51
|
Ellison PA, Olson AP, Barnhart TE, Hoffman SLV, Reilly SW, Makvandi M, Bartels JL, Murali D, DeJesus OT, Lapi SE, Bednarz B, Nickles RJ, Mach RH, Engle JW. Improved production of 76Br, 77Br and 80mBr via CoSe cyclotron targets and vertical dry distillation. Nucl Med Biol 2020; 80-81:32-36. [PMID: 31575457 PMCID: PMC9066421 DOI: 10.1016/j.nucmedbio.2019.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/11/2019] [Accepted: 09/04/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The radioisotopes of bromine are uniquely suitable radiolabels for small molecule theranostic radiopharmaceuticals but are of limited availability due to production challenges. Significantly improved methods were developed for the production and radiochemical isolation of clinical quality 76Br, 77Br, and 80mBr. The radiochemical quality of the radiobromine produced using these methods was tested through the synthesis of a novel 77Br-labeled inhibitor of poly (ADP-ribose) polymerase-1 (PARP-1), a DNA damage response protein. METHODS 76Br, 77Br, and 80mBr were produced in high radionuclidic purity via the proton irradiation of novel isotopically-enriched Co76Se, Co77Se, and Co80Se intermetallic targets, respectively. Radiobromine was isolated through thermal chromatographic distillation in a vertical furnace assembly. The 77Br-labeled PARP inhibitor was synthesized via copper-mediated aryl boronic ester radiobromination. RESULTS Cyclotron production yields were 103 ± 10 MBq∙μA-1∙h-1 for 76Br, 88 ± 10 MBq∙μA-1∙h-1 for 80mBr at 16 MeV and 17 ± 1 MBq∙μA-1∙h-1 for 77Br at 13 MeV. Radiobromide isolation yields were 76 ± 11% in a small volume of aqueous solution. The synthesized 77Br-labeled PARP-1 inhibitor had a measured apparent molar activity up to 700 GBq/μmol at end of synthesis. CONCLUSIONS A novel selenium alloy target enabled clinical-scale production of 76Br, 77Br, and 80mBr with high apparent molar activities, which was used to for the production of a new 77Br-labeled inhibitor of PARP-1. ADVANCES IN KNOWLEDGE New methods for the cyclotron production and isolation of radiobromine improved the production capacity of 77Br by a factor of three and 76Br by a factor of six compared with previous methods. IMPLICATIONS FOR PATIENT CARE Preclinical translational research of 77Br-based Auger electron radiotherapeutics, such as those targeting PARP-1, will require the production of GBq-scale 77Br, which necessitates next-generation, high-yielding, isotopically-enriched cyclotron targets, such as the novel intermetallic Co77Se.
Collapse
Affiliation(s)
- Paul A Ellison
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Aeli P Olson
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Todd E Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sabrina L V Hoffman
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sean W Reilly
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Mehran Makvandi
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jennifer L Bartels
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dhanabalan Murali
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Onofre T DeJesus
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bryan Bednarz
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Engineering Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Robert J Nickles
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Robert H Mach
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
52
|
Laird J, Lok BH, Carney B, Kossatz S, de Stanchina E, Reiner T, Poirier JT, Rudin CM. Positron-Emission Tomographic Imaging of a Fluorine 18-Radiolabeled Poly(ADP-Ribose) Polymerase 1 Inhibitor Monitors the Therapeutic Efficacy of Talazoparib in SCLC Patient-Derived Xenografts. J Thorac Oncol 2019; 14:1743-1752. [PMID: 31195178 DOI: 10.1016/j.jtho.2019.05.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/15/2019] [Accepted: 05/29/2019] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Inhibitors of poly-(ADP)-ribose polymerase (PARP) are promising therapeutics for SCLC. We tested whether PARP inhibitor (PARPi) target engagement as measured by a fluorine 18-radiolabeled PARPi ([18F]PARPi) has the potential to predict drug efficacy in vivo. METHODS Tumor growth inhibition during daily talazoparib treatment was evaluated in mice engrafted with SCLC patient-derived xenografts to evaluate talazoparib efficacy at multiple doses. Mice were intravenously injected with [18F]PARPi radiotracer at multiple timepoints after single doses of oral talazoparib to quantitatively assess the extent to which talazoparib could reduce tumor radiotracer uptake and positron-emission tomographic (PET)/computer tomographic activity. Tumors were harvested and tumor poly-(ADP) ribose level was measured by enzyme-linked immunosorbent assay. RESULTS A dose range of talazoparib with differential therapeutic efficacy was established, with significant delay in time to reach 1000 mm3 for tumors treated with 0.3 mg/kg (p = 0.02) but not 0.1 mg/kg talazoparib. On PET/computed tomography with [18F]PARPi, reduction in [18F]PARPi uptake after talazoparib dosing was consistent with talazoparib clearance, with reduction in PET activity attenuating over 24 hours. Talazoparib target engagement, measured by maximum tumor PET uptake, increased in a dose-dependent manner (3.9% versus 2.1% injected dose/g for 0.1 and 0.3 mg/kg at 3 hours post-talazoparib, p = 0.003) and correlated with PARP enzymatic activity among individual tumors as measured by total tumor poly-(ADP) ribose (p = 0.04, R = 0.62 at 1 hour post-talazoparib). CONCLUSIONS PET imaging using [18F]PARPi has the potential to be a powerful tool in treatment monitoring by assessing PARPi target engagement in real-time.
Collapse
Affiliation(s)
- James Laird
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; New York University School of Medicine, New York, New York
| | - Benjamin H Lok
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Chemistry, Hunter College and PhD Program in Chemistry, The Graduate Center of the City University of New York, New York, New York
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiology, Weill Cornell Medical College, New York, New York; Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John T Poirier
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M Rudin
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
53
|
Prevet H, Collins I. Labelled chemical probes for demonstrating direct target engagement in living systems. Future Med Chem 2019; 11:1195-1224. [PMID: 31280668 DOI: 10.4155/fmc-2018-0370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
Demonstrating target engagement in living systems can help drive successful drug discovery. Target engagement and occupancy studies in cells confirm direct binding of a ligand to its intended target protein and provide the binding affinity. Combined with biomarkers to measure the functional consequences of target engagement, these experiments can increase confidence in the relationship between in vitro pharmacology and observed biological effects. In this review, we focus on chemically and radioactively labelled probes as key reagents for performing such experiments. Using recent examples, we examine how the labelled probes have been employed in combination with unlabelled ligands to quantify target engagement in cells and in animals. Finally, we consider future developments of this emerging methodology.
Collapse
Affiliation(s)
- Hugues Prevet
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Ian Collins
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP, UK
| |
Collapse
|
54
|
Wilson TC, Xavier MA, Knight J, Verhoog S, Torres JB, Mosley M, Hopkins SL, Wallington S, Allen PD, Kersemans V, Hueting R, Smart S, Gouverneur V, Cornelissen B. PET Imaging of PARP Expression Using 18F-Olaparib. J Nucl Med 2019; 60:504-510. [PMID: 30389822 PMCID: PMC6448459 DOI: 10.2967/jnumed.118.213223] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors are increasingly being studied as cancer drugs, as single agents, or as a part of combination therapies. Imaging of PARP using a radiolabeled inhibitor has been proposed for patient selection, outcome prediction, dose optimization, genotoxic therapy evaluation, and target engagement imaging of novel PARP-targeting agents. Methods: Here, via the copper-mediated 18F-radiofluorination of aryl boronic esters, we accessed, for the first time (to our knowledge), the 18F-radiolabeled isotopolog of the Food and Drug Administration-approved PARP inhibitor olaparib. The use of the 18F-labeled equivalent of olaparib allows direct prediction of the distribution of olaparib, given its exact structural likeness to the native, nonradiolabeled drug. Results:18F-olaparib was taken up selectively in vitro in PARP-1-expressing cells. Irradiation increased PARP-1 expression and 18F-olaparib uptake in a radiation-dose-dependent fashion. PET imaging in mice showed specific uptake of 18F-olaparib in tumors expressing PARP-1 (3.2% ± 0.36% of the injected dose per gram of tissue in PSN-1 xenografts), correlating linearly with PARP-1 expression. Two hours after irradiation of the tumor (10 Gy), uptake of 18F-olaparib increased by 70% (P = 0.025). Conclusion: Taken together, we show that 18F-olaparib has great potential for noninvasive tumor imaging and monitoring of radiation damage.
Collapse
Affiliation(s)
- Thomas C. Wilson
- Department of Chemistry, University of Oxford, Oxford, United Kingdom; and
| | - Mary-Ann Xavier
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - James Knight
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Stefan Verhoog
- Department of Chemistry, University of Oxford, Oxford, United Kingdom; and
| | - Julia Baguña Torres
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Michael Mosley
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Samantha L. Hopkins
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sheena Wallington
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Phillip D. Allen
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Veerle Kersemans
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Rebekka Hueting
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sean Smart
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Bart Cornelissen
- CRUK/MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
55
|
Reilly SW, Puentes LN, Schmitz A, Hsieh CJ, Weng CC, Hou C, Li S, Kuo YM, Padakanti P, Lee H, Riad AA, Makvandi M, Mach RH. Synthesis and evaluation of an AZD2461 [ 18F]PET probe in non-human primates reveals the PARP-1 inhibitor to be non-blood-brain barrier penetrant. Bioorg Chem 2019; 83:242-249. [PMID: 30390553 PMCID: PMC6378121 DOI: 10.1016/j.bioorg.2018.10.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 01/05/2023]
Abstract
Poly(ADP-ribose)polymerase-1 inhibitor (PARPi) AZD2461 was designed to be a weak P-glycoprotein (P-gp) analogue of FDA approved olaparib. With this chemical property in mind, we utilized the AZD2461 ligand architecture to develop a CNS penetrant and PARP-1 selective imaging probe, in order to investigate PARP-1 mediated neuroinflammation and neurodegenerative diseases, such as Alzheimer's and Parkinson's. Our work led to the identification of several high-affinity PARPi, including AZD2461 congener 9e (PARP-1 IC50 = 3.9 ± 1.2 nM), which was further evaluated as a potential 18F-PET brain imaging probe. However, despite the similar molecular scaffolds of 9e and AZD2461, our studies revealed non-appreciable brain-uptake of [18F]9e in non-human primates, suggesting AZD2461 to be non-CNS penetrant.
Collapse
Affiliation(s)
- Sean W Reilly
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura N Puentes
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, PA 19104, USA
| | - Alexander Schmitz
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chia-Ju Hsieh
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chi-Chang Weng
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Catherine Hou
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shihong Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yin-Ming Kuo
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Prashanth Padakanti
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aladdin A Riad
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mehran Makvandi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
56
|
Lee DS, Cheon GJ. Nuclear Theranostics in Asia: In vivo Companion Diagnostics. Nucl Med Mol Imaging 2019; 53:1-6. [PMID: 30828392 PMCID: PMC6377583 DOI: 10.1007/s13139-019-00573-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
57
|
Strome A, Kossatz S, Zanoni DK, Rajadhyaksha M, Patel S, Reiner T. Current Practice and Emerging Molecular Imaging Technologies in Oral Cancer Screening. Mol Imaging 2018; 17:1536012118808644. [PMID: 32852263 PMCID: PMC6287312 DOI: 10.1177/1536012118808644] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oral cancer is one of the most common cancers globally. Survival rates for patients are directly correlated with stage of diagnosis; despite this knowledge, 60% of individuals are presenting with late-stage disease. Currently, the initial evaluation of a questionable lesion is performed by a conventional visual examination with white light. If a lesion is deemed suspicious, a biopsy is taken for diagnosis. However, not all lesions present suspicious under visual white light examination, and there is limited specificity in differentiating between benign and malignant transformations. Several vital dyes, light-based detection systems, and cytology evaluation methods have been formulated to aid in the visualization process, but their lack of specific biomarkers resulted in high false-positive rates and thus limits their reliability as screening and guidance tools. In this review, we will analyze the current methodologies and demonstrate the need for specific intraoral imaging agents to aid in screening and diagnosis to identify patients earlier. Several novel molecular imaging agents will be presented as, by result of their molecular targeting, they aim to have high specificity for tumor pathways and can support in identifying dysplastic/cancerous lesions and guiding visualization of biopsy sites. Imaging agents that are easy to use, inexpensive, noninvasive, and specific can be utilized to increase the number of patients who are screened and monitored in a variety of different environments, with the ultimate goal of increasing early detection.
Collapse
Affiliation(s)
- Arianna Strome
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Milind Rajadhyaksha
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Snehal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Radiology, Weill-Cornell Medical College, New York, NY, USA
| |
Collapse
|
58
|
Gonzales J, Kossatz S, Roberts S, Pirovano G, Brand C, Pérez-Medina C, Donabedian P, de la Cruz MJ, Mulder WJM, Reiner T. Nanoemulsion-Based Delivery of Fluorescent PARP Inhibitors in Mouse Models of Small Cell Lung Cancer. Bioconjug Chem 2018; 29:3776-3782. [PMID: 30354077 DOI: 10.1021/acs.bioconjchem.8b00640] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The preclinical potential of many diagnostic and therapeutic small molecules is limited by their rapid washout kinetics and consequently modest pharmacological performances. In several cases, these could be improved by loading the small molecules into nanoparticulates, improving blood half-life, in vivo uptake and overall pharmacodynamics. In this study, we report a nanoemulsion (NE) encapsulated form of PARPi-FL. As a proof of concept, we used PARPi-FL, which is a fluorescently labeled sensor for olaparib, a FDA-approved small molecule inhibitor of the nuclear enzyme poly(ADP-ribose)polymerase 1 (PARP1). Encapsulated PARPi-FL showed increased blood half-life, and delineated subcutaneous xenografts of small cell lung cancer (SCLC), a fast-progressing disease where efficient treatment options remain an unmet clinical need. Our study demonstrates an effective method for expanding the circulation time of a fluorescent PARP inhibitor, highlighting the pharmacokinetic benefits of nanoemulsions as nanocarriers and confirming the value of PARPi-FL as an imaging agent targeting PARP1 in small cell lung cancer.
Collapse
Affiliation(s)
- Junior Gonzales
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Susanne Kossatz
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Sheryl Roberts
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Giacomo Pirovano
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Christian Brand
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Department of Radiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Patrick Donabedian
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - M Jason de la Cruz
- Structural Biology Program, Sloan Kettering Institute , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Department of Radiology , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems , Eindhoven University of Technology , Eindhoven , The Netherlands
| | - Thomas Reiner
- Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Department of Radiology , Weill Cornell Medical College , New York , New York 10065 , United States
| |
Collapse
|
59
|
Reilly SW, Puentes LN, Hsieh CJ, Makvandi M, Mach RH. Altering Nitrogen Heterocycles of AZD2461 Affords High Affinity Poly(ADP-ribose) Polymerase-1 Inhibitors with Decreased P-Glycoprotein Interactions. ACS OMEGA 2018; 3:9997-10001. [PMID: 30198004 PMCID: PMC6120739 DOI: 10.1021/acsomega.8b00896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/02/2018] [Indexed: 06/08/2023]
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARPi) are targeted therapeutics with enhanced selectivity and cytotoxicity in BRCA1/2 mutant cancer cells. AZD2461, a congener of FDA approved olaparib, is a potent PARPi with high affinity for PARP-1 and nonsubstrate for P-glycoprotein (P-gp), an attractive characteristic for cancer therapeutics. Analogues of AZD2461 were synthesized and profiled in BRCA1 functional and nonfunctional cell lines, revealing compounds (2, 3, and 5) of low cytotoxicity and excellent PARP-1 affinities (∼4-8 nM). In comparison to AZD2461, these agents were found to be less stimulating of P-gp, suggesting that these compounds may be excellent candidates for neurological applications where blood brain barrier penetrance is sought.
Collapse
Affiliation(s)
- Sean W. Reilly
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Laura N. Puentes
- Department
of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, 421 Curie Boulevard, Philadelphia, Pennsylvania 19104, United States
| | - Chia-Ju Hsieh
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Mehran Makvandi
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert H. Mach
- Department
of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
60
|
Reilly SW, Puentes LN, Wilson K, Hsieh CJ, Weng CC, Makvandi M, Mach RH. Examination of Diazaspiro Cores as Piperazine Bioisosteres in the Olaparib Framework Shows Reduced DNA Damage and Cytotoxicity. J Med Chem 2018; 61:5367-5379. [PMID: 29856625 DOI: 10.1021/acs.jmedchem.8b00576] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Development of poly(ADP-ribose) polymerase inhibitors (PARPi's) continues to be an attractive area of research due to synthetic lethality in DNA repair deficient cancers; however, PARPi's also have potential as therapeutics to prevent harmful inflammation. We investigated the pharmacological impact of incorporating spirodiamine motifs into the phthalazine architecture of FDA approved PARPi olaparib. Synthesized analogues were screened for PARP-1 affinity, enzyme specificity, catalytic inhibition, DNA damage, and cytotoxicity. This work led to the identification of 10e (12.6 ± 1.1 nM), which did not induce DNA damage at similar drug concentrations as olaparib. Interestingly, several worst in class compounds with low PARP-1 affinity, including 15b (4397 ± 1.1 nM), induced DNA damage at micromolar concentrations, which can explain the cytotoxicity observed in vitro. This work provides further evidence that high affinity PARPi's can be developed without DNA damaging properties offering potential new drugs for treating inflammatory related diseases.
Collapse
Affiliation(s)
- Sean W Reilly
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Laura N Puentes
- Department of Systems Pharmacology and Translational Therapeutics , University of Pennsylvania , 421 Curie Blvd. , Philadelphia , Pennsylvania 19104 , United States
| | - Khadija Wilson
- Department of Systems Pharmacology and Translational Therapeutics , University of Pennsylvania , 421 Curie Blvd. , Philadelphia , Pennsylvania 19104 , United States
| | - Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Chi-Chang Weng
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Mehran Makvandi
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| |
Collapse
|
61
|
Kossatz S, Weber W, Reiner T. Detection and Delineation of Oral Cancer With a PARP1-Targeted Optical Imaging Agent. Mol Imaging 2018; 16:1536012117723786. [PMID: 28856922 PMCID: PMC5582799 DOI: 10.1177/1536012117723786] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
More sensitive and specific methods for early detection are imperative to improve survival rates in oral cancer. However, oral cancer detection is still largely based on visual examination and histopathology of biopsy material, offering no molecular selectivity or spatial resolution. Intuitively, the addition of optical contrast could improve oral cancer detection and delineation, but so far no molecularly targeted approach has been translated. Our fluorescently labeled small-molecule inhibitor PARPi-FL binds to the DNA repair enzyme poly(ADP-ribose)polymerase 1 (PARP1) and is a potential diagnostic aid for oral cancer delineation. Based on our preclinical work, a clinical phase I/II trial opened in March 2017 to evaluate PARPi-FL as a contrast agent for oral cancer imaging. In this commentary, we discuss why we chose PARP1 as a biomarker for tumor detection and which particular characteristics make PARPi-FL an excellent candidate to image PARP1 in optically guided applications. We also comment on the potential benefits of our molecularly targeted PARPi-FL-guided imaging approach in comparison to existing oral cancer screening adjuncts and mention the adaptability of PARPi-FL imaging to other environments and tumor types.
Collapse
Affiliation(s)
- Susanne Kossatz
- 1 Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wolfgang Weber
- 1 Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,2 Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,3 Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Thomas Reiner
- 1 Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,3 Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
62
|
Zmuda F, Blair A, Liuzzi MC, Malviya G, Chalmers AJ, Lewis D, Sutherland A, Pimlott SL. An 18F-Labeled Poly(ADP-ribose) Polymerase Positron Emission Tomography Imaging Agent. J Med Chem 2018; 61:4103-4114. [PMID: 29630818 PMCID: PMC6007963 DOI: 10.1021/acs.jmedchem.8b00138] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Indexed: 11/29/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) is involved in repair of DNA breaks and is over-expressed in a wide variety of tumors, making PARP an attractive biomarker for positron emission tomography (PET) and single photon emission computed tomography imaging. Consequently, over the past decade, there has been a drive to develop nuclear imaging agents targeting PARP. Here, we report the discovery of a PET tracer that is based on the potent PARP inhibitor olaparib (1). Our lead PET tracer candidate, [18F]20, was synthesized and evaluated as a potential PARP PET radiotracer in mice bearing subcutaneous glioblastoma xenografts using ex vivo biodistribution and PET-magnetic resonance imaging techniques. Results showed that [18F]20 could be produced in a good radioactivity yield and exhibited specific PARP binding allowing visualization of tumors over-expressing PARP. [18F]20 is therefore a potential candidate radiotracer for in vivo PARP PET imaging.
Collapse
Affiliation(s)
- Filip Zmuda
- WestCHEM,
School of Chemistry, University of Glasgow, The Joseph Black Building, Glasgow G12 8QQ, U.K.
- Wolfson
Whol Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, U.K.
| | - Adele Blair
- WestCHEM,
School of Chemistry, University of Glasgow, The Joseph Black Building, Glasgow G12 8QQ, U.K.
| | - Maria Clara Liuzzi
- WestCHEM,
School of Chemistry, University of Glasgow, The Joseph Black Building, Glasgow G12 8QQ, U.K.
- School of Medicine, College of Medical, Veterinary
and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - Gaurav Malviya
- Cancer
Research UK Beatson Institute, Glasgow G61 1BD, U.K.
| | - Anthony J. Chalmers
- Wolfson
Whol Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, U.K.
| | - David Lewis
- Cancer
Research UK Beatson Institute, Glasgow G61 1BD, U.K.
| | - Andrew Sutherland
- WestCHEM,
School of Chemistry, University of Glasgow, The Joseph Black Building, Glasgow G12 8QQ, U.K.
| | - Sally L. Pimlott
- West
of Scotland
PET Centre, Greater Glasgow and Clyde NHS
Trust, Glasgow G12 0YN, U.K.
| |
Collapse
|
63
|
Makvandi M, Pantel A, Schwartz L, Schubert E, Xu K, Hsieh CJ, Hou C, Kim H, Weng CC, Winters H, Doot R, Farwell MD, Pryma DA, Greenberg RA, Mankoff DA, Simpkins F, Mach RH, Lin LL. A PET imaging agent for evaluating PARP-1 expression in ovarian cancer. J Clin Invest 2018; 128:2116-2126. [PMID: 29509546 PMCID: PMC5919879 DOI: 10.1172/jci97992] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/28/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Poly(ADP-ribose) polymerase (PARP) inhibitors are effective in a broad population of patients with ovarian cancer; however, resistance caused by low enzyme expression of the drug target PARP-1 remains to be clinically evaluated in this context. We hypothesize that PARP-1 expression is variable in ovarian cancer and can be quantified in primary and metastatic disease using a novel PET imaging agent. METHODS We used a translational approach to describe the significance of PET imaging of PARP-1 in ovarian cancer. First, we produced PARP1-KO ovarian cancer cell lines using CRISPR/Cas9 gene editing to test the loss of PARP-1 as a resistance mechanism to all clinically used PARP inhibitors. Next, we performed preclinical microPET imaging studies using ovarian cancer patient-derived xenografts in mouse models. Finally, in a phase I PET imaging clinical trial we explored PET imaging as a regional marker of PARP-1 expression in primary and metastatic disease through correlative tissue histology. RESULTS We found that deletion of PARP1 causes resistance to all PARP inhibitors in vitro, and microPET imaging provides proof of concept as an approach to quantify PARP-1 in vivo. Clinically, we observed a spectrum of standard uptake values (SUVs) ranging from 2-12 for PARP-1 in tumors. In addition, we found a positive correlation between PET SUVs and fluorescent immunohistochemistry for PARP-1 (r2 = 0.60). CONCLUSION This work confirms the translational potential of a PARP-1 PET imaging agent and supports future clinical trials to test PARP-1 expression as a method to stratify patients for PARP inhibitor therapy. TRIAL REGISTRATION Clinicaltrials.gov NCT02637934. FUNDING Research reported in this publication was supported by the Department of Defense OC160269, a Basser Center team science grant, NIH National Cancer Institute R01CA174904, a Department of Energy training grant DE-SC0012476, Abramson Cancer Center Radiation Oncology pilot grants, the Marsha Rivkin Foundation, Kaleidoscope of Hope Foundation, and Paul Calabresi K12 Career Development Award 5K12CA076931.
Collapse
Affiliation(s)
- Mehran Makvandi
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Austin Pantel
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lauren Schwartz
- Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin Schubert
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kuiying Xu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Chia-Ju Hsieh
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Catherine Hou
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hyoung Kim
- Department of OBGYN, Division of Gynecology and Oncology
| | - Chi-Chang Weng
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Robert Doot
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Michael D. Farwell
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Daniel A. Pryma
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - David A. Mankoff
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Fiona Simpkins
- Department of OBGYN, Division of Gynecology and Oncology
| | - Robert H. Mach
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lilie L. Lin
- Department of Radiation Oncology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
64
|
Wei F, Tang L, He Y, Wu Y, Shi L, Xiong F, Gong Z, Guo C, Li X, Liao Q, Zhang W, Ni Q, Luo J, Li X, Li Y, Peng C, Chen X, Li G, Xiong W, Zeng Z. BPIFB1 (LPLUNC1) inhibits radioresistance in nasopharyngeal carcinoma by inhibiting VTN expression. Cell Death Dis 2018; 9:432. [PMID: 29568064 PMCID: PMC5864881 DOI: 10.1038/s41419-018-0409-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/13/2022]
Abstract
Bactericidal/permeability-increasing-fold-containing family B member 1 (BPIFB1, previously named LPLUNC1) is highly expressed in the nasopharynx and significantly downregulated in nasopharyngeal carcinoma (NPC). Low expression is also associated with poor prognosis in patients with NPC. Radiotherapy is a routine treatment for NPC; however, radioresistance is a major cause of treatment failure. Thus, we aimed to investigate the role of BPIFB1 in the radioresponse of NPC. Colony formation and cell survival results showed that BPIFB1 sensitized NPC cells to ionizing radiation. VTN, a previously identified BPIFB1-binding protein, was shown to induce cell proliferation and survival, G2/M phase arrest, DNA repair, activation of the ATM-Chk2 and ATR-Chk1 pathways, and anti-apoptotic effects after exposure to radiation, facilitating NPC cell radioresistance. However, BPIFB1 inhibited this VTN-mediated radioresistance, ultimately improving NPC radiosensitivity. In conclusion, this study is the first to demonstrate the functions of BPIFB1 and VTN in the NPC radioresponse. Our findings indicated that promoting BPIFB1 expression and targeting VTN might represent new therapeutic strategies for NPC.
Collapse
Affiliation(s)
- Fang Wei
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Le Tang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yi He
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yingfen Wu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Lei Shi
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qianxi Ni
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jia Luo
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cong Peng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
65
|
Jannetti SA, Carlucci G, Carney B, Kossatz S, Shenker L, Carter LM, Salinas B, Brand C, Sadique A, Donabedian PL, Cunanan KM, Gönen M, Ponomarev V, Zeglis BM, Souweidane MM, Lewis JS, Weber WA, Humm JL, Reiner T. PARP-1-Targeted Radiotherapy in Mouse Models of Glioblastoma. J Nucl Med 2018; 59:1225-1233. [PMID: 29572254 DOI: 10.2967/jnumed.117.205054] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/05/2018] [Indexed: 12/18/2022] Open
Abstract
The DNA repair enzyme poly(ADP-ribose) polymerase 1 (PARP-1) is overexpressed in glioblastoma, with overall low expression in healthy brain tissue. Paired with the availability of specific small molecule inhibitors, PARP-1 is a near-ideal target to develop novel radiotherapeutics to induce DNA damage and apoptosis in cancer cells, while sparing healthy brain tissue. Methods: We synthesized an 131I-labeled PARP-1 therapeutic and investigated its pharmacology in vitro and in vivo. A subcutaneous tumor model was used to quantify retention times and therapeutic efficacy. A potential clinical scenario, intratumoral convection-enhanced delivery, was mimicked using an orthotopic glioblastoma model combined with an implanted osmotic pump system to study local administration of 131I-PARPi (PARPi is PARP inhibitor). Results:131I-PARPi is a 1(2H)-phthalazinone, similar in structure to the Food and Drug Administration-approved PARP inhibitor AZD-2281. In vitro studies have shown that 131I-PARPi and AZD-2281 share similar pharmacologic profiles. 131I-PARPi delivered 134.1 cGy/MBq intratumoral injected activity. Doses to nontarget tissues, including liver and kidney, were significantly lower. Radiation damage and cell death in treated tumors were shown by p53 activation in U87-MG cells transfected with a p53-bioluminescent reporter. Treated mice showed significantly longer survival than mice receiving vehicle (29 vs. 22 d, P < 0.005) in a subcutaneous model. Convection-enhanced delivery demonstrated efficient retention of 131I-PARPi in orthotopic brain tumors, while quickly clearing from healthy brain tissue. Conclusion: Our results demonstrate 131I-PARPi's high potential as a therapeutic and highlight PARP's relevance as a target for radionuclide therapy. Radiation plays an integral role in brain tumor therapy, and radiolabeled PARP therapeutics could ultimately lead to improvements in the standard of care.
Collapse
Affiliation(s)
- Stephen A Jannetti
- Department of Biochemistry, Hunter College-The City University of New York, New York, New York.,Department of Biochemistry, The Graduate Center, The City University of New York, New York, New York.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Giuseppe Carlucci
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Center for Advanced Imaging Innovation and Research, New York University Langone Medical Center, New York, New York
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Chemistry, The Graduate Center, The City University of New York, New York, New York.,Department of Chemistry, Hunter College-The City University of New York, New York, New York
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Larissa Shenker
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas M Carter
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Beatriz Salinas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmad Sadique
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Patrick L Donabedian
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kristen M Cunanan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian M Zeglis
- Department of Biochemistry, Hunter College-The City University of New York, New York, New York.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pharmacology, Weill-Cornell Medical College, New York, New York.,Department of Radiology, Weill-Cornell Medical College, New York, New York
| | - Mark M Souweidane
- Department of Neurological Surgery, Weill-Cornell Medical College, New York, New York.,Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pharmacology, Weill-Cornell Medical College, New York, New York.,Department of Radiology, Weill-Cornell Medical College, New York, New York
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill-Cornell Medical College, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Department of Radiology, Weill-Cornell Medical College, New York, New York
| |
Collapse
|
66
|
Kossatz S, Carney B, Farley C, Weber WA, Drain CM, Reiner T. Direct Imaging of Drug Distribution and Target Engagement of the PARP Inhibitor Rucaparib. J Nucl Med 2018; 59:1316-1320. [PMID: 29572258 DOI: 10.2967/jnumed.117.205765] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/12/2018] [Indexed: 01/08/2023] Open
Abstract
Poly(ADP-ribose)polymerase (PARP) inhibitors have emerged as potent antitumor drugs. Here, we describe the intrinsic fluorescence properties of the clinically approved PARP inhibitor rucaparib and its potential to directly measure drug distribution and target engagement-a critical factor for understanding drug action and improving efficacy. Methods: We characterized the photophysical properties of rucaparib and determined its quantum yield and lifetime. Using confocal microscopy and flow cytometry, we imaged the intracellular distribution of rucaparib and measured uptake and release kinetics. Results: Rucaparib has an excitation/emission maximum of 355/480 nm and a quantum yield of 0.3. In vitro time-lapse imaging showed accumulation in cell nuclei within seconds of administration. Nuclear rucaparib uptake increased with higher PARP1 expression, and we determined an intracellular half-life of 6.4 h. Conclusion: The label-free, intrinsic fluorescence of rucaparib can be exploited to interrogate drug distribution and target binding, critical factors toward improving treatment efficacy and outcome.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Hunter College and Graduate Center of the City University of New York, New York, New York
| | - Christopher Farley
- Hunter College and Graduate Center of the City University of New York, New York, New York
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York; and.,Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Charles M Drain
- Hunter College and Graduate Center of the City University of New York, New York, New York
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York; and
| |
Collapse
|
67
|
Waaijer SJ, Kok IC, Eisses B, Schröder CP, Jalving M, Brouwers AH, Lub-de Hooge MN, de Vries EG. Molecular Imaging in Cancer Drug Development. J Nucl Med 2018; 59:726-732. [DOI: 10.2967/jnumed.116.188045] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/16/2018] [Indexed: 12/23/2022] Open
|
68
|
Anderson CJ, Lewis JS. Current status and future challenges for molecular imaging. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2017; 375:rsta.2017.0023. [PMID: 29038378 DOI: 10.1098/rsta.2017.0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
Molecular imaging (MI), used in its wider sense of biology at the molecular level, is a field that lies at the intersection of molecular biology and traditional medical imaging. As advances in medicine have exponentially expanded over the last few decades, so has our need to better understand the fundamental behaviour of living organisms in a non-invasive and timely manner. This commentary draws from topics the authors addressed in their presentations at the 2017 Royal Society Meeting 'Challenges for chemistry in molecular imaging', as well as a discussion of where MI is today and where it is heading in the future.This article is part of the themed issue 'Challenges for chemistry in molecular imaging'.
Collapse
Affiliation(s)
- Carolyn J Anderson
- Departments of Medicine, Radiology, Bioengineering, and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jason S Lewis
- Department of Radiology and the Program in Molecular Pharmacology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
69
|
Makvandi M, Sellmyer MA, Mach RH. Inflammation and DNA damage: Probing pathways to cancer and neurodegeneration. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017; 25:37-43. [PMID: 29233266 DOI: 10.1016/j.ddtec.2017.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/01/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023]
Abstract
Cancer and neurodegeneration represent two opposite ends of the biological spectrum but contain many common biological mechanisms. Two such mechanisms include the elevated levels of oxidative stress and DNA damage. In this brief review, we describe current approaches for imaging these biological pathways with the molecular imaging technique, Positron Emission Tomography (PET), and the potential of PET imaging studies to measure the efficacy of anticancer drugs and strategies for delaying the progression of neurodegenerative disorders.
Collapse
Affiliation(s)
- Mehran Makvandi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark A Sellmyer
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
70
|
Targeted PET imaging strategy to differentiate malignant from inflamed lymph nodes in diffuse large B-cell lymphoma. Proc Natl Acad Sci U S A 2017; 114:E7441-E7449. [PMID: 28827325 DOI: 10.1073/pnas.1705013114] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma in adults. DLBCL exhibits highly aggressive and systemic progression into multiple tissues in patients, particularly in lymph nodes. Whole-body 18F-fluodeoxyglucose positron emission tomography ([18F]FDG-PET) imaging has an essential role in diagnosing DLBCL in the clinic; however, [18F]FDG-PET often faces difficulty in differentiating malignant tissues from certain nonmalignant tissues with high glucose uptake. We have developed a PET imaging strategy for DLBCL that targets poly[ADP ribose] polymerase 1 (PARP1), the expression of which has been found to be much higher in DLBCL than in healthy tissues. In a syngeneic DLBCL mouse model, this PARP1-targeted PET imaging approach allowed us to discriminate between malignant and inflamed lymph nodes, whereas [18F]FDG-PET failed to do so. Our PARP1-targeted PET imaging approach may be an attractive addition to the current PET imaging strategy to differentiate inflammation from malignancy in DLBCL.
Collapse
|