906 results on '"Settore BIO/14"'
Search Results
2. Investigational treatments for neurodegenerative diseases caused by inheritance of gene mutations: lessons from recent clinical trials
- Author
-
BrunoP Imbimbo, Viviana Triaca, Camillo Imbimbo, and Robert Nisticò
- Subjects
amyotrophic lateral sclerosis ,presenilin 2 ,superoxide dismutase 1 ,Developmental Neuroscience ,glucocerebrosidase ,huntingtin ,presenilin 1 ,Parkinson’s disease ,Settore BIO/14 ,amyloid precursor protein ,Alzheimer’s disease ,Huntington’s disease - Published
- 2023
3. Plasma ATN(I) classification and precision pharmacology in Alzheimer's disease
- Author
-
Bruno P. Imbimbo, Mark Watling, Camillo Imbimbo, and Robert Nisticò
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,AT(N) classification ,plasma biomarkers ,Epidemiology ,Health Policy ,precision medicine ,Settore BIO/14 ,Neurology (clinical) ,Geriatrics and Gerontology ,Alzheimer's disease - Published
- 2023
4. The changing landscape of treatment options in childhood acute lymphoblastic leukaemia
- Author
-
Benjamin Micallef, Robert Nisticò, Sinan.B. Sarac, Ole W. Bjerrum, Dianne Butler, Nicolette Sammut Bartolo, Anthony Serracino-Inglott, and John Joseph Borg
- Subjects
Pharmacology ,Research ,Antineoplastic Agents/therapeutic use ,Clinical drug development ,Settore BIO/14 ,Antineoplastic Agents ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Medical Oncology ,EU regulation ,Clinical trials ,Oncology ,Drug Discovery ,Humans ,Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy ,Childhood acute lymphoblastic leukaemia ,Precision Medicine ,Child - Abstract
New paediatric acute lymphoblastic leukaemia (ALL) treatments have been developed and innovative products are in the pipeline. However, despite many active clinical trials, bridging bench science to clinical development to authorised medicines remains challenging. Research in first-line treatment continues to focus on multidrug chemotherapy with the potential addition of new targeted molecules being studied. Research in second- and third-line treatment represents a shift from cytotoxic intensification to an area of precision medicine through emergent innovative and immuno-oncology products. The collaborative research model in ALL involving different stakeholders should intensify to facilitate bench-to-bedside clinical translation for the benefit of patients.
- Published
- 2022
5. Determinants of approved acetylcholinesterase inhibitor response outcomes in Alzheimer's disease: relevance for precision medicine in neurodegenerative diseases
- Author
-
Lista, Simone, Vergallo, Andrea, Koronyo, Yosef, Koronyo-Hamaoui, Maya, Hampel, Harald, Nisticò, Robert, Initiative, Neurodegeneration Precision Medicine, Teipel, Stefan J, Lemercier, Pablo, Giorgi, Filippo Sean, Gabelle, Audrey, Garaci, Francesco, Mercuri, Nicola B, Babiloni, Claudio, and Gaire, Bhakta Prasad
- Subjects
pharmacology [Cholinesterase Inhibitors] ,Aging ,therapeutic use [Acetylcholinesterase] ,diagnosis [Alzheimer Disease] ,Settore BIO/14 ,Cholinergic system ,genetics [Alzheimer Disease] ,Biochemistry ,Disease-modifying ,Basal forebrain ,Acetylcholinesterase inhibitors ,therapeutic use [Cholinesterase Inhibitors] ,Neurology ,Humans ,drug therapy [Alzheimer Disease] ,Amyloid-β ,ddc:610 ,Precision Medicine ,Molecular Biology ,Alzheimer’s disease ,drug therapy [Neurodegenerative Diseases] ,Biotechnology - Abstract
Acetylcholinesterase inhibitors (ChEI) are the global standard of care for the symptomatic treatment of Alzheimer's disease (AD) and show significant positive effects in neurodegenerative diseases with cognitive and behavioral symptoms. Although experimental and large-scale clinical evidence indicates the potential long-term efficacy of ChEI, primary outcomes are generally heterogeneous across outpatient clinics and regional healthcare systems. Sub-optimal dosing or slow tapering, heterogeneous guidelines about the timing for therapy initiation (prodromal versus dementia stages), healthcare providers' ambivalence to treatment, lack of disease awareness, delayed medical consultation, prescription of ChEI in non-AD cognitive disorders, contribute to the negative outcomes. We present an evidence-based overview of determinants, spanning genetic, molecular, and large-scale networks, involved in the response to ChEI in patients with AD and other neurodegenerative diseases. A comprehensive understanding of cerebral and retinal cholinergic system dysfunctions along with ChEI response predictors in AD is crucial since disease-modifying therapies will frequently be prescribed in combination with ChEI. Therapeutic algorithms tailored to genetic, biological, clinical (endo)phenotypes, and disease stages will help leverage inter-drug synergy and attain optimal combined response outcomes, in line with the precision medicine model.
- Published
- 2023
6. PARP1 allows proper telomere replication through TRF1 poly (ADP-ribosyl)ation and helicase recruitment
- Author
-
C. Maresca, A. Dello Stritto, C. D’Angelo, E. Petti, A. Rizzo, E. Vertecchi, F. Berardinelli, L. Bonanni, A. Sgura, A. Antoccia, G. Graziani, A. Biroccio, E. Salvati, Maresca, C, Dello Stritto, A, D'Angelo, C, Petti, E, Rizzo, A, Vertecchi, E, Berardinelli, F, Bonanni, L, Sgura, A, Antoccia, A, Graziani, G, Biroccio, A, and Salvati, E
- Subjects
Settore BIO/14 ,Medicine (miscellaneous) ,General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular 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. Here we found that poly (ADP-ribose) polymerase 1 (PARP1) interacts and covalently PARylates TRF1 in S-phase modifying its DNA affinity. Therefore, genetic and pharmacological inhibition of PARP1 impairs the dynamic association of TRF1 and the bromodeoxyuridine incorporation at replicating telomeres. Inhibition of PARP1 also affects the recruitment of WRN and BLM helicases in TRF1 containing complexes during S-phase, triggering replication-dependent DNA-damage and telomere fragility. This work unveils an unprecedented role for PARP1 as a “surveillant” of telomere replication, which orchestrates protein dynamics at proceeding replication fork.
- Published
- 2023
7. 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
8. 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
9. Targeting of PDGF-C/NRP-1 autocrine loop as a new strategy for counteracting the invasiveness of melanoma resistant to braf inhibitors
- Author
-
Ruffini, F, Ceci, C, Atzori, Mg, Caporali, S, Levati, L, Bonmassar, L, Cappellini, Gca, D'Atri, S, Graziani, G, and Lacal, Pm
- Subjects
Pharmacology ,BRAF inhibitors ,Dabrafenib ,Extracellular matrix invasion ,Melanoma ,Neuropilin-1 ,PDGF-C ,vemurafenib (PubChem CID: 42611257) cobimetinib (PubChem CID: 16222096) ,dabrafenib (PubChem CID: 44462760)trametinib ,(PubChem CID: 11707110) ,vemurafenib (PubChem CID: 42611257) ,Settore BIO/14 - Published
- 2023
10. 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
11. Pharmacological targeting of microglia dynamics in Alzheimer's disease: Preclinical and clinical evidence
- Author
-
Atrin Yousefizadeh, Gaia Piccioni, Amira Saidi, Viviana Triaca, Dalila Mango, and Robert Nisticò
- Subjects
Pharmacology ,Neurons ,Amyloid beta-Peptides ,Drug discovery ,Settore BIO/14 ,Brain ,Neuroprotection ,Synaptic plasticity ,Neuroinflammation ,Alzheimer Disease ,Synapses ,Humans ,Microglia ,Alzheimer’s disease - Abstract
Numerous clinical trials of anti-amyloid agents for Alzheimer's disease (AD) were so far unsuccessful thereby challenging the validity of the amyloid hypothesis. This lack of progress has encouraged researchers to investigate alternative mechanisms in non-neuronal cells, among which microglia represent nowadays an attractive target. Microglia play a key role in the developing brain and contribute to synaptic remodeling in the mature brain. On the other hand, the intimate relationship between microglia and synapses led to the so-called synaptic stripping hypothesis, a process in which microglia selectively remove synapses from injured neurons. Synaptic stripping, along with the induction of a microglia-mediated chronic neuroinflammatory environment, promote the progressive synaptic degeneration in AD. Therefore, targeting microglia may pave the way for a new disease modifying approach. This review provides an overview of the pathophysiological roles of the microglia cells in AD and describes putative targets for pharmacological intervention. It also provides evidence for microglia-targeted strategies in preclinical AD studies and in early clinical trials.
- Published
- 2022
12. A Healthy Gut for a Healthy Brain: Preclinical, Clinical and Regulatory Aspects
- Author
-
Robert Nisticò, Georgios Strimpakos, Carla Petrella, Mauro Ceccanti, Giovanni Monteleone, John Joseph Borg, Stefano Farioli-Vecchioli, Giusy Ylenia Cisale, and Marco Fiore
- Subjects
0301 basic medicine ,ngf ,preclinical and clinical studies ,Autism Spectrum Disorder ,Gut–brain axis ,autism ,Gut flora ,Article ,regulatory aspects ,03 medical and health sciences ,0302 clinical medicine ,Neurodevelopmental disorder ,Humans ,Medicine ,Pharmacology (medical) ,Pathological ,Pharmacology ,biology ,alcohol ,gut-brain axis ,business.industry ,Probiotics ,aging ,Settore BIO/14 ,Brain ,brain disorders ,General Medicine ,medicine.disease ,biology.organism_classification ,Mental health ,Gastrointestinal Microbiome ,Psychiatry and Mental health ,030104 developmental biology ,Physiological Aging ,Neurology ,Dietary Supplements ,Autism ,Neurology (clinical) ,business ,Neuroscience ,Substance abuse disorder ,030217 neurology & neurosurgery - Abstract
A large body of research has shown the presence of a complex pathway of communications between the gut and the brain. It is now recognized that, through this pathway, the microbiota can influence brain homeostasis and plasticity under normal and pathological conditions. : This review aims at providing an overview of preclinical and clinical pieces of evidence supporting the possible role of gut-brain axis modulation in physiological aging, in a neurodevelopmental disorder, the autism spectrum disorders and in a substance abuse disorder, the alcohol addiction. : Since the normalization of gut flora can prevent changes in the behavior, we postulate that the gutbrain axis might represent a possible target for pharmacological and dietary strategies aimed at improving not only intestinal but also mental health. The present review also reports some regulatory considerations regarding the use of probiotics, illustrating the most debated issues about the possibility of considering probiotics not only as a food supplement but also as a “full” medicinal product.
- Published
- 2021
13. 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
14. Editorial: Neurotrophins Biodelivery to CNS: Innovative Approaches for Disease-Modifying Therapy
- Author
-
Triaca, V, Imbimbo, Bp, and Nistico', R
- Subjects
General Neuroscience ,intranasal route ,Settore BIO/14 ,focused ultrasound ,neurodegenerative diseases ,blood-brain barrier ,neurotrophins - Published
- 2022
15. Exploiting Focused Ultrasound to Aid Intranasal Drug Delivery for Brain Therapy
- Author
-
Gaetano Barbato, Robert Nisticò, and Viviana Triaca
- Subjects
Pharmacology ,clinical trials ,drug delivery ,Settore BIO/14 ,focused ultrasound ,neurodegenerative diseases ,Pharmacology (medical) ,brain circuit vulnerability - Abstract
Novel effective therapeutic strategies are needed to treat brain neurodegenerative diseases and to improve the quality of life of patients affected by Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), Amyotrophic Lateral sclerosis (ALS) as well as other brain conditions. At present no effective treatment options are available; current therapeutics for neurodegenerative diseases (NDs) improve cognitive symptoms only transiently and in a minor number of patients. Further, most of the amyloid-based phase III clinical trials recently failed in AD, in spite of promising preclinical and phase I-II clinical trials, further pinpointing the need for a better knowledge of the early mechanisms of disease as well as of more effective routes of drug administration. In fact, beyond common pathological events and molecular substrates, each of these diseases preferentially affect defined subpopulations of neurons in specific neuronal circuits (selective neuronal vulnerability), leading to the typical age-related clinical profile. In this perspective, key to successful drug discovery is a robust and reproducible biological validation of potential new molecular targets together with a concomitant set up of protocols/tools for efficient and targeted brain delivery to a specific area of interest. Here we propose and discuss Focused UltraSound aided drug administration as a specific and novel technical approach to achieve optimal concentration of the drug at the target area of interest. We will focus on drug delivery to the brain through the nasal route coupled to FUS as a promising approach to achieve neuroprotection and rescue of cognitive decline in several NDs.
- Published
- 2022
16. 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, Massimo Coletta, Tundo, G. R., Sbardella, D., Oddone, F., Grasso, G., Marini, S., Atzori, M. G., Santoro, A. M., Milardi, D., Bellia, F., Macari, G., Graziani, G., Polticelli, F., Cascio, P., Parravano, M., and Coletta, M.
- Subjects
Proteasome Endopeptidase Complex ,carfilzomib ,Proteasome ,proteasome ,insulin-degrading enzyme ,cancer ,neurodegeneration ,Settore BIO/14 ,Carfilzomib ,Antineoplastic Agents ,Biochemistry ,Insulysin ,Molecular Docking Simulation ,Humans ,Oligopeptides ,Pharmaceutical Preparations ,Proteasome Inhibitors ,Multiple Myeloma ,Insulin-degrading enzyme ,Neurodegeneration ,Settore BIO/10 ,Molecular Biology ,Cancer - 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
17. 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
18. Acid-sensing ion channel 1a: a novel target in Alzheimer’s disease?
- Author
-
Dalila, Mango and Robert, Nisticò
- Subjects
Developmental Neuroscience ,Settore BIO/14 - Published
- 2023
19. Defining and assessing intrinsic capacity in older people: A systematic review and a proposed scoring system
- Author
-
Susana López-Ortiz, Simone Lista, Saúl Peñín-Grandes, Jose Pinto-Fraga, Pedro L. Valenzuela, Robert Nisticò, Enzo Emanuele, Alejandro Lucia, and Alejandro Santos-Lozano
- Subjects
Aging ,Settore BIO/14 ,Biochemistry ,Healthy Aging ,Ageing ,Cognition ,Neurology ,Healthy ageing ,Humans ,Molecular Biology ,Geriatric ,Biomarkers ,Biotechnology ,Aged - Abstract
The World Health Organization has introduced the term 'intrinsic capacity' (IC) as a marker of healthy ageing. However, controversy exists on the definition and assessment of IC. We aimed to review the definitions and methods used for the assessment of IC in older adults. In addition, we proposed a new IC scoring method.A systematic search was performed in PubMed, Web of Science, Cochrane Library, Scopus and SPORTDiscus (up to February 10th, 2022) for studies assesing IC in older adults (60 years).Thirty-three studies were included. There is overall consensus on the definition of IC as well as on its different dimensions, that is: locomotion, vitality, sensory, cognition and psychological. However, the methods for assessing each of these five dimensions differ substantially across studies and there is no consensus on the best method to compute an eventual global compound score to evaluate IC taking into account all its different dimensions.The IC represents a highly relevant clinical concept that has been unfortunately underutilized. We propose a standardization for the assessment of each dimension of IC, with a global 0 (worst) to 10 (highest) score.
- Published
- 2021
20. Neurodegenerative Disease: What Potential Therapeutic Role of Acid-Sensing Ion Channels?
- Author
-
Dalila Mango and Robert Nisticò
- Subjects
business.industry ,Multiple sclerosis ,Mini Review ,Settore BIO/14 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Disease ,Neurotransmission ,medicine.disease ,Neuroprotection ,neuroinflammation ,Cellular and Molecular Neuroscience ,neurodegenerative disease ,ASIC1a ,Synaptic plasticity ,pH shift ,medicine ,acidosis ,business ,Neuroscience ,Acid-sensing ion channel ,Neuroinflammation ,Ion channel ,RC321-571 - Abstract
Acidic pH shift occurs in many physiological neuronal activities such as synaptic transmission and synaptic plasticity but also represents a characteristic feature of many pathological conditions including inflammation and ischemia. Neuroinflammation is a complex process that occurs in various neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and Huntington’s disease. Acid-sensing ion channels (ASICs) represent a widely expressed pH sensor in the brain that play a key role in neuroinflammation. On this basis, acid-sensing ion channel blockers are able to exert neuroprotective effects in different neurodegenerative diseases. In this review, we discuss the multifaceted roles of ASICs in brain physiology and pathology and highlight ASIC1a as a potential pharmacological target in neurodegenerative diseases.
- Published
- 2021
21. [The treatment for Fabry disease: focus on agalsidase alpha and beta]
- Author
-
Robert, Nisticò and Antonio, Pisani
- Subjects
Isoenzymes ,Male ,alpha-Galactosidase ,Settore BIO/14 ,Fabry Disease ,Humans ,Enzyme Replacement Therapy ,Female ,Recombinant Proteins - Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder resulting from the deficiency of the hydrolytic enzyme α-galactosidase A (α-Gal A), with consequent accumulation of globotrioasoylceramide in cells and tissues of the body, resulting in a multi-system pathology. Classically affected hemizygous males may display all the characteristic neurological (pain), cutaneous (angiokeratoma), renal (proteinuria, kidney failure), cardiovascular (cardiomyopathy, arrhythmia), and cerebrovascular (transient ischemic attacks, strokes) signs of the disease, while heterozygous females have symptoms ranging from very mild to severe. End-stage renal disease and cardiovascular or cerebrovascular complications limit life-expectancy of untreated patients. Demonstration of α-Gal A deficiency is the definitive method for the diagnosis of hemizygous males, while it's often inconclusive due to random X-chromosomal inactivation so that molecular testing (genotyping) of females is mandatory. The treatment options for FD are enzyme replacement therapy (ERT), and the oral pharmacological chaperone migalastat. Two different products, agalsidase alfa and agalsidase beta, have been commercially available in Europe for 20 years and they are both indicated for long-term ERT. In fact, clinical trials, observational studies and registry data have provided abundant evidence for the safety and efficacy of ERT in improving symptoms and disease progression. Agalsidase alpha and beta are two almost identical recombinant proteins although they are used clinically with a different dosage regimen. In this chapter we aim to clarify the differences between the two ERTs and how these can affect the pharmacokinetic/pharmacodynamic (PK/PD) characteristics and ultimately the risk/benefit profile. The chaperone migalastat, available in Europe since 2016, is the only oral treatment for FD, and acts stabilizing specific mutant forms of α-Gal, defined "amenable" to migalastat. A multitude of therapies are now under investigation in various phases of clinical trials. These include pegylated form of α-Gal (pegunigalsidase alpha), gene therapy (both in-vivo and ex-vivo methods), mRNA therapy (inducing production of α-Gal) and substrate reduction therapy (inhibitors of glucosylceramide synthase leading to reduction of Gb-3).
- Published
- 2021
22. Abscopal Effect and Drug-Induced Xenogenization: A Strategic Alliance in Cancer Treatment?
- Author
-
Ornella Franzese, Francesco Torino, Angelo Aquino, Enzo Bonmassar, Bernd Kaina, Elisa Giannetti, Isabella Faraoni, Anna Giuliani, Maria Pia Fuggetta, Giorgia Cioccoloni, and Liana De Vecchis
- Subjects
abscopal effect ,Review ,temozolomide ,Settore MED/06 ,immune response ,Neoplasms ,Radiation, Ionizing ,alkylating agents ,Cytotoxic T cell ,Biology (General) ,Spectroscopy ,media_common ,Settore BIO/14 ,Disease Management ,Abscopal effect ,General Medicine ,Computer Science Applications ,Chemistry ,Models, Animal ,Immunogenic cell death ,Disease Susceptibility ,xenogenization ,medicine.drug ,Drug ,Drug-Related Side Effects and Adverse Reactions ,QH301-705.5 ,media_common.quotation_subject ,Dacarbazine ,tumor neoantigens ,Antineoplastic Agents ,dacarbazine ,Catalysis ,Inorganic Chemistry ,Immune system ,Antigen ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,Radiation Injuries ,Molecular Biology ,QD1-999 ,Temozolomide ,Radiotherapy ,business.industry ,Organic Chemistry ,Immunity ,immune checkpoints ,Cancer research ,business ,Biomarkers - Abstract
The current state of cancer treatment is still far from being satisfactory considering the strong impairment of patients’ quality of life and the high lethality of malignant diseases. Therefore, it is critical for innovative approaches to be tested in the near future. In view of the crucial role that is played by tumor immunity, the present review provides essential information on the immune-mediated effects potentially generated by the interplay between ionizing radiation and cytotoxic antitumor agents when interacting with target malignant cells. Therefore, the radiation-dependent abscopal effect (i.e., a biological effect of ionizing radiation that occurs outside the irradiated field), the influence of cancer chemotherapy on the antigenic pattern of target neoplastic cells, and the immunogenic cell death (ICD) caused by anticancer agents are the main topics of this presentation. It is widely accepted that tumor immunity plays a fundamental role in generating an abscopal effect and that anticancer drugs can profoundly influence not only the host immune responses, but also the immunogenic pattern of malignant cells. Remarkably, several anticancer drugs impact both the abscopal effect and ICD. In addition, certain classes of anticancer agents are able to amplify already expressed tumor-associated antigens (TAA). More importantly, other drugs, especially triazenes, induce the appearance of new tumor neoantigens (TNA), a phenomenon that we termed drug-induced xenogenization (DIX). The adoption of the abscopal effect is proposed as a potential therapeutic modality when properly applied concomitantly with drug-induced increase in tumor cell immunogenicity and ICD. Although little to no preclinical or clinical studies are presently available on this subject, we discuss this issue in terms of potential mechanisms and therapeutic benefits. Upcoming investigations are aimed at evaluating how chemical anticancer drugs, radiation, and immunotherapies are interacting and cooperate in evoking the abscopal effect, tumor xenogenization and ICD, paving the way for new and possibly successful approaches in cancer therapy.
- Published
- 2021
23. Should we be concerned when COVID-19-positive patients take opioids to control their pain? Insights from a pharmacological point of view
- Author
-
Natoli, S, Carpenedo, R, Chine, E, Vannicola, F, Leonardis, F, and Dauri, M
- Subjects
Alanine ,SARS-CoV-2 ,Settore BIO/14 ,Anti-Inflammatory Agents ,COVID-19 ,Settore MED/09 ,Antiviral Agents ,Adenosine Monophosphate ,Dexamethasone ,COVID-19 Drug Treatment ,Analgesics, Opioid ,Settore MED/41 ,Humans ,Drug Interactions ,Chronic Pain - Abstract
The purpose of this narrative review is to discuss the available information regarding the currently utilized COVID-19 therapies (and the evidence level supporting them) and opioids for chronic pain with a focus on warnings of potential interactions between these two therapeutic approaches.Papers were retrieved from a PubMed search, using different combinations of keywords [e.g., pain treatment AND COVID-19 AND drug-drug interaction (DDI)], without limitations in terms of publication date and language.Remdesivir is an inhibitor of CYP3A4 and may increase the plasma concentration of CYP3A4 substrates (e.g., fentanyl). Dexamethasone is an inducer of CYP3A4 and glycoprotein P, thus coadministration with drugs metabolized by this isoform will lead to their increased clearance. Dexamethasone may cause hypokalemia, thus potentiating the risk of ventricular arrhythmias if it is given with opioids able to prolong the QT interval, such as oxycodone and methadone. Finally, the existing differences among opioids with regard to their impact on immune responses should also be taken into account with only tapentadol and hydromorphone appearing neutral on both cytokine production and immune parameters.Clinicians should keep in mind the frequent DDIs with drugs extensively metabolized by the CYP450 system and prefer opioids undergoing a limited hepatic metabolism. Identification and management of DDIs and dissemination of the related knowledge should be a major goal in the delivery of chronic care to ensure optimized patient outcomes and facilitate updating recommendations for COVID-19 therapy in frail populations, namely comorbid, poly-medicated patients or individuals suffering from substance use disorder.
- Published
- 2021
24. Omics sciences for systems biology in Alzheimer's disease: State-of-the-art of the evidence
- Author
-
Pedro L. Valenzuela, Alejandro Lucia, Harald Hampel, Massimo Corbo, Mark Mapstone, Nicholas T. Seyfried, Giulia Maria Sancesario, Pablo Lemercier, Enzo Emanuele, Francesco Garaci, Andrea Urbani, Simone Lista, George Perry, Filippo Baldacci, Nicola Toschi, Erica Modeste, Allan I. Levey, Andrea Vergallo, and Robert Nisticò
- Subjects
0301 basic medicine ,Aging ,Systems biology ,Alzheimer's disease ,Biomarker signatures ,Multiple omics ,Pathophysiology ,Precision medicine ,Genomics ,Humans ,Metabolomics ,Precision Medicine ,Alzheimer Disease ,Systems Biology ,Computational biology ,Biology ,Proteomics ,Fisiología humana ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Molecular Biology ,Epigenomics ,Biología molecular ,Enfermedad de alzheimer ,Settore BIO/14 ,Omics ,Biología de sistemas ,030104 developmental biology ,Proteostasis ,Biomarcadores ,Neurology ,Alzheimer’s disease ,030217 neurology & neurosurgery ,Biological network ,Biotechnology - Abstract
Alzheimer's disease (AD) is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in biological alterations and disease spatial-temporal progression. Human in-vivo and post-mortem studies point out a failure of multi-level biological networks underlying AD pathophysiology, including proteostasis (amyloid-β and tau), synaptic homeostasis, inflammatory and immune responses, lipid and energy metabolism, oxidative stress. Therefore, a holistic, systems-level approach is needed to fully capture AD multi-faceted pathophysiology. Omics sciences - genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics - embedded in the systems biology (SB) theoretical and computational framework can generate explainable readouts describing the entire biological continuum of a disease. Such path in Neurology is encouraged by the promising results of omics sciences and SB approaches in Oncology, where stage-driven pathway-based therapies have been developed in line with the precision medicine paradigm. Multi-omics data integrated in SB network approaches will help detect and chart AD upstream pathomechanistic alterations and downstream molecular effects occurring in preclinical stages. Finally, integrating omics and neuroimaging data - i.e., neuroimaging-omics - will identify multi-dimensional biological signatures essential to track the clinical-biological trajectories, at the subpopulation or even individual level. Sin financiación 10.895 JCR (2021) Q1, 25/195 Cell Biology 3.523 SJR (2021) Q1, 1/35 Aging No data IDR 2021 UEM
- Published
- 2021
25. Saffron and Its Major Ingredients’ Effect on Colon Cancer Cells with Mismatch Repair Deficiency and Microsatellite Instability
- Author
-
Amr Amin, Hassan Brim, Françoise Praz, Aaminah Farrukh, Akbar Soleimani, Chandraprabha Murali, Hassan Ashktorab, Grazia Graziani, United Arab Emirates University (UAEU), The University of Chicago Medicine [Chicago], Howard University College of Medicine [Washington, DC, USA], Centre de Recherche Saint-Antoine (CR Saint-Antoine), Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Saint-Antoine [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Department of System Medicine, Università degli Studi di Roma Tor Vergata [Roma], Centre de Recherche Saint-Antoine (CRSA), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), and Gestionnaire, HAL Sorbonne Université 5
- Subjects
Colorectal cancer ,Pharmaceutical Science ,Organic chemistry ,DNA Mismatch Repair ,Analytical Chemistry ,Crocin ,chemistry.chemical_compound ,0302 clinical medicine ,QD241-441 ,Drug Discovery ,0303 health sciences ,DNA damage and repair ,Settore BIO/14 ,MLH1 ,apoptosis ,safranal ,3. Good health ,Safranal ,Chemistry (miscellaneous) ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Molecular Medicine ,Microsatellite Instability ,saffron ,Context (language use) ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,colorectal cancer ,Biology ,HCT116 ,Article ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,medicine ,Humans ,Physical and Theoretical Chemistry ,neoplasms ,030304 developmental biology ,Plant Extracts ,Microsatellite instability ,medicine.disease ,Crocus ,HCT116 Cells ,MSH3 ,digestive system diseases ,crocin ,chemistry ,Apoptosis ,Cancer research - 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
26. Lithium as a Treatment for Alzheimer’s Disease: The Systems Pharmacology Perspective
- Author
-
Harald Hampel, Hugo Geerts, Andrea Vergallo, Félix Hernández, Simone Lista, George Perry, Robert Nisticò, Dalila Mango, Jesús Avila, Sorbonne Université, and Fondation pour la Recherche sur Alzheimer
- Subjects
0301 basic medicine ,Drug ,Lithium (medication) ,media_common.quotation_subject ,Lithium ,GSK3 ,Alzheimer’s disease ,lithium ,neurotoxicity ,post-translational modification ,precision medicine ,systems pharmacology ,tau ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,Antimanic Agents ,GSK-3 ,Neurotoxicity ,Animals ,Humans ,Medicine ,media_common ,Systems pharmacology ,Glycogen Synthase Kinase 3 beta ,business.industry ,Mechanism (biology) ,General Neuroscience ,Precision medicine ,Settore BIO/14 ,Long-term potentiation ,General Medicine ,medicine.disease ,Psychiatry and Mental health ,Clinical Psychology ,030104 developmental biology ,Post-translational modification ,Tau ,Geriatrics and Gerontology ,Lithium Chloride ,business ,Neuroscience ,Metabolic Networks and Pathways ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Systems pharmacology is a novel framework for drug research that models traditional and innovative pharmacological parameters and provides the overall efficacy and safety profile of a drug across body systems and complex, non-linear, molecular interactions. Lithium chloride, a pharmacological compound approved for the therapy of psychiatric disorders, represents a poorly explored compound for the treatment of Alzheimer's disease (AD). Lithium has been shown to reduce downstream effects associated with the aberrant overactivation of certain molecular pathways, such as glycogen synthase kinase 3 subunit β (GSK3-β)-related pathways, involved in AD-related pathophysiology. It seems that overactivation and overexpression of GSK3-β lead to an impairment of long-term potentiation and amyloid-β induced neurotoxicity that can be normalized using lithium. Moreover, a growing body of evidence has demonstrated that lithium's GSK3-β inhibitory effect prevents tau phosphorylation in mouse models of tauopathies. Clinical data have been inconclusive, partly due to methodological limitations. The lack of studies exploring the dynamics of protein misfolding in AD and investigating the specific tau-isoforms appearing prior to the accumulation of neurofibrillary tangles calls for new and optimized clinical trials. Advanced computer modeling based on a formal implementation of quantitative parameters and basic enzymatic insights into a mechanism-based model would present a good start to tackle these non-linear interactions. This innovative approach will pave the way for developing "molecularly" biomarker-guided targeted therapies, i.e., treatments specifically adapted ("tailored") to the individual, consistently with the primary objectives and key conceptual points of precision medicine and precision pharmacology., Program “PHOENIX” led by the Sorbonne University Foundation and sponsored by la Fondation pour la Recherche sur Alzheimer.
- Published
- 2019
27. 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
28. 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
29. 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
30. The β-Secretase BACE1 in Alzheimer’s Disease
- Author
-
John Hardy, Riqiang Yan, Akihiko Koyama, Harald Hampel, Lisa Yarenis, Andrea Vergallo, Robert Nisticò, Simone Lista, Johannes Streffer, Michael C. Irizarry, Iryna Voytyuk, Massimo Corbo, Maarten Timmers, Michael Willem, Amir Abbas Tahami Monfared, Bart De Strooper, John Zhou, Bruce Albala, Ann De Vos, Bruno P. Imbimbo, Teiji Kimura, Robert Vassar, Neeraj Kumar Singh, Eugeen Vanmechelen, Lynn D. Kramer, Naoto Watanabe, Alzheimer Precision Medicine [CHU Pitié-Salpétriêre] (GRC 21 AMP), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Sorbonne Université - Faculté de Médecine (SU FM), Sorbonne Université (SU), Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), University College of London [London] (UCL), Ludwig-Maximilians-Universität München (LMU), European Brain Research Institute [Rome, Italy] (EBRI), Università degli Studi di Roma Tor Vergata [Roma], University of Antwerp (UA), University of Cambridge [UK] (CAM), McGill University = Université McGill [Montréal, Canada], Institut de la Mémoire et de la Maladie d'Alzheimer [Paris] (IM2A), Service de Neurologie [CHU Pitié-Salpêtrière], IFR70-CHU Pitié-Salpêtrière [AP-HP], Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), and Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Disease ,Germline ,Article ,03 medical and health sciences ,Amyloid beta-Protein Precursor ,Mice ,0302 clinical medicine ,Clinical trials ,Alzheimer Disease ,mental disorders ,Amyloid precursor protein ,Animals ,Aspartic Acid Endopeptidases ,Humans ,Biological Psychiatry ,chemistry.chemical_classification ,Mice, Knockout ,BACE1 inhibitors ,Amyloid beta-Peptides ,biology ,Soluble amyloid ,Synaptic ,Settore BIO/14 ,Phenotype ,3. Good health ,030104 developmental biology ,Enzyme ,chemistry ,Synaptic plasticity ,Knockout mouse ,biology.protein ,Alzheimer’s disease ,Biomarkers ,Biomarker (medicine) ,Human medicine ,Amyloid Precursor Protein Secretases ,Neuroscience ,030217 neurology & neurosurgery - Abstract
BACE1 (beta-site amyloid precursor protein cleaving enzyme 1) was initially cloned and characterized in 1999. It is required for the generation of all monomeric forms of amyloid-β (Aβ), including Aβ42, which aggregates into bioactive conformational species and likely initiates toxicity in Alzheimer's disease (AD). BACE1 concentrations and rates of activity are increased in AD brains and body fluids, thereby supporting the hypothesis that BACE1 plays a critical role in AD pathophysiology. Therefore, BACE1 is a prime drug target for slowing down Aβ production in early AD. Besides the amyloidogenic pathway, BACE1 has other substrates that may be important for synaptic plasticity and synaptic homeostasis. Indeed, germline and adult conditional BACE1 knockout mice display complex neurological phenotypes. Despite BACE1 inhibitor clinical trials conducted so far being discontinued for futility or safety reasons, BACE1 remains a well-validated therapeutic target for AD. A safe and efficacious compound with high substrate selectivity as well as a more accurate dose regimen, patient population, and disease stage may yet be found. Further research should focus on the role of Aβ and BACE1 in physiological processes and key pathophysiological mechanisms of AD. The functions of BACE1 and the homologue BACE2, as well as the biology of Aβ in neurons and glia, deserve further investigation. Cellular and molecular studies of BACE1 and BACE2 knockout mice coupled with biomarker-based human research will help elucidate the biological functions of these important enzymes and identify their substrates and downstream effects. Such studies will have critical implications for BACE1 inhibition as a therapeutic approach for AD. ispartof: BIOLOGICAL PSYCHIATRY vol:89 issue:8 pages:745-756 ispartof: location:United States status: published
- Published
- 2021
31. Exercise interventions in Alzheimer's disease: A systematic review and meta-analysis of randomized controlled trials
- Author
-
Pedro L. Valenzuela, Nicola Biagio Mercuri, María M. Seisdedos, Susana López-Ortiz, Javier S. Morales, Tomás Vega, Alejandro Lucia, Robert Nisticò, Simone Lista, Alejandro Santos-Lozano, and Adrián Castillo-García
- Subjects
Aging ,medicine.medical_specialty ,Enfermedad del sistema nervioso ,Physical exercise ,Disease ,Biochemistry ,law.invention ,Cognition ,Randomized controlled trial ,law ,Alzheimer Disease ,Medicine ,Aerobic exercise ,Humans ,Molecular Biology ,Exercise ,Aged ,Randomized Controlled Trials as Topic ,Aged, 80 and over ,Enfermedad de alzheimer ,business.industry ,Settore BIO/14 ,Physical Activity ,Deporte ,Alzheimer's disease ,Ejercicio físico ,Confidence interval ,Test (assessment) ,Exercise Therapy ,Efectos fisiológicos ,Neurology ,Meta-analysis ,Physical therapy ,Quality of Life ,business ,Biotechnology - Abstract
Aims: To assess the potential multi-domain benefits of exercise interventions on patients with Alzheimer's disease (AD), as well as to determine the specific effects of different exercise modalities (aerobic, strength, or combined training). Methods: A systematic search was conducted in PubMed and Web of Science until March 2021 for randomized controlled trials assessing the effect of exercise interventions (compared with no exercise) on patients with AD. Outcomes included cognitive function (mini-mental state examination [MMSE] test), physical function (e.g., 6-minute walking test [6MWT]), functional independence (Barthel index), and neuropsychiatric symptoms (Neuropsychiatric Inventory [NPI]). A random-effects meta-analysis was conducted. Results: 28 studies (total n = 1337 participants, average age 79-90 years) were included in the systematic review, of which 21 could be meta-analyzed. Although considerable heterogeneity was found, exercise interventions induced several significant benefits, including in Barthel index (n = 147 patients, mean difference [MD]=8.36 points, 95% confidence interval [CI]=0.63-16.09), 6MWT (n = 369, MD=84 m, 95% CI=44-133)), and NPI (n = 263, MD=-4.4 points, 95% CI=-8.42 to -0.38). Benefits were also found in the MMSE test, albeit significance was only reached for aerobic exercise (n = 187, MD=2.31 points, 95% CI 0.45-4.27). Conclusions: Exercise interventions appear to exert multi-domain benefits in patients with AD. Sin financiación 10.895 JCR (2021) Q1, 25/195 Cell Biology 3.523 SJR (2021) Q1, 1/35 Aging No data IDR 2021 UEM
- Published
- 2021
32. Targeting microglia‐synapse interactions in alzheimer’s disease
- Author
-
Dalila Mango, Amira Saidi, Gaia Piccioni, Massimo Corbo, and Robert Nisticò
- Subjects
Context (language use) ,Review ,Biology ,Catalysis ,Inorganic Chemistry ,Synapse ,lcsh:Chemistry ,Neuroinflammation ,Alzheimer Disease ,Immunologic ,Receptors ,medicine ,Alzheimer’s disease ,Cytokines ,Long‐term potentiation ,Microglia ,Synaptic Plasticity ,Animals ,Humans ,Inflammation ,Membrane Glycoproteins ,Neuronal Plasticity ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Receptors, Immunologic ,Synapses ,Physical and Theoretical Chemistry ,Molecular Biology ,lcsh:QH301-705.5 ,Spectroscopy ,long-term potentiation ,TREM2 ,Organic Chemistry ,Settore BIO/14 ,Granulocyte-Macrophage Colony-Stimulating Factor ,Cognition ,Long-term potentiation ,General Medicine ,Computer Science Applications ,medicine.anatomical_structure ,lcsh:Biology (General) ,lcsh:QD1-999 ,Synaptic plasticity ,Neuroscience - Abstract
In this review, we focus on the emerging roles of microglia in the brain, with particular attention to synaptic plasticity in health and disease. We present evidence that ramified microglia, classically believed to be “resting” (i.e., inactive), are instead strongly implicated in dynamic and plastic processes. Indeed, there is an intimate relationship between microglia and neurons at synapses which modulates activity-dependent functional and structural plasticity through the release of cytokines and growth factors. These roles are indispensable to brain development and cognitive function. Therefore, approaches aimed at maintaining the ramified state of microglia might be critical to ensure normal synaptic plasticity and cognition. On the other hand, inflammatory signals associated with Alzheimer’s disease are able to modify the ramified morphology of microglia, thus leading to synapse loss and dysfunction, as well as cognitive impairment. In this context, we highlight microglial TREM2 and CSF1R as emerging targets for disease-modifying therapy in Alzheimer’s disease (AD) and other neurodegenerative disorders.
- Published
- 2021
33. 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
34. Biological Mechanism-based Neurology and Psychiatry: a BACE1/2 and Downstream Pathway Model
- Author
-
Simone Lista, Harald Hampel, Giuseppe Caruso, Robert Nisticò, Gaia Piccioni, Nicola B. Mercuri, Filippo Sean Giorgi, Fabio Ferrarelli, Pablo Lemercier, Filippo Caraci, Andrea Vergallo, and null Neurodegeneration Precision Medicine Initiative
- Subjects
Pharmacology ,medicine.medical_specialty ,Neurology ,Systems biology ,neurology ,precision medicine ,Settore BIO/14 ,β-site amyloid precursor protein cleaving enzyme (BACE) ,Translational research ,systems biology ,General Medicine ,Disease ,Precision medicine ,medicine.disease ,psychiatry ,Psychiatry and Mental health ,Drug development ,medicine ,Autism ,Pharmacology (medical) ,Neurology (clinical) ,Psychiatry ,systems pharmacology ,Systems pharmacology - Abstract
In oncology, comprehensive omics and functional enrichment studies have led to an extensive profiling of (epi)genetic and neurobiological alterations that can be mapped onto a single tumor’s clinical phenotype and divergent clinical phenotypes expressing common pathophysiological pathways. Consequently, molecular pathway-based therapeutic interventions for different cancer typologies, namely tumor type- and site-agnostic treatments, have been developed, encouraging the real-world implementation of a paradigm shift in medicine. Given the breakthrough nature of the new-generation translational research and drug development in oncology, there is an increasing rationale to transfertilize this blueprint to other medical fields, including psychiatry and neurology. In order to illustrate the emerging paradigm shift in neuroscience, we provide a state-of-the-art review of translational studies on the β-site amyloid precursor protein cleaving enzyme (BACE) and its most studied downstream effector, neuregulin, which are molecular orchestrators of distinct biological pathways involved in several neurological and psychiatric diseases. This body of data aligns with the evidence of a shared genetic/biological architecture among Alzheimer’s disease, schizoaffective disorder, and autism spectrum disorders. To facilitate a forward-looking discussion about a potential first step towards the adoption of biological pathway-based, clinical symptom-agnostic, categorization models in clinical neurology and psychiatry for precision medicine solutions, we engage in a speculative intellectual exercise gravitating around BACE-related science, which is used as a paradigmatic case here. We draw a perspective whereby pathway-based therapeutic strategies could be catalyzed by highthroughput techniques embedded in systems-scaled biology, neuroscience, and pharmacology approaches that will help overcome the constraints of traditional descriptive clinical symptom and syndrome-focused constructs in neurology and psychiatry.
- Published
- 2021
35. Involvement of Bradykinin Receptor 2 in Nerve Growth Factor Neuroprotective Activity
- Author
-
Sebastiano Cavallaro, Sonia Piccinin, Simona Capsoni, Robert Nisticò, Pietro Calissano, Maria Teresa Ciotti, Carla Petrella, Cinzia Severini, Roberta Possenti, and Delio Mercanti
- Subjects
Receptor, Bradykinin B2 ,Cell Survival ,Primary Cell Culture ,Socio-culturale ,Mice, Transgenic ,Pharmacology ,Hippocampal formation ,s disease animal models ,Neuroprotection ,nerve growth factor (NGF), bradykinin receptor 2 (B2R), microglial cells, LTP, Alzheimer’s disease animal models ,Article ,Mice ,bradykinin receptor 2 (B2R) ,Downregulation and upregulation ,microglial cells ,Alzheimer Disease ,Nerve Growth Factor ,medicine ,Animals ,Alzheimer’ ,lcsh:QH301-705.5 ,Administration, Intranasal ,Cells, Cultured ,Cerebral Cortex ,Neuronal Plasticity ,biology ,Chemistry ,Alzheimer’s disease animal models ,Settore BIO/14 ,Long-term potentiation ,General Medicine ,nerve growth factor (NGF) ,Rats ,Up-Regulation ,Disease Models, Animal ,Neuroprotective Agents ,Nerve growth factor ,lcsh:Biology (General) ,Gene Expression Regulation ,Mechanism of action ,nervous system ,Synaptic plasticity ,biology.protein ,Microglia ,medicine.symptom ,LTP ,Neurotrophin - Abstract
Neurotrophin nerve growth factor (NGF) has been demonstrated to upregulate the gene expression of bradykinin receptor 2 (B2R) on sensory neurons, thus facilitating nociceptive signals. The aim of the present study is to investigate the involvement of B2R in the NGF mechanism of action in nonsensory neurons in vitro by using rat mixed cortical primary cultures (CNs) and mouse hippocampal slices, and in vivo in Alzheimer&rsquo, s disease (AD) transgenic mice (5xFAD) chronically treated with NGF. A significant NGF-mediated upregulation of B2R was demonstrated by microarray, Western blot, and immunofluorescence analysis in CNs, indicating microglial cells as the target of this modulation. The B2R involvement in the NGF mechanism of action was also demonstrated by using a selective B2R antagonist which was able to reverse the neuroprotective effect of NGF in CNs, as revealed by viability assay, and the NGF-induced long-term potentiation (LTP) in hippocampal slices. To confirm in vitro observations, B2R upregulation was observed in 5xFAD mouse brain following chronic intranasal NGF treatment. This study demonstrates for the first time that B2R is a key element in the neuroprotective activity and synaptic plasticity mediated by NGF in brain cells.
- Published
- 2020
- Full Text
- View/download PDF
36. 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
37. 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
38. Reply to the reply of the authors of the review article entitled 'Management of status epilepticus in adults. Position paper of the Italian League against Epilepsy'
- Author
-
Armando Magrelli, Giuseppe Capovilla, and Robert Nisticò
- Subjects
Adult ,medicine.medical_specialty ,Epilepsy ,Settore BIO/14 ,Status epilepticus ,League ,medicine.disease ,Review article ,Behavioral Neuroscience ,Status Epilepticus ,Italy ,Neurology ,medicine ,Humans ,Position paper ,Neurology (clinical) ,medicine.symptom ,Psychology ,Psychiatry - Published
- 2020
39. Role of ASIC1a in Normal and Pathological Synaptic Plasticity
- Author
-
Mango, D and Nisticò, R
- Subjects
Acid Sensing Ion Channels ,Neuronal Plasticity ,Memory ,LTD ,Settore BIO/14 ,Animals ,Humans ,Acid-sensing ion channel ,LTP ,Nervous System Diseases ,CA1 Region, Hippocampal ,Synaptic Transmission - Abstract
Acid-sensing ion channels (ASICs), members of the degenerin/epithelial Na+ channel superfamily, are broadly distributed in the mammalian nervous system where they play important roles in a variety of physiological processes, including neurotransmission and memory-related behaviors. In the last few years, we and others have investigated the role of ASIC1a in different forms of synaptic plasticity especially in the CA1 area of the hippocampus. This review summarizes the latest research linking ASIC1a to synaptic function either in physiological or pathological conditions. A better understanding of how these channels are regulated in brain circuitries relevant to synaptic plasticity and memory may offer novel targets for pharmacological intervention in neuropsychiatric and neurological disorders.
- Published
- 2020
40. 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
41. 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
42. 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
43. 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
44. Cerebrospinal fluid and serum D-serine concentrations are unaltered across the whole clinical spectrum of Alzheimer's disease
- Author
-
Andrea Mancini, Lucilla Parnetti, Alessia Casamassa, Paolo Eusebi, Masumi Katane, Tommaso Nuzzo, Lorenzo Gaetani, Francesco Errico, Paolo Calabresi, Hiroshi Homma, Robert Nisticò, Mattia Miroballo, Alessandro Usiello, Nuzzo, T., Miroballo, M., Casamassa, A., Mancini, A., Gaetani, L., Nistico, R., Eusebi, P., Katane, M., Homma, H., Calabresi, P., Errico, F., Parnetti, L., Usiello, A., Nuzzo, T, Miroballo, M, Casamassa, A, Mancini, A, Gaetani, L, Nisticò, R, Eusebi, P, Katane, M, Homma, H, Calabresi, P, Errico, F, and Parnetti, L
- Subjects
0301 basic medicine ,Male ,D-amino acid ,Amyloid beta-Peptide ,Biochemistry ,Analytical Chemistry ,0302 clinical medicine ,Cerebrospinal fluid ,Blood serum ,Serine ,80 and over ,Receptor ,Aged, 80 and over ,Amyloidosis ,Postpartum Period ,Settore BIO/14 ,Brain ,Alzheimer's disease ,Prognosis ,Settore MED/26 - NEUROLOGIA ,Organ Specificity ,Biomarker (medicine) ,D-amino acids ,Female ,Tauopathy ,Human ,medicine.medical_specialty ,Prognosi ,Clinical Dementia Rating ,Biophysics ,tau Proteins ,03 medical and health sciences ,Biomarker ,Dementia ,Mild cognitive impairment ,Alzheimer Disease ,Internal medicine ,medicine ,Humans ,Molecular Biology ,Aged ,Aspartic Acid ,Amyloid beta-Peptides ,business.industry ,tau Protein ,medicine.disease ,030104 developmental biology ,Endocrinology ,business ,030217 neurology & neurosurgery ,Biomarkers - Abstract
The diagnosis of Alzheimer's disease (AD) relies on the presence of amyloidosis and tauopathy, as reflected in cerebrospinal fluid (CSF), independently from the clinical stage. Recently, CSF d-serine has been proposed as a possible new AD biomarker, reflecting dysfunctional activation of neuronal glutamatergic N-methyl-d-aspartate receptor (NMDAR). In this study, we measured blood serum and CSF concentration of two NMDAR modulators, such as d-serine and d-aspartate, in a cohort of drug-free subjects encompassing the whole AD clinical spectrum. In addition, we also analyzed d-serine levels in a cohort of post-mortem AD and control cortex samples. We reported unaltered serum and CSF concentrations of d-serine and d-aspartate in AD patients both during the AD progression and compared to non-demented controls. Accordingly, no correlation was detected between serum or CSF d-serine content and mini-mental state examination or Clinical Dementia Rating. Similarly, cortical d-serine levels were also unaltered in post-mortem samples of AD patients. Overall, our results failed to confirm previous findings indicating the CSF d-serine as a novel biomarker for AD.
- Published
- 2020
45. 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
46. Effects of intranasally-delivered pro-nerve growth factors on the septo-hippocampal system in healthy and diabetic rats
- Author
-
Marzia Soligo 1, Virginia Protto 1, Antonio Chiaretti 2, Sonia Piccinin 3, Maria Egle De Stefano 4, Robert Nisticò 3, 5, Luisa Bracci-Laudiero 1, 6, and Luigi Manni 1
- Subjects
0301 basic medicine ,medicine.medical_specialty ,long-term potentiation (DG-LTP) ,Hippocampal neurogenesis ,Hippocampus ,Hippocampal formation ,Diabetes Mellitus, Experimental ,pro nerve growth factor (proNGF) ,Rats, Sprague-Dawley ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Drug Delivery Systems ,Organ Culture Techniques ,Internal medicine ,Nerve Growth Factor ,medicine ,Animals ,Dentate-gyrus long-term potentiation (DG-LTP) ,Cholinergic neuron ,Protein Precursors ,Dentate-gyrus ,Administration, Intranasal ,Pharmacology ,Basal forebrain ,Cholinergic Fibers ,Cholinergic system ,Diabetes ,Rat ,business.industry ,Neurogenesis ,Settore BIO/14 ,Rats ,030104 developmental biology ,Endocrinology ,Nerve growth factor ,Settore MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,Cholinergic ,Female ,Septum of Brain ,Nerve Net ,business ,030217 neurology & neurosurgery - Abstract
Pro-nerve growth factor (proNGF) is the predominant form of NGF in the brain and its levels increase in neurodegenerative diseases. The balance between NGF receptors may explain the contradictory biological activities of proNGF. However, the specific role of the two main proNGF variants is mostly unexplored. proNGF-A is prevalently expressed in healthy brain, while proNGF-B content increases in the neuro-degenerating brain. Recently we have investigated in vitro the biological action of native mouse proNGF variants. To gain further insights into the specific functions of the two proNGFs, here we intranasally delivered mouse-derived proNGF-A and proNGF-B to the brain parenchyma of healthy and diabetic rats, the latter characterized by dysfunction in spatial learning and memory, in the septo-hippocampal circuitry and by relative increase in proNGF-B hippocampal levels. Exogenous proNGF-B induces depression of hippocampal DG-LTP and impairment of hippocampal neurogenesis in healthy animals, with concomitant decrease in basal forebrain cholinergic neurons and cholinergic fibers projecting to the hippocampus. proNGF-A, while ineffective in healthy animals, rescues the diabetes-induced impairment in DG-LTP and hippocampal neurogenesis, promoting the concomitant recovery of the basal forebrain cholinergic phenotype. Our experimental evidences suggest that the balance between different proNGFs may influence the development and progression of neurodegenerative diseases.
- Published
- 2020
47. A Path Toward Precision Medicine for Neuroinflammatory Mechanisms in Alzheimer's Disease
- Author
-
Harald Hampel, Filippo Caraci, A. Claudio Cuello, Giuseppe Caruso, Robert Nisticò, Massimo Corbo, Filippo Baldacci, Nicola Toschi, Francesco Garaci, Patrizia A. Chiesa, Steven R. Verdooner, Leyla Akman-Anderson, Félix Hernández, Jesús Ávila, Enzo Emanuele, Pedro L. Valenzuela, Alejandro Lucía, Mark Watling, Bruno P. Imbimbo, Andrea Vergallo, Simone Lista, Gestionnaire, Hal Sorbonne Université, Alzheimer Precision Medicine [CHU Pitié-Salpétriêre] (GRC 21 APM), CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), University of Catania [Italy], Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), McGill University = Université McGill [Montréal, Canada], University of Oxford, Università degli Studi di Roma Tor Vergata [Roma], European Brain Research Institute [Rome, Italy] (EBRI), Institut du Cerveau = Paris Brain Institute (ICM), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Institut de la Mémoire et de la Maladie d'Alzheimer [CHU Pitié-Salpétriêre] (IM2A), Service de Neurologie [CHU Pitié-Salpêtrière], IFR70-CHU Pitié-Salpêtrière [AP-HP], University of Pisa - Università di Pisa, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School [Boston] (HMS)-Massachusetts General Hospital [Boston], Harvard Medical School [Boston] (HMS), NeuroVision Imaging, Centro de Biología Molecular Severo Ochoa [Madrid] (CBMSO), Universidad Autónoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas [Madrid] (CSIC), Centro de Investigacion Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III [Madrid] (ISC), Universidad de Alcalá - University of Alcalá (UAH), Universidad Europea de Madrid = European University of Madrid (UEM), Hospital Universitario 12 de Octubre [Madrid], Alzheimer Precision Medicine [CHU Pitié-Salpétriêre] (GRC 21 AMP), Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), University of Oxford [Oxford], Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Institut de la Mémoire et de la Maladie d'Alzheimer [Paris] (IM2A), Sorbonne Université (SU), Service de neurologie 1 [CHU Pitié-Salpétrière], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Massachusetts General Hospital [Boston]-Harvard Medical School [Boston] (HMS), Universidad Autonoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas [Madrid] (CSIC), Universidad Europea de Madrid, Sorbonne Université, Fondation pour la Recherche sur Alzheimer, and AXA Research Fund
- Subjects
0301 basic medicine ,Proteomics ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Enfermedad del sistema nervioso ,microglia ,Review ,Genética humana ,neuroinflammation ,Tratamiento médico ,0302 clinical medicine ,Enfermedad de Alzheimer ,Medicine ,Immunology and Allergy ,Cell Self Renewal ,Terapeútica ,Antiinflammatory therapy ,Microglia ,Neurodegeneration ,Settore BIO/14 ,systems biology ,Alzheimer's disease ,3. Good health ,medicine.anatomical_structure ,Cytokines ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Neurogenic Inflammation ,Alzheimer’s disease ,Signal Transduction ,lcsh:Immunologic diseases. Allergy ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,Systems biology ,precision medicine ,Immunology ,anti-inflammatory therapy ,biomarkers ,neuroinflammatory pathways ,Proinflammatory cytokine ,03 medical and health sciences ,Alzheimer Disease ,Animals ,Humans ,Neuroinflammation ,business.industry ,TREM2 ,Mechanism (biology) ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,medicine.disease ,Precision medicine ,Lipid Metabolism ,030104 developmental biology ,business ,lcsh:RC581-607 ,Neuroscience ,030215 immunology ,Genome-Wide Association Study - Abstract
Neuroinflammation commences decades before Alzheimer's disease (AD) clinical onset and represents one of the earliest pathomechanistic alterations throughout the AD continuum. Large-scale genome-wide association studies point out several genetic variants—TREM2, CD33, PILRA, CR1, MS4A, CLU, ABCA7, EPHA1, and HLA-DRB5-HLA-DRB1—potentially linked to neuroinflammation. Most of these genes are involved in proinflammatory intracellular signaling, cytokines/interleukins/cell turnover, synaptic activity, lipid metabolism, and vesicle trafficking. Proteomic studies indicate that a plethora of interconnected aberrant molecular pathways, set off and perpetuated by TNF-α, TGF-β, IL-1β, and the receptor protein TREM2, are involved in neuroinflammation. Microglia and astrocytes are key cellular drivers and regulators of neuroinflammation. Under physiological conditions, they are important for neurotransmission and synaptic homeostasis. In AD, there is a turning point throughout its pathophysiological evolution where glial cells sustain an overexpressed inflammatory response that synergizes with amyloid-β and tau accumulation, and drives synaptotoxicity and neurodegeneration in a self-reinforcing manner. Despite a strong therapeutic rationale, previous clinical trials investigating compounds with anti-inflammatory properties, including non-steroidal anti-inflammatory drugs (NSAIDs), did not achieve primary efficacy endpoints. It is conceivable that study design issues, including the lack of diagnostic accuracy and biomarkers for target population identification and proof of mechanism, may partially explain the negative outcomes. However, a recent meta-analysis indicates a potential biological effect of NSAIDs. In this regard, candidate fluid biomarkers of neuroinflammation are under analytical/clinical validation, i.e., TREM2, IL-1β, MCP-1, IL-6, TNF-α receptor complexes, TGF-β, and YKL-40. PET radio-ligands are investigated to accomplish in vivo and longitudinal regional exploration of neuroinflammation. Biomarkers tracking different molecular pathways (body fluid matrixes) along with brain neuroinflammatory endophenotypes (neuroimaging markers), can untangle temporal–spatial dynamics between neuroinflammation and other AD pathophysiological mechanisms. Robust biomarker–drug codevelopment pipelines are expected to enrich large-scale clinical trials testing new-generation compounds active, directly or indirectly, on neuroinflammatory targets and displaying putative disease-modifying effects: novel NSAIDs, AL002 (anti-TREM2 antibody), anti-Aβ protofibrils (BAN2401), and AL003 (anti-CD33 antibody). As a next step, taking advantage of breakthrough and multimodal techniques coupled with a systems biology approach is the path to pursue for developing individualized therapeutic strategies targeting neuroinflammation under the framework of precision medicine., Sorbonne University Foundation and sponsored by la Fondation pour la Recherche sur Alzheimer. HH is an employee of Eisai Inc. During his previous work (until April 2019), he was supported by the AXA Research Fund, the Fondation partenariale Sorbonne Université and the Fondation pour la Recherche sur Alzheimer, Paris, France
- Published
- 2020
48. 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
49. 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
50. Reply to the article 'Management of status epilepticus in adults. Position paper of the Italian League against Epilepsy'
- Author
-
Armando Magrelli, Robert Nisticò, and Giuseppe Capovilla
- Subjects
Adult ,medicine.medical_specialty ,Lacosamide ,MEDLINE ,Status epilepticus ,League ,03 medical and health sciences ,Behavioral Neuroscience ,Epilepsy ,0302 clinical medicine ,Status Epilepticus ,medicine ,Humans ,030212 general & internal medicine ,Psychiatry ,business.industry ,Settore BIO/14 ,medicine.disease ,Review article ,Neurology ,Italy ,Position paper ,Neurology (clinical) ,Levetiracetam ,medicine.symptom ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
We would like to comment on the review article entitled “Manage- ment of status epilepticus in adults. Position paper of the Italian League against Epilepsy” [1]. This article updates the previous article published by the same Italian League against Epilepsy in 2006. In particular, the updating regards some drugs (levetiracetam, lacosamide, midazolam, etc.) that were not available at that time, even if some of these have not yet obtained the specific indication to treat status epilepticus (SE) and their use is considered off-label.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.