Bio


Dr. Habte, originally from Eritrea, got his MSc and Ph.D. from Stockholm University, Sweden, in applied Physics. Prior to his recruitment in 2008 as an imaging scientist at Stanford University, Dr. Habte has worked on various research centered on imaging system development, including new radiation detector design, readout electronics, Monte Carlo simulations and computations tools. In his current position, as director of a small animal imaging facility at Stanford, Dr. Habte run and oversee the operations and services of the imaging facility. His work requires close collaboration with researchers and investigators to help solve any issues related to the in vivo imaging and quantitation of images acquired with different modalities including PET/SPECT/CT, PET/MRI, MRI, Optical Imaging, Ultrasound, Photoacoustic and other Cell, or Ex-vivo imaging systems. In addition, Dr. Habte is actively involved in various collaborative and independent research. Some of the recent and ongoing projects include integrated image data management system for preclinical and clinical translation, algorithm development for image segmentation, validation and optimization of image acquisition and analysis for preclinical and theranostic applications.

Current Role at Stanford


Director, Stanford Center for Innovation, in In vivo Imaging

Honors & Awards


  • Justice, Diversity, Equity and Inclusion Seed Grants Award in Radiology, Stanford, Department of Radiology (2021)
  • Small Equipment Grant, Stanford School of Medicine (2019)
  • Most cited paper award (2006-2008), Physica Medica (2009)
  • Travel award IEEE nuclear science symposium and medical imaging conference., IEEE (2006)
  • Travel award IEEE nuclear science symposium and medical imaging conference., IEEE (2006)
  • Travel Award Annual Conference of the National Society of Black Physicists, National Society of Black Physicists (2003)
  • UNESCO scholarship and travel award to attend the 1997 CERN School of Computing, CERN (1997)
  • Full Ph.D. scholarship award, Swedish International Development Agency (SIDA) (1995-2001)
  • Scholarship and travel award to attend a post-graduate course, State of Israel (1993)
  • Scholarship to attend international workshops at the, International Centre for Theoretical Physics, Italy (1998, 1991, 1994)

Education & Certifications


  • Ph.D., Stockholm University, Sweden, Applied and Instrumentation Physics (2001)
  • M.Sc, Stockholm University, Sweden, Applied Physics, (1996)
  • B. Sc, University of Asmara, Eritrea, Physics (1988)

All Publications


  • Immune cell identity behind the Ktrans mapping of mouse glioblastoma. Magnetic resonance imaging Zhang, Y., Keunen, O., Golebiewska, A., Gerosa, M., Wang, J., Ghobadi, S. N., Huang, A., Hou, Q., Habte, F. G., Li, N., Grant, G., Paulmurugan, R., Lee, K. S., Wintermark, M. 2023

    Abstract

    Dynamic contrast-enhanced MR imaging (DCE-MRI) can assess the integrity of the blood brain barrier (BBB) and has been used in GBM patients to determine glioma grade, predict prognosis, evaluate treatment response, and differentiate treatment-induced effect from recurrence. The volume transfer constant Ktrans is the most frequently used metric in tumor assessment. Based on previous studies that a higher WHO grade of brain tumor was associated with greater impairments of immunity and that Ktrans value was associated with the pathological grading, the relationship between differential composition of immune cells in GBM tissue and dynamic changes in Ktrans mapping was anticipated in this study. The present study utilized an orthotopic allograft model of GBM in which mouse GL26 cells are implanted into Ccr2RFP/wtCx3cr1GFP/wt mice on a C57 background. The brain tumors exhibited heterogenous Ktrans values with the coefficients of variation (CV) above 75%, or relatively homogeneous Ktrans maps with CV values below 50%. The Ktrans values of homogeneous tumors ranged between 0.02/min-0.32/min with a median value of 0.10/min. The immune cell composition defined by quantitative immunohistochemistry and cell sorting was compared between the tumors with Ktrans values above 0.10/min (higher Ktrans) or below 0.10/min (lower Ktrans). Histological analysis showed that tumors with higher Ktrans values exhibited greater numbers of CCR2pos cells (257.60 ± 16.42/mm2 vs 203.23 ± 12.20/mm2, p = 0.04) and an increased ratio of CCR2pos cells to CX3CR1pos cells (1.20 ± 0.02 vs 0.38 ± 0.04, p = 0.001), the numbers of CX3CR1pos cells did not differ significantly based on Ktrans values (219.70 ± 16.20/mm2 vs 250.38 ± 21.20/mm2, p = 0.19). Flowcytometry analysis showed that tumors with higher Ktrans values (above 0.1/min) were associated with greater numbers of both overall monocytes (54.93 ± 6.81% vs 29.75 ± 3.54%, p = 0.01) and inflammatory monocytes (72.38 ± 1.49% vs 59.52 ± 2.44%, p = 0.001). In contrast, tumors with lower Ktrans values (below 0.1/min) exhibited greater numbers of patrolling monocytes (75.65 ± 4.14% vs 63 ± 6.94%, p = 0.05). In the tumors with lower Ktrans values, all three types of tumor associated cells, including patrolling monocytes, inflammatory monocytes, and microglia cells possessed a higher proportion of cells at pro-inflammatory status (41.77 ± 6.13% vs 25.06 ± 6.72%, p = 0.05; 27.50 ± 2.11% vs 20.62 ± 1.87%, p = 0.03; and 55.80 ± 9.88% vs 31.12 ± 7.31%, p = 0.05), inflammatory monocytes showed fewer anti-inflammatory cells (1.25 ± 0.62% vs 3.16 ± 3.56%, p = 0.04). Taken together, differences in Ktrans values were associated with differential immune cell phenotypes and polarizations. Ktrans mapping may therefore represent a novel approach for defining the immune status of GBM.

    View details for DOI 10.1016/j.mri.2023.06.008

    View details for PubMedID 37353182

  • Molecular Identity Changes of Tumor-Associated Macrophages and Microglia After Magnetic Resonance Imaging-Guided Focused Ultrasound-Induced Blood-Brain Barrier Opening in a Mouse Glioblastoma Model. Ultrasound in medicine & biology Zhang, Y., Wang, J., Ghobadi, S. N., Zhou, H., Huang, A., Gerosa, M., Hou, Q., Keunen, O., Golebiewska, A., Habte, F. G., Grant, G. A., Paulmurugan, R., Lee, K. S., Wintermark, M. 2023

    Abstract

    An orthotopically allografted mouse GL26 glioma model (Ccr2RFP/wt-Cx3cr1GFP/wt) was used to evaluate the effect of transient, focal opening of the blood-brain barrier (BBB) on the composition of tumor-associated macrophages and microglia (TAMs). BBB opening was induced by magnetic resonance imaging (MRI)-guided focused ultrasound (MRgFUS) combined with microbubbles. CX3CR1-GFP cells and CCR2-RFP cells in brain tumors were quantified in microscopic images. Tumors in animals treated with a single session of MRgFUS did not exhibit significant changes in cell numbers when compared with tumors in animals not receiving FUS. However, tumors that received two or three sessions of MRgFUS had significantly increased amounts of both CX3CR1-GFP and CCR2-RFP cells. The effect of MRgFUS on immune cell composition was also characterized and quantified using flow cytometry. Glioma implantation resulted in increased amounts of lymphocytes, monocytes and neutrophils in the brain parenchyma. Tumors administered MRgFUS exhibited increased numbers of monocytes and monocyte-derived TAMs. In addition, MRgFUS-treated tumors exhibited more CD80+ cells in monocytes and microglia. In summary, transient, focal opening of the BBB using MRgFUS combined with microbubbles can activate the homing and differentiation of monocytes and induce a shift toward a more pro-inflammatory status of the immune environment in glioblastoma.

    View details for DOI 10.1016/j.ultrasmedbio.2022.12.006

    View details for PubMedID 36717283

  • Multimodal imaging of capsid and cargo reveals differential brain targeting and liver detargeting of systemically-administered AAVs. Biomaterials Seo, J. W., Ajenjo, J., Wu, B., Robinson, E., Raie, M. N., Wang, J., Tumbale, S. K., Buccino, P., Anders, D. A., Shen, B., Habte, F. G., Beinat, C., James, M. L., Reyes, S. T., Ravindra Kumar, S., Miles, T. F., Lee, J. T., Gradinaru, V., Ferrara, K. W. 2022: 121701

    Abstract

    The development of gene delivery vehicles with high organ specificity when administered systemically is a critical goal for gene therapy. We combine optical and positron emission tomography (PET) imaging of 1) reporter genes and 2) capsid tags to assess the temporal and spatial distribution and transduction of adeno-associated viruses (AAVs). AAV9 and two engineered AAV vectors (PHP.eB and CAP-B10) that are noteworthy for maximizing blood-brain barrier transport were compared. CAP-B10 shares a modification in the 588 loop with PHP.eB, but also has a modification in the 455 loop, added with the goal of reducing off-target transduction. PET and optical imaging revealed that the additional modifications retained brain receptor affinity. In the liver, the accumulation of AAV9 and the engineered AAV capsids was similar (15% of the injected dose per cc and not significantly different between capsids at 21h). However, the engineered capsids were primarily internalized by Kupffer cells rather than hepatocytes, and liver transduction was greatly reduced. PET reporter gene imaging after engineered AAV systemic injection provided a non-invasive method to monitor AAV-mediated protein expression over time. Through comparison with capsid tagging, differences between brain localization and transduction were revealed. In summary, AAV capsids bearing imaging tags and reporter gene payloads create a unique and powerful platform to assay the pharmacokinetics, cellular specificity and protein expression kinetics of AAV vectors in vivo, a key enabler for the field of gene therapy.

    View details for DOI 10.1016/j.biomaterials.2022.121701

    View details for PubMedID 35985893

  • Web-Based Application for Biomedical Image Registry, Analysis, and Translation (BiRAT). Tomography (Ann Arbor, Mich.) Pemmaraju, R., Minahan, R., Wang, E., Schadl, K., Daldrup-Link, H., Habte, F. 2022; 8 (3): 1453-1462

    Abstract

    Imaging has become an invaluable tool in preclinical research for its capability to non-invasively detect and monitor disease and assess treatment response. With the increased use of preclinical imaging, large volumes of image data are being generated requiring critical data management tools. Due to proprietary issues and continuous technology development, preclinical images, unlike DICOM-based images, are often stored in an unstructured data file in company-specific proprietary formats. This limits the available DICOM-based image management database to be effectively used for preclinical applications. A centralized image registry and management tool is essential for advances in preclinical imaging research. Specifically, such tools may have a high impact in generating large image datasets for the evolving artificial intelligence applications and performing retrospective analyses of previously acquired images. In this study, a web-based server application is developed to address some of these issues. The application is designed to reflect the actual experimentation workflow maintaining detailed records of both individual images and experimental data relevant to specific studies and/or projects. The application also includes a web-based 3D/4D image viewer to easily and quickly view and evaluate images. This paper briefly describes the initial implementation of the web-based application.

    View details for DOI 10.3390/tomography8030117

    View details for PubMedID 35736865

  • Engineered Cell-Derived Vesicles Displaying Targeting Peptide and Functionalized with Nanocarriers for Therapeutic microRNA Delivery to Triple-Negative Breast Cancer in Mice. Advanced healthcare materials Bose, R. J., Kumar, U. S., Garcia-Marques, F., Zeng, Y., Habte, F., McCarthy, J. R., Pitteri, S., Massoud, T. F., Paulmurugan, R. 2021: e2101387

    Abstract

    Polymeric nanocarriers (PNCs) can be used to deliver therapeutic microRNAs (miRNAs) to solid cancers. However, the ability of these nanocarriers to specifically target tumors remains a challenge. Alternatively, extracellular vesicles (EVs) derived from tumor cells show homotypic affinity to parent cells, but loading sufficient amounts of miRNAs into EVs is difficult. Here, we investigate whether uPAR-targeted delivery of nanococktails containing PNCs loaded with therapeutic antimiRNAs, and coated with uPA engineered extracellular vesicles (uPA-eEVs) can elicit synergistic antitumor responses. The uPA-eEVs coating on PNCs increases natural tumor targeting affinities, thereby enhancing the antitumor activity of antimiRNA nanococktails. The systemic administration of uPA-eEV-PNCs nanococktail showed a robust tumor tropism, which significantly enhanced the combinational antitumor effects of antimiRNA-21 and antimiRNA-10b, and led to significant tumor regression and extension of progression free survival for syngeneic 4T1 tumor-bearing mice. In addition, the uPA-eEV-PNCs-antimiRNAs nanococktail plus low dose doxorubicin resulted in a synergistic antitumor effect as evidenced by inhibition of tumor growth, reduction of lung metastases, and extension of survival of 4T1 tumor-bearing mice. Our targeted combinational nanococktail strategy could be readily translated to the clinical setting by using autologous cancer cells that have flexibility for ex vivo expansion and genetic engineering. This article is protected by copyright. All rights reserved.

    View details for DOI 10.1002/adhm.202101387

    View details for PubMedID 34879180

  • Non-invasive, neurotoxic surgery reduces seizures in a rat model of temporal lobe epilepsy. Experimental neurology Zhang, Y., Buckmaster, P. S., Qiu, L., Wang, J., Keunen, O., Ghobadi, S. N., Huang, A., Hou, Q., Li, N., Narang, S., Habte, F. G., Bertram, E. H., Lee, K. S., Wintermark, M. 2021: 113761

    Abstract

    Surgery can be highly effective for treating certain cases of drug resistant epilepsy. The current study tested a novel, non-invasive, surgical strategy for treating seizures in a rat model of temporal lobe epilepsy. The surgical approach uses magnetic resonance-guided, low-intensity focused ultrasound (MRgFUS) in combination with intravenous microbubbles to open the blood-brain barrier (BBB) in a transient and focal manner. During the period of BBB opening, a systemically administered neurotoxin (Quinolinic Acid: QA) that is normally impermeable to the BBB gains access to a targeted area in the brain, destroying neurons where the BBB has been opened. This strategy is termed Precise Intracerebral Non-invasive Guided Surgery (PING). Spontaneous recurrent seizures induced by pilocarpine were monitored behaviorally prior to and after PING or under control conditions. Seizure frequency in untreated animals or animals treated with MRgFUS without QA exhibited expected seizure rate fluctuations frequencies between the monitoring periods. In contrast, animals treated with PING targeting the intermediate-temporal aspect of the hippocampus exhibited substantial reductions in seizure frequency, with convulsive seizures being eliminated entirely in two animals. These findings suggest that PING could provide a useful alternative to invasive surgical interventions for treating drug resistant epilepsy, and perhaps for treating other neurological disorders in which aberrant neural circuitries play a role.

    View details for DOI 10.1016/j.expneurol.2021.113761

    View details for PubMedID 33991523

  • High-Throughput Whole-Plate Imaging of Cells for Multiple Biological Applications. Methods in molecular biology (Clifton, N.J.) Sukumar, U. K., Habte, F., Massoud, T. F., Paulmurugan, R. 2021; 2274: 367-384

    Abstract

    Advanced multipurpose cell imaging systems along with integrated rapid quantitation software can enhance and expedite cancer cell culture studies in a variety of applications. Though accurate cell culture studies are an important and necessary component of nearly all cancer biomarker detection and therapy studies, the methods we currently use are of low-throughput, time consuming, and lack accuracy. Hence, it is important to improve several features of the assays to increase the accuracy of their quantitative outputs in most studies. In general, we perform cell culture analysis semimanually by counting a small aliquot of suspended cells using a hemocytometer or viewing a small area of cells on a plate using a bright-field microscope, and then extrapolate the counts or observations to estimate the values for the total numbers of cells. The fundamental problem with this process lies in using techniques, such as extrapolation, which inherently introduces intrasample variability while collecting the cells by enzymatic trypsinization for these assays that are affecting cell growth and other downstream assessments. Fluorescence (FL) microscopy-based assays are also used to image and count cells for various applications, including cell viability, proliferation, apoptosis, cell death, transfection efficiency, protein expression, stem cell properties, colony formation, cytotoxicity, drug dose-response, and treatment efficacy studies. These methods are not optimal for many researches, as they require real-time visualization under a microscope plus manual analysis to determine the final results. Owing to long exposure times for cells under fluorescent light of a microscope, the cells may be exposed to suboptimal conditions that affect cell growth, and with occasional photobleaching of the expressed FL probes. Alternatively, the use of cell imaging systems that integrate both advanced bright-field and FL imaging for cell counting and quantification can be useful. In this protocol, we discuss the advantages of a high-throughput cell imaging system using a whole-plate imaging format when used in various bioimaging studies by highlighting a few applications of the system. The system is designed to fundamentally improve the accuracy and time of cell culture analysis while also allowing us to perform the assay without trypsinization, thus avoiding the need to replicate multiple wells for monitoring cell growth over time.

    View details for DOI 10.1007/978-1-0716-1258-3_30

    View details for PubMedID 34050486

  • Effects of Non-invasive, Targeted, Neuronal Lesions on Seizures in a Mouse Model of Temporal Lobe Epilepsy. Ultrasound in medicine & biology Zhang, Y., Zhou, H., Qu, H., Liao, C., Jiang, H., Huang, S., Ghobadi, S. N., Telichko, A., Li, N., Habte, F. G., Doyle, T., Woznak, J. P., Bertram, E. H., Lee, K. S., Wintermark, M. 2020

    Abstract

    Surgery to treat drug-resistant epilepsy can be quite effective but remains substantially underutilized. A pilot study was undertaken to test the feasibility of using a non-invasive, non-ablative, approach to produce focal neuronal loss to treat seizures in a rodent model of temporal lobe epilepsy. In this study, spontaneous, recurrent seizures were established in a mouse model of pilocarpine-induced status epilepticus. After post-status epilepticus stabilization, baseline behavioral seizures were monitored for 30 d. Non-invasive opening of the blood-brain barrier targeting the hippocampus was then produced by using magnetic resonance-guided, low-intensity focused ultrasound, through which a neurotoxin (quinolinic acid) administered intraperitoneally gained access to the brain parenchyma to produce focal neuronal loss. Behavioral seizures were then monitored for 30 d after this procedure, and brains were subsequently prepared for histologic analysis of the sites of neuronal loss. The average frequency of behavioral seizures in all animals (n = 11) was reduced by 21.2%. Histologic analyses along the longitudinal axis of the hippocampus revealed that most of the animals (n = 8) exhibited neuronal loss located primarily in the intermediate aspect of the hippocampus, while sparing the septal aspect. Two other animals with damage to the intermediate hippocampus also exhibited prominent bilateral damage to the septal aspect of the hippocampus. A final animal had negligible neuronal loss overall. Notably, the site of neuronal loss along the longitudinal axis of the hippocampus influenced seizure outcomes. Animals that did not have bilateral damage to the septal hippocampus displayed a mean decrease in seizure frequency of 27.7%, while those with bilateral damage to the septal hippocampus actually increased seizure frequency by 18.7%. The animal without neuronal loss exhibited an increase in seizure frequency of 19.6%. The findings indicate an overall decrease in seizure frequency in treated animals. And, the site of neuronal loss along the longitudinal axis of the hippocampus appears to play a key role in reducing seizure activity. These pilot data are promising, and they encourage additional and more comprehensive studies examining the effects of targeted, non-invasive, neuronal lesions for the treatment of epilepsy.

    View details for DOI 10.1016/j.ultrasmedbio.2020.01.008

    View details for PubMedID 32081583

  • Trop2 is a driver of metastatic prostate cancer with neuroendocrine phenotype via PARP1. Proceedings of the National Academy of Sciences of the United States of America Hsu, E. C., Rice, M. A., Bermudez, A. n., Marques, F. J., Aslan, M. n., Liu, S. n., Ghoochani, A. n., Zhang, C. A., Chen, Y. S., Zlitni, A. n., Kumar, S. n., Nolley, R. n., Habte, F. n., Shen, M. n., Koul, K. n., Peehl, D. M., Zoubeidi, A. n., Gambhir, S. S., Kunder, C. A., Pitteri, S. J., Brooks, J. D., Stoyanova, T. n. 2020

    Abstract

    Resistance to androgen deprivation therapy, or castration-resistant prostate cancer (CRPC), is often accompanied by metastasis and is currently the ultimate cause of prostate cancer-associated deaths in men. Recently, secondary hormonal therapies have led to an increase of neuroendocrine prostate cancer (NEPC), a highly aggressive variant of CRPC. Here, we identify that high levels of cell surface receptor Trop2 are predictive of recurrence of localized prostate cancer. Moreover, Trop2 is significantly elevated in CRPC and NEPC, drives prostate cancer growth, and induces neuroendocrine phenotype. Overexpression of Trop2 induces tumor growth and metastasis while loss of Trop2 suppresses these abilities in vivo. Trop2-driven NEPC displays a significant up-regulation of PARP1, and PARP inhibitors significantly delay tumor growth and metastatic colonization and reverse neuroendocrine features in Trop2-driven NEPC. Our findings establish Trop2 as a driver and therapeutic target for metastatic prostate cancer with neuroendocrine phenotype and suggest that high Trop2 levels could identify cancers that are sensitive to Trop2-targeting therapies and PARP1 inhibition.

    View details for DOI 10.1073/pnas.1905384117

    View details for PubMedID 31932422

  • Reconstructed Apoptotic Bodies as Targeted "Nano Decoys" to Treat Intracellular Bacterial Infections within Macrophages and Cancer Cells. ACS nano Bose, R. J., Tharmalingam, N. n., Garcia Marques, F. J., Sukumar, U. K., Natarajan, A. n., Zeng, Y. n., Robinson, E. n., Bermudez, A. n., Chang, E. n., Habte, F. n., Pitteri, S. J., McCarthy, J. R., Gambhir, S. S., Massoud, T. F., Mylonakis, E. n., Paulmurugan, R. n. 2020

    Abstract

    Staphylococcus aureus (S. aureus) is a highly pathogenic facultative anaerobe that in some instances resides as an intracellular bacterium within macrophages and cancer cells. This pathogen can establish secondary infection foci, resulting in recurrent systemic infections that are difficult to treat using systemic antibiotics. Here, we use reconstructed apoptotic bodies (ReApoBds) derived from cancer cells as "nano decoys" to deliver vancomycin intracellularly to kill S. aureus by targeting inherent "eat me" signaling of ApoBds. We prepared ReApoBds from different cancer cells (SKBR3, MDA-MB-231, HepG2, U87-MG, and LN229) and used them for vancomycin delivery. Physicochemical characterization showed ReApoBds size ranges from 80 to 150 nm and vancomycin encapsulation efficiency of 60 ± 2.56%. We demonstrate that the loaded vancomycin was able to kill intracellular S. aureus efficiently in an in vitro model of S. aureus infected RAW-264.7 macrophage cells, and U87-MG (p53-wt) and LN229 (p53-mt) cancer cells, compared to free-vancomycin treatment (P < 0.001). The vancomycin loaded ReApoBds treatment in S. aureus infected macrophages showed a two-log-order higher CFU reduction than the free-vancomycin treatment group. In vivo studies revealed that ReApoBds can specifically target macrophages and cancer cells. Vancomycin loaded ReApoBds have the potential to kill intracellular S. aureus infection in vivo in macrophages and cancer cells.

    View details for DOI 10.1021/acsnano.0c00921

    View details for PubMedID 32347709

  • Evaluation of integrin alphavbeta6 cystine knot PET tracers to detect cancer and idiopathic pulmonary fibrosis. Nature communications Kimura, R. H., Wang, L., Shen, B., Huo, L., Tummers, W., Filipp, F. V., Guo, H. H., Haywood, T., Abou-Elkacem, L., Baratto, L., Habte, F., Devulapally, R., Witney, T. H., Cheng, Y., Tikole, S., Chakraborti, S., Nix, J., Bonagura, C. A., Hatami, N., Mooney, J. J., Desai, T., Turner, S., Gaster, R. S., Otte, A., Visser, B. C., Poultsides, G. A., Norton, J., Park, W., Stolowitz, M., Lau, K., Yang, E., Natarajan, A., Ilovich, O., Srinivas, S., Srinivasan, A., Paulmurugan, R., Willmann, J., Chin, F. T., Cheng, Z., Iagaru, A., Li, F., Gambhir, S. S. 2019; 10 (1): 4673

    Abstract

    Advances in precision molecular imaging promise to transform our ability to detect, diagnose and treat disease. Here, we describe the engineering and validation of a new cystine knot peptide (knottin) that selectively recognizes human integrin alphavbeta6 with single-digit nanomolar affinity. We solve its 3D structure by NMR and x-ray crystallography and validate leads with 3 different radiolabels in pre-clinical models of cancer. We evaluate the lead tracer's safety, biodistribution and pharmacokinetics in healthy human volunteers, and show its ability to detect multiple cancers (pancreatic, cervical and lung) in patients at two study locations. Additionally, we demonstrate that the knottin PET tracers can also detect fibrotic lung disease in idiopathic pulmonary fibrosis patients. Our results indicate that these cystine knot PET tracers may have potential utility in multiple disease states that are associated with upregulation of integrin alphavbeta6.

    View details for DOI 10.1038/s41467-019-11863-w

    View details for PubMedID 31611594

  • Intranasal delivery of targeted polyfunctional gold-iron oxide nanoparticles loaded with therapeutic microRNAs for combined theranostic multimodality imaging and presensitization of glioblastoma to temozolomide. Biomaterials Sukumar, U. K., Bose, R. J., Malhotra, M., Babikir, H. A., Afjei, R., Robinson, E., Zeng, Y., Chang, E., Habte, F., Sinclair, R., Gambhir, S. S., Massoud, T. F., Paulmurugan, R. 2019; 218: 119342

    Abstract

    The prognosis for glioblastoma (GBM) remains depressingly low. The biological barriers of the brain present a major challenge to achieving adequate drug concentrations for GBM therapy. To address this, we explore the potential of the nose-to-brain direct transport pathway to bypass the blood-brain barrier, and to enable targeted delivery of theranostic polyfunctional gold-iron oxide nanoparticles (polyGIONs) surface loaded with therapeutic miRNAs (miR-100 and antimiR-21) to GBMs in mice. These nanoformulations would thus allow presensitization of GBM cells to the systemically delivered chemotherapy drug temozolomide (TMZ), as well as in vivo multimodality molecular and anatomic imaging of nanoparticle delivery, trafficking, and treatment effects. First, we synthesized GIONs coated with beta-cyclodextrin-chitosan (CD-CS) hybrid polymer, and co-loaded with miR-100 and antimiR-21. Then we decorated their surface with PEG-T7 peptide using CD-adamantane host-guest chemistry. The resultant polyGIONs showed efficient miRNA loading with enhanced serum stability. We characterized them for particle size, PDI, polymer functionalization, charge and release using dynamic light scattering analysis, TEM and qRT-PCR. For in vivo intranasal delivery, we used U87-MG GBM cell-derived orthotopic xenograft models in mice. Intranasal delivery resulted in efficient accumulation of Cy5-miRNAs in mice treated with T7-targeted polyGIONs, as demonstrated by in vivo optical fluorescence and MR imaging. We measured the therapeutic response of these FLUC-EGFP labelled U87-MG GBMs using bioluminescence imaging. Overall, there was a significant increase in survival of mice co-treated with T7-polyGIONs loaded with miR-100/antimiR-21 plus systemic TMZ, compared to the untreated control group, or the animals receiving non-targeted polyGIONs-miR-100/antimiR-21, or TMZ alone. Once translated clinically, this novel theranostic nanoformulation and its associated intranasal delivery strategy will have a strong potential to potentiate the effects of TMZ treatment in GBM patients.

    View details for DOI 10.1016/j.biomaterials.2019.119342

    View details for PubMedID 31326657

  • Nanomedicine for Spontaneous Brain Tumors: A Companion Clinical Trial ACS NANO Arami, H., Patel, C. B., Madsen, S. J., Dickinson, P. J., Davis, R. M., Zeng, Y., Sturges, B. K., Woolard, K. D., Habte, F. G., Akin, D., Sinclair, R., Gambhir, S. S. 2019; 13 (3): 2858–69
  • Nanomedicine for Spontaneous Brain Tumors: A Companion Clinical Trial. ACS nano Arami, H., Patel, C. B., Madsen, S. J., Dickinson, P. J., Davis, R. M., Zeng, Y., Sturges, B. K., Woolard, K. D., Habte, F. G., Akin, D., Sinclair, R., Gambhir, S. S. 2019

    Abstract

    Nanoparticles' enhanced permeation and retention (EPR) variations due to tumor heterogeneity in naturally occurring brain tumors are commonly neglected in preclinical nanomedicine studies. Recent pathological studies have shown striking similarities between brain tumors in humans and dogs, indicating that canine brain tumors may be a valuable model to evaluate nanoparticles' EPR in this context. We recruited canine clinical cases with spontaneous brain tumors to investigate nanoparticles' EPR in different brain tumor pathologies using surface-enhanced Raman spectroscopy (SERS). We used gold nanoparticles due to their surface plasmon effect that enables their sensitive and microscopic resolution detection using the SERS technique. Raman microscopy of the resected tumors showed heterogeneous EPR of nanoparticles into oligodendrogliomas and meningiomas of different grades, without any detectable traces in necrotic parts of the tumors or normal brain. Raman observations were confirmed by scanning electron microscopy (SEM) and X-ray elemental analyses, which enabled localization of individual nanoparticles embedded in tumor tissues. Our results demonstrate nanoparticles' EPR and its variations in clinically relevant, spontaneous brain tumors. Such heterogeneities should be considered alongside routine preoperative imaging and histopathological analyses in order to accelerate clinical management of brain tumors using nanomedicine approaches.

    View details for PubMedID 30714717

  • TESTING DIFFERENT COMBINATIONS OF ACOUSTIC PRESSURE AND DOSES OF QUINOLINIC ACID FOR INDUCTION OF FOCAL NEURON LOSS IN MICE USING TRANSCRANIAL LOW-INTENSITY FOCUSED ULTRASOUND ULTRASOUND IN MEDICINE AND BIOLOGY Zhang, Y., Liao, C., Qu, H., Huang, S., Jiang, H., Zhou, H., Abrams, E., Habte, F. G., Yuan, L., Bertram, E. H., Lee, K. S., Pauly, K., Buckmaster, P. S., Wintermark, M. 2019; 45 (1): 129–36
  • Tumor Cell-Derived Extracellular Vesicle-Coated Nanocarriers: An Efficient Theranostic Platform for the Cancer-Specific Delivery of Anti-miR-21 and Imaging Agents ACS NANO Bose, R. C., Kumar, S., Zeng, Y., Afjei, R., Robinson, E., Lau, K., Bermudez, A., Habte, F., Pitteri, S. J., Sinclair, R., Willmann, J. K., Massoud, T. F., Gambhir, S. S., Paulmurugan, R. 2018; 12 (11): 10817–32
  • Testing Different Combinations of Acoustic Pressure and Doses of Quinolinic Acid for Induction of Focal Neuron Loss in Mice Using Transcranial Low-Intensity Focused Ultrasound. Ultrasound in medicine & biology Zhang, Y., Liao, C., Qu, H., Huang, S., Jiang, H., Zhou, H., Abrams, E., Habte, F. G., Yuan, L., Bertram, E. H., Lee, K. S., Pauly, K. B., Buckmaster, P. S., Wintermark, M. 2018

    Abstract

    The goal of this study was to test different combinations of acoustic pressure and doses of quinolinic acid (QA) for producing a focal neuronal lesion in the murine hippocampus without causing unwanted damage to adjacent brain structures. Sixty male CD-1 mice were divided into 12 groups that underwent magnetic resonance-guided focused ultrasound at high (0.67 MPa), medium (0.5 MPa) and low (0.33 MPa) acoustic peak negative pressures and received QA at high (0.012 mmol), medium (0.006 mmol) and low (0.003 mmol) dosages. Neuronal loss occurred only when magnetic resonance-guided focused ultrasound with adequate acoustic power (0.67 or 0.5 MPa) was combined with QA. The animals subjected to the highest acoustic power had larger lesions than those treated with medium acoustic power, but two mice had evidence of bleeding. When the intermediate acoustic power was used, medium andhigh dosages of QA produced lesions larger than those produced by the low dosage.

    View details for PubMedID 30309748

  • Quantification of Cerenkov Luminescence Imaging (CLI) Comparable With 3-D PET Standard Measurements MOLECULAR IMAGING Habte, F., Natarajan, A., Paik, D. S., Gambhir, S. 2018; 17
  • Dosimetry Prediction for Clinical Translation of 64Cu-Pembrolizumab ImmunoPET Targeting Human PD-1 Expression. Scientific reports Natarajan, A., Patel, C. B., Habte, F., Gambhir, S. S. 2018; 8 (1): 633

    Abstract

    The immune checkpoint programmed death 1 receptor (PD-1) expressed on some tumor-infiltrating lymphocytes, and its ligand (PD-L1) expressed on tumor cells, enable cancers to evade the immune system. Blocking PD-1 with the monoclonal antibody pembrolizumab is a promising immunotherapy strategy. Thus, noninvasively quantifying the presence of PD-1 expression in the tumor microenvironment prior to initiation of immune checkpoint blockade may identify the patients likely to respond to therapy. We have developed a 64Cu-pembrolizumab radiotracer and evaluated human dosimetry. The tracer was utilized to image hPD-1 levels in two subcutaneous mouse models: (a) 293 T/hPD-1 cells xenografted into NOD-scid IL-2Rγnull mice (NSG/293 T/hPD-1) and (b) human peripheral blood mononuclear cells engrafted into NSG bearing A375 human melanoma tumors (hNSG/A375). In each mouse model two cohorts were evaluated (hPD-1 blockade with pembrolizumab [blk] and non-blocked [nblk]), for a total of four groups (n = 3-5/group). The xenograft-to-muscle ratio in the NSG/293 T/hPD-1 model at 24 h was significantly increased in the nblk group (7.0 ± 0.5) compared to the blk group (3.4 ± 0.9), p = 0.01. The radiotracer dosimetry evaluation (PET/CT ROI-based and ex vivo) in the hNSG/A375 model revealed the highest radiation burden to the liver. In summary, we validated the 64Cu-pembrolizumab tracer's specific hPD-1 receptor targeting and predicted human dosimetry.

    View details for DOI 10.1038/s41598-017-19123-x

    View details for PubMedID 29330552

  • Dosimetry Prediction for Clinical Translation of 64Cu-Pembrolizumab ImmunoPET Targeting Human PD-1 Expression Scientific Reports Natarajan, A., Patel, C. B., Habte, F., Gambhir, S. S. 2018
  • Tumor Cell-Derived Extracellular Vesicle-Coated Nanocarriers: An Efficient Theranostic Platform for the Cancer-Specific Delivery of Anti-miR-21 and Imaging Agents. ACS nano Jc Bose, R. n., Uday Kumar, S. n., Zeng, Y. n., Afjei, R. n., Robinson, E. n., Lau, K. n., Bermudez, A. n., Habte, F. n., Pitteri, S. J., Sinclair, R. n., Willmann, J. K., Massoud, T. F., Gambhir, S. S., Paulmurugan, R. n. 2018

    Abstract

    MicroRNAs are critical regulators of cancer initiation, progression, and dissemination. Extensive evidence suggests that the inhibition of over-expressed oncogenic miRNA function can be a robust strategy for anticancer therapy. However, in vivo targeted delivery of miRNA therapeutics to various types of cancers remains a major challenge. Inspired by their natural synthesis and cargo delivery capabilities, researchers have exploited tumor cell-derived extracellular vesicles (TEVs) for the cancer-targeted delivery of therapeutics and theranostics. Here, we investigate a TEV-based nanoplatform for multimodal miRNA delivery and phototherapy treatments as well as the magnetic resonance imaging of cancer. We demonstrated loading of anti-miR-21 that blocks the function of endogenous oncogenic miR-21 over-expressed in cancer cells into and subsequent delivery by TEVs derived from 4T1 cells. We also produced Cy5-anti-miR-21-loaded TEVs from two other cancer cell lines (HepG2 and SKBR3) and confirmed their robust homologous and heterologous transfection efficiency and intracellular Cy5-anti-miR-21 delivery. Additionally, TEV-mediated anti-miR-21 delivery attenuated doxorubicin (DOX) resistance in breast cancer cells with a 3-fold higher cell kill efficiency than in cells treated with DOX alone. We then investigated TEVs as a biomimetic source for the functionalization of gold-iron oxide nanoparticles (GIONs) and demonstrated nanotheranostic properties of TEV-GIONs in vitro. TEV-GIONs demonstrated excellent T2 contrast in in vitro magnetic resonance (MR) imaging and resulted in efficient photothermal effect in 4T1 cells. We also evaluated the biodistribution and theranostic property of anti-miR-21 loaded TEV-GIONs in vivo by labeling with indocyanine green near-infrared dye. We further validated the tumor specific accumulation of TEV-GIONs using MR imaging. Our findings demonstrate that the distribution pattern of the TEV-anti-miR-21-GIONs correlated well with the tumor-targeting capability as well as the activity and efficacy obtained in response to doxorubicin combination treatments. TEVs and TEV-GIONs are promising nanotheranostics for future applications in cancer molecular imaging and therapy.

    View details for PubMedID 30346694

  • Quantification of Cerenkov Luminescence Imaging (CLI) Comparable With 3-D PET Standard Measurements. Molecular imaging Habte, F., Natarajan, A., Paik, D. S., Gambhir, S. S. 2018; 17: 1536012118788637

    Abstract

    Cerenkov luminescence imaging (CLI) is commonly performed using two-dimensional (2-D) conventional optical imaging systems for its cost-effective solution. However, quantification of CLI comparable to conventional three-dimensional positron emission tomography (PET) is challenging using these systems due to both the high attenuation of Cerenkov radiation (CR) on mouse tissue and nonexisting depth resolution of CLI using 2-D imaging systems (2-D CLI). In this study, we developed a model that estimates effective tissue attenuation coefficient and corrects the tissue attenuation of CLI signal intensity independent of tissue depth and size. To evaluate this model, we used several thin slices of ham as a phantom and placed a radionuclide (89Zr and 64Cu) inside the phantom at different tissue depths and sizes (2, 7, and 12 mm). We performed 2-D CLI and MicroPET/CT (Combined small animal PET and Computed Tomography (CT)) imaging of the phantom and in vivo mouse model after administration of 89Zr tracer. Estimates of the effective tissue attenuation coefficient (mueff) for 89Zr and 64Cu were 2.4 and 2.6 cm-1, respectively. The computed unit conversion factor to %ID/g from 2-D CLI signal was 2.74 * 10-3 muCi/radiance estimated from phantom study. After applying tissue attenuation correction and unit conversion to the in vivo animal study, an average quantification difference of 10% for spleen and 35% for liver was obtained compared to PET measurements. The proposed model provides comparable quantification accuracy to standard PET system independent of deep tissue CLI signal attenuation.

    View details for PubMedID 30043654

  • Reporter gene imaging of targeted T cell immunotherapy in recurrent glioma. Science translational medicine Keu, K. V., Witney, T. H., Yaghoubi, S., Rosenberg, J., Kurien, A., Magnusson, R., Williams, J., Habte, F., Wagner, J. R., Forman, S., Brown, C., Allen-Auerbach, M., Czernin, J., Tang, W., Jensen, M. C., Badie, B., Gambhir, S. S. 2017; 9 (373)

    Abstract

    High-grade gliomas are aggressive cancers that often become rapidly fatal. Immunotherapy using CD8(+) cytotoxic T lymphocytes (CTLs), engineered to express both herpes simplex virus type 1 thymidine kinase (HSV1-TK) and interleukin-13 (IL-13) zetakine chimeric antigen receptor (CAR), is a treatment strategy with considerable potential. To optimize this and related immunotherapies, it would be helpful to monitor CTL viability and trafficking to glioma cells. We show that noninvasive positron emission tomography (PET) imaging with 9-[4-[(18)F]fluoro-3-(hydroxymethyl)butyl]guanine ([(18)F]FHBG) can track HSV1-tk reporter gene expression present in CAR-engineered CTLs. [(18)F]FHBG imaging was safe and enabled the longitudinal imaging of T cells stably transfected with a PET reporter gene in patients. Further optimization of this imaging approach for monitoring in vivo cell trafficking should greatly benefit various cell-based therapies for cancer.

    View details for DOI 10.1126/scitranslmed.aag2196

    View details for PubMedID 28100832

    View details for PubMedCentralID PMC5260938

  • Multimodality Molecular Imaging of Cardiac Cell Transplantation: Part II. In Vivo Imaging of Bone Marrow Stromal Cells in Swine with PET/CT and MR Imaging. Radiology Parashurama, N., Ahn, B., Ziv, K., Ito, K., Paulmurugan, R., Willmann, J. K., Chung, J., Ikeno, F., Swanson, J. C., Merk, D. R., Lyons, J. K., Yerushalmi, D., Teramoto, T., Kosuge, H., Dao, C. N., Ray, P., Patel, M., Chang, Y., Mahmoudi, M., Cohen, J. E., Goldstone, A. B., Habte, F., Bhaumik, S., Yaghoubi, S., Robbins, R. C., Dash, R., Yang, P. C., Brinton, T. J., Yock, P. G., McConnell, M. V., Gambhir, S. S. 2016; 280 (3): 826-836

    Abstract

    Purpose To quantitatively determine the limit of detection of marrow stromal cells (MSC) after cardiac cell therapy (CCT) in swine by using clinical positron emission tomography (PET) reporter gene imaging and magnetic resonance (MR) imaging with cell prelabeling. Materials and Methods Animal studies were approved by the institutional administrative panel on laboratory animal care. Seven swine received 23 intracardiac cell injections that contained control MSC and cell mixtures of MSC expressing a multimodality triple fusion (TF) reporter gene (MSC-TF) and bearing superparamagnetic iron oxide nanoparticles (NP) (MSC-TF-NP) or NP alone. Clinical MR imaging and PET reporter gene molecular imaging were performed after intravenous injection of the radiotracer fluorine 18-radiolabeled 9-[4-fluoro-3-(hydroxyl methyl) butyl] guanine ((18)F-FHBG). Linear regression analysis of both MR imaging and PET data and nonlinear regression analysis of PET data were performed, accounting for multiple injections per animal. Results MR imaging showed a positive correlation between MSC-TF-NP cell number and dephasing (dark) signal (R(2) = 0.72, P = .0001) and a lower detection limit of at least approximately 1.5 × 10(7) cells. PET reporter gene imaging demonstrated a significant positive correlation between MSC-TF and target-to-background ratio with the linear model (R(2) = 0.88, P = .0001, root mean square error = 0.523) and the nonlinear model (R(2) = 0.99, P = .0001, root mean square error = 0.273) and a lower detection limit of 2.5 × 10(8) cells. Conclusion The authors quantitatively determined the limit of detection of MSC after CCT in swine by using clinical PET reporter gene imaging and clinical MR imaging with cell prelabeling. (©) RSNA, 2016 Online supplemental material is available for this article.

    View details for DOI 10.1148/radiol.2016151150

    View details for PubMedID 27332865

  • Multimodality Molecular Imaging of Cardiac Cell Transplantation: Part I. Reporter Gene Design, Characterization, and Optical in Vivo Imaging of Bone Marrow Stromal Cells after Myocardial Infarction. Radiology Parashurama, N., Ahn, B., Ziv, K., Ito, K., Paulmurugan, R., Willmann, J. K., Chung, J., Ikeno, F., Swanson, J. C., Merk, D. R., Lyons, J. K., Yerushalmi, D., Teramoto, T., Kosuge, H., Dao, C. N., Ray, P., Patel, M., Chang, Y., Mahmoudi, M., Cohen, J. E., Goldstone, A. B., Habte, F., Bhaumik, S., Yaghoubi, S., Robbins, R. C., Dash, R., Yang, P. C., Brinton, T. J., Yock, P. G., McConnell, M. V., Gambhir, S. S. 2016; 280 (3): 815-825

    Abstract

    Purpose To use multimodality reporter-gene imaging to assess the serial survival of marrow stromal cells (MSC) after therapy for myocardial infarction (MI) and to determine if the requisite preclinical imaging end point was met prior to a follow-up large-animal MSC imaging study. Materials and Methods Animal studies were approved by the Institutional Administrative Panel on Laboratory Animal Care. Mice (n = 19) that had experienced MI were injected with bone marrow-derived MSC that expressed a multimodality triple fusion (TF) reporter gene. The TF reporter gene (fluc2-egfp-sr39ttk) consisted of a human promoter, ubiquitin, driving firefly luciferase 2 (fluc2), enhanced green fluorescent protein (egfp), and the sr39tk positron emission tomography reporter gene. Serial bioluminescence imaging of MSC-TF and ex vivo luciferase assays were performed. Correlations were analyzed with the Pearson product-moment correlation, and serial imaging results were analyzed with a mixed-effects regression model. Results Analysis of the MSC-TF after cardiac cell therapy showed significantly lower signal on days 8 and 14 than on day 2 (P = .011 and P = .001, respectively). MSC-TF with MI demonstrated significantly higher signal than MSC-TF without MI at days 4, 8, and 14 (P = .016). Ex vivo luciferase activity assay confirmed the presence of MSC-TF on days 8 and 14 after MI. Conclusion Multimodality reporter-gene imaging was successfully used to assess serial MSC survival after therapy for MI, and it was determined that the requisite preclinical imaging end point, 14 days of MSC survival, was met prior to a follow-up large-animal MSC study. (©) RSNA, 2016 Online supplemental material is available for this article.

    View details for DOI 10.1148/radiol.2016140049

    View details for PubMedID 27308957

  • A Thermo-Sensitive Delivery Platform for Topical Administration of Inflammatory Bowel Disease Therapies. Gastroenterology Sinha, S. R., Nguyen, L. P., Inayathullah, M., Malkovskiy, A., Habte, F., Rajadas, J., Habtezion, A. 2015; 149 (1): 52-55 e2

    Abstract

    Systemic therapies for inflammatory bowel disease are associated with increased risk of infections and malignancies. Topical therapies reduce systemic exposure, but can be difficult to retain or have limited proximal distribution. To mitigate these issues, we developed a thermo-sensitive platform, using a polymer-based system that is liquid at room temperature but turns into a viscous gel upon reaching body temperature. Following rectal administration to mice with dextran sulphate sodium-induced colitis, the platform carrying budesonide or mesalamine becomes more viscoelastic near body temperature. Mice given the drug-containing platform gained more weight and had reduced histologic and biologic features of colitis than mice given the platform alone or liquid drugs via enema. Image analysis showed that enemas delivered with and without the platform reached similar distances in the colons of mice, but greater colonic retention was achieved by using the platform.

    View details for DOI 10.1053/j.gastro.2015.04.002

    View details for PubMedID 25863215

  • Cu-64-Labeled Divalent Cystine Knot Peptide for Imaging Carotid Atherosclerotic Plaques JOURNAL OF NUCLEAR MEDICINE Jiang, L., Tu, Y., Kimura, R. H., Habte, F., Chen, H., Cheng, K., Shi, H., Gambhir, S. S., Cheng, Z. 2015; 56 (6): 939-944

    Abstract

    The rupture of vulnerable atherosclerotic plaques that lead to stroke and myocardial infarction may be induced by macrophage infiltration and augmented by the expression of integrin αvβ3. Indeed, atherosclerotic angiogenesis may be a promising marker of inflammation. In this study, an engineered integrin αvβ3-targeting PET probe, (64)Cu-NOTA-3-4A, derived from a divalent knottin miniprotein was evaluated in a mouse model for carotid atherosclerotic plaques.Atherosclerotic plaques in BALB/C mice, maintained on a high-fat diet, were induced with streptozotocin injection and carotid artery ligation and verified by MR imaging. Knottin 3-4A was synthesized by solid-phase peptide synthesis chemistry and coupled to 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) before radiolabeling with (64)Cu. PET probe stability in mouse serum was evaluated. Mice with carotid atherosclerotic plaques were injected via the tail vein with (64)Cu-NOTA-3-4A or (18)F-FDG, followed by small-animal PET/CT imaging at different time points. Receptor targeting specificity of the probe was verified by coinjection of c(RGDyK) administered in molar excess. Subsequently, carotid artery dissection and immunofluorescence staining were performed to evaluate target expression.(64)Cu-NOTA-3-4A was synthesized in high radiochemical purity and yield and demonstrated molecular stability in both phosphate-buffered saline and mouse serum at 4 h. Small-animal PET/CT showed that (64)Cu-NOTA-3-4A accumulated at significantly higher levels in the neovasculature of carotid atherosclerotic plaques (7.41 ± 1.44 vs. 0.67 ± 0.23 percentage injected dose/gram, P < 0.05) than healthy or normal vessels at 1 h after injection. (18)F-FDG also accumulated in atherosclerotic lesions at 0.5 and 1 h after injection but at lower plaque-to-normal tissue ratios than (64)Cu-NOTA-3-4A. For example, plaque-to-normal carotid artery ratios for (18)F-FDG and (64)Cu-NOTA-3-4A at 1 h after injection were 3.75 and 14.71 (P < 0.05), respectively. Furthermore, uptake of (64)Cu-NOTA-3-4A in atherosclerotic plaques was effectively blocked (∼90% at 1 h after injection) by coinjection of c(RGDyK). Immunostaining confirmed integrin αvβ3 expression in both the infiltrating macrophages and the neovasculature of atherosclerotic plaques.(64)Cu-NOTA-3-4A demonstrates specific accumulation in carotid atherosclerotic plaques in which macrophage infiltration and angiogenesis are responsible for elevated integrin αvβ3 levels. Therefore, (64)Cu-NOTA-3-4A may demonstrate clinical utility as a PET probe for atherosclerosis imaging or for the evaluation of therapies used to treat atherosclerosis.

    View details for DOI 10.2967/jnumed.115.155176

    View details for Web of Science ID 000355570300026

    View details for PubMedID 25908832

  • Semiquantitative Analysis of the Biodistribution of the Combined F-18-NaF and F-18-FDG Administration for PET/CT Imaging JOURNAL OF NUCLEAR MEDICINE Minamimoto, R., Mosci, C., Jamali, M., Barkhodari, A., Habte, F., Jackson, T., Mittra, E., Gambhir, S. S., Iagaru, A. 2015; 56 (5): 688-694

    Abstract

    In this study we evaluated the biodistribution of the (18)F-/(18)F-FDG administration compared to separate (18)F-NaF and (18)F-FDG. We also estimated the interaction of (18)F-NaF and (18)F-FDG in the (18)F-/(18)F-FDG administration by semiquantitative analysis.We retrospectively analyzed data of 49 patients (male 39, female 10; mean ± SD age: 59.3 ± 15.2 years) who had separate (18)F-FDG PET/CT and (18)F-NaF PET/CT, as well as the (18)F-/(18)F-FDG PET/CT sequentially. The most common primary diagnosis was prostate cancer (n = 28), followed by sarcoma (n = 9) and breast cancer (n = 6). The mean standardized uptake values (SUVmean) were recorded for 18 organs in all patients, while maximum SUV (SUVmax) and SUVmean were recorded for all the identified malignant lesions. We also estimated the (18)F-/(18)F-FDG uptake by sum of (18)F-FDG uptake and adjusted (18)F-NaF uptake based on the ratio of (18)F-NaF injected dose in (18)F-/(18)F-FDG PET/CT. Lastly, we compared the results in order to explore the interaction of (18)F-FDG and (18)F-NaF uptake in the (18)F-/(18)F-FDG scan.The (18)F-/(18)F-FDG uptake in the cerebral cortex, cerebellum, parotid grand, myocardium and bowel mostly reflect the (18)F-FDG uptake, while the uptake in the other analyzed structures is influenced by both the (18)F-FDG and the (18)F-NaF uptake. The (18)F-/(18)F-FDG uptake in extra skeletal lesions shows no significant difference when compared to the uptake from the separate (18)F-FDG scan. The (18)F-/(18)F-FDG uptake in skeletal lesions reflected mostly the (18)F-NaF uptake. Tumor to background (T/B) ratio of (18)F-/(18)F-FDG in extra skeletal lesions showed no significant difference when compared with that from (18)F-FDG alone (P = 0.73). For skeletal lesions, T/B ratio of (18)F-/(18)F-FDG was lower than that from (18)F-NaF alone (P <0.001); however, this difference did not result in missed skeletal lesions on the (18)F-/(18)F-FDG scan.The understanding of the biodistribution of radiopharmaceuticals and the lesions uptake of the (18)F-/(18)F-FDG scan, as well as the variations compared to the uptake on the separate (18)F-FDG PET/CT and (18)F-NaF PET/CT are valuable for more in depth evaluation of the combined scanning technique.

    View details for DOI 10.2967/jnumed.115.153767

    View details for Web of Science ID 000353831000013

    View details for PubMedID 25840978

  • PET Imaging Carotid Atherosclerostic Plaque Using Divalent Knottin Jiang, L., Tu, Y., Kimura, R., Habte, F., Chen, H., Cheng, K., Shi, H., Gambhir, S., Cheng, Z. SOC NUCLEAR MEDICINE INC. 2015
  • Optical coherence contrast imaging using gold nanorods in living mice eyes CLINICAL AND EXPERIMENTAL OPHTHALMOLOGY de la Zerda, A., Prabhulkar, S., Perez, V. L., Ruggeri, M., Paranjape, A. S., Habte, F., Gambhir, S. S., Awdeh, R. M. 2015; 43 (4): 358-366

    Abstract

    Optical coherence tomography (OCT) is a powerful imaging modality to visualize tissue structures, with axial image pixel resolution as high as 1.6 μm in tissue. However, OCT is intrinsically limited to providing structural information as the OCT contrast is produced by optically scattering tissues.Gold nanorods (GNRs) were injected into the anterior chamber (AC) and cornea of mice eyes which could create a significant OCT signal and hence could be used as a contrast agent for in vivo OCT imaging.A dose of 30 nM of GNRs (13 nm in diameter and 45 nm in length) were injected to the AC of mice eyes and produced an OCT contrast nearly 50-fold higher than control mice injected with saline. Furthermore, the lowest detectable concentration of GNRs in living mice AC was experimentally estimated to be as low as 120 pM.The high sensitivity and low toxicity of GNRs brings great promise for OCT to uniquely become a high-resolution molecular imaging modality.

    View details for DOI 10.1111/ceo.12299

    View details for Web of Science ID 000356810200009

    View details for PubMedID 24533647

  • A simple model for deep tissue attenuation correction and large organ analysis of Cerenkov luminescence imaging Medical Imaging - Physics of Medical Imaging Habte, F., Natarajan, A., Paik, D. S., Gambhir, S. S. SPIE-INT SOC OPTICAL ENGINEERING. 2014

    View details for DOI 10.1117/12.2043879

    View details for Web of Science ID 000338775800154

  • Impact of a Multiple Mice Holder on Quantitation of High-Throughput MicroPET Imaging With and Without Ct Attenuation Correction. Molecular imaging and biology Habte, F., Ren, G., Doyle, T. C., Liu, H., Cheng, Z., Paik, D. S. 2013; 15 (5): 569-575

    Abstract

    PURPOSE: The aim of this study is to evaluate the impact of scanning multiple mice simultaneously on image quantitation, relative to single mouse scans on both a micro-positron emission tomography/computed tomography (microPET/CT) scanner (which utilizes CT-based attenuation correction to the PET reconstruction) and a dedicated microPET scanner using an inexpensive mouse holder "hotel." METHODS: We developed a simple mouse holder made from common laboratory items that allows scanning multiple mice simultaneously. It is also compatible with different imaging modalities to allow multiple mice and multi-modality imaging. For this study, we used a radiotracer ((64)Cu-GB170) with a relatively long half-life (12.7 h), selected to allow scanning at times after tracer uptake reaches steady state. This also reduces the effect of decay between sequential imaging studies, although the standard decay corrections were performed. The imaging was also performed using a common tracer, 2-deoxy-2-[(18) F]fluoro-D-glucose (FDG), although the faster decay and faster pharmacokinetics of FDG may introduce greater biological variations due to differences in injection-to-scan timing. We first scanned cylindrical mouse phantoms (50 ml tubes) both in a groups of four at a time (multiple mice mode) and then individually (single mouse mode), using microPET/CT and microPET scanners to validate the process. Then, we imaged a first set of four mice with subcutaneous tumors (C2C12Ras) in both single- and multiple-mice imaging modes. Later, a second set of four normal mice were injected with FDG and scanned 1 h post-injection. Immediately after completion of the scans, ex vivo biodistribution studies were performed on all animals to provide a "gold-standard" to compare quantitative values obtained from PET. A semi-automatic threshold-based region of interest tool was used to minimize operator variability during image analysis. RESULTS: Phantom studies showed less than 4.5 % relative error difference between the single- and multiple-mice imaging modes of PET imaging with CT-based attenuation correction and 18.4 % without CT-based attenuation correction. In vivo animal studies (n = 4) showed <5 % (for (64)Cu, p > 0.686) and <15 % (for FDG, p > 0.4 except for brain image data p = 0.029) relative mean difference with respect to percent injected dose per gram (%ID/gram) between the single- and multiple-mice microPET imaging mode when CT-based attenuation correction is performed. Without CT-based attenuation correction, we observed relative mean differences of about 11 % for (64)Cu and 15 % for FDG. CONCLUSION: Our results confirmed the potential use of a microPET/CT scanner for multiple mice simultaneous imaging without significant sacrifice in quantitative accuracy as well as in image quality. Thus, the use of the mouse "hotel" is an aid to increasing instrument throughput on small animal scanners with minimal loss of quantitative accuracy.

    View details for DOI 10.1007/s11307-012-0602-y

    View details for PubMedID 23479323

  • Evaluation of Zr-89-rituximab Tracer by Cerenkov Luminescence Imaging and Correlation with PET in a Humanized Transgenic Mouse Model to Image NHL MOLECULAR IMAGING AND BIOLOGY Natarajan, A., Habte, F., Liu, H., Sathirachinda, A., Hu, X., Cheng, Z., Nagamine, C. M., Gambhir, S. S. 2013; 15 (4): 468-475

    Abstract

    PURPOSE: This research aimed to study the use of Cerenkov luminescence imaging (CLI) for non-Hodgkin's lymphoma (NHL) using (89)Zr-rituximab positron emission tomography (PET) tracer with a humanized transgenic mouse model that expresses human CD20 and the correlation of CLI with PET. PROCEDURES: Zr-rituximab (2.6 MBq) was tail vein-injected into transgenic mice that express the human CD20 on their B cells (huCD20TM). One group (n = 3) received 2 mg/kg pre-dose (blocking) of cold rituximab 2 h prior to tracer; a second group (n = 3) had no pre-dose (non-blocking). CLI was performed using a cooled charge-coupled device optical imager. We also performed PET imaging and ex vivo studies in order to confirm the in vivo CLI results. At each time point (4, 24, 48, 72, and 96 h), two groups of mice were imaged in vivo and ex vivo with CLI and PET, and at 96 h, organs were measured by gamma counter. RESULTS: huCD20 transgenic mice injected with (89)Zr-rituximab demonstrated a high-contrast CLI image compared to mice blocked with a cold dose. At various time points of 4-96 h post-radiotracer injection, the in vivo CLI signal intensity showed specific uptake in the spleen where B cells reside and, hence, the huCD20 biomarker is present at very high levels. The time-activity curve of dose decay-corrected CLI intensity and percent injected dose per gram of tissue of PET uptake in the spleen were increased over the time period (4-96 h). At 96 h, the (89)Zr-rituximab uptake ratio (non-blocking vs blocking) counted (mean ± standard deviation) for the spleen was 1.5 ± 0.6 for CLI and 1.9 ± 0.3 for PET. Furthermore, spleen uptake measurements (non-blocking and blocking of all time points) of CLI vs PET showed good correlation (R (2) = 0.85 and slope = 0.576), which also confirmed the corresponding correlations parameter value (R (2) = 0.834 and slope = 0.47) obtained for ex vivo measurements. CONCLUSIONS: CLI and PET of huCD20 transgenic mice injected with (89)Zr-rituximab demonstrated that the tracer was able to target huCD20-expressing B cells. The in vivo and ex vivo tracer uptake corresponding to the CLI radiance intensity from the spleen is in good agreement with PET. In this report, we have validated the use of CLI with PET for NHL imaging in huCD20TM.

    View details for DOI 10.1007/s11307-013-0624-0

    View details for Web of Science ID 000321972500014

    View details for PubMedID 23471750

  • In situ study of the impact of inter- and intra-reader variability on region of interest (ROI) analysis in preclinical molecular imaging. American journal of nuclear medicine and molecular imaging Habte, F., Budhiraja, S., Keren, S., Doyle, T. C., Levin, C. S., Paik, D. S. 2013; 3 (2): 175-181

    Abstract

    We estimated reader-dependent variability of region of interest (ROI) analysis and evaluated its impact on preclinical quantitative molecular imaging. To estimate reader variability, we used five independent image datasets acquired each using microPET and multispectral fluorescence imaging (MSFI). We also selected ten experienced researchers who utilize molecular imaging in the same environment that they typically perform their own studies. Nine investigators blinded to the data type completed the ROI analysis by drawing ROIs manually that delineate the tumor regions to the best of their knowledge and repeated the measurements three times, non-consecutively. Extracted mean intensities of voxels within each ROI are used to compute the coefficient of variation (CV) and characterize the inter- and intra-reader variability. The impact of variability was assessed through random samples iterated from normal distributions for control and experimental groups on hypothesis testing and computing statistical power by varying subject size, measured difference between groups and CV. The results indicate that inter-reader variability was 22.5% for microPET and 72.2% for MSFI. Additionally, mean intra-reader variability was 10.1% for microPET and 26.4% for MSFI. Repeated statistical testing showed that a total variability of CV < 50% may be needed to detect differences < 50% between experimental and control groups when six subjects (n = 6) or more are used and statistical power is adequate (80%). Surprisingly high variability has been observed mainly due to differences in the ROI placement and geometry drawn between readers, which may adversely affect statistical power and erroneously lead to negative study outcomes.

    View details for PubMedID 23526701

  • A Brain Tumor Molecular Imaging Strategy using a New Triple-Modality MRI-Photoacoustic-Raman Nanoparticle Conference on Photons Plus Ultrasound - Imaging and Sensing de la Zerda, A., Kircher, M. F., Jokerst, J. V., Zavaleta, C. L., Kempen, P. J., Mittra, E., Pitter, K., Huang, R., Campos, C., Habte, F., Sinclair, R., Brennan, C. W., Mellinghoff, I. K., Holland, E. C., Gambhir, S. S. SPIE-INT SOC OPTICAL ENGINEERING. 2013

    View details for DOI 10.1117/12.2001719

    View details for Web of Science ID 000322832800007

  • Remodeling of Endogenous Mammary Epithelium by Breast Cancer Stem Cells STEM CELLS Parashurama, N., Lobo, N. A., Ito, K., Mosley, A. R., Habte, F. G., Zabala, M., Smith, B. R., Lam, J., Weissman, I. L., Clarke, M. F., Gambhir, S. S. 2012; 30 (10): 2114-2127

    Abstract

    Poorly regulated tissue remodeling results in increased breast cancer risk, yet how breast cancer stem cells (CSC) participate in remodeling is unknown. We performed in vivo imaging of changes in fluorescent, endogenous duct architecture as a metric for remodeling. First, we quantitatively imaged physiologic remodeling of primary branches of the developing and regenerating mammary tree. To assess CSC-specific remodeling events, we isolated CSC from MMTV-Wnt1 (mouse mammary tumor virus long-term repeat enhancer driving Wnt1 oncogene) breast tumors, a well studied model in which tissue remodeling affects tumorigenesis. We confirm that CSC drive tumorigenesis, suggesting a link between CSC and remodeling. We find that normal, regenerating, and developing gland maintain a specific branching pattern. In contrast, transplantation of CSC results in changes in the branching patterns of endogenous ducts while non-CSC do not. Specifically, in the presence of CSC, we identified an increased number of branches, branch points, ducts which have greater than 40 branches (5/33 for CSC and 0/39 for non-CSC), and histological evidence of increased branching. Moreover, we demonstrate that only CSC implants invade into surrounding stroma with structures similar to developing mammary ducts (nine for CSC and one for non-CSC). Overall, we demonstrate a novel approach for imaging physiologic and pathological remodeling. Furthermore, we identify unique, CSC-specific, remodeling events. Our data suggest that CSC interact with the microenvironment differently than non-CSC, and that this could eventually be a therapeutic approach for targeting CSC.

    View details for DOI 10.1002/stem.1205

    View details for Web of Science ID 000308928300005

    View details for PubMedID 22899386

  • Development of a Novel Long-Lived ImmunoPET Tracer for Monitoring Lymphoma Therapy in a Humanized Transgenic Mouse Model BIOCONJUGATE CHEMISTRY Natarajan, A., Habte, F., Gambhir, S. S. 2012; 23 (6): 1221-1229

    Abstract

    Positron emission tomography (PET) is an attractive imaging tool to localize and quantify tracer biodistribution. ImmunoPET with an intact mAb typically requires two to four days to achieve optimized tumor-to-normal ratios. Thus, a positron emitter with a half-life of two to four days such as zirconium-89 [(89)Zr] (t(1/2): 78.4 h) is ideal. We have developed an antibody-based, long-lived immunoPET tracer (89)Zr-Desferrioxamine-p-SCN (Df-Bz-NCS)-rituximab (Zr-iPET) to image tumor for longer durations in a humanized CD20-expressing transgenic mouse model. To optimize the radiolabeling efficiency of (89)Zr with Df-Bz-rituximab, multiple radiolabelings were performed. Radiochemical yield, purity, immunoreactivity, and stability assays were carried out to characterize the Zr-iPET for chemical and biological integrity. This tracer was used to image transgenic mice that express the human CD20 on their B cells (huCD20TM). Each huCD20TM mouse received a 7.4 MBq/dose. One group (n = 3) received a 2 mg/kg predose (blocking) of cold rituximab 2 h prior to (89)Zr-iPET; the other group (n = 3) had no predose (nonblocking). Small animal PET/CT was used to image mice at 1, 4, 24, 48, 72, and 120 h. Quality assurance of the (89)Zr-iPET demonstrated NCS-Bz-Df: antibody ratio (c/a: 1.5 ± 0.31), specific activity (0.44-1.64 TBq/mol), radiochemical yield (>70%), and purity (>98%). The Zr-iPET immunoreactivity was >80%. At 120 h, Zr-iPET uptake (% ID/g) as mean ± STD for blocking and nonblocking groups in spleen was 3.2 ± 0.1% and 83.3 ± 2.0% (p value <0.0013.). Liver uptake was 1.32 ± 0.05% and 0.61 ± 0.001% (p value <0.0128) for blocking and nonblocking, respectively. The small animal PET/CT image shows the spleen specific uptake of Zr-iPET in mice at 120 h after tracer injection. Compared to the liver, the spleen specific uptake of Zr-iPET is very high due to the expression of huCD20. We optimized the radiolabeling efficiency of (89)Zr with Df-Bz-rituximab. These radioimmunoconjugate lots were stable up to 5 days in serum in vitro. The present study showed that (89)Zr is well-suited for mAbs to image cancer over an extended period of time (up to 5 days).

    View details for DOI 10.1021/bc300039r

    View details for Web of Science ID 000305358700015

    View details for PubMedCentralID PMC3459285

  • A brain tumor molecular imaging strategy using a new triple-modality MRI-photoacoustic-Raman nanoparticle NATURE MEDICINE Kircher, M. F., de la Zerda, A., Jokerst, J. V., Zavaleta, C. L., Kempen, P. J., Mittra, E., Pitter, K., Huang, R., Campos, C., Habte, F., Sinclair, R., Brennan, C. W., Mellinghoff, I. K., Holland, E. C., Gambhir, S. S. 2012; 18 (5): 829-U235

    Abstract

    The difficulty in delineating brain tumor margins is a major obstacle in the path toward better outcomes for patients with brain tumors. Current imaging methods are often limited by inadequate sensitivity, specificity and spatial resolution. Here we show that a unique triple-modality magnetic resonance imaging-photoacoustic imaging-Raman imaging nanoparticle (termed here MPR nanoparticle) can accurately help delineate the margins of brain tumors in living mice both preoperatively and intraoperatively. The MPRs were detected by all three modalities with at least a picomolar sensitivity both in vitro and in living mice. Intravenous injection of MPRs into glioblastoma-bearing mice led to MPR accumulation and retention by the tumors, with no MPR accumulation in the surrounding healthy tissue, allowing for a noninvasive tumor delineation using all three modalities through the intact skull. Raman imaging allowed for guidance of intraoperative tumor resection, and a histological correlation validated that Raman imaging was accurately delineating the brain tumor margins. This new triple-modality-nanoparticle approach has promise for enabling more accurate brain tumor imaging and resection.

    View details for DOI 10.1038/nm.2721

    View details for Web of Science ID 000303763500053

    View details for PubMedID 22504484

    View details for PubMedCentralID PMC3422133

  • Enhancing Pixelated Fast-Neutron Block Detector Performance Using a Slotted Light Guide Habte, F., Blackston, M. A., Hausladen, P. A., Fabris, L., IEEE IEEE. 2009: 2403-+
  • A Fieldable-Prototype, Large-Area, Gamma-Ray Imager for Orphan Source Search IEEE TRANSACTIONS ON NUCLEAR SCIENCE Ziock, K. P., Fabris, L., Carr, D., Collins, J., Cunningham, M., Habte, F., Karnowski, T., Marchant, W. 2008; 55 (6): 3643-3653
  • The Use of Gamma-Ray Imaging to Improve Portal Monitor Performance IEEE TRANSACTIONS ON NUCLEAR SCIENCE Ziock, K. P., Collins, J., Cunningham, M., Fabris, L., Gee, T., Goddard, J., Habte, F., Karnowski, T. 2008; 55 (6): 3654-3664
  • Prototype parallel readout system for position sensitive PMT based gamma ray imaging systems IEEE TRANSACTIONS ON NUCLEAR SCIENCE Habte, F., Olcott, P. D., Levin, C. S., Foudray, A. M. 2007; 54 (1): 60-65
  • Positioning annihilation photon interactions in a thin LSO crystal sheet with a position-sensitive avalanche photodiode IEEE TRANSACTIONS ON NUCLEAR SCIENCE Foudray, A. M., Habte, F., Levin, C. S., Olcott, P. D. 2006; 53 (5): 2549-2556
  • Count rate studies of a box-shaped PET breast imaging system comprised of position sensitive avalanche photodiodes utilizing Monte Carlo simulation Workshop on Nuclear Radiology of Breast Cancer Foudray, A. M., Habte, F., Chinn, G., Zhang, J., Levin, C. S. IST EDITORIALI POLGRAFICI INT. 2006: 64–67

    Abstract

    We are investigating a high-sensitivity, high-resolution positron emission tomography (PET) system for clinical use in the detection, diagnosis and staging of breast cancer. Using conventional figures of merit, design parameters were evaluated for count rate performance, module dead time, and construction complexity. The detector system modeled comprises extremely thin position-sensitive avalanche photodiodes coupled to lutetium oxy-orthosilicate scintillation crystals. Previous investigations of detector geometries with Monte Carlo indicated that one of the largest impacts on sensitivity is local scintillation crystal density when considering systems having the same average scintillation crystal densities (same crystal packing fraction and system solid-angle coverage). Our results show the system has very good scatter and randoms rejection at clinical activity ranges ( approximately 200 muCi).

    View details for Web of Science ID 000245817500018

    View details for PubMedID 17645997

  • Fully 3-D List-Mode OSEM Accelerated by Graphics Processing Units 15th International Workshop on Room-Temperature Semiconductor X- and Gamma-Ray Detectors/ 2006 IEEE Nuclear Science Symposium Pratx, G., Chinn, G., Habte, F., Olcott, P., Levin, C. IEEE. 2006: 2196–2202
  • Impact of high energy resolution detectors on the performance of a PET system dedicated to breast cancer imaging Workshop on Nuclear Radiology of Breast Cancer Levin, C. S., Foudray, A. M., Habte, F. IST EDITORIALI POLGRAFICI INT. 2006: 28–34

    Abstract

    We are developing a high resolution, high sensitivity PET camera dedicated to breast cancer imaging. We are studying two novel detector technologies for this imaging system: a scintillation detector comprising layers of small lutetium oxyorthosilicate (LSO) crystals coupled to new position sensitive avalanche photodiodes (PSAPDs), and a pure semiconductor detector comprising cadmium zinc telluride (CZT) crystal slabs with thin anode and cathode strips deposited in orthogonal directions on either side of each slab. Both detectors achieve 1 mm spatial resolution with 3-5 mm directly measured photon interaction depth resolution, which promotes uniform reconstructed spatial resolution throughout a compact, breast-size field of view. Both detector types also achieve outstanding energy resolution (<3% and <12%, respectively for LSO-PSAPD and CZT at 511 keV). This paper studies the effects that this excellent energy resolution has on the expected system performance. Results indicate the importance that high energy resolution and narrow energy window settings have in reducing background random as well as scatter coincidences without compromising statistical quality of the dedicated breast PET data. Simulations predict that using either detector type the excellent performance and novel arrangement of these detectors proposed for the system facilitate approximately 20% instrument sensitivity at the system center and a peak noise-equivalent count rate of >4 kcps for 200 microCi in a simulated breast phantom.

    View details for Web of Science ID 000245817500011

    View details for PubMedID 17645990

  • Evaluation of a dual-panel PET camera design to breast cancer imaging Workshop on Nuclear Radiology of Breast Cancer Zhang, J., Chinn, G., Foudray, A. M., Habte, F., Olcott, P., Levin, C. S. IST EDITORIALI POLGRAFICI INT. 2006: 94–98

    Abstract

    We are developing a novel, portable dual-panel positron emission tomography (PET) camera dedicated to breast cancer imaging. With a sensitive area of approximately 150 cm(2), this camera is based on arrays of lutetium oxyorthosilicate (LSO) crystals (1x1x3 mm(3)) coupled to 11x11-mm(2) position-sensitive avalanche photodiodes (PSAPD). GATE open source software was used to perform Monte Carlo simulations to optimize the parameters for the camera design. The noise equivalent counting (NEC) rate, together with the true, scatter, and random counting rates were simulated at different time and energy windows. Focal plane tomography (FPT) was used for visualizing the tumors at different depths between the two detector panels. Attenuation and uniformity corrections were applied to images.

    View details for Web of Science ID 000245817500026

    View details for PubMedID 17646005

  • Investigation of scintillation light multiplexing for PET detectors based on position sensitive avalanche photodiodes Nuclear Science Symposium/Medical Imaging Conference Habte, F., Olcott, P. D., Levin, C. S., Foudray, A. M. IEEE. 2005: 2027–2030