Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Umesh B. Gangadharmath is active.

Publication


Featured researches published by Umesh B. Gangadharmath.


Alzheimers & Dementia | 2013

[18F]T807, a novel tau positron emission tomography imaging agent for Alzheimer's disease

Chunfang Xia; Janna Arteaga; Gang Chen; Umesh B. Gangadharmath; Luis F. Gomez; Dhanalakshmi Kasi; Chung Lam; Qianwa Liang; Changhui Liu; Vani P. Mocharla; Fanrong Mu; Anjana Sinha; Helen Su; A. Katrin Szardenings; Joseph C. Walsh; Eric Wang; Chul Yu; Wei Zhang; Tieming Zhao; Hartmuth C. Kolb

We wished to develop a highly selective positron emission tomography (PET) imaging agent targeting PHF‐tau in human Alzheimers disease (AD) brains.


Journal of Alzheimer's Disease | 2012

A Highly Selective and Specific PET Tracer for Imaging of Tau Pathologies

Wei Zhang; Janna Arteaga; Daniel Kurt Cashion; Gang Chen; Umesh B. Gangadharmath; Luis F. Gomez; Dhanalakshmi Kasi; Chung Lam; Qianwa Liang; Changhui Liu; Vani P. Mocharla; Fanrong Mu; Anjana Sinha; A. Katrin Szardenings; Eric Wang; Joseph C. Walsh; Chunfang Xia; Chul Yu; Tieming Zhao; Hartmuth C. Kolb

Senile plaques and neurofibrillary tangles are prominent neuropathological hallmarks in Alzheimers disease and are considered to be targets for therapeutic intervention as well as biomarkers for diagnostic in vivo imaging agents. While there are a number of amyloid-β positron emission tomography (PET) tracers currently in different stages of clinical development and commercialization, there have been very few reports on imaging agents selectively targeting tau aggregates. In search of [18F]-PET tracers that possess great binding affinity and selectivity toward tau tangles, we tested more than 900 compounds utilizing a unique screening process. A competitive autoradiography assay was set up to test compounds for binding to native tau tangles and amyloid-β plaques on human brain tissue sections. In our in vitro assays, the 18F labeled compound [18F]-T808 displayed a high level of binding affinity and good selectivity for tau aggregates over amyloid-β plaques. [18F]-T808 showed rapid uptake and washout in rodent brains. Our in vitro and preclinical in vivo studies suggest that [18F]-T808 possesses suitable properties and characteristics to be a specific and selective PET probe for imaging of paired helical filament tau in human brains.


Journal of Medicinal Chemistry | 2012

A New Class of Highly Potent Matrix Metalloproteinase Inhibitors Based on Triazole-Substituted Hydroxamates: (Radio)Synthesis and in Vitro and First in Vivo Evaluation

Verena Hugenberg; Hans-Jörg Breyholz; Burkhard Riemann; Sven Hermann; Otmar Schober; Michael Schäfers; Umesh B. Gangadharmath; Vani P. Mocharla; Hartmuth C. Kolb; Joseph C. Walsh; Wei Zhang; Klaus Kopka; Stefan Wagner

In vivo imaging of MMPs is of great (pre)clinical interest and can potentially be realized with modern three-dimensional and noninvasive in vivo molecular imaging techniques such as positron emission tomography (PET). Consequently, MMP inhibitors (MMPIs) radiolabeled with positron emitting nuclides (e.g., (18)F) represent a suitable tool for the visualization of activated MMPs with PET. On the basis of our previous work and results regarding radiolabeled and unlabeled derivatives of the nonselective MMPIs, we discovered a new class of fluorinated MMPIs with a triazole-substituted hydroxamate substructure. These novel MMPIs are characterized by an increased hydrophilicity compared with the lead structures and excellent MMP inhibition potencies for MMP-2, MMP-8, MMP-9, and MMP-13 (IC(50) = 0.006-107 nM). Therefore, one promising fluorinated triazole-substituted hydroxamate (30b) was selected and resynthesised as its (18)F-labeled version to yield the potential PET radioligand [(18)F]30b. The biodistribution behavior of this novel compound was investigated with small animal PET.


Circulation-cardiovascular Imaging | 2015

Noninvasive Molecular Imaging of Apoptosis in a Mouse Model of Anthracycline-Induced Cardiotoxicity

Helen Su; Natalia Gorodny; Luis F. Gomez; Umesh B. Gangadharmath; Fanrong Mu; Gang Chen; Joseph C. Walsh; Katrin Szardenings; Hartmuth C. Kolb; Balaji Tamarappoo

Background—Anthracycline-induced cardiotoxicity and myocardial dysfunction may be associated with apoptosis. Caspase 3 catalyzes a terminal step in apoptosis, and its expression may serve as a marker of cardiomyocyte apoptosis. We synthesized 18F-CP18, a caspase-3 substrate and evaluated cardiac 18F-CP18 uptake in a mouse model of anthracycline cardiotoxicity. Methods and Results—For 12 weeks, mice were injected with doxorubicin, 3 mg/kg/week, or vehicle (control). Left ventricular fractional shortening was quantified by echocardiography. CP18 uptake after intravenous injection of 250 &mgr;Ci of 18F-CP18, 24 hours post-doxorubicin treatment was quantified by microPET, autoradiography, and gamma counting. Apoptosis was assessed by enzymatic assay of myocardial caspase 3 and TUNEL staining of tissue sections. Compared with controls, at 6 and 12 weeks of doxorubicin treatment, fractional shortening was reduced (20.7%±2.5% versus 31%±3.5%, P=0.010; and 20.3%±3.1% versus 32.4%±2.1%, P=0.011). Doxorubicin treatment was associated with increased 18F-CP18 uptake in %ID/g by gamma counting from 0.36±0.01 (week 1) to 0.78±0.01 (week 12), P=0.003. A similar increase in 18F-CP18 uptake was observed by microPET (0.41±0.04 versus 0.73±0.1, P=0.014) and autoradiography (1.1±0.3 versus 2.8±0.2 P=0.001). Caspase 3 enzymatic activity and apoptosis by TUNEL staining were also increased after 12 weeks of doxorubicin compared with weeks 1 and 3. CP18 uptake in controls was relatively unchanged at weeks 1, 3, and 12. Conclusions—In a mouse model of cardiotoxicity, doxorubicin treatment is associated with increased myocardial caspase 3 expression and an increase in CP18 uptake. 18F-CP18 may be useful for detection of anthracycline-induced myocardial apoptosis.


Journal of Nuclear Cardiology | 2014

Atherosclerotic plaque uptake of a novel integrin tracer 18F-Flotegatide in a mouse model of atherosclerosis

Helen Su; Natalia Gorodny; Luis F. Gomez; Umesh B. Gangadharmath; Fanrong Mu; Gang Chen; Joseph C. Walsh; Katrin Szardenings; Daniel S. Berman; Hartmuth C. Kolb; Balaji Tamarappoo

IntroductionRupture of unstable atherosclerotic plaque that leads to stroke and myocardial infarction may be induced by macrophage infiltration and neovessel formation. A tracer that selectively binds to integrin αvβ3 a protein expressed by macrophages and neovascular endothelium may identify rupture prone plaque.Methods18F-labeled “R-G-D” containing tripeptide (Flotegatide), a click chemistry derived radiotracer that binds to integrin αvβ3 was injected in ApoE knockout mice fed a high fat diet. Uptake of Flotegatide by atherosclerotic plaque was visualized by micro-PET, autoradiography, and correlated to histologic markers of inflammation and angiogenesis.ResultsWe found that Flotegatide preferentially binds to aortic plaque in an ApoE knockout mouse model of atherosclerosis. The tracer’s uptake is strongly associated with presence of histologic markers for macrophage infiltration and integrin expression. There is a weaker but detectable association between Flotegatide uptake and presence of an immunohistochemical marker for neovascularization.DiscussionWe hypothesize that Flotegatide may be a useful tracer for visualization of inflamed plaque in clinical subjects with atherosclerosis and may have potential for detecting vulnerable plaque.


Molecular Imaging and Biology | 2013

Evaluation of [18F]-CP18 as a PET Imaging Tracer for Apoptosis

Helen Su; Gang Chen; Umesh B. Gangadharmath; Luis F. Gomez; Qianwa Liang; Fanrong Mu; Vani P. Mocharla; A. Katrin Szardenings; Joseph C. Walsh; Chunfang Xia; Chul Yu; Hartmuth C. Kolb

PurposeWe identified and validated [18F]-CP18, a DEVD (the caspase 3 substrate recognition motif) containing substrate-based compound as an imaging tracer for caspase-3 activity in apoptotic cells.ProceduresCP18 was radiolabeled with fluorine-18 using click chemistry. The affinity and selectivity of CP18 for caspase-3 were evaluated in vitro. The biodistribution and metabolism pattern of [18F]-CP18 were assessed in vivo. [18F]-CP18 positron emission tomography (PET) scans were performed in a dexamethasone-induced thymic apoptosis mouse model. After imaging, the mice were sacrificed, and individual organs were collected, measured in a gamma counter, and tested for caspase-3 activity.ResultsIn vitro enzymatic caspase-3 assay demonstrated specific cleavage of CP18. In vivo, [18F]-CP18 is predominantly cleared through the kidneys and urine, and is rapidly eliminated from the bloodstream. There was a sixfold increase in caspase activity and a fourfold increase of [18F]-CP18 retention in the dexamethasone-induced thymus of treated versus control mice.ConclusionsWe report the use [18F]-CP18 as a PET tracer for imaging apoptosis. Our data support further development of this tracer for clinical PET applications.


Molecular Imaging and Biology | 2013

In Vitro and In Vivo Evaluation of the Caspase-3 Substrate-Based Radiotracer [ 18 F]-CP18 for PET Imaging of Apoptosis in Tumors

Chunfang Xia; Gang Chen; Umesh B. Gangadharmath; Luis F. Gomez; Qianwa Liang; Fanrong Mu; Vani P. Mocharla; Helen Su; A. Katrin Szardenings; Joseph C. Walsh; Tieming Zhao; Hartmuth C. Kolb

PurposeA novel caspase-3 substrate-based probe [18F]-CP18 was evaluated as an in vivo positron emission tomography (PET) imaging agent for monitoring apoptosis in tumors.MethodsUptake of [18F]-CP18 in cell assays and tumors was measured. Caspase-3/7 activities in cell lysates and tumor homogenates were determined. Autoradiography,Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and cleaved caspase-3 immunostaining were performed on adjacent tumor sections to identify areas of apoptosis.ResultsThe in vitro cell assays showed caspase-3-dependent uptake of [18F]-CP18 in tumor cells when treated with an apoptosis inducer. The in vivo microPET imaging signal of [18F]-CP18 in xenograft tumors correlated with the ex vivo caspase-3/7 activities in these tumors. Furthermore, tumor autoradiographies of [18F]-CP18 in tumor sections matched adjacent sections stained by TUNEL and caspase-3 immunohistochemistry (IHC).Conclusions[18F]-CP18 demonstrated high affinity and selectivity for activated caspase-3 both in vitro and in vivo, and the results support [18F]-CP18 as a promising new PET imaging agent for apoptosis.


Archive | 2010

IMAGING AGENTS FOR DETECTING NEUROLOGICAL DISORDERS

Umesh B. Gangadharmath; Hartmuth C. Kolb; Peter J.H. Scott; Joseph C. Walsh; Wei Zhang; Anna Katrin Szardenings; Anjana Sinha; Gang Chen; Eric Wang; Vani P. Mocharia; Chul Yu; Changhui Liu; Daniel Kurt Cashion; Dhanalakshmi Kasi


Journal of Medicinal Chemistry | 2013

Inverse 1,2,3-Triazole-1-yl-ethyl Substituted Hydroxamates as Highly Potent Matrix Metalloproteinase Inhibitors: (Radio)synthesis, in Vitro and First in Vivo Evaluation

Verena Hugenberg; Burkhard Riemann; Sven Hermann; Otmar Schober; Michael Schäfers; Katrin Szardenings; Artem Lebedev; Umesh B. Gangadharmath; Hartmuth C. Kolb; Joseph C. Walsh; Wei Zhang; Klaus Kopka; Stefan Wagner


Archive | 2008

Development of molecular imaging probes for carbonic anhydrase-IX using click chemistry

Hartmuth C. Kolb; Joseph C. Walsh; Dhanalakshmi Kasi; Vani P. Mocharla; Bing Wang; Umesh B. Gangadharmath; Brian A. Duclos; Kai Chen; Wei Zhang; Gang Chen; Henry C. Padgett; Farhad Karimi; Peter J.H. Scott; Zhiyong Gao; Qianwa Liang; Thomas Lee Collier; Tieming Zhao; Chunfang Xia

Collaboration


Dive into the Umesh B. Gangadharmath's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gang Chen

Janssen Pharmaceutica

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge