EA-containing fruit juice or extracts in a variety of solid tumour models. The EA oral administration as supportive therapy to standard chemotherapy has been recently evaluated in small clinical studies with colorectal or prostate cancer patients. Novel formulations with improved solubility and bioavailability are expected to fully develop the therapeutic potential of EA derivatives in the near future.
- Published
- 2018
49. Beneficial and Detrimental Effects of Antiretroviral Therapy on HIV-Associated Immunosenescence
- Author
-
Maria Luisa Barbaccia, Ornella Franzese, Grazia Graziani, and Enzo Bonmassar
- Subjects
0301 basic medicine ,CD28 ,Immunosenescence ,T cells ,Inflammation ,Antiretroviral therapy ,HIV ,Telomerase ,Virus ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Drug Discovery ,Medicine ,Pharmacology (medical) ,030212 general & internal medicine ,Pharmacology ,business.industry ,Incidence (epidemiology) ,Settore BIO/14 ,General Medicine ,030104 developmental biology ,Infectious Diseases ,Oncology ,Ageing ,Immunology ,Life expectancy ,medicine.symptom ,business - Abstract
Since the introduction of highly active antiretroviral therapy more than 2 decades ago, HIV-related deaths have dramatically decreased and HIV infection has become a chronic disease. Due to the inability of antiretroviral drugs to eradicate the virus, treatment of HIV infection requires a systemic lifelong therapy. However, even when successfully treated, HIV patients still show increased incidence of age-associated co-morbidities compared with uninfected individuals. Virus- induced immunosenescence, a process characterized by a progressive decline of immune system function, contributes to the premature ageing observed in HIV patients. Although antiretroviral therapy has significantly improved both the quality and length of patient lives, the life expectancy of treated patients is still shorter compared with that of uninfected individuals. In particular, while antiretroviral therapy can contrast some features of HIV-associated immunosenescence, several anti-HIV agents may themselves contribute to other aspects of immune ageing. Moreover, older HIV patients tend to have a worse immunological response to the antiviral therapy. In this review we will examine the available evidence on the role of antiretroviral therapy in the control of the main features regulating immunosenescence.
- Published
- 2018
50. Poly(ADP-ribose) polymerase inhibitor olaparib hampers placental growth factor-driven activation of myelomonocytic cells
- Author
-
Pedro Miguel Lacal, Maria Grazia Atzori, Federica Ruffini, Grazia Graziani, and Lucio Tentori
- Subjects
0301 basic medicine ,Cancer Research ,cell migration ,Angiogenesis ,Poly ADP ribose polymerase ,Cellular differentiation ,Apoptosis ,HL-60 Cells ,Poly(ADP-ribose) Polymerase Inhibitors ,Poly (ADP-Ribose) Polymerase Inhibitor ,Monocytes ,Piperazines ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Movement ,PARP inhibitor, PlGF, VEGFR‑1, myelomonocytic cells, cell migration, melanoma ,melanoma ,medicine ,Humans ,Myeloid Cells ,Placenta Growth Factor ,Vascular Endothelial Growth Factor Receptor-1 ,Monocyte ,Settore BIO/14 ,NF-kappa B ,Antibodies, Monoclonal ,Cell Differentiation ,myelomonocytic cells ,General Medicine ,VEGFR‑1 ,PARP inhibitor ,030104 developmental biology ,medicine.anatomical_structure ,PlGF ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Phthalazines - Abstract
The vascular endothelial growth factor receptor-1 (VEGFR-1) is a tyrosine kinase receptor activated by the angiogenic factors VEGF‑A and placental growth factor (PlGF). While VEGF‑A binds to both VEGFR‑1 and VEGFR‑2, PlGF interacts exclusively with VEGFR‑1 triggering a signaling pathway involved in: i) tumor‑associated angiogenesis; ii) chemotaxis and invasion of the extracellular matrix (ECM) by cancer cells; and iii) mobilization of bone marrow‑derived myeloid cells that generate tumor‑associated macrophages. By using a novel anti‑VEGFR‑1 monoclonal antibody (D16F7 mAb), which hampers receptor activation without avoiding ligand binding, we recently demonstrated that VEGFR‑1 blockade reduced myeloid progenitor mobilization and monocyte/macrophage cell infiltration of tumor grafts in vivo. Since poly(ADP‑ribose) polymerase (PARP)‑1 exerts a pro‑inflammatory role favoring monocyte activation, in the present study we investigated whether the PARP inhibitor (PARPi) olaparib hampers PlGF‑induced activation of human myelomonocytic cells. HL‑60 cells induced to differentiate towards the monocytic/macrophage lineage were tested and the results were confirmed in freshly isolated monocytes obtained from healthy donors. Cells were treated with olaparib, at clinically achievable concentrations, before exposure to PlGF and were analyzed for migration and ECM invasion in response to PlGF. Olaparib effects were compared to those obtained with D16F7 mAb used as single agent or in combination with the PARPi. The results indicate that differentiated HL‑60 cells and monocytes expressed VEGFR‑1 and migrated in response to PlGF. Moreover, olaparib and D16F7 inhibited PlGF‑induced chemotaxis and ECM invasion in a dose‑dependent manner and with similar efficacy. However, in combination studies the PARPi and D16F7 did not exert synergistic effects. Olaparib also hampered PlGF‑induced monocyte adhesion to fibronectin, while it did not affect NF‑κB activation in response to the angiogenic factor. These data suggest that olaparib likely interferes with the same pathway affected by the anti‑VEGFR‑1 mAb and that inhibition of PlGF-induced monocyte activation may contribute to PARPi antitumor activity.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.