Honors & Awards


  • Alavi-Mandell Award, Society of Nuclear Medicine and Molecular Imaging (2020)
  • Stanford Cancer Institute Fellowship, Stanford Cancer Institute (2020)
  • Brain Imaging Council Young Investigator Award, Society of Nuclear Medicine and Molecular Imaging (2019)
  • The R J W Le Fèvre Research Travelling Scholarship, The School of Chemistry, University of Sydney (2013)
  • The John A Lamberton Scholarship, The University of Sydney (2010-2014)
  • The University Postgraduate Award Scholarship, The University of Sydney (2010-2014)
  • SIRO Top-up Scholarship, CSIRO Materials Science and Engineering, Ian Wark Laboratory (2010-2013)
  • Agnes Campbell Postgraduate Prizes, The University of Sydney (2009, 2010, 2011, 2012, 2013, 2014)

Boards, Advisory Committees, Professional Organizations


  • Member, Society of Neuro-Oncology (2017 - Present)
  • Member, Society of Nuclear Medicine and Molecular Imaging (2017 - Present)
  • Member, World Molecular Imaging Society (2015 - Present)
  • Member, Royal Australian Chemical Society (2009 - Present)

Professional Education


  • PhD, The University of Sydney, Medicinal Chemistry (2014)
  • BSc (Hons), The University of Sydney, Organic Chemistry and Pharmacology (2009)

Current Research and Scholarly Interests


The focus of my research is to develop novel imaging and treatment strategies to detect and better manage cancer. This approach relies first on the identification and validation of molecular targets and biomarkers that are linked with underlying the underlying biology driving the initiation and progression of cancers. We then develop novel small molecule based radiotracers to monitor fundamental molecular and cellular processes occurring in living subjects using positron emission tomography (PET) with the goal of improving cancer diagnosis and management. We additionally develop novel peptide based theragnostic agents for stratification of patients with high receptor expression, treatment with targeted radionuclide therapy, and subsequent monitoring of treatment response. Our overall goal is to develop multiple clinically translatable strategies to improve cancer diagnosis, management, and outcomes.

Clinical Trials


  • [18F]DASA-23 and PET Scan in Evaluating Pyruvate Kinase M2 Expression in Patients With Intracranial Tumors or Recurrent Glioblastoma and Healthy Volunteers Not Recruiting

    This phase I trial studies how well [18F]DASA-23 and positron emission tomography (PET) scan work in evaluating pyruvate kinase M2 (PKM2) expression in patients with intracranial tumors or recurrent glioblastoma and healthy volunteers. PKM2 regulates brain tumor metabolism, a key factor in glioblastoma growth. [18F]DASA-23 is a radioactive substance with the ability to monitor PKM2 activity. A PET scan is a procedure in which a small amount of a radioactive substance, such as [18F]DASA-23, is injected into a vein, and a scanner is used to make detailed, computerized pictures of areas inside the body where the substance is used. Tumor cells usually pick up more of these radioactive substances, allowing them to be found. Giving [18F]DASA-23 with a PET scan may help doctors evaluate PKM2 expression in healthy volunteers and in participants with intracranial tumors or recurrent glioblastoma.

    Stanford is currently not accepting patients for this trial. For more information, please contact Mark M. Santos, 650-498-5189.

    View full details

All Publications


  • Carbon-11 Labelling of Benzenesulfonamide Analogues as Novel Radiotracers to Visualize the Pore-forming Activity of Perforin Zhang, B., Pandrala, M., Shen, B., Beinat, C. ELSEVIER SCIENCE INC. 2023: S221
  • [<SUP>18</SUP>F]hGTS13 for imaging of x<sub>C</sub><SUP>-</SUP> transporter activity in high-grade glioma Kendrili, M., Acosta, C., Recht, L., Beinat, C. ELSEVIER SCIENCE INC. 2023: S163-S164
  • Synthesis and preclinical evaluation of [<SUP>18</SUP>F]DASA-10 a second generation PKM2 specific radiotracer Beinat, C., Malek, R., Kendrili, M., Acosta, C., Habte, F., Recht, L. ELSEVIER SCIENCE INC. 2023: S35
  • PET imaging of focused-ultrasound enhanced delivery of AAVs into the murine brain. Theranostics Ajenjo, J., Seo, J. W., Foiret, J., Wu, B., Raie, M. N., Wang, J., Fite, B. Z., Zhang, N., Malek, R., Beinat, C., Malik, N., Anders, D. A., Ferrara, K. W. 2023; 13 (15): 5151-5169

    Abstract

    Rationale: Despite recent advances in the use of adeno-associated viruses (AAVs) as potential vehicles for genetic intervention of central and peripheral nervous system-associated disorders, gene therapy for the treatment of neuropathology in adults has not been approved to date. The currently FDA-approved AAV-vector based gene therapies rely on naturally occurring serotypes, such as AAV2 or AAV9, which display limited or no transport across the blood-brain barrier (BBB) if systemically administered. Recently developed engineered AAV variants have shown broad brain transduction and reduced off-target liver toxicity in non-human primates (NHPs). However, these vectors lack spatial selectivity for targeted gene delivery, a potentially critical limitation for delivering therapeutic doses in defined areas of the brain. The use of microbubbles, in conjunction with focused ultrasound (FUS), can enhance regional brain AAV transduction, but methods to assess transduction in vivo are needed. Methods: In a murine model, we combined positron emission tomography (PET) and optical imaging of reporter gene payloads to non-invasively assess the spatial distribution and transduction efficiency of systemically administered AAV9 after FUS and microbubble treatment. Capsid and reporter probe accumulation are reported as percent injected dose per cubic centimeter (%ID/cc) for in vivo PET quantification, whereas results for ex vivo assays are reported as percent injected dose per gram (%ID/g). Results: In a study spanning accumulation and transduction, mean AAV9 accumulation within the brain was 0.29 %ID/cc without FUS, whereas in the insonified region of interest of FUS-treated mice, the spatial mean and maximum reached ~2.3 %ID/cc and 4.3 %ID/cc, respectively. Transgene expression assessed in vivo by PET reporter gene imaging employing the pyruvate kinase M2 (PKM2)/[18F]DASA-10 reporter system increased up to 10-fold in the FUS-treated regions, as compared to mice receiving AAVs without FUS. Systemic injection of AAV9 packaging the EF1A-PKM2 transgene followed by FUS in one hemisphere resulted in 1) an average 102-fold increase in PKM2 mRNA concentration compared to mice treated with AAVs only and 2) a 12.5-fold increase in the insonified compared to the contralateral hemisphere of FUS-treated mice. Conclusion: Combining microbubbles with US-guided treatment facilitated a multi-hour BBB disruption and stable AAV transduction in targeted areas of the murine brain. This unique platform has the potential to provide insight and aid in the translation of AAV-based therapies for the treatment of neuropathologies.

    View details for DOI 10.7150/thno.85549

    View details for PubMedID 37908737

    View details for PubMedCentralID PMC10614693

  • Development of [18F]DASA-10 for enhanced imaging of pyruvate kinase M2. Nuclear medicine and biology Kendirli, M. T., Malek, R., Silveira, M. B., Acosta, C., Zhang, S., Azevedo, C., Nagy, S. C., Habte, F., James, M. L., Recht, L. D., Beinat, C. 2023; 124-125: 108382

    Abstract

    The aim of this study was to develop a positron emission tomography (PET) radiotracer for measuring pyruvate kinase M2 (PKM2) with improved physicochemical and pharmacokinetic properties compared to [18F]DASA-23.First, we synthesized [18F]DASA-10 and tested its uptake and retention compared to [18F]DASA-23 in human and mouse glioma cell lines. We then confirmed the specificity of [18F]DASA-10 by transiently modulating the expression of PKM2 in DU145 and HeLa cells. Next, we determined [18F]DASA-10 pharmacokinetics in healthy nude mice using PET imaging and subsequently assessed the ability of [18F]DASA-10 versus [18F]DASA-23 to enable in vivo detection of intracranial gliomas in syngeneic C6 rat models of glioma.[18F]DASA-10 demonstrated excellent cellular uptake and retention with values significantly higher than [18F]DASA-23 in all cell lines and timepoints investigated. [18F]DASA-10 showed a 73 % and 65 % reduced uptake respectively in DU145 and HeLa cells treated with PKM2 siRNA as compared to control siRNA treated cells. [18F]DASA-10 showed favorable biodistribution and pharmacokinetic properties and a significantly improved tumor-to-brain ratio in rat C6 glioma models relative to [18F]DASA-23 (3.2 ± 0.8 versus 1.6 ± 0.3, p = 0.01).[18F]DASA-10 is a new PET radiotracer for molecular imaging of PKM2 with potential to overcome the prior limitations observed with [18F]DASA-23. [18F]DASA-10 shows promise for clinical translation to enable imaging of brain malignancies owing to its low background signal in the healthy brain.

    View details for DOI 10.1016/j.nucmedbio.2023.108382

    View details for PubMedID 37634399

  • Clinical Radiosynthesis and Translation of [18F]OP-801: A Novel Radiotracer for Imaging Reactive Microglia and Macrophages. ACS chemical neuroscience Jackson, I. M., Carlson, M. L., Beinat, C., Malik, N., Kalita, M., Reyes, S., Azevedo, E. C., Nagy, S. C., Alam, I. S., Sharma, R., La Rosa, S. A., Moradi, F., Cleland, J., Shen, B., James, M. L. 2023

    Abstract

    Positron emission tomography (PET) is a powerful tool for studying neuroinflammatory diseases; however, current PET biomarkers of neuroinflammation possess significant limitations. We recently reported a promising dendrimer PET tracer ([18F]OP-801), which is selectively taken up by reactive microglia and macrophages. Here, we describe further important characterization of [18F]OP-801 in addition to optimization and validation of a two-step clinical radiosynthesis. [18F]OP-801 was found to be stable in human plasma for 90 min post incubation, and human dose estimates were calculated for 24 organs of interest; kidneys and urinary bladder wall without bladder voiding were identified as receiving the highest absorbed dose. Following optimization detailed herein, automated radiosynthesis and quality control (QC) analyses of [18F]OP-801 were performed in triplicate in suitable radiochemical yield (6.89 ± 2.23% decay corrected), specific activity (37.49 ± 15.49 GBq/mg), and radiochemical purity for clinical imaging. Importantly, imaging mice with tracer (prepared using optimized methods) 24 h following the intraperitoneal injection of liposaccharide resulted in the robust brain PET signal. Cumulatively, these data enable clinical translation of [18F]OP-801 for imaging reactive microglia and macrophages in humans. Data from three validation runs of the clinical manufacturing and QC were submitted to the Food and Drug Administration (FDA) as part of a Drug Master File (DMF). Subsequent FDA approval to proceed was obtained, and a phase 1/2 clinical trial (NCT05395624) for first-in-human imaging in healthy controls and patients with amyotrophic lateral sclerosis is underway.

    View details for DOI 10.1021/acschemneuro.3c00028

    View details for PubMedID 37310119

  • 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

  • Radiosynthesis and initial preclinical evaluation of [11C]AZD1283 as a potential P2Y12R PET radiotracer. Nuclear medicine and biology Jackson, I. M., Buccino, P. J., Azevedo, E. C., Carlson, M. L., Luo, A. S., Deal, E. M., Kalita, M., Reyes, S. T., Shao, X., Beinat, C., Nagy, S. C., Chaney, A. M., Anders, D. A., Scott, P. J., Smith, M., Shen, B., James, M. L. 2022

    Abstract

    INTRO: Chronic neuroinflammation and microglial dysfunction are key features of many neurological diseases, including Alzheimer's Disease and multiple sclerosis. While there is unfortunately a dearth of highly selective molecular imaging biomarkers/probes for studying microglia in vivo, P2Y12R has emerged as an attractive candidate PET biomarker being explored for this purpose. Importantly, P2Y12R is selectively expressed on microglia in the CNS and undergoes dynamic changes in expression according to inflammatory context (e.g., toxic versus beneficial/healing states), thus having the potential to reveal functional information about microglia in living subjects. Herein, we identified a high affinity, small molecule P2Y12R antagonist (AZD1283) to radiolabel and assess as a candidate radiotracer through in vitro assays and in vivo positron emission tomography (PET) imaging of both wild-type and total knockout mice and a non-human primate.METHODS: First, we evaluated the metabolic stability and passive permeability of non-radioactive AZD1283 in vitro. Next, we radiolabeled [11C]AZD1283 with radioactive precursor [11C]NH4CN and determined stability in formulation and human plasma. Finally, we investigated the in vivo stability and kinetics of [11C]AZD1283 via dynamic PET imaging of naive wild-type mice, P2Y12R knockout mouse, and a rhesus macaque.RESULTS: We determined the half-life of AZD1283 in mouse and human liver microsomes to be 37 and>160min, respectively, and predicted passive CNS uptake with a small amount of active efflux, using a Caco-2 assay. Our radiolabeling efforts afforded [11C]AZD1283 in an activity of 12.69±10.64mCi with high chemical and radiochemical purity (>99%) and molar activity of 1142.84±504.73mCi/mumol (average of n=3). Of note, we found [11C]AZD1283 to be highly stable in vitro, with >99% intact tracer present after 90min of incubation in formulation and 60min of incubation in human serum. PET imaging revealed negligible brain signal in healthy wild-type mice (n=3) and a P2Y12 knockout mouse (0.55±0.37%ID/g at 5min post injection). Strikingly, high signal was detected in the liver of all mice within the first 20min of administration (peak uptake=58.28±18.75%ID/g at 5min post injection) and persisted for the remaining duration of the scan. Ex vivo gamma counting of mouse tissues at 60min post-injection mirrored in vivo data with a mean %ID/g of 0.9%±0.40, 0.02%±0.01, and 106±29.70% in the blood, brain, and liver, respectively (n=4). High performance liquid chromatography (HPLC) analysis of murine blood and liver metabolite samples revealed a single radioactive peak (relative area under peak: 100%), representing intact tracer. Finally, PET imaging of a rhesus macaque also revealed negligible CNS uptake/binding in monkey brain (peak uptake=0.37 Standard Uptake Values (SUV)).CONCLUSION: Despite our initial encouraging liver microsome and Caco-2 monolayer data, in addition to the observed high stability of [11C]AZD1283 in formulation and human serum, in vivo brain uptake was negligible and rapid accumulation was observed in the liver of both naive wildtype and P2Y12R knockout mice. Liver signal appeared to be independent of both metabolism and P2Y12R expression due to the confirmation of intact tracer in this tissue for both wildtype and P2Y12R knockout mice. In Rhesus Macaque, negligible uptake of [11C]AZD1283 brain indicates a lack of potential for translation or its further investigation in vivo. P2Y12R is an extremely promising potential PET biomarker, and the data presented here suggests encouraging metabolic stability for this scaffold; however, the mechanism of liver uptake in mice should be elucidated prior to further analogue development.

    View details for DOI 10.1016/j.nucmedbio.2022.05.001

    View details for PubMedID 35680502

  • A Clinical PET Imaging Tracer ([18F]DASA-23) to Monitor Pyruvate Kinase M2 Induced Glycolytic Reprogramming in Glioblastoma. Clinical cancer research : an official journal of the American Association for Cancer Research Beinat, C., Patel, C. B., Haywood, T., Murty, S., Naya, L., Castillo, J. B., Reyes, S. T., Phillips, M., Buccino, P., Shen, B., Park, J. H., Koran, M. E., Alam, I. S., James, M. L., Holley, D., Halbert, K., Gandhi, H., He, J. Q., Granucci, M., Johnson, E., Liu, D. D., Uchida, N., Sinha, R., Chu, P., Born, D. E., Warnock, G. I., Weissman, I., Hayden Gephart, M., Khalighi, M. M., Massoud, T. F., Iagaru, A., Davidzon, G., Thomas, R., Nagpal, S., Recht, L. D., Gambhir, S. S. 2021

    Abstract

    PURPOSE: Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, a key process of cancer metabolism. PKM2 is preferentially expressed by glioblastoma (GBM) cells with minimal expression in healthy brain. We describe the development, validation, and translation of a novel positron emission tomography (PET) tracer to study PKM2 in GBM. We evaluated 1-((2-fluoro-6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in cell culture, mouse models of GBM, healthy human volunteers, and GBM patients.EXPERIMENTAL DESIGN: [18F]DASA-23 was synthesized with a molar activity of 100.47 {plus minus} 29.58 GBq/mol and radiochemical purity >95%. We performed initial testing of [18F]DASA-23 in GBM cell culture and human GBM xenografts implanted orthotopically into mice. Next we produced [18F]DASA-23 under FDA oversight, and evaluated it in healthy volunteers, and a pilot cohort of glioma patients.RESULTS: In mouse imaging studies, [18F]DASA-23 clearly delineated the U87 GBM from surrounding healthy brain tissue and had a tumor-to-brain ratio (TBR) of 3.6 {plus minus} 0.5. In human volunteers, [18F]DASA-23 crossed the intact blood-brain barrier and was rapidly cleared. In GBM patients, [18F]DASA-23 successfully outlined tumors visible on contrast-enhanced magnetic resonance imaging (MRI). The uptake of [18F]DASA-23 was markedly elevated in GBMs compared to normal brain, and it identified a metabolic non-responder within 1-week of treatment initiation.CONCLUSIONS: We developed and translated [18F]DASA-23 as a new tracer that demonstrated the visualization of aberrantly expressed PKM2 for the first time in human subjects. These results warrant further clinical evaluation of [18F]DASA-23 to assess its utility for imaging therapy-induced normalization of aberrant cancer metabolism.

    View details for DOI 10.1158/1078-0432.CCR-21-0544

    View details for PubMedID 34475101

  • Initial Clinical Evaluation of [F-18]DASA-23, a PET Imaging Tracer for Evaluation of Aberrantly Expressed Pyruvate Kinase M2 in Glioblastoma Beinat, C., Patel, C., Haywood, T., Naya, L., Castillo, J., Shen, B., Massoud, T., Iagaru, A., Davidzon, G., Recht, L., Gambhir, S. SOC NUCLEAR MEDICINE INC. 2021
  • Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [18F]DASA-23, a non-invasive probe of pyruvate kinase M2 (PKM2) expression Neoplasia Patel, C. B., Beinat, C., Xie, Y., Chang, E., Gambhir, S. S. 2021; 23 (1): 58-67
  • Minicircles for a two-step blood biomarker and PET imaging early cancer detection strategy. Journal of controlled release : official journal of the Controlled Release Society Robinson, E. R., Gowrishankar, G., D'Souza, A., Kheirolomoom, A., Haywood, T., Hori, S. S., Chuang, H. Y., Zeng, Y., Tumbale, S., Aalipour, A., Beinat, C., Alam, I. S., Sathirachinda, A., Kanada, M., Paulmurugan, R., Ferrara, K. W., Gambhir, S. S. 2021

    Abstract

    Early cancer detection can dramatically increase treatment options and survival rates for patients, yet detection of early-stage tumors remains difficult. Here, we demonstrate a two-step strategy to detect and locate cancerous lesions by delivering tumor-activatable minicircle (MC) plasmids encoding a combination of blood-based and imaging reporter genes to tumor cells. We genetically engineered the MCs, under the control of the pan-tumor-specific Survivin promoter, to encode: 1) Gaussia Luciferase (GLuc), a secreted biomarker that can be easily assayed in blood samples; and 2) Herpes Simplex Virus Type 1 Thymidine Kinase mutant (HSV-1 sr39TK), a PET reporter gene that can be used for highly sensitive and quantitative imaging of the tumor location. We evaluated two methods of MC delivery, complexing the MCs with the chemical transfection agent jetPEI or encapsulating the MCs in extracellular vesicles (EVs) derived from a human cervical cancer HeLa cell line. MCs delivered by EVs or jetPEI yielded significant expression of the reporter genes in cell culture versus MCs delivered without a transfection agent. Secreted GLuc correlated with HSV-1 sr39TK expression with R2 = 0.9676. MC complexation with jetPEI delivered a larger mass of MC for enhanced transfection, which was crucial for in vivo animal studies, where delivery of MCs via jetPEI resulted in GLuc and HSV-1 sr39TK expression at significantly higher levels than controls. To the best of our knowledge, this is the first report of the PET reporter gene HSV-1 sr39TK delivered via a tumor-activatable MC to tumor cells for an early cancer detection strategy. This work explores solutions to endogenous blood-based biomarker and molecular imaging limitations of early cancer detection strategies and elucidates the delivery capabilities and limitations of EVs.

    View details for DOI 10.1016/j.jconrel.2021.05.026

    View details for PubMedID 34029631

  • Ultra-high-frequency radio-frequency acoustic molecular imaging with saline nanodroplets in living subjects. Nature nanotechnology Chen, Y. S., Zhao, Y. n., Beinat, C. n., Zlitni, A. n., Hsu, E. C., Chen, D. H., Achterberg, F. n., Wang, H. n., Stoyanova, T. n., Dionne, J. n., Gambhir, S. S. 2021

    Abstract

    Molecular imaging is a crucial technique in clinical diagnostics but it relies on radioactive tracers or strong magnetic fields that are unsuitable for many patients, particularly infants and pregnant women. Ultra-high-frequency radio-frequency acoustic (UHF-RF-acoustic) imaging using non-ionizing RF pulses allows deep-tissue imaging with sub-millimetre spatial resolution. However, lack of biocompatible and targetable contrast agents has prevented the successful in vivo application of UHF-RF-acoustic imaging. Here we report our development of targetable nanodroplets for UHF-RF-acoustic molecular imaging of cancers. We synthesize all-liquid nanodroplets containing hypertonic saline that are stable for at least 2 weeks and can produce high-intensity UHF-RF-acoustic signals. Compared with concentration-matched iron oxide nanoparticles, our nanodroplets produce at least 1,600 times higher UHF-RF-acoustic signals at the same imaging depth. We demonstrate in vivo imaging using the targeted nanodroplets in a prostate cancer xenograft mouse model expressing gastrin release protein receptor (GRPR), and show that targeting specificity is increased by more than 2-fold compared with untargeted nanodroplets or prostate cancer cells not expressing this receptor.

    View details for DOI 10.1038/s41565-021-00869-5

    View details for PubMedID 33782588

  • PET reporter gene imaging and ganciclovir-mediated ablation of chimeric antigen receptor T-cells in solid tumors. Cancer research Murty, S., Labanieh, L., Murty, T., Gowrishankar, G., Haywood, T., Alam, I. S., Beinat, C., Robinson, E., Aalipour, A., Klysz, D. D., Cochran, J. R., Majzner, R. G., Mackall, C. L., Gambhir, S. S. 2020

    Abstract

    Imaging strategies to monitor chimeric antigen receptor (CAR) T-cell biodistribution and proliferation harbor the potential to facilitate clinical translation for the treatment of both liquid and solid tumors. Additionally, the potential adverse effects of CAR T-cells highlight the need for mechanisms to modulate CAR T-cell activity. The herpes simplex virus type 1 thymidine kinase (HSV1-tk) gene has previously been translated as a positron emission tomography (PET) reporter gene for imaging of T-cell trafficking in brain tumor patients. The HSV1-TK enzyme can act as a suicide gene of transduced cells through treatment with the prodrug ganciclovir (GCV). Here we report the molecular engineering, imaging, and GCV-mediated destruction of B7H3 CAR T-cells incorporating a mutated version of the HSV1-tk gene (sr39tk) with improved enzymatic activity for GCV. The sr39tk gene did not affect B7H3 CAR T-cell functionality and in vitro and in vivo studies in osteosarcoma models showed no significant effect on B7H3 CAR T-cell antitumor activity. PET/CT imaging with 9-(4-[18F]-fluoro-3-[hydroxymethyl]butyl)guanine [18F]FHBG of B7H3-sr39tk CAR T-cells in an orthotopic model of osteosarcoma revealed tumor homing and systemic immune expansion. Bioluminescence and PET imaging of B7H3-sr39tk CAR T-cells confirmed complete tumor ablation with intraperitoneal GCV administration. This imaging and suicide ablation system can provide insight into CAR T-cell migration and proliferation during clinical trials while serving as a suicide switch to limit potential toxicities.

    View details for DOI 10.1158/0008-5472.CAN-19-3579

    View details for PubMedID 32958548

  • Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [F-18]DASA-23 MOLECULAR IMAGING AND BIOLOGY Beinat, C., Patel, C. B., Xie, Y., Gambhir, S. S. 2020; 22 (1): 124–33
  • Human biodistribution and radiation dosimetry of [18F]DASA-23, a PET probe targeting pyruvate kinase M2. European journal of nuclear medicine and molecular imaging Beinat, C. n., Patel, C. B., Haywood, T. n., Shen, B. n., Naya, L. n., Gandhi, H. n., Holley, D. n., Khalighi, M. n., Iagaru, A. n., Davidzon, G. n., Gambhir, S. S. 2020

    Abstract

    To assess the safety, biodistribution, and radiation dosimetry of the novel positron emission tomography (PET) radiopharmaceutical 1-((2-fluoro-6-[[18F]]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA-23) in healthy volunteers.We recruited 5 healthy volunteers who provided a written informed consent. Volunteers were injected with 295.0 ± 8.2 MBq of [18F]DASA-23 intravenously. Immediately following injection, a dynamic scan of the brain was acquired for 15 min. This was followed by serial whole-body PET/MRI scans acquired up to 3 h post-injection. Blood samples were collected at regular intervals, and vital signs monitored pre- and post-radiotracer administration. Regions of interest were drawn around multiple organs, time-activity curves were calculated, and organ uptake and dosimetry were estimated with OLINDA/EXM (version 1.1) software.All subjects tolerated the PET/MRI examination, without adverse reactions to [18F]DASA-23. [18F]DASA-23 passively crossed the blood-brain barrier, followed by rapid clearance from the brain. High accumulation of [18F]DASA-23 was noted in organs such as the gallbladder, liver, small intestine, and urinary bladder, suggesting hepatobiliary and urinary clearance. The effective dose of [18F]DASA-23 was 23.5 ± 5.8 μSv/MBq.We successfully completed a pilot first-in-human study of [18F]DASA-23. Our results indicate that [18F]DASA-23 can be used safely in humans to evaluate pyruvate kinase M2 levels. Ongoing studies are evaluating the ability of [18F]DASA-23 to visualize intracranial malignancies, NCT03539731.ClinicalTrials.gov, NCT03539731 (registered 28 May 2018).

    View details for DOI 10.1007/s00259-020-04687-0

    View details for PubMedID 31938892

  • Intravital imaging reveals synergistic effect of CAR T-cells and radiation therapy in a preclinical immunocompetent glioblastoma model Oncoimmunology Murty, S., Haile, S. T., Beinat, C., Aalipour, A., Alam, I. S., Murty, T., Shaffer, T. M., Patel, C. B., Graves, E. E., Mackall, C. L., Gambhir, S. S. 2020; 9 (1)
  • EVALUATION OF [18F]DASA-23 FOR NON-INVASIVE MEASUREMENT OF ABERRANTLY EXPRESSED PYRUVATE KINASE M2 IN GLIOMA: FIRST-IN-HUMAN STUDY Patel, C., Beinat, C., Haywood, T., Murty, S., Xie, Y., Recht, L., Nagpal, S., Thomas, R., Khalighi, M., Gandhi, H., Holley, D., Gambhir, S. OXFORD UNIV PRESS INC. 2019: 169
  • TUMOR TREATING FIELDS LEADS TO CHANGES IN MEMBRANE PERMEABILITY AND INCREASED PENETRATION BY ANTI-GLIOMA DRUGS Chang, E., Patel, C., Beinat, C., Young, C., Flores, T., Zeng, Y., Joubert, L., Arami, H., Natarajan, A., Sinclair, R., Gambhir, S. OXFORD UNIV PRESS INC. 2019: 93
  • Evaluation of [18F]DASA-23 for non-invasive measurement of aberrantly expressed pyruvate kinase M2 in glioblastoma: preclinical and first in human studies Beinat, C., Patel, C., Haywood, T., Murty, S., Alam, I., Xie, Y., Gandhi, H., Holley, D., Gambhir, S. SOC NUCLEAR MEDICINE INC. 2019
  • Engineered immune cells as highly sensitive cancer diagnostics NATURE BIOTECHNOLOGY Aalipour, A., Chuang, H., Murty, S., D'Souza, A. L., Park, S., Gulati, G. S., Patel, C. B., Beinat, C., Simonetta, F., Martinic, I., Gowrishankar, G., Robinson, E. R., Aalipour, E., Zhian, Z., Gambhir, S. S. 2019; 37 (5): 531-+
  • Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [18F]DASA-23. Molecular imaging and biology : MIB : the official publication of the Academy of Molecular Imaging Beinat, C., Patel, C. B., Xie, Y., Gambhir, S. S. 2019

    Abstract

    PURPOSE: Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, the key process of tumor metabolism. PKM2 is found in high levels in glioblastoma (GBM) cells with marginal expression within healthy brain tissue, rendering it a key biomarker of GBM metabolic re-programming. Our group has reported the development of a novel radiotracer, 1-((2-fluoro- 6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA- 23), to non-invasively detect PKM2 levels with positron emission tomography (PET).PROCEDURE: U87 human GBM cells were treated with the IC50 concentration of various agents used in the treatment of GBM, including alkylating agents (temozolomide, carmustine, lomustine, procarbazine), inhibitor of topoisomerase I (irinotecan), vascular endothelial and epidermal growth factor receptor inhibitors (cediranib and erlotinib, respectively) anti-metabolite (5-fluorouracil), microtubule inhibitor (vincristine), and metabolic agents (dichloroacetate and IDH1 inhibitor ivosidenib). Following drug exposure for three or 6days (n=6 replicates per condition), the radiotracer uptake of [18F]DASA-23 and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) was assessed. Changes in PKM2 protein levels were determined via Western blot and correlated to radiotracer uptake.RESULTS: Significant interactions were found between the treatment agent (n=12 conditions total comprised 11 drugs and vehicle) and the duration of treatment (3- or 6-day exposure to each drug) on the cellular uptake of [18F]DASA-23 (p=0.0001). The greatest change in the cellular uptake of [18F]DASA-23 was found after exposure to alkylating agents (p<0. 0001) followed by irinotecan (p=0. 0012), erlotinib (p=0. 02), and 5-fluorouracil (p=0. 005). Correlation of PKM2 protein levels and [18F]DASA-23 cellular uptake revealed a moderate correlation (r=0.44, p=0.15).CONCLUSIONS: These proof of principle studies emphasize the superiority of [18F]DASA-23 to [18F]FDG in detecting the glycolytic response of GBM to multiple classes of anti-neoplastic drugs in cell culture. A clinical trial evaluating the diagnostic utility of [18F]DASA-23 PET in GBM patients (NCT03539731) is ongoing.

    View details for PubMedID 30989436

  • The characterization of 18F-hGTS13 for molecular imaging of xC- transporter activity with positron emission tomography. Journal of nuclear medicine : official publication, Society of Nuclear Medicine Beinat, C. n., Gowrishankar, G. n., Shen, B. n., Alam, I. S., Robinson, E. n., Haywood, T. n., Patel, C. B., Azevedo, E. C., Castillo, J. n., Ilovich, O. n., Koglin, N. n., Schmitt-Willich, H. n., Berndt, M. n., Mueller, A. n., Zerna, M. n., Srinivasan, A. n., Gambhir, S. S. 2019

    Abstract

    Purpose: The aim of this study was development of an improved positron emission tomography (PET) radiotracer for measuring xC- activity with increased tumor uptake and reduced uptake in inflammatory cells compared to (S)-4-(3-18F-Fluoropropyl)-L-glutamic acid (18F-FSPG). Experimental design: A racemic glutamate derivative, 18F-hGTS13 was evaluated in cell culture and animal tumor models. 18F-hGTS13 was separated into C5-epimers and the corresponding 18F-hGTS13-isomer1 and 18F-hGTS13-isomer2 evaluated in H460 tumor bearing rats. Preliminary studies investigate the cellular uptake of 18F-hGTS13-isomer2 in multiple immune cell populations and states. Results:18F-hGTS13 demonstrated excellent H460 tumor visualization with high tumor-to-background ratios, confirmed by ex vivo biodistribution studies. Tumor associated radioactivity of 18F-hGTS13 (7.5±0.9%ID/g, n = 3) was significantly higher than with 18F-FSPG (4.6±0.7%ID/g, n = 3, P = 0.01). 18F-hGTS13-isomer2 exhibited excellent H460 tumor visualization (6.3±1.1%ID/g, n-3), and significantly reduced uptake in multiple immune cell populations relative to 18F-FSPG. 18F-hGTS13-isomer2 exhibited increased liver uptake relative to 18F-FSPG (4.6±0.8%ID/g vs. 0.7±0.01%ID/g) limiting its application in hepatocellular carcinoma. Conclusion:18F-hGTS13-isomer2 is a new PET radiotracer for molecular imaging of xC- activity which may provide information regarding tumor oxidation states. 18F-hGTS13-isomer2 has potential for clinical translation for imaging cancers of the thorax due to the low background signal in healthy tissue.

    View details for DOI 10.2967/jnumed.119.225870

    View details for PubMedID 31171595

  • Engineered immune cells as highly sensitive cancer diagnostics. Nature biotechnology Aalipour, A. n., Chuang, H. Y., Murty, S. n., D'Souza, A. L., Park, S. M., Gulati, G. S., Patel, C. B., Beinat, C. n., Simonetta, F. n., Martinić, I. n., Gowrishankar, G. n., Robinson, E. R., Aalipour, E. n., Zhian, Z. n., Gambhir, S. S. 2019

    Abstract

    Endogenous biomarkers remain at the forefront of early disease detection efforts, but many lack the sensitivities and specificities necessary to influence disease management. Here, we describe a cell-based in vivo sensor for highly sensitive early cancer detection. We engineer macrophages to produce a synthetic reporter on adopting an M2 tumor-associated metabolic profile by coupling luciferase expression to activation of the arginase-1 promoter. After adoptive transfer in colorectal and breast mouse tumor models, the engineered macrophages migrated to the tumors and activated arginase-1 so that they could be detected by bioluminescence imaging and luciferase measured in the blood. The macrophage sensor detected tumors as small as 25-50 mm3 by blood luciferase measurements, even in the presence of concomitant inflammation, and was more sensitive than clinically used protein and nucleic acid cancer biomarkers. Macrophage sensors also effectively tracked the immunological response in muscle and lung models of inflammation, suggesting the potential utility of this approach in disease states other than cancer.

    View details for PubMedID 30886438

  • Positron emission tomography reporter gene strategy for use in the central nervous system PNAS Haywood, T., Beinat, C., Gowrishankar, G., Patel, C. B., Alam, I. S., Murty, S., Gambhir, S. S. 2019

    View details for DOI 10.1073/pnas.1901645116

  • A NOVEL METABOLIC PET TRACER STRATEGY TO DETERMINE EARLY EFFECTS OF TUMOR TREATING FIELDS (TTFIELDS) Patel, C., Beinat, C., Xie, Y., Haywood, T., Murty, S., Chang, E., Gambhir, S. OXFORD UNIV PRESS INC. 2018: 32
  • COMPARISON OF THREE METABOLIC PET RADIOTRACERS IN GLIOBLASTOMA: CELL CULTURE AND ANIMAL STUDIES Beinat, C., Patel, C., Murty, S., Haywood, T., Park, J., Xie, Y., Gambhir, S. OXFORD UNIV PRESS INC. 2018: 34
  • EVALUATION OF GLYCOLYTIC RESPONSE TO SEVEN CLASSES OF ANTI-GLIOBLASTOMA DRUGS BY NON-INVASIVE MEASUREMENT OF PYRUVATE KINASE M2 Beinat, C., Patel, C., Xie, Y., Gambhir, S. OXFORD UNIV PRESS INC. 2018: 33–34
  • A novel synthesis of 6''-[18 F]-fluoromaltotriose as a PET tracer for imaging bacterial infection. Journal of labelled compounds & radiopharmaceuticals Namavari, M. n., Gowrishankar, G. n., Srinivasan, A. n., Gambhir, S. S. 2018

    Abstract

    The aim of this study was to develop a positron emission tomography (PET) tracer to visualize and monitor therapeutic response to bacterial infections. In our continued efforts to find maltose based PET tracers that can image bacterial infections, we have designed and prepared 6''-[18 F]fluoromaltotriose as a second generation PET imaging tracer targeting the maltodextrin transporter of bacteria. We have developed methods to synthesize 6''-deoxy-6''-[18 F]fluoro-α-D-glucopyranosyl-(1-4)-O-α-D-glucopyranosyl-(1-4)-O-D-glucopyranose (6''-[18 F]-fluoromaltotriose) as a bacterial infection PET imaging agent. 6''-[18 F]fluoromaltotriose was prepared from precursor, 2'',3'',4''-tri-O-acetyl-6''-O-nosyl-α-D-glucopyranosyl-(1-4)-O-2',3',6'-tri-O-acetyl-α-D-glucopyranosyl-(1-4)-1,2,3,6-tetra-O-acetyl-D-glucopyranose (per-O-acetyl-6''-O-nosyl-maltotriose 4). This method utilizes the reaction between precursor 4 and anhydrous [18 F]KF/Kryptofix 2.2.2 in Dimethylformamide (DMF) at 85o C for 10 minutes to yield per-O-acetyl-6''-deoxy-6-'' [18 F]-fluoromaltotriose (7). Successive acidic and basic hydrolysis of the acetyl protecting groups in 7 produced 6''-[18 F]fluoromaltotriose (8). Also, cold 6''- [19 F]fluoromaltotriose was prepared from per-O-acetyl-6''-hydroxymaltotriose via a DAST reaction followed by a basic hydrolysis. A successful synthesis of 6''-[18 F]-fluoromaltotriose has been accomplished in 8±1.2 % radiochemical yield (decay corrected). Total synthesis time was 120 min. Serum stability of 6''-[18 F]fluoromaltotriose at 37o C indicated that 6''-[18 F]-fluoromaltotriose remained intact up to 2 h. In conclusion, we have successfully synthesized 6''-[18 F]-fluoromaltotriose via direct fluorination of an appropriate precursor of a protected maltotriose.

    View details for PubMedID 29314161

  • The Utility of [18F]DASA-23 for Molecular Imaging of Prostate Cancer with Positron Emission Tomography. Molecular imaging and biology : MIB : the official publication of the Academy of Molecular Imaging Beinat, C. n., Haywood, T. n., Chen, Y. S., Patel, C. B., Alam, I. S., Murty, S. n., Gambhir, S. S. 2018

    Abstract

    There is a strong, unmet need for superior positron emission tomography (PET) imaging agents that are able to measure biochemical processes specific to prostate cancer. Pyruvate kinase M2 (PKM2) catalyzes the concluding step in glycolysis and is a key regulator of tumor growth and metabolism. Elevation of PKM2 expression was detected in Gleason 8-10 tumors compared to Gleason 6-7 carcinomas, indicating that PKM2 may potentially be a marker of aggressive prostate cancer. We have recently reported the development of a PKM2-specific radiopharmaceutical [18F]DASA-23 and herein describe its evaluation in cell culture and preclinical models of prostate cancer.The cellular uptake of [18F]DASA-23 was evaluated in a panel of prostate cancer cell lines and compared to that of [18F]FDG. The specificity of [18F]DASA-23 to measure PKM2 levels in cell culture was additionally confirmed through the use of PKM2-specific siRNA. PET imaging studies were then completed utilizing subcutaneous prostate cancer xenografts using either PC3 or DU145 cells in mice.[18F]DASA-23 uptake values over 60-min incubation period in PC3, LnCAP, and DU145 respectively were 23.4 ± 4.5, 18.0 ± 2.1, and 53.1 ± 4.6 % tracer/mg protein. Transient reduction in PKM2 protein expression with siRNA resulted in a 50.1 % reduction in radiotracer uptake in DU145 cells. Small animal PET imaging revealed 0.86 ± 0.13 and 1.6 ± 0.2 % ID/g at 30 min post injection of radioactivity in DU145 and PC3 subcutaneous tumor bearing mice respectively.Herein, we evaluated a F-18-labeled PKM2-specific radiotracer, [18F]DASA-23, for the molecular imaging of prostate cancer with PET. [18F]DASA-23 revealed rapid and extensive uptake levels in cellular uptake studies of prostate cancer cells; however, there was only modest tumor uptake when evaluated in mouse subcutaneous tumor models.

    View details for PubMedID 29736561

  • F]DASA-23 for Imaging Tumor Glycolysis Through Noninvasive Measurement of Pyruvate Kinase M2. Molecular imaging and biology Beinat, C., Alam, I. S., James, M. L., Srinivasan, A., Gambhir, S. S. 2017

    Abstract

    A hallmark of cancer is metabolic reprogramming, which is exploited by cancer cells to ensure rapid growth and survival. Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, a key step in tumor metabolism and growth. Recently, we reported the radiosynthesis of the first positron emission tomography tracer for visualizing PKM2 in vivo-i.e., [(11)C]DASA-23. Due to the highly promising imaging results obtained with [(11)C]DASA-23 in rodent model glioblastoma, we set out to generate an F-18-labeled version of this tracer, with the end goal of clinical translation in mind. Herein, we report the radiosynthesis of 1-((2-fluoro-6-[(18)F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([(18)F]DASA-23) and our initial investigation of its binding properties in cancer cells.We synthesized [(18)F]DASA-23 via fluorination of 1-((2-fluoro-6-nitrophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine (10) with K[(18)F]F/K2.2.2 in N,N-dimethylformamide at 110 °C for 20 min. Subsequently, we evaluated uptake of [(18)F]DASA-23 in HeLa cervical adenocarcinoma cells and in vitro stability in human and mouse serum.We successfully prepared [(18)F]DASA-23 in 2.61 ± 1.54 % radiochemical yield (n = 10, non-decay corrected at end of synthesis) with a specific activity of 2.59 ± 0.44 Ci/μmol. Preliminary cell uptake experiments revealed high uptake in HeLa cells, which was effectively blocked by pretreating cells with the structurally distinct PKM2 activator, TEPP-46. [(18)F]DASA-23 remained intact in human and mouse serum up to 120 min.Herein, we have identified a F-18-labeled PKM2 specific radiotracer which shows potential for in vivo imaging. The promising cell uptake results reported herein warrant the further evaluation of [(18)F]DASA-23 for its ability to detect and monitor cancer noninvasively.

    View details for DOI 10.1007/s11307-017-1068-8

    View details for PubMedID 28236227

  • receptor. EJNMMI research Palner, M., Beinat, C., Banister, S., Zanderigo, F., Park, J. H., Shen, B., Hjoernevik, T., Jung, J. H., Lee, B. C., Kim, S. E., Fung, L., Chin, F. T. 2016; 6 (1): 80-?

    Abstract

    The availability of GABAA receptor binding sites in the brain can be assessed by positron emission tomography (PET) using the radioligand, [(18)F]flumazenil. However, the brain uptake and binding of this PET radioligand are influenced by anesthetic drugs, which are typically needed in preclinical imaging studies and clinical imaging studies involving patient populations that do not tolerate relatively longer scan times. The objective of this study was to examine the effects of anesthesia on the binding of [(18)F]flumazenil to GABAA receptors in mice.Brain and whole blood radioactivity concentrations were measured ex vivo by scintillation counting or in vivo by PET in four groups of mice following administration of [(18)F]flumazenil: awake mice and mice anesthetized with isoflurane, dexmedetomidine, or ketamine/dexmedetomidine. Dynamic PET recordings were obtained for 60 min in mice anesthetized by either isoflurane or ketamine/dexmedetomidine. Static PET recordings were obtained at 25 or 55 min after [(18)F]flumazenil injection in awake or dexmedetomidine-treated mice acutely anesthetized with isoflurane. The apparent distribution volume (VT*) was calculated for the hippocampus and frontal cortex from either the full dynamic PET scans using an image-derived input function or from a series of ex vivo experiments using whole blood as the input function.PET images showed persistence of high [(18)F]flumazenil uptake (up to 20 % ID/g) in the brains of mice scanned under isoflurane or ketamine/dexmedetomidine anesthesia, whereas uptake was almost indiscernible in late samples or static scans from awake or dexmedetomidine-treated animals. The steady-state VT* was twofold higher in hippocampus of isoflurane-treated mice and dexmedetomidine-treated mice than in awake mice.Anesthesia has pronounced effects on the binding and blood-brain distribution of [(18)F]flumazenil. Consequently, considerable caution must be exercised in the interpretation of preclinical and clinical PET studies of GABAA receptors involving the use of anesthesia.

    View details for PubMedID 27826950

  • Effects of common anesthetic agents on [F-18] flumazenil binding to the GABA(A) receptor EJNMMI RESEARCH Palner, M., Beinat, C., Banister, S., Zanderigo, F., Park, J. H., Shen, B., Hjoernevik, T., Jung, J. H., Lee, B. C., Kim, S. E., Fung, L., Chin, F. T. 2016; 6

    Abstract

    The availability of GABAA receptor binding sites in the brain can be assessed by positron emission tomography (PET) using the radioligand, [(18)F]flumazenil. However, the brain uptake and binding of this PET radioligand are influenced by anesthetic drugs, which are typically needed in preclinical imaging studies and clinical imaging studies involving patient populations that do not tolerate relatively longer scan times. The objective of this study was to examine the effects of anesthesia on the binding of [(18)F]flumazenil to GABAA receptors in mice.Brain and whole blood radioactivity concentrations were measured ex vivo by scintillation counting or in vivo by PET in four groups of mice following administration of [(18)F]flumazenil: awake mice and mice anesthetized with isoflurane, dexmedetomidine, or ketamine/dexmedetomidine. Dynamic PET recordings were obtained for 60 min in mice anesthetized by either isoflurane or ketamine/dexmedetomidine. Static PET recordings were obtained at 25 or 55 min after [(18)F]flumazenil injection in awake or dexmedetomidine-treated mice acutely anesthetized with isoflurane. The apparent distribution volume (VT*) was calculated for the hippocampus and frontal cortex from either the full dynamic PET scans using an image-derived input function or from a series of ex vivo experiments using whole blood as the input function.PET images showed persistence of high [(18)F]flumazenil uptake (up to 20 % ID/g) in the brains of mice scanned under isoflurane or ketamine/dexmedetomidine anesthesia, whereas uptake was almost indiscernible in late samples or static scans from awake or dexmedetomidine-treated animals. The steady-state VT* was twofold higher in hippocampus of isoflurane-treated mice and dexmedetomidine-treated mice than in awake mice.Anesthesia has pronounced effects on the binding and blood-brain distribution of [(18)F]flumazenil. Consequently, considerable caution must be exercised in the interpretation of preclinical and clinical PET studies of GABAA receptors involving the use of anesthesia.

    View details for DOI 10.1186/s13550-016-0235-2

    View details for Web of Science ID 000387828200001

  • The Recent Development of alpha(7) Nicotinic Acetylcholine Receptor (nAChR) Ligands as Therapeutic Candidates for the Treatment of Central Nervous System (CNS) Diseases CURRENT PHARMACEUTICAL DESIGN Beinat, C., Banister, S. D., Herrera, M., Kassiou, M. 2016; 22 (14): 2134-2151

    Abstract

    Homomeric α7 nicotinic acetylcholine receptors (nAChRs) are implicated in the regulation of cognitive processes such as memory and attention and have potential as therapeutic targets for the treatment of the cognitive deficits associated with schizophrenia. Though numerous α7 nAChR agonists have been developed, and several have progressed to clinical trials, these are derived from few common chemotypes. Consequently, many of these α7 nAChR clinical candidates share unfavorable side-effect profile. SEN12333 represents a novel chemotype for the development of α7 nAChR agonists, and exploration of this scaffold has produced structurally diverse ligands with promising pharmacological properties. This review will summarize structure-affinity and -activity relationships surrounding analogs of SEN12333.

    View details for DOI 10.2174/1381612822666160127114125

    View details for Web of Science ID 000375146900016

    View details for PubMedID 26818858

  • Pharmacology of Indole and Indazole Synthetic Cannabinoid Designer Drugs AB-FUBINACA, ADB-FUBINACA, AB-PINACA, ADB-PINACA, 5F-AB-PINACA, 5F-ADB-PINACA, ADBICA, and 5F-ADBICA ACS CHEMICAL NEUROSCIENCE Banister, S. D., Moir, M., Stuart, J., Kevin, R. C., Wood, K. E., Longworth, M., Wilkinson, S. M., Beinat, C., Buchanan, A. S., Glass, M., Connor, M., McGregor, I. S., Kassiou, M. 2015; 6 (9): 1546-1559

    Abstract

    Synthetic cannabinoid (SC) designer drugs based on indole and indazole scaffolds and featuring l-valinamide or l-tert-leucinamide side chains are encountered with increasing frequency by forensic researchers and law enforcement agencies and are associated with serious adverse health effects. However, many of these novel SCs are unprecedented in the scientific literature at the time of their discovery, and little is known of their pharmacology. Here, we report the synthesis and pharmacological characterization of AB-FUBINACA, ADB-FUBINACA, AB-PINACA, ADB-PINACA, 5F-AB-PINACA, 5F-ADB-PINACA, ADBICA, 5F-ADBICA, and several analogues. All synthesized SCs acted as high potency agonists of CB1 (EC50 = 0.24-21 nM) and CB2 (EC50 = 0.88-15 nM) receptors in a fluorometric assay of membrane potential, with 5F-ADB-PINACA showing the greatest potency at CB1 receptors. The cannabimimetic activities of AB-FUBINACA and AB-PINACA in vivo were evaluated in rats using biotelemetry. AB-FUBINACA and AB-PINACA dose-dependently induced hypothermia and bradycardia at doses of 0.3-3 mg/kg, and hypothermia was reversed by pretreatment with a CB1 (but not CB2) antagonist, indicating that these SCs are cannabimimetic in vivo, consistent with anecdotal reports of psychoactivity in humans.

    View details for DOI 10.1021/acschemneuro.5b00112

    View details for Web of Science ID 000361505100006

    View details for PubMedID 26134475

  • Effects of bioisosteric fluorine in synthetic cannabinoid designer drugs JWH-018, AM-2201, UR-144, XLR-11, PB-22, 5F-PB-22, APICA, and STS-135. ACS chemical neuroscience Banister, S. D., Stuart, J., Kevin, R. C., Edington, A., Longworth, M., Wilkinson, S. M., Beinat, C., Buchanan, A. S., Hibbs, D. E., Glass, M., Connor, M., McGregor, I. S., Kassiou, M. 2015; 6 (8): 1445-58

    Abstract

    Synthetic cannabinoid (SC) designer drugs featuring bioisosteric fluorine substitution are identified by forensic chemists and toxicologists with increasing frequency. Although terminal fluorination of N-pentyl indole SCs is sometimes known to improve cannabinoid type 1 (CB1) receptor binding affinity, little is known of the effects of fluorination on functional activity of SCs. This study explores the in vitro functional activities of SC designer drugs JWH-018, UR-144, PB-22, and APICA, and their respective terminally fluorinated analogues AM-2201, XLR-11, 5F-PB-22, and STS-135 at human CB1 and CB2 receptors using a FLIPR membrane potential assay. All compounds demonstrated agonist activity at CB1 (EC50 = 2.8-1959 nM) and CB2 (EC50 = 6.5-206 nM) receptors, with the fluorinated analogues generally showing increased CB1 receptor potency (∼2-5 times). Additionally, the cannabimimetic activities and relative potencies of JWH-018, AM-2201, UR-144, XLR-11, PB-22, 5F-PB-22, APICA, and STS-135 in vivo were evaluated in rats using biotelemetry. All SCs dose-dependently induced hypothermia and reduced heart rate at doses of 0.3-10 mg/kg. There was no consistent trend for increased potency of fluorinated SCs over the corresponding des-fluoro SCs in vivo. Based on magnitude and duration of hypothermia, the SCs were ranked for potency (PB-22 > 5F-PB-22 = JWH-018 > AM-2201 > APICA = STS-135 = XLR-11 > UR-144).

    View details for DOI 10.1021/acschemneuro.5b00107

    View details for PubMedID 25921407

  • Effects of Bioisosteric Fluorine in Synthetic Cannabinoid Designer Drugs JWH-018, AM-2201, UR-144, XLR-11, PB-22, 5F-PB-22, APICA, and STS-135 ACS CHEMICAL NEUROSCIENCE Banister, S. D., Stuart, J., Kevin, R. C., Edington, A., Longworth, M., Wilkinson, S. M., Beinat, C., Buchanan, A. S., Hibbs, D. E., Glass, M., Connor, M., McGregor, I. S., Kassiou, M. 2015; 6 (8): 1445-1458
  • Structure-activity relationships of synthetic cannabinoid designer drug RCS-4 and its regioisomers and C4 homologues FORENSIC TOXICOLOGY Banister, S. D., Stuart, J., Conroy, T., Longworth, M., Manohar, M., Beinat, C., Wilkinson, S. M., Kevin, R. C., Hibbs, D. E., Glass, M., Connor, M., McGregor, I. S., Kassiou, M. 2015; 33 (2): 355-366
  • The Therapeutic Potential of alpha(7) Nicotinic Acetylcholine Receptor (alpha(7) nAChR) Agonists for the Treatment of the Cognitive Deficits Associated with Schizophrenia CNS DRUGS Beinat, C., Banister, S. D., Herrera, M., Law, V., Kassiou, M. 2015; 29 (7): 529-542

    Abstract

    Homomeric α7 nicotinic acetylcholine receptors (α7 nAChRs) have implications in the regulation of cognitive processes such as memory and attention, and have shown promise as a therapeutic target for the treatment of the cognitive deficits associated with schizophrenia. Multiple α7 nAChR agonists have entered human trials; however, unfavorable side effects and pharmacokinetic issues have hindered the development of a clinical α7 nAChR agonist. Currently, EVP-6124 is in phase III clinical trials, and several other α7 nAChR agonists (GTS-21 and AQW051) are in earlier stages of development. This review will summarize the recent advances and failures of α7 nAChR agonists in clinical trials for the treatment of the aforementioned pathology.

    View details for DOI 10.1007/s40263-015-0260-0

    View details for Web of Science ID 000360567900002

    View details for PubMedID 26242477

  • Structure-activity relationship studies of SEN12333 analogues: Determination of the optimal requirements for binding affinities at alpha 7 nAChRs through incorporation of known structural motifs EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY Beinat, C., Reekie, T., Banister, S. D., O'Brien-Brown, J., Xie, T., Olson, T. T., Xiao, Y., Harvey, A., O'Connor, S., Coles, C., Grishin, A., Kolesik, P., Tsanaktsidis, J., Kassiou, M. 2015; 95: 277-301

    Abstract

    Alpha7 nicotinic acetylcholine receptors (nAChRs) have implications in the regulation of cognitive processes such as memory and attention and have been identified as a promising therapeutic target for the treatment of the cognitive deficits associated with schizophrenia and Alzheimer's disease (AD). Structure affinity relationship studies of the previously described α7 agonist SEN12333 (8), have resulted in the identification of compound 45, a potent and selective agonist of the α7 nAChR with enhanced affinity and improved physicochemical properties over the parent compound (SEN12333, 8).

    View details for DOI 10.1016/j.ejmech.2015.03.025

    View details for Web of Science ID 000354139900025

    View details for PubMedID 25827398

  • Ether analogues of DPA-714 with subnanomolar affinity for the translocator protein (TSPO) EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY Banister, S. D., Beinat, C., Wilkinson, S. M., Shen, B., Bartoli, C., Selleri, S., Da Pozzo, E., Martini, C., Chin, F. T., Kassiou, M. 2015; 93: 392-400

    Abstract

    Sixteen new phenyl alkyl ether derivatives (12, 14-28) of the 5,7-dimethylpyrazolo[1,5-a]pyrimidin-3-ylacetamide (DPA) class were synthesized and evaluated in a competition binding assay against [(3)H]PK11195 using 18 kDa translocator protein (TSPO) derived from rat kidney mitochondrial fractions. All analogues showed superior binding affinities for TSPO compared to DPA-713 (5) and DPA-714 (6). Picomolar affinities were observed for this class of TSPO ligands in this assay for the first time, with phenethyl ether 28 showing the greatest affinity (Ki = 0.13 nM). Additionally, all analogues increased pregnenolone biosynthesis (134-331% above baseline) in a rat C6 glioma cell steroidogenesis assay.

    View details for DOI 10.1016/j.ejmech.2015.02.004

    View details for Web of Science ID 000351646100040

    View details for PubMedID 25725375

  • Recent Advances in the Development of Sigma-1 Receptor Ligands Australian Journal of Chemistry Manohar, M., Banister, S. D., Beinat, C., O'Brien-Brown, J., Kassiou, M. 2015; 68 (4): 600-609

    View details for DOI 10.1071/CH14590

  • Ether analogues of DPA-714 with subnanomolar affinity for the translocator protein (TSPO) European Journal of Medicinal Chemistry Banister, S. D., Beinat, C., Wilkinson, S. M., Shen, B., Bartoli, C., Selleri, S., Da Pozzo, E., Martini, C., Chin, F. T., Kassiou, M. 2015; 93: 392-400
  • Investigations of amide bond variation and biaryl modification in analogues of alpha 7 nAChR agonist SEN12333 EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY Beinat, C., Reekie, T., Hibbs, D., Xie, T., Olson, T. T., Xiao, Y., Harvey, A., O'Connor, S., Coles, C., Tsanaktsidis, J., Kassiou, M. 2014; 84: 200-205

    Abstract

    Several lines of experimental evidence support the involvement of the α7 nAChR in schizophrenia and Alzheimer's disease. Modulators of the α7 nAChR have been extensively reviewed for the treatment of the cognitive deficits associated with these pathologies. SEN12333 represents a novel α7 nAChR agonist chemotype with potential for reduced side effects but requiring further SAR exploration. The present work investigates the amide bond of SEN12333, specifically its connectivity and replacement with the tetrazole functionality, a known cis amide isostere. The results reveal the original amide bond connectivity of SEN12333 to be favorable for binding affinity and agonist activity at α7 nAChRs. The use of a tetrazole isostere completely abolishes affinity and functional activity and suggests that SEN12333 binds in a linear conformation. Results reported herein also suggest the pyridine nitrogen within the terminal aromatic ring of SEN12333 is not essential for binding affinity or functional activity. Further SAR investigations involving manipulation of other moieties contained within SEN12333 are warranted.

    View details for DOI 10.1016/j.ejmech.2014.07.029

    View details for Web of Science ID 000341464500022

    View details for PubMedID 25019477

  • Structure-activity relationships of N-substituted 4-(trifluoromethoxy) benzamidines with affinity for GluN2B-containing NMDA receptors BIOORGANIC & MEDICINAL CHEMISTRY LETTERS Beinat, C., Banister, S. D., Hoban, J., Tsanaktsidis, J., Metaxas, A., Windhorst, A. D., Kassiou, M. 2014; 24 (3): 828-830

    Abstract

    GluN2B subtype-selective NMDA antagonists represent promising therapeutic targets for the symptomatic treatment of multiple CNS pathologies. A series of N-benzyl substituted benzamidines were synthesised and the benzyl ring was further replaced with various polycyclic moieties. Compounds were evaluated for activity at GluN2B containing NMDA receptors where analogues 9, 12, 16 and 18 were the most potent of the series, replacement of the benzyl ring with polycycles resulted in a complete loss of activity.

    View details for DOI 10.1016/j.bmcl.2013.12.087

    View details for Web of Science ID 000330120800022

    View details for PubMedID 24412068

  • A practical synthesis of (1S,4S)-2,5-diazabicyclo[2.2.1]heptane TETRAHEDRON LETTERS Beinat, C., Banister, S. D., McErlean, C. S., Kassiou, M. 2013; 54 (39): 5345-5347
  • Consequences of linker length alteration of the alpha 7 nicotinic acetylcholine receptor (nAChR) agonist, SEN12333 BIOORGANIC & MEDICINAL CHEMISTRY LETTERS Beinat, C., Banister, S. D., van Prehn, S., Doddareddy, M. R., Hibbs, D., Sako, M., Chebib, M., Thao Tran, T., Al-Muhtasib, N., Xiao, Y., Kassiou, M. 2012; 22 (7): 2380-2384

    Abstract

    A series of ligands based on SEN12333, containing either contracted or elongated alkyl chains, were synthesized and evaluated in molecular docking studies against a homology model of the α7 nicotinic acetylcholine receptor (nAChR) subtype. The predicted binding of all ligands was highly similar, with the exception of the analog containing a 5 methylene unit spacer. However, in vitro competition binding assays revealed that the ligands possessed dissimilar binding affinities, with a K(i) range of more than an order of magnitude (K(i)=0.50 to >10 μM), and only SEN12333 itself exhibited functional activity at the α7 nAChR.

    View details for DOI 10.1016/j.bmcl.2012.02.052

    View details for Web of Science ID 000301846100002

    View details for PubMedID 22410083

  • Trishomocubane as a scaffold for the development of selective dopamine transporter (DAT) ligands BIOORGANIC & MEDICINAL CHEMISTRY LETTERS Banister, S. D., Moussa, I. A., Beinat, C., Reynolds, A. J., Schiavini, P., Jorgensen, W. T., Kassiou, M. 2011; 21 (1): 38-41

    Abstract

    In our continued exploration of trishomocubane derivatives with central nervous system (CNS) activity, N-arylalkyl-8-aminopentacyclo[5.4.0.0(2,6).0(3,10).0(5,9)]undecanes (10-13) displaying affinity for the sigma (σ) receptor were also found, in several cases, to interact with the dopamine transporter (DAT). Compound 12 was identified as the first trishomocubane-derived high affinity DAT ligand (K(i) = 1.2 nM), with greater than 8300-fold selectivity over the monoamine transporters NET and SERT, and only low to moderate affinity for σ(1) and σ(2) receptors.

    View details for DOI 10.1016/j.bmcl.2010.11.075

    View details for Web of Science ID 000285544400002

    View details for PubMedID 21146989

  • Design, Synthesis, and Structure-Affinity Relationships of Regioisomeric N-Benzyl Alkyl Ether Piperazine Derivatives as sigma-1 Receptor Ligands JOURNAL OF MEDICINAL CHEMISTRY Moussa, I. A., Banister, S. D., Beinat, C., Giboureau, N., Reynolds, A. J., Kassiou, M. 2010; 53 (16): 6228-6239

    Abstract

    A series of N-(benzofuran-2-ylmethyl)-N'-benzylpiperazines bearing alkyl or fluoroalkyl aryl ethers were synthesized and evaluated at various central nervous system receptors. Examination of in vitro sigma1 {[3H]+-pentazocine} and sigma2 ([3H]DTG) receptor binding profiles of piperazines 11-13 and 25-36 revealed several highly potent and sigma1 selective ligands, notably, N-(benzofuran-2-ylmethyl)-N'-(4'-methoxybenzyl)piperazine (13, Ki=2.7 nM, sigma2/sigma1=38) and N-(benzofuran-2-ylmethyl)-N'-(4'-(2''-fluoroethoxy)benzyl)piperazine (30, Ki=2.6 nM, sigma2/sigma1=187). Structural features for optimal sigma1 receptor affinity and selectivity over the sigma2 receptor were identified. On the basis of its favorable log D value, 13 was selected as a candidate for the development of a sigma1 receptor positron emission tomography radiotracer. [11C]13 showed high uptake in the brain and other sigma receptor-rich organs of a Papio hamadryas baboon. The in vivo evaluation of [11C]13 indicates that this radiotracer is a suitable candidate for imaging the sigma1 receptor in neurodegenerative processes.

    View details for DOI 10.1021/jm100639f

    View details for Web of Science ID 000280962700028

    View details for PubMedID 20662542

  • Insights into Structure-Activity Relationships and CNS Therapeutic Applications of NR2B Selective Antagonists - See more at: http://www.eurekaselect.com/72752/article#sthash.FVkeRGWN.dpuf Current Medicinal Chemistry Beinat, C., Banister, S., Moussa, I., Reynolds, A. J., McErlean, C. S., Kassiou, M. 2010 ; 17 (34): 4166-90
  • Development of Vesicular Acetylcholine Transporter Ligands: Molecular Probes for Alzheimers Disease - See more at: http://www.eurekaselect.com/71948/article#sthash.dsWtg2ps.dpuf Current Bioactive Compounds Giboureau, N., Aumann, K. M., Beinat, C., Kassiou, M. 2010; 27: 129-155