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

Publication year range
1.
Malar J ; 18(1): 94, 2019 Mar 22.
Article in English | MEDLINE | ID: mdl-30902051

ABSTRACT

Over the last two decades there has been a renaissance in the pipeline of new drugs targeting malaria, with the launch of new products that help save the lives of children throughout the world. In addition, there is a wealth of new molecules both entering and progressing through clinical development. These bring hope for a new generation of simpler and more effective cures that could overcome the emerging threat of drug resistance. In addition, there is hope that some of these medicines will have prophylactic activity and can be used to protect vulnerable populations, given the absence of a highly effective vaccine. Switzerland has played a key role in the development of these medicines. First, the country has a long history of understanding the biology of parasites and the pharmacology of drug responses through the leadership of the Swiss Tropical and Public Health Institute in Basel. Second, the highly successful Swiss pharmaceutical industry brings, beyond excellence, a strong interest in neglected diseases, building on work at Hoffmann-La Roche in the last century and with more recent products from Novartis and other Swiss companies. Third, the emergence of product-development-partnerships, in this case led by the Medicines for Malaria Venture, based in Geneva, has helped to catalyze the development of new medicines and bring the community together within Switzerland and beyond. Finally, this progress would not have been possible without the engagement of the Swiss people and the support of the federal government through the Swiss Agency for Development and Cooperation (SDC), the State Secretariat of Education, Research and Innovation (SERI) and the Swiss Republic and Canton of Geneva.


Subject(s)
Antimalarials/history , Drug Industry/history , International Cooperation/history , Malaria/drug therapy , History, 20th Century , History, 21st Century , Humans , Switzerland
2.
J Vector Borne Dis ; 56(1): 15-24, 2019.
Article in English | MEDLINE | ID: mdl-31070161

ABSTRACT

The global fight against malaria requires continual development of new tools. Collaborations in India have played a key role in MMV's partnerships to discover, develop and deliver new medicines. Over the last decade, India has become a focal point of global medicinal chemistry, and combined with investments in basic science, this has led to the discovery of new potential drugs. India also brings significant experience to drug development, in clinical trials, but also in formulation and manufacturing. Finally, innovative new approaches in case management have streamlined impact at the level of communities and the patients.


Subject(s)
Communicable Disease Control/trends , Malaria/drug therapy , Malaria/prevention & control , Antimalarials/therapeutic use , Communicable Disease Control/methods , Drug Discovery/statistics & numerical data , Drug Discovery/trends , Global Health , Humans , India/epidemiology , Malaria Vaccines
3.
J Biol Chem ; 292(12): 4942-4952, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28159843

ABSTRACT

Reversible tyrosine phosphorylation is a widespread post-translational modification mechanism underlying cell physiology. Thus, understanding the mechanisms responsible for substrate selection by kinases and phosphatases is central to our ability to model signal transduction at a system level. Classical protein-tyrosine phosphatases can exhibit substrate specificity in vivo by combining intrinsic enzymatic specificity with the network of protein-protein interactions, which positions the enzymes in close proximity to their substrates. Here we use a high throughput approach, based on high density phosphopeptide chips, to determine the in vitro substrate preference of 16 members of the protein-tyrosine phosphatase family. This approach helped identify one residue in the substrate binding pocket of the phosphatase domain that confers specificity for phosphopeptides in a specific sequence context. We also present a Bayesian model that combines intrinsic enzymatic specificity and interaction information in the context of the human protein interaction network to infer new phosphatase substrates at the proteome level.


Subject(s)
Phosphopeptides/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Bayes Theorem , Binding Sites , Humans , Models, Biological , Molecular Docking Simulation , Phosphopeptides/chemistry , Phosphorylation , Protein Conformation , Protein Domains , Protein Interaction Maps , Protein Tyrosine Phosphatases/chemistry , Substrate Specificity
4.
Malar J ; 17(1): 462, 2018 Dec 10.
Article in English | MEDLINE | ID: mdl-30526594

ABSTRACT

Reaching the overall goal of eliminating malaria requires halting disease transmission. One approach to blocking transmission is to prevent passage of the parasite to a mosquito, by preventing formation or transmission of gametocytes. An alternative approach, pioneered in the veterinary field, is to use endectocides, which are molecules that render vertebrate blood meals toxic for the mosquito vector, also killing the parasite. Field studies and modelling suggest that reducing the lifespan of the mosquito may significantly reduce transmission, given the lengthy maturation process of the parasite. To guide the development of new endectocides, or the reformulation of existing molecules, it is important to construct a framework of the required attributes, commonly called the target candidate profile. Here, using a combination of insights from current endectocides, mathematical models of the malaria transmission dynamics, and known impacts of vector control, a target candidate profile (TCP-6) and a regulatory strategy are proposed for a transmission reducing agent. The parameters chosen can be used to assess the potential of a new medicine, independent of whether it has classical endectocide activity, reduces the insect and parasite lifespan or any combination of all three, thereby constituting an 'endectocidal transmission blocking' paradigm.


Subject(s)
Anopheles , Antiparasitic Agents/pharmacology , Malaria , Mosquito Vectors , Plasmodium , Animals , Anopheles/drug effects , Anopheles/parasitology , Humans , Insecticides/pharmacology , Malaria/prevention & control , Malaria/transmission , Mosquito Vectors/drug effects , Mosquito Vectors/parasitology , Plasmodium/drug effects , Plasmodium/pathogenicity
5.
Malar J ; 17(1): 402, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30384848

ABSTRACT

Over the last 15 years, the majority of malaria drug discovery and development efforts have focused on new molecules and regimens to treat patients with uncomplicated or severe disease. In addition, a number of new molecular scaffolds have been discovered which block the replication of the parasite in the liver, offering the possibility of new tools for oral prophylaxis or chemoprotection, potentially with once-weekly dosing. However, an intervention which requires less frequent administration than this would be a key tool for the control and elimination of malaria. Recent progress in HIV drug discovery has shown that small molecules can be formulated for injections as native molecules or pro-drugs which provide protection for at least 2 months. Advances in antibody engineering offer an alternative approach whereby a single injection could potentially provide protection for several months. Building on earlier profiles for uncomplicated and severe malaria, a target product profile is proposed here for an injectable medicine providing long-term protection from this disease. As with all of such profiles, factors such as efficacy, cost, safety and tolerability are key, but with the changing disease landscape in Africa, new clinical and regulatory approaches are required to develop prophylactic/chemoprotective medicines. An overall framework for these approaches is suggested here.


Subject(s)
Antimalarials , Drug Development , Drug Discovery , Injections, Intravenous , Malaria/prevention & control , Humans
7.
Malar J ; 16(1): 26, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28086874

ABSTRACT

A decade of discovery and development of new anti-malarial medicines has led to a renewed focus on malaria elimination and eradication. Changes in the way new anti-malarial drugs are discovered and developed have led to a dramatic increase in the number and diversity of new molecules presently in pre-clinical and early clinical development. The twin challenges faced can be summarized by multi-drug resistant malaria from the Greater Mekong Sub-region, and the need to provide simplified medicines. This review lists changes in anti-malarial target candidate and target product profiles over the last 4 years. As well as new medicines to treat disease and prevent transmission, there has been increased focus on the longer term goal of finding new medicines for chemoprotection, potentially with long-acting molecules, or parenteral formulations. Other gaps in the malaria armamentarium, such as drugs to treat severe malaria and endectocides (that kill mosquitoes which feed on people who have taken the drug), are defined here. Ultimately the elimination of malaria requires medicines that are safe and well-tolerated to be used in vulnerable populations: in pregnancy, especially the first trimester, and in those suffering from malnutrition or co-infection with other pathogens. These updates reflect the maturing of an understanding of the key challenges in producing the next generation of medicines to control, eliminate and ultimately eradicate malaria.


Subject(s)
Antimalarials/isolation & purification , Antimalarials/pharmacology , Drug Discovery/trends , Malaria/drug therapy , Malaria/prevention & control , Clinical Trials as Topic , Drug Evaluation, Preclinical/trends , Humans
8.
Antimicrob Agents Chemother ; 60(6): 3669-75, 2016 06.
Article in English | MEDLINE | ID: mdl-27044554

ABSTRACT

Effective progression of candidate antimalarials is dependent on optimal dosing in clinical studies, which is determined by a sound understanding of pharmacokinetics and pharmacodynamics (PK/PD). Recently, two important translational models for antimalarials have been developed: the NOD/SCID/IL2Rγ(-/-) (NSG) model, whereby mice are engrafted with noninfected and Plasmodium falciparum-infected human erythrocytes, and the induced blood-stage malaria (IBSM) model in human volunteers. The antimalarial mefloquine was used to directly measure the PK/PD in both models, which were compared to previously published trial data for malaria patients. The clinical part was a single-center, controlled study using a blood-stage Plasmodium falciparum challenge inoculum in volunteers to characterize the effectiveness of mefloquine against early malaria. The study was conducted in three cohorts (n = 8 each) using different doses of mefloquine. The characteristic delay in onset of action of about 24 h was seen in both NSG and IBSM systems. In vivo 50% inhibitory concentrations (IC50s) were estimated at 2.0 µg/ml and 1.8 µg/ml in the NSG and IBSM models, respectively, aligning with 1.8 µg/ml reported previously for patients. In the IBSM model, the parasite reduction ratios were 157 and 195 for the 10- and 15-mg/kg doses, within the range of previously reported clinical data for patients but significantly lower than observed in the mouse model. Linking mouse and human challenge models to clinical trial data can accelerate the accrual of critical data on antimalarial drug activity. Such data can guide large clinical trials required for development of urgently needed novel antimalarial combinations. (This trial was registered at the Australian New Zealand Clinical Trials Registry [http://anzctr.org.au] under registration number ACTRN12612000323820.).


Subject(s)
Antimalarials/pharmacokinetics , Malaria, Falciparum/drug therapy , Mefloquine/pharmacokinetics , Plasmodium falciparum/drug effects , Adult , Animals , Antimalarials/blood , Antimalarials/pharmacology , Cohort Studies , Disease Models, Animal , Drug Administration Schedule , Drug Dosage Calculations , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , Healthy Volunteers , Humans , Inhibitory Concentration 50 , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Male , Mefloquine/blood , Mefloquine/pharmacology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Plasmodium falciparum/growth & development
9.
J Antimicrob Chemother ; 71(9): 2620-7, 2016 09.
Article in English | MEDLINE | ID: mdl-27272721

ABSTRACT

OBJECTIVES: OZ439, or artefenomel, is an investigational synthetic ozonide antimalarial with similar potency, but a significantly improved pharmacokinetic profile, compared with artemisinins. We wished to measure key pharmacokinetic and pharmacodynamic parameters and the pharmacokinetic/pharmacodynamic relationship of artefenomel in humans to guide the drug's further development as combination therapy in patients. PATIENTS AND METHODS: We tested artefenomel in the human induced blood-stage malaria (IBSM) model. Plasmodium infection was monitored by quantitative PCR (qPCR) and upon reaching 1000 parasites/mL single doses of 100, 200 and 500 mg of artefenomel were administered orally with evaluation of drug exposure and parasitaemia until rescue treatment after 16 days or earlier, if required. RESULTS: A single 100 mg dose had only a transient effect, while the 200 mg dose resulted in a significant reduction in parasitaemia before early recrudescence. At the highest (500 mg) dose, initial clearance of parasites below the limit of detection of qPCR was observed, with a 48 h parasite reduction ratio (PRR48) >10 000 and a parasite clearance half-life of 3.6 h (95% CI 3.4-3.8 h). However, at this dose, recrudescence was seen in four of eight subjects 6-10 days after treatment. Pharmacokinetic/pharmacodynamic modelling predicted an MIC of 4.1 ng/mL. CONCLUSIONS: These results confirm the antimalarial potential of artefenomel for use in a single-exposure combination therapy. The observations from this study support and will assist further clinical development of artefenomel.


Subject(s)
Adamantane/analogs & derivatives , Antimalarials/pharmacology , Antimalarials/pharmacokinetics , Malaria, Falciparum/drug therapy , Peroxides/pharmacology , Peroxides/pharmacokinetics , Plasmodium falciparum/drug effects , Adamantane/administration & dosage , Adamantane/pharmacokinetics , Adamantane/pharmacology , Administration, Oral , Adolescent , Adult , Antimalarials/administration & dosage , Cohort Studies , Female , Healthy Volunteers , Humans , Male , Microbial Sensitivity Tests , Parasite Load , Peroxides/administration & dosage , Real-Time Polymerase Chain Reaction , Treatment Outcome , Young Adult
10.
Malar J ; 15: 469, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27624471

ABSTRACT

BACKGROUND: Ferroquine (SSR97193) is a candidate anti-malarial currently undergoing clinical trials for malaria. To better understand its pharmacokinetic (PK) and pharmacodynamic (PD) parameters the compound was tested in the experimentally induced blood stage malaria infection model in volunteers. METHODS: Male and non-pregnant female aged 18-50 years were screened for this phase II, controlled, single-centre clinical trial. Subjects were inoculated with ~1800 viable Plasmodium falciparum 3D7A-infected human erythrocytes, and treated with a single-dose of 800 mg ferroquine. Blood samples were taken at defined time-points to measure PK and PD parameters. The blood concentration of ferroquine and its active metabolite, SSR97213, were measured on dry blood spot samples by ultra-performance liquid chromatography with tandem mass spectrometry (LC-MS/MS). Parasitaemia and emergence of gametocytes were monitored by quantitative PCR. Safety was determined by recording adverse events and monitoring clinical laboratory assessments during the course of the study. RESULTS: Eight subjects were enrolled into the study, inoculated with infected erythrocytes and treated with 800 mg ferroquine. Ferroquine was rapidly absorbed with maximal exposure after 4-8 and 4-12 h exposure for SSR97213. Non-compartmental PK analysis resulted in estimates for half-lives of 10.9 and 23.8 days for ferroquine and SSR97213, respectively. Parasite clearance as reported by parasite reduction ratio was 162.9 (95 % CI 141-188) corresponding to a parasite clearance half-life of 6.5 h (95 % CI: 6.4-6.7 h). PK/PD modelling resulted in a predicted minimal parasiticidal concentration of 20 ng/mL, and the single dosing tested in this study was predicted to maintain an exposure above this threshold for 454 h (37.8 days). Although ferroquine was overall well tolerated, transient elevated transaminase levels were observed in three subjects. Paracetamol was the only concomitant treatment among the two out of these three subjects that may have played a role in the elevated transaminases levels. No clinically significant ECG abnormalities were observed. CONCLUSIONS: The parameters and PK/PD model derived from this study pave the way to the further rational development of ferroquine as an anti-malarial partner drug. The safety of ferroquine has to be further explored in controlled human trials. Trial registration anzctr.org.au (registration number: ACTRN12613001040752), registered 18/09/2013.


Subject(s)
Aminoquinolines/administration & dosage , Antimalarials/administration & dosage , Ferrous Compounds/administration & dosage , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Adolescent , Adult , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Blood/parasitology , Blood Chemical Analysis , Chromatography, Liquid , Female , Ferrous Compounds/pharmacokinetics , Ferrous Compounds/pharmacology , Healthy Volunteers , Humans , Male , Metallocenes , Middle Aged , Parasite Load , Plasmodium falciparum/isolation & purification , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry , Treatment Outcome , Young Adult
12.
Proc Natl Acad Sci U S A ; 107(36): 15774-9, 2010 Sep 07.
Article in English | MEDLINE | ID: mdl-20798033

ABSTRACT

Protein tyrosine phosphatases (PTPs) are regulated through reversible oxidation of the active-site cysteine. Previous studies have implied soluble reactive oxygen species (ROS), like H(2)O(2), as the mediators of PTP oxidation. The potential role(s) of peroxidized lipids in PTP oxidation have not been described. This study demonstrates that increases in cellular lipid peroxides, induced by disruption of glutathione peroxidase 4, induce cellular PTP oxidation and reduce the activity of PDGF receptor targeting PTPs. These effects were accompanied by site-selective increased PDGF beta-receptor phosphorylation, sensitive to 12/15-lipoxygenase (12/15-LOX) inhibitors, and increased PDGF-induced cytoskeletal rearrangements. Importantly, the 12/15-LOX-derived 15-OOH-eicosatetraenoic acid lipid peroxide was much more effective than H(2)O(2) in induction of in vitro PTP oxidation. Our study thus establishes that lipid peroxides are previously unrecognized inducers of oxidation of PTPs. This identifies a pathway for control of receptor tyrosine kinase signaling, which might also be involved in the etiology of diseases associated with increased lipid peroxidation.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Lipid Peroxides/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Enzyme Activation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Mice , Oxidation-Reduction , Phospholipid Hydroperoxide Glutathione Peroxidase , Phosphorylation , Reactive Oxygen Species/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism
13.
J Mol Cell Cardiol ; 52(6): 1257-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22446161

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) regulates tyrosine kinase receptor-mediated responses, and especially negatively influences insulin sensitivity, thus PTP1B inhibitors (PTP1Bi) are currently evaluated in the context of diabetes. We recently revealed another important target for PTP1Bi, consisting in endothelial protection. The present study was designed to test whether reduction of PTP1B activity may be beneficial in chronic heart failure (CHF). We evaluated the impact of either a 2 month pharmacological inhibition, or a gene deletion of PTP1B (PTP1B(-/-)) in CHF mice (2 months post-myocardial infarction). PTP1Bi and PTP1B deficiency reduced adverse LV remodeling, and improved LV function, as shown by the increased LV fractional shortening and cardiac output (measured by echocardiography), the increased LV end systolic pressure, and the decreased LV end diastolic pressure, at identical infarct sizes. This was accompanied by reduced cardiac fibrosis, myocyte hypertrophy and cardiac expression of ANP. In vitro vascular studies performed in small mesenteric artery segments showed a restored endothelial function (i.e. improved NO-dependent, flow-mediated dilatation, increased eNOS phosphorylation) after either pharmacological inhibition or gene deletion. PTP1B(-/-) CHF also displayed an improved insulin sensitivity (assessed by euglycemic-hyperinsulinemic clamp studies), when compared to wild-type CHF associated with an increased insulin mediated mesenteric artery dilation. Thus, chronic pharmacological inhibition or gene deletion of PTP1B improves cardiac dysfunction and cardiac remodeling in the absence of changes in infarct size. Thus this enzyme may be a new therapeutic target in CHF. Diabetic patients with cardiac complications may potentially benefit from PTP1B inhibition via two different mechanisms, reduced diabetic complications, and reduced heart failure.


Subject(s)
Gene Deletion , Heart Failure/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Animals , Disease Models, Animal , Echocardiography , Enzyme Inhibitors/pharmacology , Gene Expression , Heart Failure/drug therapy , Heart Failure/physiopathology , Hemodynamics , Insulin Resistance , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase/genetics , Ventricular Remodeling
14.
Cell Commun Signal ; 10(1): 19, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22784513

ABSTRACT

Protein-tyrosine phosphatases (PTPs) are important regulators of cellular signaling and changes in PTP activity can contribute to cell transformation. Little is known about the role of PTPs in Acute Myeloid Leukemia (AML). The aim of this study was therefore to establish a PTP expression profile in AML cells and to explore the possible role of FLT3 ITD (Fms-like tyrosine kinase 3 with internal tandem duplication), an important oncoprotein in AML for PTP gene expression. PTP mRNA expression was analyzed in AML cells from patients and in cell lines using a RT-qPCR platform for detection of transcripts of 92 PTP genes. PTP mRNA expression was also analyzed based on a public microarray data set for AML patients. Highly expressed PTPs in AML belong to all PTP subfamilies. Very abundantly expressed PTP genes include PTPRC, PTPN2, PTPN6, PTPN22, DUSP1, DUSP6, DUSP10, PTP4A1, PTP4A2, PTEN, and ACP1. PTP expression was further correlated with the presence of FLT3 ITD, focusing on a set of highly expressed dual-specificity phosphatases (DUSPs). Elevated expression of DUSP6 in patients harboring FLT3 ITD was detected in this analysis. The mechanism and functional role of FLT3 ITD-mediated upregulation of DUSP6 was then explored using pharmacological inhibitors of FLT3 ITD signal transduction and si/shRNA technology in human and murine cell lines. High DUSP6 expression was causally associated with the presence of FLT3 ITD and dependent on FLT3 ITD kinase activity and ERK signaling. DUSP6 depletion moderately increased ERK1/2 activity but attenuated FLT3 ITD-dependent cell proliferation of 32D cells. In conclusion, DUSP6 may play a contributing role to FLT3 ITD-mediated cell transformation.

15.
ACS Infect Dis ; 8(4): 713-720, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35286809

ABSTRACT

The current Covid-19 pandemic has underlined the need for a more coordinated and forward-looking investment in the search for new medicines targeting emerging health care threats. Repositioning currently approved drugs is a popular approach to any new emerging disease, but it represents a first wave of response. Behind this would be a second wave of more specifically designed therapies based on activities against specific molecular targets or in phenotypic assays. Following the successful deployment and uptake of previous open access compound collections, we assembled the Pandemic Response Box, a collection of 400 compounds to facilitate drug discovery in emerging infectious disease. These are based on public domain information on chemotypes currently in discovery and early development which have been shown to have useful activities and were prioritized by medicinal chemistry experts. They are freely available to the community as a pharmacological test set with the understanding that data will be shared rapidly in the public domain.


Subject(s)
COVID-19 Drug Treatment , Pandemics , Disease Outbreaks , Drug Discovery , Humans
16.
J Neurosci Res ; 88(12): 2546-57, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20544820

ABSTRACT

Oligodendrocytes generate and maintain myelin, which is essential for axonal function and protection of the mammalian central nervous system. To advance our molecular understanding of differentiation by these cells, we screened libraries of pharmacologically active compounds and identified inducers of differentiation of Oli-neu, a stable cell line of mouse oligodendrocyte precursors (OPCs). We identified four broad classes of inducers, namely, forskolin/cAMP (protein kinase A activators), steroids (glucocorticoids and retinoic acid), ErbB2 inhibitors, and nucleoside analogs, and confirmed the activity of these compounds on rat primary oligodendrocyte precursors and mixed cortical cultures. We also analyzed transcriptional responses in the chemically induced mouse and rat OPC differentiation processes and compared these with earlier studies. We confirm the view that ErbB2 is a natural signaling component that is required for OPC proliferation, whereas ErbB2 inhibition or genetic knockdown results in OPC differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/drug effects , Cerebral Cortex/metabolism , Oligodendroglia/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Animals , Animals, Newborn , Biomarkers/analysis , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Colforsin/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Oligodendroglia/cytology , RNA Interference/physiology , Rats , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology
17.
Acta Neuropathol ; 119(2): 157-75, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19936768

ABSTRACT

Gliomas are a diverse group of brain tumors of glial origin. Most are characterized by diffuse infiltrative growth in the surrounding brain. In combination with their refractive nature to chemotherapy this makes it almost impossible to cure patients using combinations of conventional therapeutic strategies. The drastically increased knowledge about the molecular underpinnings of gliomas during the last decade has elicited high expectations for a more rational and effective therapy for these tumors. Most studies on the molecular pathways involved in glioma biology thus far had a strong focus on growth factor receptor protein tyrosine kinase (PTK) and phosphatidylinositol phosphatase signaling pathways. Except for the tumor suppressor PTEN, much less attention has been paid to the PTK counterparts, the protein tyrosine phosphatase (PTP) superfamily, in gliomas. PTPs are instrumental in the reversible phosphorylation of tyrosine residues and have emerged as important regulators of signaling pathways that are linked to various developmental and disease-related processes. Here, we provide an overview of the current knowledge on PTP involvement in gliomagenesis. So far, the data point to the potential implication of receptor-type (RPTPdelta, DEP1, RPTPmicro, RPTPzeta) and intracellular (PTP1B, TCPTP, SHP2, PTPN13) classical PTPs, dual-specific PTPs (MKP-1, VHP, PRL-3, KAP, PTEN) and the CDC25B and CDC25C PTPs in glioma biology. Like PTKs, these PTPs may represent promising targets for the development of novel diagnostic and therapeutic strategies in the treatment of high-grade gliomas.


Subject(s)
Brain Neoplasms/enzymology , Glioma/enzymology , Protein Tyrosine Phosphatases/metabolism , Signal Transduction/physiology , Animals , Humans
18.
PLoS Negl Trop Dis ; 14(1): e0007860, 2020 01.
Article in English | MEDLINE | ID: mdl-31999695

ABSTRACT

In the past two decades there has been a significant expansion in the number of new therapeutic monoclonal antibodies (mAbs) that are approved by regulators. The discovery of these new medicines has been driven primarily by new approaches in inflammatory diseases and oncology, especially in immuno-oncology. Other recent successes have included new antibodies for use in viral diseases, including HIV. The perception of very high costs associated with mAbs has led to the assumption that they play no role in prophylaxis for diseases of poverty. However, improvements in antibody-expression yields and manufacturing processes indicate this is a cost-effective option for providing protection from many types of infection that should be revisited. Recent technology developments also indicate that several months of protection could be achieved with a single dose. Moreover, new methods in B cell sorting now enable the systematic identification of high-quality antibodies from humanized mice, or patients. This Review discusses the potential for passive immunization against schistosomiasis, fungal infections, dengue, and other neglected diseases.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neglected Diseases/drug therapy , Animals , Dengue/drug therapy , Drug Development , Humans , Immunization, Passive , Mice , Mycoses/drug therapy , Schistosomiasis/drug therapy , Tropical Medicine
19.
Structure ; 15(4): 499-509, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17437721

ABSTRACT

Tyrosine kinases and phosphatases establish the crucial balance of tyrosine phosphorylation in cellular signaling, but creating specific inhibitors of protein Tyr phosphatases (PTPs) remains a challenge. Here, we report the development of a potent, selective inhibitor of Mycobacterium tuberculosis PtpB, a bacterial PTP that is secreted into host cells where it disrupts unidentified signaling pathways. The inhibitor, (oxalylamino-methylene)-thiophene sulfonamide (OMTS), showed an IC(50) of 440 +/- 50 nM and >60-fold specificity for PtpB over six human PTPs. The 2 A resolution crystal structure of PtpB in complex with OMTS revealed a large rearrangement of the enzyme, with some residues shifting >27 A relative to the PtpB:PO(4) complex. Extensive contacts with the catalytic loop provide a potential basis for inhibitor selectivity. Two OMTS molecules bound adjacent to each other, raising the possibility of a second substrate phosphotyrosine binding site in PtpB. The PtpB:OMTS structure provides an unanticipated framework to guide inhibitor improvement.


Subject(s)
Enzyme Inhibitors/chemistry , Mycobacterium tuberculosis/enzymology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Sulfonamides/chemistry , Thiophenes/chemistry , Enzyme Inhibitors/pharmacology , Kinetics , Structure-Activity Relationship , Sulfonamides/pharmacology , Thiophenes/pharmacology
20.
Mol Immunol ; 45(14): 3756-66, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18614237

ABSTRACT

T cell receptor signaling processes are controlled by the integrated actions of families of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPases). Several distinct cytosolic protein tyrosine phosphatases have been described that are able to negatively regulate TCR signaling pathways, including SHP-1, SHP-2, PTPH1, and PEP. Using PTPase substrate-trapping mutants and wild type enzymes, we determined that PTPN4/PTP-MEG1, a PTPH1-family member, could complex and dephosphorylate the ITAMs of the TCR zeta subunit. In addition, the substrate-trapping derivative augmented basal and TCR-induced activation of NF-kappaB in T cells. To characterize the contribution of this PTPase in T cells, we developed PTPN4-deficient mice. T cell development and TCR signaling events were comparable between wild type and PTPN4-deficient animals. The magnitude and duration of TCR-regulated ITAM phosphorylation, as well as overall protein phosphorylation, was unaltered in the absence of PTPN4. Finally, Th1- and Th2-derived cytokines and in vivo immune responses to Listeria monocytogenes were equivalent between wild type and PTPN4-deficient mice. These findings suggest that additional PTPases are involved in controlling ITAM phosphorylations.


Subject(s)
NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 4/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Jurkat Cells , Kidney/cytology , Mice , NF-kappa B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 4/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 4/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 4/metabolism , Receptors, Antigen, T-Cell/immunology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL