Network


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

Hotspot


Dive into the research topics where Jothilingam Sivapackiam is active.

Publication


Featured researches published by Jothilingam Sivapackiam.


Methods of Molecular Biology | 2010

Targeted chemotherapy in drug-resistant tumors, noninvasive imaging of P-glycoprotein-mediated functional transport in cancer, and emerging role of Pgp in neurodegenerative diseases.

Jothilingam Sivapackiam; Seth T. Gammon; Scott E. Harpstrite; Vijay Sharma

Multidrug resistance (MDR) mediated by overexpression of P-glycoprotein (Pgp) is one of the best characterized transporter-mediated barriers to successful chemotherapy in cancer patients and is also a rapidly emerging target in the progression of neurodegenerative disorders such as Alzheimers and Parkinsons diseases. Therefore, strategies capable of delivering chemotherapeutic agents into drug-resistant tumors and targeted radiopharmaceuticals acting as ultrasensitive molecular imaging probes for detecting functional Pgp expression in vivo could be expected to play a vital role in systemic biology as personalized medicine gains momentum in the twenty-first century. While targeted therapy could be expected to deliver optimal doses of chemotherapeutic drugs into the desired targets, the interrogation of Pgp-mediated transport activity in vivo via noninvasive imaging techniques (SPECT and PET) would be beneficial in stratification of patient populations likely to benefit from a given therapeutic treatment, thereby assisting management of drug resistance in cancer and treatment of neurodegenerative diseases. Both strategies could play a vital role in advancement of personalized treatments in cancer and neurodegenerative diseases. Via this tutorial, authors make an attempt in outlining these strategies and discuss their strengths and weaknesses.


PLOS ONE | 2014

A Generator-produced gallium-68 radiopharmaceutical for PET imaging of myocardial perfusion

Vijay Sharma; Jothilingam Sivapackiam; Scott E. Harpstrite; Julie L. Prior; Hannah Gu; Nigam P. Rath; David Piwnica-Worms

Lipophilic cationic technetium-99m-complexes are widely used for myocardial perfusion imaging (MPI). However, inherent uncertainties in the supply chain of molybdenum-99, the parent isotope required for manufacturing 99Mo/99mTc generators, intensifies the need for discovery of novel MPI agents incorporating alternative radionuclides. Recently, germanium/gallium (Ge/Ga) generators capable of producing high quality 68Ga, an isotope with excellent emission characteristics for clinical PET imaging, have emerged. Herein, we report a novel 68Ga-complex identified through mechanism-based cell screening that holds promise as a generator-produced radiopharmaceutical for PET MPI.


Nuclear Medicine and Biology | 2016

67/68Galmydar: A metalloprobe for monitoring breast cancer resistance protein (BCRP)-mediated functional transport activity

Jothilingam Sivapackiam; Scott E. Harpstrite; Julie L. Prior; Stephen Mattingly; Vijay Sharma

INTRODUCTION For stratification of chemotherapeutic choices, radiopharmaceuticals capable of imaging breast cancer resistance protein (BCRP/ABCG2)-mediated functional transport are desired. To accomplish this objective, Galmydar, a fluorescent and moderately hydrophobic Ga(III) cationic complex and its (67/68)Ga-radiolabeled counterparts were interrogated in HEK293 cells stably transfected with BCRP and their WT counterparts transfected with empty vector. Additionally, the sensitivity and specificity of (68)Ga-Galmydar to evaluate functional expression of BCRP at the blood-brain barrier (BBB) was investigated in gene-knockout mdr1a/1b(-/-) (double knockout, dKO) and mdr1a/1b(-/-)ABCG2(-/-) (triple knockout, tKO) mouse models. METHODS For radiotracer uptake assays and live cell fluorescence imaging, either (67)Ga-Galmydar or its unlabeled counterpart was incubated in HEK293 cells transfected with BCRP (HEK293/BCRP) and their WT counterparts at 37°C under a continuous flux of CO2 (5%) in the presence or absence of Ko143, a potent BCRP antagonist, and cellular uptake was measured to assess the sensitivity of Galmydar to probe BCRP-mediated functional transport activity in cellulo. For assessing the potential of Galmydar to enable diagnostic imaging of targeted tissues in vivo, the (67)Ga-radiolabeled counterpart was incubated in either human serum albumin or human serum at 37°C and the percentage of unbound (67)Ga-Galmydar was determined. To evaluate the sensitivity of (68)Ga-Galmydar for molecular imaging of BCRP-mediated efflux activity in vivo, microPET/CT brain imaging was performed in dKO and tKO mice and their age-matched WT counterparts, 60min post-intravenous injection. RESULTS (67)Ga-Galmydar shows uptake profiles in HEK293 cells inversely proportional to BCRP expression, and antagonist (Ko143) induced accumulation in HEK293/BCRP cells, thus indicating target sensitivity and specificity. Furthermore, employing the fluorescent characteristics of Galmydar, optical imaging in HEK293/BCRP cells shows an excellent correlation with the radiotracer cellular accumulation data. (67)Ga-Galmydar shows > 85% unbound fraction and presence of parental compound in human serum. Finally, microPET/CT imaging shows higher retention of (68)Ga-Galmydar in brains of dKO and tKO mice compared to their age-matched WT counterparts, 60min post-intravenous tail-vein injection. CONCLUSIONS Combined data indicate that Galmydar could provide a template scaffold for development of a PET tracer for imaging BCRP-mediated functional transport activity in vivo.


EJNMMI research | 2015

Synthesis, characterization, and preclinical validation of a PET radiopharmaceutical for interrogating Aβ (β-amyloid) plaques in Alzheimer’s disease

Guruswami S.M. Sundaram; Dhruva Dhavale; Julie L. Prior; Jothilingam Sivapackiam; Richard Laforest; Paul T. Kotzbauer; Vijay Sharma

BackgroundPET radiopharmaceuticals capable of imaging β-amyloid (Aβ) plaque burden in the brain could offer highly valuable diagnostic tools for clinical studies of Alzheimer’s disease. To further supplement existing armamentarium of FDA-approved agents as well as those under development, and to correlate multiphoton-imaging data reported earlier, herein, we describe preclinical validation of a PET tracer.MethodsA novel PET radiopharmaceutical (18F-7B) was synthesized and characterized. To assess its affinity for Aβ, binding assays with Aβ1-42 fibrils, Alzheimer’s disease (AD) homogenates, and autoradiography studies and their IHC correlations were performed. For assessing its overall pharmacokinetic profiles in general and its ability to cross the blood-brain barrier (BBB) in particular, biodistribution studies in normal mice were performed. Finally, for evaluating potential for 18F-7B to serve as a targeted Aβ probe, the microPET/CT imaging was performed in age-matched amyloid precursor protein/presenilin-1 (APP/PS1) mice and wild-type (WT) counterparts.ResultsThe radiotracer 18F-7B shows saturable binding to autopsy-confirmed AD homogenates (Kd = 17.7 nM) and Aβ1-42 fibrils (Kd = 61 nM). Preliminary autoradiography studies show binding of 18F-7B to cortical Aβ plaques in autopsy-confirmed AD tissue sections, inhibition of that binding by unlabeled counterpart 7A-indicating specificity, and a good correlation of tracer binding with Aβ immunostaining. The agent indicates high initial penetration into brains (7.23 ± 0.47%ID/g; 5 min) of normal mice, thus indicating a 5-min/120-min brain uptake clearance ratio of 4.7, a benchmark value (>4) consistent with the ability of agents to traverse the BBB to enable PET brain imaging. Additionally, 18F-7B demonstrates the presence of parental species in human serum. Preliminary microPET/CT imaging demonstrates significantly higher retention of 18F-7B in brains of transgenic mice compared with their WT counterparts, consistent with expected binding of the radiotracer to Aβ plaques, present in APP/PS1 mice, compared with their age-matched WT counterparts lacking those Aβ aggregates.ConclusionsThese data offer a platform scaffold conducive to further optimization for developing new PET tracers to study Aβ pathophysiology in vitro and in vivo.


Nuclear Medicine and Biology | 2017

68Ga-Galmydar: Biodistribution and Radiation Dosimetry Studies in Rodents

Jothilingam Sivapackiam; Richard Laforest; Vijay Sharma

INTRODUCTION 68Ga[Ga]-Galmydar is an avid transport substrate of ABCB1 (P-Glycoprotein; 170kDa plasma membrane protein), breast cancer resistance protein (BCRP; ABCG2; 72kDa), penetrates human epidermal carcinoma (KB3-1), breast cancer (MCF7), embryonic kidney (HEK 293) tumor cells and rat cardiomyoblasts, and localizes within the mitochondria of tumor and myocardium cells. 68Ga[Ga]-Galmydar excretes from blood pool quickly, and shows stable retention within rat myocardium in vivo for extended periods, therefore, the agent shows potential to enable myocardial perfusion imaging. The PET tracer also demonstrates ability to probe viability of the blood brain barrier (BBB) in WT mice compared with their mdr1a/1b(-/-) (dKO) and mdr1a/1b/ABCG2(-/-/-) (t-KO) counterparts. Herein, we report dosimetry data for 68Ga[Ga]-Galmydar in mice, and extrapolate that information to determine effective dose (ED) for human studies. METHODS To further assess safety profiles of 68Ga[Ga]-Galmydar for enabling its deployment as a PET imaging probe for biomedical imaging in vivo, we estimated human radiation dosimetry extrapolated from mice biodistribution data. To accomplish this objective, 68Ga[Ga]-Galmydar was injected intravenously into tails, mice were euthanized, organs harvested (5min, 15min, 30min, 60min, 120min), counted, radiation doses to each organ, and whole body were also determined. RESULTS The effective dose (ED) have been found to be 0.021mGy/MBq in males and 0.023mGy/MBq in females. The highest radiation dose was estimated to the kidneys with a value of 0.17mGy/MBq for males and 0.19mGy/MBq for females with contribution from activity in the urine prior to excretion. The critical organ in humans has been determined to be the gall bladder. These data provide preliminary projections on human dosimetry derived from rodent estimates thus providing platform for further validation of dosimetry analysis in human subjects. CONCLUSIONS Combined data obtained from radiation dosimetry studies in mice indicate that 68Ga[Ga]-Galmydar would be safe for further evaluation of dosimetry toxicity and myocardial perfusion PET imaging in humans.


Dalton Transactions | 2010

Synthesis, molecular structure, and validation of metalloprobes for assessment of MDR1 P-glycoprotein-mediated functional transport

Jothilingam Sivapackiam; Scott E. Harpstrite; Julie L. Prior; Hannah Gu; Nigam P. Rath; Vijay Sharma


Archive | 2015

HETEROCYCLIC MOLECULES FOR BIOMEDICAL IMAGING AND THERAPEUTIC APPLICATIONS

G.S.M. Sundaram; Jothilingam Sivapackiam; Vijay Sharma


Medicinal Chemistry | 2010

Synthesis, Characterization, and Molecular Structure of a Novel Zinc (II) Complex: Assessment of Impact of MDR1Pgp Expression on its Cytotoxic Activity

Scott E. Harpstrite; Julie L. Prior; Jothilingam Sivapackiam; Silvia D. Collins; Nigam P. Rath; Vijay Sharma


MedChemComm | 2017

67Ga-metalloprobes: monitoring the impact of geometrical isomers on accumulation profiles in rat cardiomyoblasts and human breast carcinoma cells

Jothilingam Sivapackiam; Scott E. Harpstrite; Nigam P. Rath; Vijay Sharma


Archive | 2014

Molécules hétérocycliques pour l'imagerie biomédicale et leurs applications thérapeutiques

Vijay Sharma; Jothilingam Sivapackiam; Gsm Sundaram

Collaboration


Dive into the Jothilingam Sivapackiam's collaboration.

Top Co-Authors

Avatar

Vijay Sharma

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Scott E. Harpstrite

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Julie L. Prior

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Nigam P. Rath

University of Missouri–St. Louis

View shared research outputs
Top Co-Authors

Avatar

David Piwnica-Worms

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hannah Gu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Richard Laforest

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Dhruva Dhavale

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

G.S.M. Sundaram

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Guruswami S.M. Sundaram

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge