Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.143
Filter
Add more filters

Publication year range
2.
Semin Cancer Biol ; 74: 62-78, 2021 09.
Article in English | MEDLINE | ID: mdl-33609665

ABSTRACT

Cancer is a disease that evolves continuously with unpredictable outcomes. Although conventional chemotherapy can display significant antitumor effects, the lack of specificity and poor bioavailability remain major concerns in cancer therapy. Moreover, with the advent of novel anti-cancer gene therapies, there is an urgent need for drug delivery vectors capable of bypassing cellular barriers and efficiently transferring therapeutic cargo to recipient cells. A number of drug delivery systems have been proposed to overcome these limitations, but their successful clinical translation has been hampered by the onset of unexpected side effects and associated toxicities. The application of extracellular vesicles (EVs), a class of naturally released, cell-derived particles, as drug delivery vectors presents a breakthrough in nanomedicine, taking into account their biocompatibility and natural role in intercellular communication. Combining the advantageous intrinsic properties of EVs with surface functionalization and the encapsulation of drugs allows for a new class of engineered EVs that serve as effective therapeutic carriers. Here, we describe the various successful approaches involving the application of engineered EVs as bio-derived drug delivery vectors in cancer therapy. The latest and most effective strategies of engineering EVs to improve drug loading, stealth properties and tumour targeting capabilities of EVs are debated. Finally, current obstacles and future perspectives of smart engineered EVs are discussed.


Subject(s)
Bioengineering/methods , Drug Carriers , Drug Delivery Systems/methods , Extracellular Vesicles , Neoplasms/drug therapy , Animals , Bioengineering/trends , Drug Delivery Systems/trends , Humans
3.
Semin Cancer Biol ; 74: 45-61, 2021 09.
Article in English | MEDLINE | ID: mdl-33609664

ABSTRACT

Extracellular vesicles (EVs) are a class of cell-derived lipid-bilayer membrane vesicles secreted by almost all mammalian cells and involved in intercellular communication by shuttling various biological cargoes. Over the last decade, EVs - namely exosomes and microvesicles - have been extensively explored as next-generation nanoscale drug delivery systems (DDSs). This is in large due to their endogenous origin, which enables EVs to circumvent some of the limitations associated with existing cancer therapy approaches (i.e. by preventing recognition by the immune system and improving selectivity towards tumor tissue). However, successful translation of these cell-derived vesicles into clinical applications has been hindered by several factors, among which the loading of exogenous therapeutic molecules still represents a great challenge. In order to address this issue and to further advance these biologically-derived systems as drug carriers, EV-biohybrid nano-DDSs, obtained through the fusion of EVs with conventional synthetic nano-DDSs, have recently been proposed as a valuable alternative as DDSs. Building on the idea of "combining the best of both worlds", a combination of these two unique entities aims to harness the beneficial properties associated with both EVs and conventional nano-DDSs, while overcoming the flaws of the individual components. These biohybrid systems also provide a unique opportunity for exploitation of new synergisms, often leading to improved therapeutic outcomes, thus paving the way for advancements in cancer therapy. This review aims to describe the recent developments of EV-biohybrid nano-DDSs in cancer therapy, to highlight the most promising results and breakthroughs, as well as to provide a glimpse on the possible intrinsic targeting mechanisms of EVs that can be bequeathed to their hybrid systems. Finally, we also provide some insights in the future perspectives of EV-hybrid DDSs.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/administration & dosage , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Extracellular Vesicles , Neoplasms/drug therapy , Animals , Humans , Nanotechnology/methods , Nanotechnology/trends
4.
Ann Neurol ; 90(2): 177-188, 2021 08.
Article in English | MEDLINE | ID: mdl-34219266

ABSTRACT

Inflammasomes are multimeric protein complexes that can sense a plethora of microbe- and damage-associated molecular signals. They play important roles in innate immunity and are key regulators of inflammation in health and disease. Inflammasome-mediated processing and secretion of proinflammatory cytokines such as interleukin (IL) 1ß and IL-18 and induction of pyroptosis, a proinflammatory form of cell death, have been associated with the development and progression of common immune-mediated and degenerative central nervous system (CNS) diseases such as Alzheimer disease, multiple sclerosis, brain injury, stroke, epilepsy, Parkinson disease, and amyotrophic lateral sclerosis. A growing number of pharmacological compounds inhibiting inflammasome activation and signaling show therapeutic efficacy in preclinical models of the aforementioned disease conditions. Here, we illustrate regulatory mechanisms of inflammasome activation during CNS homeostasis and tissue injury. We highlight the evidence for inflammasome activation as a mechanistic underpinning in a wide range of CNS diseases and critically discuss the promise and potential limitations of therapeutic strategies that aim to inhibit the inflammasome components in neurological disorders. ANN NEUROL 2021;90:177-188.


Subject(s)
Drug Delivery Systems/methods , Inflammasomes/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Nervous System Diseases/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Drug Delivery Systems/trends , Humans , Inflammasomes/metabolism , Inflammation Mediators/metabolism , Nervous System Diseases/metabolism , Treatment Outcome
5.
Mol Pharmacol ; 100(4): 348-355, 2021 10.
Article in English | MEDLINE | ID: mdl-34330821

ABSTRACT

Cisplatin is a platinum-based drug, which remains among the most efficacious anticancer treatment options. Unfortunately, use of cisplatin is hindered by dose-limiting toxicities, including irreversible hearing loss, which can grossly affect patient quality of life. Cisplatin-induced ototoxicity is the result of cochlear hair cell damage through a mechanism that is poorly understood. However, cisplatin cytotoxicity is reliant on intracellular accumulation, a process that is largely dependent on the presence of particular membrane transporters. This review will provide an update on our current understanding of the various transporters known to be involved in the disposition and cytotoxicity of platinum drugs or their metabolites, as well as their role in mediating cisplatin-induced hearing loss. We also provide a summary of the successes and opportunities in therapeutically targeting membrane transporters to alleviate platinum-induced hearing loss. Moreover, we describe how this approach could be used to reduce the severity or onset of other adverse events associated with exposure to various forms of platinum drugs, without diminishing antitumor efficacy. SIGNIFICANCE STATEMENT: Cisplatin-induced hearing loss is a dose-limiting and irreversible adverse event with no current preventative or curative treatment measures. Pharmacological targeting of membrane transporters that regulate platinum uptake into cochlear hair cells, if conducted appropriately, may alleviate this devastating side effect and could be applied to alleviate other platinum-induced toxicities.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Drug Delivery Systems/trends , Hearing Loss/chemically induced , Hearing Loss/metabolism , Membrane Transport Proteins/metabolism , Hearing Loss/prevention & control , Humans , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism
6.
Mol Pharmacol ; 99(1): 29-38, 2021 01.
Article in English | MEDLINE | ID: mdl-32321734

ABSTRACT

Bone marrow-derived hematopoietic stem/progenitor cells are vasculogenic and play an important role in endothelial health and vascular homeostasis by participating in postnatal vasculogenesis. Progenitor cells are mobilized from bone marrow niches in response to remote ischemic injury and migrate to the areas of damage and stimulate revascularization largely by paracrine activation of angiogenic functions in the peri-ischemic vasculature. This innate vasoprotective mechanism is impaired in certain chronic clinical conditions, which leads to the development of cardiovascular complications. Members of the renin-angiotensin system-angiotensin-converting enzymes (ACEs) ACE and ACE2, angiotensin II (Ang II), Ang-(1-7), and receptors AT1 and Mas-are expressed in vasculogenic progenitor cells derived from humans and rodents. Ang-(1-7), generated by ACE2, is known to produce cardiovascular protective effects by acting on Mas receptor and is considered as a counter-regulatory mechanism to the detrimental effects of Ang II. Evidence has now been accumulating in support of the activation of the ACE2/Ang-(1-7)/Mas receptor pathway by pharmacologic or molecular maneuvers, which stimulates mobilization of progenitor cells from bone marrow, migration to areas of vascular damage, and revascularization of ischemic areas in pathologic conditions. This minireview summarizes recent studies that have enhanced our understanding of the physiology and pharmacology of vasoprotective axis in bone marrow-derived progenitor cells in health and disease. SIGNIFICANCE STATEMENT: Hematopoietic stem progenitor cells (HSPCs) stimulate revascularization of ischemic areas. However, the reparative potential is diminished in certain chronic clinical conditions, leading to the development of cardiovascular diseases. ACE2 and Mas receptor are key members of the alternative axis of the renin-angiotensin system and are expressed in HSPCs. Accumulating evidence points to activation of ACE2 or Mas receptor as a promising approach for restoring the reparative potential, thereby preventing the development of ischemic vascular diseases.


Subject(s)
Angiotensin I/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Cardiovascular Diseases/metabolism , Drug Delivery Systems/trends , Hematopoietic Stem Cells/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/metabolism , Cardiovascular Diseases/drug therapy , Drug Delivery Systems/methods , Hematopoietic Stem Cells/drug effects , Humans , Proto-Oncogene Mas , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/drug effects , Stem Cells/metabolism
7.
Stroke ; 52(8): 2697-2706, 2021 08.
Article in English | MEDLINE | ID: mdl-34162215

ABSTRACT

New therapeutic approaches are required for secondary prevention of residual vascular risk after stroke. Diverse sources of evidence support a causal role for inflammation in the pathogenesis of stroke. Randomized controlled trials of anti-inflammatory agents have reported benefit for secondary prevention in patients with coronary disease. We review the data from observational studies supporting a role for inflammation in pathogenesis of stroke, overview randomized controlled trials of anti-inflammatory therapy in cardiac disease and discuss the potential implications for stroke prevention therapy.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Drug Delivery Systems/trends , Secondary Prevention/methods , Stroke/prevention & control , Animals , Colchicine/administration & dosage , Drug Delivery Systems/methods , Gout Suppressants/administration & dosage , Humans , Inflammation/diagnosis , Inflammation/epidemiology , Inflammation/prevention & control , Mendelian Randomization Analysis/methods , Observational Studies as Topic/methods , Risk Factors , Secondary Prevention/trends , Stroke/diagnosis , Stroke/epidemiology
8.
J Neurochem ; 158(6): 1381-1393, 2021 09.
Article in English | MEDLINE | ID: mdl-33930191

ABSTRACT

Neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease, are devastating diseases in the elderly world, which are closely associated with progressive neuronal loss induced by a variety of genetic and/or environmental factors. Unfortunately, currently available treatments for neurodegenerative disorders can only relieve the symptoms but not modify the pathological processes. Over the past decades, our group by collaborating with Profs. Yuan-Ping Pang and Paul R. Carlier has developed three series of homo/hetero dimeric acetylcholinesterase inhibitors derived from tacrine and/or huperzine A. The representative dimers bis(3)-Cognitin (B3C), bis(12)-hupyridone, and tacrine(10)-hupyridone might possess disease-modifying effects through the modulation of N-methyl-d-aspartic acid receptors, the activation of myocyte enhancer factor 2D gene transcription, and the promotion of neurotrophic factor secretion. In this review, we summarize that the representative dimers, such as B3C, provide neuroprotection against a variety of neurotoxins via multiple targets, including the inhibitions of N-methyl-d-aspartic acid receptor with pathological-activated potential, neuronal nitric oxide synthase, and ß-amyloid cascades synergistically. More importantly, B3C might offer disease-modifying potentials by activating myocyte enhancer factor 2D transcription, inducing neuritogenesis, and promoting the expressions of neurotrophic factors in vitro and in vivo. Taken together, the novel dimers might offer synergistic disease-modifying effects, proving that dimerization might serve as one of the strategies to develop new generation of therapeutics for neurodegenerative disorders.


Subject(s)
Acetylcholinesterase/metabolism , Alkaloids/administration & dosage , Cholinesterase Inhibitors/administration & dosage , Drug Delivery Systems/methods , Neurodegenerative Diseases/drug therapy , Sesquiterpenes/administration & dosage , Tacrine/administration & dosage , Alkaloids/chemistry , Animals , Cholinesterase Inhibitors/chemistry , Drug Combinations , Drug Delivery Systems/trends , Humans , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/enzymology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/chemistry , Sesquiterpenes/chemistry , Tacrine/chemistry
9.
J Neuroinflammation ; 18(1): 84, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33785039

ABSTRACT

Chronic pain affects the life quality of the suffering patients and posts heavy problems to the health care system. Conventional medications are usually insufficient for chronic pain management and oftentimes results in many adverse effects. The NLRP3 inflammasome controls the processing of proinflammatory cytokine interleukin 1ß (IL-1ß) and is implicated in a variety of disease conditions. Recently, growing number of evidence suggests that NLRP3 inflammasome is dysregulated under chronic pain condition and contributes to pathogenesis of chronic pain. This review provides an up-to-date summary of the recent findings of the involvement of NLRP3 inflammasome in chronic pain and discussed the expression and regulation of NLRP3 inflammasome-related signaling components in chronic pain conditions. This review also summarized the successful therapeutic approaches that target against NLRP3 inflammasome for chronic pain treatment.


Subject(s)
Chronic Pain/drug therapy , Chronic Pain/metabolism , Drug Delivery Systems/methods , Furans/administration & dosage , Indenes/administration & dosage , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sulfonamides/administration & dosage , Animals , Drug Delivery Systems/trends , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
10.
Neurochem Res ; 46(11): 2800-2831, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34282491

ABSTRACT

Cerebral ischemic injury is a leading cause of death and long-term disability throughout the world. Peroxisome proliferator-activated receptor gamma (PPAR-É£) is a ligand-activated nuclear transcription factor that is a member of the PPAR family. PPAR-É£ has been shown in several in vitro and in vivo models to prevent post-ischemic inflammation and neuronal damage by negatively controlling the expression of genes modulated by cerebral ischemic injury, indicating a neuroprotective effect during cerebral ischemic injury. A extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the mechanistic role of Peroxisome proliferator activated receptor gamma and its modulation in Cerebral ischemic injury. PPAR-É£ can interact with specific DNA response elements to control gene transcription and expression when triggered by its ligand. It regulates lipid metabolism, improves insulin sensitivity, modulates antitumor mechanisms, reduces oxidative stress, and inhibits inflammation. This review article provides insights on the current state of research into the neuroprotective effects of PPAR-É£ in cerebral ischemic injury, as well as the cellular and molecular mechanisms by which these effects are modulated, such as inhibition of inflammation, reduction of oxidative stress, suppression of pro-apoptotic production, modulation of transcription factors, and restoration of injured tissue through neurogenesis and angiogenesis.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Drug Delivery Systems/methods , Neuroprotective Agents/administration & dosage , PPAR gamma/agonists , PPAR gamma/metabolism , Animals , Brain/drug effects , Brain/metabolism , Drug Delivery Systems/trends , Humans , Neurons/drug effects , Neurons/metabolism
11.
Circ Res ; 125(10): 884-906, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31556812

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling with aberrant pulmonary artery smooth muscle cells (PASMCs) proliferation, endothelial dysfunction, and extracellular matrix remodeling. OBJECTIVE: Right ventricular (RV) failure is an important prognostic factor in PAH. Thus, we need to elucidate a novel therapeutic target in both PAH and RV failure. METHODS AND RESULTS: We performed microarray analysis in PASMCs from patients with PAH (PAH-PASMCs) and controls. We found a ADAMTS8 (disintegrin and metalloproteinase with thrombospondin motifs 8), a secreted protein specifically expressed in the lung and the heart, was upregulated in PAH-PASMCs and the lung in hypoxia-induced pulmonary hypertension (PH) in mice. To elucidate the role of ADAMTS8 in PH, we used vascular smooth muscle cell-specific ADAMTS8-knockout mice (ADAMTSΔSM22). Hypoxia-induced PH was attenuated in ADAMTSΔSM22 mice compared with controls. ADAMTS8 overexpression increased PASMC proliferation with downregulation of AMPK (AMP-activated protein kinase). In contrast, deletion of ADAMTS8 reduced PASMC proliferation with AMPK upregulation. Moreover, deletion of ADAMTS8 reduced mitochondrial fragmentation under hypoxia in vivo and in vitro. Indeed, PASMCs harvested from ADAMTSΔSM22 mice demonstrated that phosphorylated DRP-1 (dynamin-related protein 1) at Ser637 was significantly upregulated with higher expression of profusion genes (Mfn1 and Mfn2) and improved mitochondrial function. Moreover, recombinant ADAMTS8 induced endothelial dysfunction and matrix metalloproteinase activation in an autocrine/paracrine manner. Next, to elucidate the role of ADAMTS8 in RV function, we developed a cardiomyocyte-specific ADAMTS8 knockout mice (ADAMTS8ΔαMHC). ADAMTS8ΔαMHC mice showed ameliorated RV failure in response to chronic hypoxia. In addition, ADAMTS8ΔαMHC mice showed enhanced angiogenesis and reduced RV ischemia and fibrosis. Finally, high-throughput screening revealed that mebendazole, which is used for treatment of parasite infections, reduced ADAMTS8 expression and cell proliferation in PAH-PASMCs and ameliorated PH and RV failure in PH rodent models. CONCLUSIONS: These results indicate that ADAMTS8 is a novel therapeutic target in PAH.


Subject(s)
ADAMTS Proteins/deficiency , Heart Failure/metabolism , Pulmonary Arterial Hypertension/metabolism , Ventricular Dysfunction, Right/metabolism , ADAMTS Proteins/antagonists & inhibitors , ADAMTS Proteins/genetics , Adult , Animals , Cells, Cultured , Drug Delivery Systems/trends , Female , Heart Failure/drug therapy , Heart Failure/pathology , Humans , Male , Mebendazole/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/pathology , Random Allocation , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/pathology
12.
Pharmacol Res ; 165: 105432, 2021 03.
Article in English | MEDLINE | ID: mdl-33508434

ABSTRACT

Heart failure causes significant morbidity and mortality worldwide. The underlying mechanisms and pathological changes associated with heart failure are exceptionally complex. Despite recent advances in heart failure research, treatment outcomes remain poor. The sirtuin family member sirtuin-3 (SIRT3) is involved in several key biological processes, including ATP production, catabolism, and reactive oxygen species detoxification. In addition to its role in metabolism, SIRT3 regulates cell death and survival and has been implicated in the pathogenesis of cardiovascular diseases. Emerging evidence also shows that SIRT3 can protect cardiomyocytes from hypertrophy, ischemia-reperfusion injury, cardiac fibrosis, and impaired angiogenesis. In this review article, we summarize the recent advances in SIRT3 research and discuss the role of SIRT3 in heart failure. We also discuss the potential use of SIRT3 as a therapeutic target in heart failure.


Subject(s)
Cardiotonic Agents/administration & dosage , Cardiotonic Agents/metabolism , Drug Delivery Systems , Heart Failure/drug therapy , Heart Failure/metabolism , Sirtuin 3/metabolism , Animals , Drug Delivery Systems/trends , Humans , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
13.
Pharm Res ; 38(5): 739-757, 2021 May.
Article in English | MEDLINE | ID: mdl-33903976

ABSTRACT

The current perspective reviews the biopharmaceutical market until end of 2020 and highlights the transforming biopharmaceutical landscape during the recent decade. In particular, the rise of biosimilars and the development of new therapeutic modalities through recent advancement in molecular biology research sustainably change the product scenery. The present manuscript describes opportunities for pharmaceutical technical development, highlighting concepts such as product differentiation to succeed in a competitive product landscape. Product differentiation offers the opportunity for numerous life-cycle options and market exclusivity through incremental improvements in standard of care treatment. In particular, different formulation options and formulation-device combinations are described, focusing on systemic delivery of monoclonal antibody products and patient-centered development. The concept of product differentiation is exemplified in a case study about HER2+ breast cancer therapy, underlining pharmaceutical technical solutions and major improvements for the patient.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Biological Products/therapeutic use , Breast Neoplasms/drug therapy , Drug Development/trends , Drug Industry/organization & administration , Antibodies, Monoclonal/pharmacology , Biological Products/pharmacology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Drug Compounding/methods , Drug Compounding/trends , Drug Delivery Systems/methods , Drug Delivery Systems/trends , Drug Development/organization & administration , Drug Industry/trends , Female , Humans , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Survival Rate
14.
J Cardiovasc Pharmacol ; 77(1): 1-3, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33079828

ABSTRACT

ABSTRACT: The involvement of the vascular endothelium in the complications of coronavirus disease 2019 is now recognized. Chief among these are pulmonary endotheliitis, cytokine storm, endotoxic shock, and cardiovascular collapse. This Perspectives article is focused on therapeutical strategies to reduce the risk of these complications by targeting the vascular endothelium as a part of the overall treatment of coronavirus disease 2019.


Subject(s)
COVID-19/metabolism , Cytokine Release Syndrome/metabolism , Drug Delivery Systems/methods , Endothelium, Vascular/metabolism , Angiotensin II/administration & dosage , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/metabolism , Cytokine Release Syndrome/drug therapy , Drug Delivery Systems/trends , Endothelium, Vascular/drug effects , Humans , COVID-19 Drug Treatment
15.
Drug Resist Updat ; 53: 100721, 2020 12.
Article in English | MEDLINE | ID: mdl-33132205

ABSTRACT

Coronaviridae is a peculiar viral family, with a very large RNA genome and characteristic appearance, endowed with remarkable tendency to transfer from animals to humans. Since the beginning of the 21st century, three highly transmissible and pathogenic coronaviruses have crossed the species barrier and caused deadly pneumonia, inflicting severe outbreaks and causing human health emergencies of inconceivable magnitude. Indeed, in the past two decades, two human coronaviruses emerged causing serious respiratory illness: severe acute respiratory syndrome coronavirus (SARS-CoV-1) and Middle Eastern respiratory syndrome coronavirus (MERS-CoV), causing more than 10,000 cumulative cases, with mortality rates of 10 % for SARS-CoV-1 and 34.4 % for MERS-CoV. More recently, the severe acute respiratory syndrome coronavirus virus 2 (SARS-CoV-2) has emerged in China and has been identified as the etiological agent of the recent COVID-19 pandemic outbreak. It has rapidly spread throughout the world, causing nearly 22 million cases and ∼ 770,000 deaths worldwide, with an estimated mortality rate of ∼3.6 %, hence posing serious challenges for adequate and effective prevention and treatment. Currently, with the exception of the nucleotide analogue prodrug remdesivir, and despite several efforts, there is no known specific, proven, pharmacological treatment capable of efficiently and rapidly inducing viral containment and clearance of SARS-CoV-2 infection as well as no broad-spectrum drug for other human pathogenic coronaviruses. Another confounding factor is the paucity of molecular information regarding the tendency of coronaviruses to acquire drug resistance, a gap that should be filled in order to optimize the efficacy of antiviral drugs. In this light, the present review provides a systematic update on the current knowledge of the marked global efforts towards the development of antiviral strategies aimed at coping with the infection sustained by SARS-CoV-2 and other human pathogenic coronaviruses, displaying drug resistance profiles. The attention has been focused on antiviral drugs mainly targeting viral protease, RNA polymerase and spike glycoprotein, that have been tested in vitro and/or in clinical trials as well as on promising compounds proven to be active against coronaviruses by an in silico drug repurposing approach. In this respect, novel insights on compounds, identified by structure-based virtual screening on the DrugBank database endowed by multi-targeting profile, are also reported. We specifically identified 14 promising compounds characterized by a good in silico binding affinity towards, at least, two of the four studied targets (viral and host proteins). Among which, ceftolozane and NADH showed the best multi-targeting profile, thus potentially reducing the emergence of resistant virus strains. We also focused on potentially novel pharmacological targets for the development of compounds with anti-pan coronavirus activity. Through the analysis of a large set of viral genomic sequences, the current review provides a comprehensive and specific map of conserved regions across human coronavirus proteins which are essential for virus replication and thus with no or very limited tendency to mutate. Hence, these represent key druggable targets for novel compounds against this virus family. In this respect, the identification of highly effective and innovative pharmacological strategies is of paramount importance for the treatment and/or prophylaxis of the current pandemic but potentially also for future and unavoidable outbreaks of human pathogenic coronaviruses.


Subject(s)
Antiviral Agents/administration & dosage , Coronavirus Infections/drug therapy , Drug Delivery Systems/methods , SARS-CoV-2/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , COVID-19/metabolism , Coronavirus Infections/metabolism , Drug Delivery Systems/trends , Humans , Protease Inhibitors/administration & dosage , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , SARS-CoV-2/metabolism , COVID-19 Drug Treatment
16.
Proc Natl Acad Sci U S A ; 115(51): 12887-12895, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30559181

ABSTRACT

Bacterial infections have been traditionally controlled by antibiotics and vaccines, and these approaches have greatly improved health and longevity. However, multiple stakeholders are declaring that the lack of new interventions is putting our ability to prevent and treat bacterial infections at risk. Vaccine and antibiotic approaches still have the potential to address this threat. Innovative vaccine technologies, such as reverse vaccinology, novel adjuvants, and rationally designed bacterial outer membrane vesicles, together with progress in polysaccharide conjugation and antigen design, have the potential to boost the development of vaccines targeting several classes of multidrug-resistant bacteria. Furthermore, new approaches to deliver small-molecule antibacterials into bacteria, such as hijacking active uptake pathways and potentiator approaches, along with a focus on alternative modalities, such as targeting host factors, blocking bacterial virulence factors, monoclonal antibodies, and microbiome interventions, all have potential. Both vaccines and antibacterial approaches are needed to tackle the global challenge of antimicrobial resistance (AMR), and both areas have the underpinning science to address this need. However, a concerted research agenda and rethinking of the value society puts on interventions that save lives, by preventing or treating life-threatening bacterial infections, are needed to bring these ideas to fruition.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Vaccines/therapeutic use , Drug Resistance, Bacterial , Bacteria/drug effects , Bacteria/immunology , Drug Delivery Systems/trends , Humans , Medical Overuse/trends
17.
Drug Dev Res ; 82(2): 145-148, 2021 04.
Article in English | MEDLINE | ID: mdl-33137245

ABSTRACT

The anticancer and antimicrobial drugs customarily suffer a functional inefficacy due to a limited delivery to the target site, active cellular efflux, in addition to the inadequacy of carrier system. Metal nanoparticles possess unique physicochemical properties as drug delivery vehicles, for delivering the drugs susceptible to cellular efflux pumps. However, a direct physiological exposure of nanoparticle surface after releasing the carrier drug poses serious concerns. The polysaccharides with enhanced biotolerance used for encapsulating the cargo drug molecules, when loaded on the nanoparticle surface presents a perspective drug delivery system combining the physiological benevolence of the former and theranostic/efflux pump evading features of the latter. The present commentary highlight the importance of metal nanoparticle-loaded polysaccharides as perspective drug delivery system.


Subject(s)
Drug Delivery Systems/methods , Metal Nanoparticles/administration & dosage , Polysaccharides/administration & dosage , Animals , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Drug Carriers/administration & dosage , Drug Carriers/chemical synthesis , Drug Carriers/metabolism , Drug Delivery Systems/trends , Humans , Metal Nanoparticles/chemistry , Polysaccharides/chemical synthesis , Polysaccharides/metabolism
18.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34360621

ABSTRACT

Metabolic syndrome (MetS) is a set of complex, chronic inflammatory conditions that are characterized by central obesity and associated with an increased risk of cardiovascular diseases. In recent years, microRNAs (miRNAs) have become an important type of endocrine factors, which play crucial roles in maintaining energy balance and metabolic homeostasis. However, its unfavorable properties such as easy degradation in blood and off-target effect are still a barrier for clinical application. Nanosystem based delivery possess strong protection, high bioavailability and control release rate, which is beneficial for success of gene therapy. This review first describes the current progress and advances on miRNAs associated with MetS, then provides a summary of the therapeutic potential and targets of miRNAs in metabolic organs. Next, it discusses recent advances in the functionalized development of classic delivery systems (exosomes, liposomes and polymers), including their structures, properties, functions and applications. Furthermore, this work briefly discusses the intelligent strategies used in emerging novel delivery systems (selenium nanoparticles, DNA origami, microneedles and magnetosomes). Finally, challenges and future directions in this field are discussed provide a comprehensive overview of the future development of targeted miRNAs delivery for MetS treatment. With these contributions, it is expected to address and accelerate the development of effective NA delivery systems for the treatment of MetS.


Subject(s)
Drug Delivery Systems/methods , Genetic Therapy/methods , Metabolic Syndrome/therapy , MicroRNAs/therapeutic use , Nanostructures , Drug Delivery Systems/trends , Exosomes , Humans , Liposomes
19.
Int J Mol Sci ; 22(10)2021 May 17.
Article in English | MEDLINE | ID: mdl-34067699

ABSTRACT

Gene therapy has the potential to become a staple of 21st-century medicine. However, to overcome the limitations of existing gene-delivery therapies, that is, poor stability and inefficient and delivery and accumulation of nucleic acids (NAs), safe drug-delivery systems (DDSs) allowing the prolonged circulation and expression of the administered genes in vivo are needed. In this review article, the development of DDSs over the past 70 years is briefly described. Since synthetic DDSs can be recognized and eliminated as foreign substances by the immune system, new approaches must be found. Using the body's own cells as DDSs is a unique and exciting strategy and can be used in a completely new way to overcome the critical limitations of existing drug-delivery approaches. Among the different circulatory cells, red blood cells (RBCs) are the most abundant and thus can be isolated in sufficiently large quantities to decrease the complexity and cost of the treatment compared to other cell-based carriers. Therefore, in the second part, this article describes 70 years of research on the development of RBCs as DDSs, covering the most important RBC properties and loading methods. In the third part, it focuses on RBCs as the NA delivery system with advantages and drawbacks discussed to decide whether they are suitable for NA delivery in vivo.


Subject(s)
Drug Delivery Systems/methods , Drug Delivery Systems/trends , Erythrocytes/metabolism , Drug Carriers/metabolism , Erythrocytes/physiology , Humans , Nanoparticles/administration & dosage , Nucleic Acids/chemistry , Nucleic Acids/metabolism , Pharmaceutical Preparations/metabolism
20.
Molecules ; 26(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803294

ABSTRACT

Tannic acid is a chief gallo-tannin belonging to the hydrolysable tannins extracted from gall nuts and other plant sources. A myriad of pharmaceutical and biological applications in the medical field has been well recognized to tannic acid. Among these effects, potential anticancer activities against several solid malignancies such as liver, breast, lung, pancreatic, colorectal and ovarian cancers have been reported. Tannic acid was found to play a maestro-role in tuning several oncological signaling pathways including JAK/STAT, RAS/RAF/mTOR, TGF-ß1/TGF-ß1R axis, VEGF/VEGFR and CXCL12/CXCR4 axes. The combinational beneficial effects of tannic acid with other conventional chemotherapeutic drugs have been clearly demonstrated in literature such as a synergistic anticancer effect and enhancement of the chemo-sensitivity in several resistant cases. Yet, clinical applications of tannic acid have been limited owing to its poor lipid solubility, low bioavailability, off-taste, and short half-life. To overcome such obstacles, novel drug delivery systems have been employed to deliver tannic acid with the aim of improving its applications and/or efficacy against cancer cells. Among these drug delivery systems are several types of organic and metallic nanoparticles. In this review, the authors focus on the molecular mechanisms of tannic acid in tuning several neoplastic diseases as well as novel drug delivery systems that can be used for its clinical applications with an attempt to provide a systemic reference to promote the development of tannic acid as a cheap drug and/or drug delivery system in cancer management.


Subject(s)
Hydrolyzable Tannins/pharmacology , Neoplasms/drug therapy , Tannins/pharmacology , Antineoplastic Agents/pharmacology , Drug Delivery Systems/trends , Humans , Hydrolyzable Tannins/therapeutic use , Nanoparticles , Tannins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL