Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Invest Radiol ; 57(10): 655-663, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36069439

ABSTRACT

OBJECTIVES: In an effort to exploit the elevated need for phospholipids displayed by cancer cells relative to normal cells, we have developed tumor-targeted alkylphosphocholines (APCs) as broad-spectrum cancer imaging and therapy agents. Radioactive APC analogs have exhibited selective uptake and prolonged tumor retention in over 50 cancer types in preclinical models, as well as over 15 cancer types in over a dozen clinical trials. To push the structural limits of this platform, we recently added a chelating moiety capable of binding gadolinium and many other metals for cancer-targeted magnetic resonance imaging (MRI), positron emission tomography imaging, and targeted radionuclide therapy. The aim of this work was to synthesize, characterize, and validate the tumor selectivity of a new broad-spectrum, tumor-targeted, macrocyclic MRI chelate, Gd-NM600, in xenograft and orthotopic tumor models. A secondary aim was to identify and track the in vivo chemical speciation and spatial localization of this new chelate Gd-NM600 in order to assess its Gd deposition properties. MATERIALS AND METHODS: T1 relaxivities of Gd-NM600 were characterized in water and plasma at 1.5 T and 3.0 T. Tumor uptake and subcellular localization studies were performed using transmission electron microscopy. We imaged 8 different preclinical models of human cancer over time and compared the T1-weighted imaging results to that of a commercial macrocyclic Gd chelate, Gd-DOTA. Finally, matrix-assisted laser desorption and ionization-mass spectrometry imaging was used to characterize and map the tissue distribution of the chemical species of Gd-NM600. RESULTS: Gd-NM600 exhibits high T1 relaxivity (approximately 16.4 s-1/mM at 1.5 T), excellent tumor uptake (3.95 %ID/g at 48 hours), prolonged tumor retention (7 days), and MRI conspicuity. Moreover, minimal tumor uptake saturability of Gd-NM600 was observed. Broad-spectrum tumor-specific uptake was demonstrated in 8 different human cancer models. Cancer cell uptake of Gd-NM600 via endosomal internalization and processing was revealed with transmission electron microscopy. Importantly, tissue mass spectrometry imaging successfully interrogated the spatial localization and chemical speciation of Gd compounds and also identified breakdown products of Gd species. CONCLUSIONS: We have introduced a new macrocyclic cancer-targeted Gd chelate that achieves broad-spectrum tumor uptake and prolonged retention. Furthermore, we have demonstrated in vivo stability of Gd-NM600 by ultrahigh resolution MS tissue imaging. A tumor-targeted contrast agent coupled with the enhanced imaging resolution of MRI relative to positron emission tomography may transform oncologic imaging.


Subject(s)
Contrast Media , Neoplasms , Chelating Agents , Contrast Media/chemistry , Gadolinium , Humans , Magnetic Resonance Imaging , Neoplasms/diagnostic imaging
3.
Mol Pharm ; 16(8): 3350-3360, 2019 08 05.
Article in English | MEDLINE | ID: mdl-31082240

ABSTRACT

Alkylphosphocholine (APC) analogs are a novel class of broad-spectrum tumor-targeting agents that can be used for both diagnosis and treatment of cancer. The potential for clinical translation for APC analogs will strongly depend on their pharmacokinetic (PK) profiles. The aim of this work was to understand how the chemical structures of various APC analogs impact binding and PK. To achieve this aim, we performed in silico docking analysis, in vitro and in vivo partitioning experiments, and in vivo PK studies. Our results have identified 7 potential high-affinity binding sites of these compounds on human serum albumin (HSA) and suggest that the size of the functional group directly influences the albumin binding, partitioning, and PK. Namely, the bulkier the functional groups, the weaker the agent binds to albumin, the more the agent partitions onto lipoproteins, and the less time the agent spends in circulation. The results of these experiments provide novel molecular insights into the binding, partitioning, and PK of this class of compounds and similar molecules as well as suggest pharmacological strategies to alter their PK profiles. Importantly, our methodology may provide a way to design better drugs by better characterizing the PK profile for lead compound optimization.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Drug Design , Molecular Docking Simulation , Phosphorylcholine/pharmacokinetics , Serum Albumin, Human/metabolism , Animals , Antineoplastic Agents/chemistry , Humans , Lipoproteins/metabolism , Mice , Mice, Nude , Models, Biological , Neoplasms/blood , Neoplasms/drug therapy , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry
5.
Neurosurgery ; 82(5): E113-E114, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29669124
8.
Nat Rev Clin Oncol ; 14(6): 347-364, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28094261

ABSTRACT

Over the past two decades, synergistic innovations in imaging technology have resulted in a revolution in which a range of biomedical applications are now benefiting from fluorescence imaging. Specifically, advances in fluorophore chemistry and imaging hardware, and the identification of targetable biomarkers have now positioned intraoperative fluorescence as a highly specific real-time detection modality for surgeons in oncology. In particular, the deeper tissue penetration and limited autofluorescence of near-infrared (NIR) fluorescence imaging improves the translational potential of this modality over visible-light fluorescence imaging. Rapid developments in fluorophores with improved characteristics, detection instrumentation, and targeting strategies led to the clinical testing in the early 2010s of the first targeted NIR fluorophores for intraoperative cancer detection. The foundations for the advances that underline this technology continue to be nurtured by the multidisciplinary collaboration of chemists, biologists, engineers, and clinicians. In this Review, we highlight the latest developments in NIR fluorophores, cancer-targeting strategies, and detection instrumentation for intraoperative cancer detection, and consider the unique challenges associated with their effective application in clinical settings.


Subject(s)
Fluorescent Dyes , Neoplasms/diagnostic imaging , Spectroscopy, Near-Infrared/methods , Drug Discovery/methods , Drug Discovery/trends , Humans , Indocyanine Green , Intraoperative Care , Levulinic Acids , Methylene Blue , Neoplasms/surgery , Aminolevulinic Acid
9.
10.
CNS Oncol ; 5(4): 223-31, 2016 10.
Article in English | MEDLINE | ID: mdl-27616199

ABSTRACT

The following is a special report on alkylphosphocholine analogs as targeted imaging and therapy agents for cancer, and their potential role in diagnosis and treatment in glioblastoma and brain metastases. These novel cancer-targeting agents display impressive tumor avidity with low background in the normal brain, and multimodal diagnostic imaging and therapy capabilities. The use of these agents may significantly improve diagnosis, treatment and post-treatment follow-up in patients with brain malignancies.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use , Brain Neoplasms/diagnostic imaging , Disease Management , Glioblastoma/diagnostic imaging , Humans , Multimodal Imaging
12.
Mol Pharm ; 13(9): 3341-9, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27421304

ABSTRACT

Cancer-targeting alkylphosphocholine (APC) analogues are being clinically developed for diagnostic imaging, intraoperative visualization, and therapeutic applications. These APC analogues derived from chemically synthesized phospholipid ethers were identified and optimized for cancer-targeting specificity using extensive structure-activity studies. While they strongly label human brain cancers associated with disrupted blood-brain barriers (BBB), APC permeability across intact BBB remains unknown. Three of our APC analogues, CLR1404 (PET radiotracer), CLR1501 (green fluorescence), and CLR1502 (near-infrared fluorescence), were tested for permeability across a BBB model composed of human induced pluripotent stem cell-derived brain microvascular endothelial cells (iPSC-derived BMECs). This in vitro BBB system has reproducibly consistent high barrier integrity marked by high transendothelial electrical resistance (TEER > 1500 Ω-cm(2)) and functional expression of drug efflux transporters. The radioiodinated and fluorescent APC analogues demonstrated fairly low permeability across the iPSC-BMEC (35 ± 5.7 (CLR1404), 54 ± 3.2 (CLR1501), and 26 ± 4.9 (CLR1502) × 10(-5) cm/min) compared with BBB-impermeable sucrose (13 ± 2.5) and BBB-permeable diazepam (170 ± 29). Only the fluorescent APC analogues (CLR1501, CLR1502) underwent BCRP and MRP polarized drug efflux transport in the brain-to-blood direction of the BBB model, and this efflux can be specifically blocked with pharmacological inhibition. None of the tested APC analogues appeared to undergo substantial P-gp transport. Limited permeability of the APC analogues across an intact BBB into normal brain likely contributes to the high tumor to background ratios observed in initial human trials. Moreover, addition of fluorescent moieties to APCs resulted in greater BMEC efflux via MRP and BCRP, and may affect fluorescence-guided applications. Overall, the characterization of APC analogue permeability across human BBB is significant for advancing future brain tumor-targeted applications of these agents.


Subject(s)
Blood-Brain Barrier/metabolism , Induced Pluripotent Stem Cells/metabolism , Phosphorylcholine/analogs & derivatives , Antineoplastic Agents/metabolism , Cell Differentiation/physiology , Cells, Cultured , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/cytology
17.
Neurosurgery ; 76(2): 115-23; discussion 123-4, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25549194

ABSTRACT

BACKGROUND: 5-Aminolevulinic acid (5-ALA)-induced tumor fluorescence aids brain tumor resections but is not approved for routine use in the United States. We developed and describe testing of 2 novel fluorescent, cancer-selective alkylphosphocholine analogs, CLR1501 (green) and CLR1502 (near infrared), in a proof-of-principle study for fluorescence-guided glioma surgery. OBJECTIVE: To demonstrate that CLR1501 and CLR1502 are cancer cell-selective fluorescence agents in glioblastoma models and to compare tumor-to-normal brain (T:N) fluorescence ratios with 5-ALA. METHODS: CLR1501, CLR1502, and 5-ALA were administered to mice with magnetic resonance imaging-verified orthotopic U251 glioblastoma multiforme- and glioblastoma stem cell-derived xenografts. Harvested brains were imaged with confocal microscopy (CLR1501), the IVIS Spectrum imaging system (CLR1501, CLR1502, and 5-ALA), or the Fluobeam near-infrared fluorescence imaging system (CLR1502). Imaging and quantitative analysis of T:N fluorescence ratios were performed. RESULTS: Excitation/emission peaks are 500/517 nm for CLR1501 and 760/778 nm for CLR1502. The observed T:N ratio for CLR1502 (9.28±1.08) was significantly higher (P<.01) than for CLR1501 (3.51±0.44 on confocal imaging; 7.23±1.63 on IVIS imaging) and 5-ALA (4.81±0.92). Near-infrared Fluobeam CLR1502 imaging in a mouse xenograft model demonstrated high- contrast tumor visualization compatible with surgical applications. CONCLUSION: CLR1501 (green) and CLR1502 (near infrared) are novel tumor-selective fluorescent agents for discriminating tumor from normal brain. CLR1501 exhibits a tumor-to-brain fluorescence ratio similar to that of 5-ALA, whereas CLR1502 has a superior tumor-to-brain fluorescence ratio. This study demonstrates the potential use of CLR1501 and CLR1502 in fluorescence-guided tumor surgery.


Subject(s)
Brain Neoplasms/surgery , Fluorescent Dyes , Glioma/surgery , Indoles , Phosphorylcholine , Aminolevulinic Acid , Animals , Flow Cytometry , Heterografts , Humans , Magnetic Resonance Imaging , Mice , Microscopy, Confocal , Phosphorylcholine/analogs & derivatives , Spectroscopy, Near-Infrared
19.
Phys Med ; 30(4): 503-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24662096

ABSTRACT

This study investigates the superficial dose from FFF beams in comparison with the conventional flattened ones using a Monte Carlo (MC) method. Published phase-space files which incorporated real geometry of a TrueBeam accelerator were used for the dose calculation in phantom and clinical cases. The photon fluence on the central axis is 3 times that of a flattened beam for a 6 MV FFF beam and 5 times for a 10 MV beam. The mean energy across the field in air at the phantom surface is 0.92-0.95 MeV for the 6 MV FFF beam and 1.18-1.30 MeV for the corresponding flattened beam. At 10 MV, the values are 1.52-1.72 and 2.15-2.87 MeV for the FFF and flattened beams, respectively. The phantom dose at the depth of 1 mm in the 6 MV FFF beam is 6% ± 2.5% (of the maximum dose) higher compared to the flattened beam for a 25 × 25 cm(2) field and 14.6% ± 1.9% for the 2 × 2 cm(2) field. For the 10 MV beam, the corresponding differences are 3.4% ± 1.5% and 10.7% ± 0.6%. The skin dose difference at selected points on the patient's surface between the plans using FFF and flattened beams in the head-and-neck case was 6.5% ± 2.3% (1SD), and for the breast case it was 6.4% ± 2.3%. The Monte Carlo simulations showed that due to the lower mean energy in the FFF beam, the clinical superficial dose is higher without the flattening filter compared to the flattened beam.


Subject(s)
Monte Carlo Method , Radiation Dosage , Radiotherapy, Intensity-Modulated/methods , Breast Neoplasms/radiotherapy , Head and Neck Neoplasms/radiotherapy , Humans , Particle Accelerators , Phantoms, Imaging , Radiotherapy Dosage , Radiotherapy, Intensity-Modulated/adverse effects , Radiotherapy, Intensity-Modulated/instrumentation , Skin/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...