Leo G. Flores
University of Texas MD Anderson Cancer Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Leo G. Flores.
Cell | 2012
Chia Hsin Chan; Chien Feng Li; Wei Lei Yang; Yuan Gao; Szu Wei Lee; Zizhen Feng; Hsuan Ying Huang; Kelvin K.-C. Tsai; Leo G. Flores; Yiping Shao; John D. Hazle; Dihua Yu; Wenyi Wei; Dos D. Sarbassov; Mien Chie Hung; Keiichi I. Nakayama; Hui Kuan Lin
Akt kinase plays a central role in cell growth, metabolism, and tumorigenesis. The TRAF6 E3 ligase orchestrates IGF-1-mediated Akt ubiquitination and activation. Here, we show that Akt ubiquitination is also induced by activation of ErbB receptors; unexpectedly, and in contrast to IGF-1 induced activation, the Skp2 SCF complex, not TRAF6, is a critical E3 ligase for ErbB-receptor-mediated Akt ubiquitination and membrane recruitment in response to EGF. Skp2 deficiency impairs Akt activation, Glut1 expression, glucose uptake and glycolysis, and breast cancer progression in various tumor models. Moreover, Skp2 overexpression correlates with Akt activation and breast cancer metastasis and serves as a marker for poor prognosis in Her2-positive patients. Finally, Skp2 silencing sensitizes Her2-overexpressing tumors to Herceptin treatment. Our study suggests that distinct E3 ligases are utilized by diverse growth factors for Akt activation and that targeting glycolysis sensitizes Her2-positive tumors to Herceptin treatment.
Cancer Research | 2011
Wei Lu; Marites P. Melancon; Chiyi Xiong; Qian Huang; Andrew M. Elliott; Shaoli Song; Rui Zhang; Leo G. Flores; Juri G. Gelovani; Lihong V. Wang; Geng Ku; R. Jason Stafford; Chun Li
Advancements in nanotechnology have made it possible to create multifunctional nanostructures that can be used simultaneously to image and treat cancers. For example, hollow gold nanospheres (HAuNS) have been shown to generate intense photoacoustic signals and induce efficient photothermal ablation (PTA) therapy. In this study, we used photoacoustic tomography, a hybrid imaging modality, to assess the intravenous delivery of HAuNS targeted to integrins that are overexpressed in both glioma and angiogenic blood vessels in a mouse model of glioma. Mice were then treated with near-infrared laser, which elevated tumor temperature by 20.7°C. We found that PTA treatment significantly prolonged the survival of tumor-bearing mice. Taken together, these results show the feasibility of using a single nanostructure for image-guided local tumor PTA therapy with photoacoustic molecular imaging.
Journal of Clinical Investigation | 2011
Fernanda I. Staquicini; Michael G. Ozawa; Catherine A. Moya; Wouter Driessen; E. Magda Barbu; Hiroyuki Nishimori; Suren Soghomonyan; Leo G. Flores; Xiaowen Liang; Vincenzo Paolillo; Mian M. Alauddin; James P. Basilion; Frank B. Furnari; Oliver Bögler; Frederick F. Lang; Kenneth D. Aldape; Gregory N. Fuller; Magnus Höök; Juri G. Gelovani; Richard L. Sidman; Webster K. Cavenee; Renata Pasqualini; Wadih Arap
The management of CNS tumors is limited by the blood-brain barrier (BBB), a vascular interface that restricts the passage of most molecules from the blood into the brain. Here we show that phage particles targeted with certain ligand motifs selected in vivo from a combinatorial peptide library can cross the BBB under normal and pathological conditions. Specifically, we demonstrated that phage clones displaying an iron-mimic peptide were able to target a protein complex of transferrin and transferrin receptor (TfR) through a non-canonical allosteric binding mechanism and that this functional protein complex mediated transport of the corresponding viral particles into the normal mouse brain. We also showed that, in an orthotopic mouse model of human glioblastoma, a combination of TfR overexpression plus extended vascular permeability and ligand retention resulted in remarkable brain tumor targeting of chimeric adeno-associated virus/phage particles displaying the iron-mimic peptide and carrying a gene of interest. As a proof of concept, we delivered the HSV thymidine kinase gene for molecular-genetic imaging and targeted therapy of intracranial xenografted tumors. Finally, we established that these experimental findings might be clinically relevant by determining through human tissue microarrays that many primary astrocytic tumors strongly express TfR. Together, our combinatorial selection system and results may provide a translational avenue for the targeted detection and treatment of brain tumors.
Proceedings of the National Academy of Sciences of the United States of America | 2011
Hsin Hsien Yeh; Kazuma Ogawa; Julius Balatoni; Asutosh Pal; Carlos Gonzalez-Lepera; Aleksandr Shavrin; Suren Soghomonyan; Leo G. Flores; Daniel Young; Andrei Volgin; Amer Najjar; Victor Krasnykh; William P. Tong; Mian M. Alauddin; Juri G. Gelovani
The importance of the EGF receptor (EGFR) signaling pathway in the development and progression of nonsmall cell lung carcinomas (NSCLC) is widely recognized. Gene sequencing studies revealed that a majority of tumors responding to EGFR kinase inhibitors harbor activating mutations in the EGFR kinase domain. This underscores the need for novel biomarkers and diagnostic imaging approaches to identify patients who may benefit from particular therapeutic agents and approaches with improved efficacy and safety profiles. To this goal, we developed 4-[(3-iodophenyl)amino]-7-{2-[2-{2-(2-[2-{2-([18F]fluoroethoxy)-ethoxy}-ethoxy]-ethoxy)-ethoxy}-ethoxy]-quinazoline-6-yl-acrylamide ([18F]F-PEG6-IPQA), a radiotracer with increased selectivity and irreversible binding to the active mutant L858R EGFR kinase. We show that PET with [18F]F-PEG6-IPQA in tumor-bearing mice discriminates H3255 NSCLC xenografts expressing L858R mutant EGFR from H441 and PC14 xenografts expressing EGFR or H1975 xenografts with L858R/T790M dual mutation in EGFR kinase domain, which confers resistance to EGFR inhibitors (i.e., gefitinib). The T790M mutation precludes the [18F]F-PEG6-IPQA from irreversible binding to EGFR. These results suggest that PET with [18F]F-PEG6-IPQA could be used for the selection of NSCLC patients for individualized therapy with small molecular inhibitors of EGFR kinase that are currently used in the clinic and have a similar structure (i.e., iressa, gefitinib, and erlotinib).
The Journal of Nuclear Medicine | 2011
Chiyi Xiong; Miao Huang; Rui Zhang; Shaoli Song; Wei Lu; Leo G. Flores; Juri G. Gelovani; Chun Li
Many solid tumors overexpress EphB4 receptor, a member of the ephrin receptor tyrosine kinase family. Noninvasive imaging of EphB4 could potentially increase early detection rates, monitor response to therapy directed against EphB4, and improve patient outcomes. The purpose of this study was to evaluate a novel 64Cu-labeled peptide with high receptor binding affinity for PET of EphB4 receptors. Methods: The EphB4-binding peptide TNYLFSPNGPIARAW (TNYL-RAW) was conjugated with fluorescein isothiocyanate (FITC) and DOTA. DOTA-TNYL-RAW was labeled with 64Cu with high labeling efficiency. The binding affinity of TNYL-RAW and its derivatives to purified recombinant EphB4 was determined using surface plasmon resonance technology. In vitro binding of both FITC-TNYL-RAW and 64Cu-DOTA-TNYL-RAW to cancer cells was assessed by fluorescent microscopy and a radioactivity count method. In vivo biodistribution and small-animal PET/CT were performed in mice bearing EphB4-expressing CT26 and PC-3M tumors as well as EphB4-negative A549 tumors. Results: TNYL-RAW and its derivatives displayed high binding affinity to EphB4, with equilibrium dissociation constant of 1.98–23 nM. In vitro, both FITC-TNYL-RAW and 64Cu-DOTA-TNYL-RAW were selectively taken up by CT26 and PC-3M cells but not by A549 cells. Binding of FITC-TNYL-RAW and 64Cu-DOTA-TNYL-RAW to CT26 and PC-3M cells could be blocked by an excess amount of TNYL-RAW. In vivo, 64Cu-DOTA-TNYL-RAW showed significantly higher uptake in PC-3M tumors than in A549 tumors, with percentages of injected dose per gram of tumor of 0.84 ± 0.09 and 0.44 ± 0.09 at 24 h after radiotracer injection, respectively. Small-animal PET/CT clearly revealed deposition of 64Cu-DOTA-TNYL-RAW in CT26 and PC-3M tumors but not in A549 tumors. Furthermore, uptake of 64Cu-DOTA-TNYL-RAW in both CT26 and PC-3M tumors could be blocked by cold TNYL-RAW. Conclusion: The expression of EphB4 receptors can be noninvasively interrogated by small-animal PET/CT using 64Cu-DOTA-TNYL-RAW.
NeuroImage | 2013
Hsien-Hsien Yeh; Mei Tian; Rainer Hinz; Daniel Young; Alexander Shavrin; Leo G. Flores; Julius Balatoni; Suren Soghomonyan; Hwan J. Jeong; Ashutosh Pal; Rajesh Uthamanthil; James N. Jackson; Ryuichi Nishii; Hiroshi Mizuma; Hirotaka Onoe; Shinya Kagawa; Tatsuya Higashi; Nobuyoshi Fukumitsu; Mian M. Alauddin; William P. Tong; Karl Herholz; Juri G. Gelovani
Epigenetic modifications mediated by histone deacetylases (HDACs) play important roles in the mechanisms of different neurologic diseases and HDAC inhibitors (HDACIs) have shown promise in therapy. However, pharmacodynamic profiles of many HDACIs in the brain remain largely unknown due to the lack of validated methods for noninvasive imaging of HDAC expression-activity. In this study, dynamic PET/CT imaging was performed in 4 rhesus macaques using [(18)F]FAHA, a novel HDAC substrate, and [(18)F]fluoroacetate, the major radio-metabolite of [(18)F]FAHA, and fused with corresponding MR images of the brain. Quantification of [(18)F]FAHA accumulation in the brain was performed using a customized dual-tracer pharmacokinetic model. Immunohistochemical analyses of brain tissue revealed the heterogeneity of expression of individual HDACs in different brain structures and cell types and confirmed that PET/CT/MRI with [(18)F]FAHA reflects the level of expression-activity of HDAC class IIa enzymes. Furthermore, PET/CT/MRI with [(18)F]FAHA enabled non-invasive, quantitative assessment of pharmacodynamics of HDAC inhibitor SAHA in the brain.
The Journal of Nuclear Medicine | 2011
Shaoli Song; Chiyi Xiong; Min Zhou; Wei Lu; Qian Huang; Geng Ku; Jun Zhao; Leo G. Flores; Yicheng Ni; Chun Li
The purpose of this study was to investigate the potential application of small-molecular-weight 64Cu-labeled bis-DOTA-hypericin in the noninvasive assessment of response to photothermal ablation therapy. Methods: Bis-DOTA-hypericin was labeled with 64Cu with high efficiency (>95% without purification). Nine mice bearing subcutaneous human mammary BT474 tumors were used. Five mice were injected intratumorally with semiconductor CuS nanoparticles, followed by near-infrared laser irradiation 24 h later (12 W/cm2 for 3 min), and 4 mice were not treated (control group). All mice were intravenously injected with 64Cu-bis-DOTA-hypericin (24 h after laser treatment in treated mice). Small-animal PET images were acquired at 2, 6, and 24 h after radiotracer injection. All mice were killed immediately after the imaging session for biodistribution and histology study. In vitro cell uptake and surface plasmon resonance studies were performed to validate the small-animal PET results. Results: 64Cu-bis-DOTA-hypericin uptake was significantly higher in the treatment group than in the control group. The percentage injected dose per gram of tissue in the treated and control groups was 1.72 ± 0.43 and 0.76 ± 0.19, respectively (P = 0.017), at 24 h after injection. Autoradiography and histology results were consistent with selective uptake of the radiotracer in the necrotic zone of the tumor induced by photothermal ablation therapy. In vitro results showed that treated BT474 cells had a higher uptake of 64Cu-bis-DOTA-hypericin than nontreated cells. Surface plasmon resonance study showed that bis-DOTA-hypericin had higher binding affinity to phosphatidylserine and phosphatidylethanolamine than to phosphatidylcholine. Conclusion: 64Cu-bis-DOTA-hypericin has a potential to image thermal therapy–induced tumor cell damage. The affinity of 64Cu-bis-DOTA-hypericin for injured tissues may be attributed to the breakdown of the cell membrane and exposure of phosphatidylserine or phosphatidylethanolamine to the radiotracer, which binds selectively to these phospholipids.
Nuclear Medicine and Biology | 2012
Nashaat Turkman; Aleksander Shavrin; Vincenzo Paolillo; Hsin Hsien Yeh; Leo G. Flores; Suren Soghomonian; Brian Rabinovich; Andrei Volgin; Juri G. Gelovani; Mian M. Alauddin
INTRODUCTION The cannabinoid receptor type 2 (CB(2)) is an important target for development of drugs and imaging agents for diseases, such as neuroinflammation, neurodegeneration and cancer. Recently, we reported synthesis and results of in vitro receptor binding of a focused library of fluorinated 2-oxoquinoline derivatives as CB(2) receptor ligands. Some of the compounds demonstrated to be good CB(2)-specific ligands with Ki values in the nanomolar to subnanomolar concentrations; therefore, we pursued the development of their (18)F-labeled analogues that should be useful for positron emission tomography (PET) imaging of CB(2) receptor expression. Here, we report the radiosynthesis of two (18)F-labeled 2-oxoquinoline derivatives and the preliminary in vitro and ex vivo evaluation of one compound as a CB(2)-specific radioligand. METHODS 4-[(18)F]fluorobenzyl amine [(18)F]-3 was prepared by radiofluorination of 4-cyano-N,N,N-trimethylanilinium triflate salt followed by reduction with LiAlH(4) and then coupled with acid chlorides 11 and 12 to afford [(18)F]-13 and [(18)F]-14. In vitro CB(2) receptor binding assay was performed using U87 cells transduced with CB(2) and CB(1) receptor. Ex vivo autoradiography was performed with [(18)F]-14 on spleen and on CB(2)- and CB(1)-expressing and wild-type U87 subcutaneous tumors grown in mice. RESULTS The radiochemical yields of [(18)F]-13 and [(18)F]-14 were 10%-15.0% with an average of 12% (n=10); radiochemical purity was >99% with specific activity 1200 mCi/μmol. The dissociation constant Kd for [(18)F]-14 was 3.4 nM. Ex vivo autoradiography showed accumulation of [(18)F]-14 in the CB(2)-expressing tumor. CONCLUSION Two new [(18)F]-labeled CB(2) ligands have been synthesized. Compound [(18)F]-14 appears to be a potential PET imaging agent for the assessment of CB(2) receptor expression; however, poor solubility restrain its use in vivo.
Annals of Nuclear Medicine | 1998
Shigeki Nagamachi; Seishi Jdmnouchi; Takeshi Kurose; Takashi Ohnishi; Leo G. Flores; Hiroshi Nakahara; Shigemi Futami; Shozo Tamura; Shigeru Matsukura
PurposeTo investigate the influence of diabetic myocardial damage (suspected myocardial damage; SMD) diagnosed by201T1-SPECT and diabetic cardiac autonomic neuropathy (AN) on myocardial MIBG uptake in patients with non-insulin-dependent diabetes mellitus (NIDDM).Subjects and MethodsEighty-seven diabetic patients divided into four subgroups: 23 with SMD (+) AN (+); 19 with SMD (+) AN (−); 27 with SMD (−) AN (+); 18 with SMD (−) AN (−), and 10 controls were studied. Both planar and SPECT images were taken at 30 minutes (early) and 3 hours (delayed) after123I-MIBG injection. The heart to mediastinum uptake ratio (H/M) and washout ratio of123I-MIBG (WR) were obtained from both planar images. On SPECT images, the total uptake score (TUS) was obtained by the 5 point score method by dividing the myocardium into 20 segments on visual analysis. Similarly, the difference between the201Tl image and the123I-MIBG image in TUS was taken as the difference in the total uptake score (ΔTUS) representing cardiac sympathetic denervation without SMD.ResultsOn both early and delayed planar images, the mean H/M value in the subgroups of diabetic patients was significantly lower in the SMD (+) AN (+) group than in the control group, but among those subgroups, there was statistically significant difference between the SMD (+) AN (+) and SMD (−) AN (−) groups only on the delayed images. Regarding the WR value, there was no statistically significant difference among subjects. On SPECT image analysis, the diabetic sub-group with AN or SMD had statistically significant lower values for TUS than those of the control group. Among diabetics, there was a statistically significant differences between SMD [+] AN [+] and SMD [−] AN [−] on both early and delayed images. Similarly, the SMD [+] AN [−] group also had significantly lower values than those of SMD [−] AN [−] on early images. Regarding ΔTUS, there was a statistically significant differences between AN [+] subgroups and controls. Similarly, the mean value for ΔTUS was much higher in AN [+] subgroups than in AN [−] subgroups with or without SMD in diabetes mellitus.Conclusion123I-MIBG myocardial uptake is affected by both SMD and cardiac autonomie neuropathy. Based on the finding that ΔTUS was much higher in AN [+] subgroups and there was no statistically significant difference between SMD [+] AN [+] and SMD [−] AN [+] subgroups, a decrease in myocardial123I-MIBG uptake might progress independently of SMD.
PLOS ONE | 2009
Leo G. Flores; Susanna Bertolini; Hsin Hsin Yeh; Daniel Young; Uday Mukhopadhyay; Ashutosh Pal; Yunming Ying; Andrei Volgin; Aleksandr Shavrin; Suren Soghomonyan; William P. Tong; William G. Bornmann; Mian M. Alauddin; Craig D. Logsdon; Juri G. Gelovani
Background Early diagnosis of pancreatic carcinoma with highly sensitive diagnostic imaging methods could save lives of many thousands of patients, because early detection increases resectability and survival rates. Current non-invasive diagnostic imaging techniques have inadequate resolution and sensitivity for detection of small size (∼2–3 mm) early pancreatic carcinoma lesions. Therefore, we have assessed the efficacy of positron emission tomography and computer tomography (PET/CT) imaging with β-O-D-galactopyranosyl-(1,4′)-2′-deoxy-2′-[18F]fluoroethyl-D-glucopyranose ([18F]FEDL) for detection of less than 3 mm orthotopic xenografts of L3.6pl pancreatic carcinomas in mice. [18F]FEDL is a novel radioligand of hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein (HIP/PAP), which is overexpressed in peritumoral pancreatic acinar cells. Methodology/Principal Findings Dynamic PET/CT imaging demonstrated rapid accumulation of [18F]FEDL in peritumoral pancreatic tissue (4.04±2.06%ID/g), bi-exponential blood clearance with half-lives of 1.65±0.50 min and 14.14±3.60 min, and rapid elimination from other organs and tissues, predominantly by renal clearance. Using model-independent graphical analysis of dynamic PET data, the average distribution volume ratio (DVR) for [18F]FEDL in peritumoral pancreatic tissue was estimated as 3.57±0.60 and 0.94±0.72 in sham-operated control pancreas. Comparative analysis of quantitative autoradiographic images and densitometry of immunohistochemically stained and co-registered adjacent tissue sections demonstrated a strong linear correlation between the magnitude of [18F]FEDL binding and HIP/PAP expression in corresponding regions (r = 0.88). The in situ analysis demonstrated that at least a 2–4 fold apparent lesion size amplification was achieved for submillimeter tumors and to nearly half a murine pancreas for tumors larger than 3 mm. Conclusion/Significance We have demonstrated the feasibility of detection of early pancreatic tumors by non-invasive imaging with [18F]FEDL PET/CT of tumor biomarker HIP/PAP over-expressed in peritumoral pancreatic tissue. Non-invasive non-invasive detection of early pancreatic carcinomas with [18F]FEDL PET/CT imaging should aid the guidance of biopsies and additional imaging procedures, facilitate the resectability and improve the overall prognosis.