Journal articles on the topic 'Drug imaging'

To see the other types of publications on this topic, follow the link: Drug imaging.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Drug imaging.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

T. Varkey, Jaya. "Peptides-Incorporated Nanoparticles for Imaging and Drug Delivery Applications." Journal of Pharmaceutical and Medicinal Chemistry 2, no. 2 (2016): 145–48. http://dx.doi.org/10.21088/jpmc.2395.6615.2216.4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
2

Malhotra, Rhea, and Ajay Singh. "Imaging of drug mules." Emergency Radiology 28, no. 4 (March 18, 2021): 809–14. http://dx.doi.org/10.1007/s10140-021-01924-3.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Kitson, Sean. "Squaryl Molecular Metaphors – Application to Rational Drug Design and Imaging Agents." Journal of Diagnostic Imaging in Therapy 4, no. 1 (May 3, 2017): 35–75. http://dx.doi.org/10.17229/jdit.2017-0503-029.

Full text
APA, Harvard, Vancouver, ISO, and other styles
4

Koshikawa, N., A. Omata, M. Masubuchi, Y. Okazaki, J. Kataoka, K. Matsunaga, H. Kato, A. Toyoshima, Y. Wakabayashi, and T. Kobayashi. "Activation imaging of drugs with hybrid Compton camera: A proof-of-concept study." Applied Physics Letters 121, no. 19 (November 7, 2022): 193701. http://dx.doi.org/10.1063/5.0116570.

Full text
Abstract:
The visualization of drugs is essential for cancer treatment. Although several methods for visualizing drugs have been proposed, a versatile method that can be easily applied to various drugs has not yet been established. Therefore, we propose “activation imaging,” in which a drug is irradiated with thermal neutrons and becomes radioactive, enabling visualization using emitted x rays and/or gamma rays. Activation imaging does not require the conjugation of specific tracers with drugs. Therefore, it can be easily applied to a variety of drugs, drug carriers (e.g., metal nanoparticles), and contrast agents. In this study, neutron activation, gamma-ray spectroscopy, and imaging of drug carriers, anticancer drug, and contrast agents were performed. Gold nanoparticles (AuNPs) and platinum nanoparticles were used as drug carriers, cisplatin was used as an anticancer drug, and gadoteridol and iohexol were used as contrast agents. As a neutron source, the RIKEN accelerator-driven compact neutron source II (RANS-II) was utilized. The imaging was performed using a hybrid Compton camera (HCC). The HCC can visualize x rays and gamma rays ranging from a few keV to nearly 1 MeV, which enables the imaging of various x rays and gamma rays emitted from the activated drugs. As a result, the gamma-ray spectra indicated the generation of radioisotopes through neutron irradiation, and AuNPs and iohexol were visualized.
APA, Harvard, Vancouver, ISO, and other styles
5

Ban, Weikang, Yuyang You, and Zhihong Yang. "Imaging Technologies for Cerebral Pharmacokinetic Studies: Progress and Perspectives." Biomedicines 10, no. 10 (September 30, 2022): 2447. http://dx.doi.org/10.3390/biomedicines10102447.

Full text
Abstract:
Pharmacokinetic assessment of drug disposition processes in vivo is critical in predicting pharmacodynamics and toxicology to reduce the risk of inappropriate drug development. The blood–brain barrier (BBB), a special physiological structure in brain tissue, hinders the entry of targeted drugs into the central nervous system (CNS), making the drug concentrations in target tissue correlate poorly with the blood drug concentrations. Additionally, once non-CNS drugs act directly on the fragile and important brain tissue, they may produce extra-therapeutic effects that may impair CNS function. Thus, an intracerebral pharmacokinetic study was developed to reflect the disposition and course of action of drugs following intracerebral absorption. Through an increasing understanding of the fine structure in the brain and the rapid development of analytical techniques, cerebral pharmacokinetic techniques have developed into non-invasive imaging techniques. Through non-invasive imaging techniques, molecules can be tracked and visualized in the entire BBB, visualizing how they enter the BBB, allowing quantitative tools to be combined with the imaging system to derive reliable pharmacokinetic profiles. The advent of imaging-based pharmacokinetic techniques in the brain has made the field of intracerebral pharmacokinetics more complete and reliable, paving the way for elucidating the dynamics of drug action in the brain and predicting its course. The paper reviews the development and application of imaging technologies for cerebral pharmacokinetic study, represented by optical imaging, radiographic autoradiography, radionuclide imaging and mass spectrometry imaging, and objectively evaluates the advantages and limitations of these methods for predicting the pharmacodynamic and toxic effects of drugs in brain tissues.
APA, Harvard, Vancouver, ISO, and other styles
6

Jaklevic, Mary Chris. "Imaging Drug for Prostate Cancer." JAMA 325, no. 3 (January 19, 2021): 214. http://dx.doi.org/10.1001/jama.2020.26384.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Perkins, Alan, and Malcolm Frier. "Radionuclide Imaging in Drug Development." Current Pharmaceutical Design 10, no. 24 (September 1, 2004): 2907–21. http://dx.doi.org/10.2174/1381612043383476.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Nicholls, Stephen J., Srinivasa Kalidindi, Keon-Woong Moon, and Steven E. Nissen. "Atherosclerosis imaging in drug development." Expert Opinion on Drug Discovery 2, no. 9 (August 29, 2007): 1241–50. http://dx.doi.org/10.1517/17460441.2.9.1241.

Full text
APA, Harvard, Vancouver, ISO, and other styles
9

Baleato-Gonzalez, Sandra, Roberto Garcia-Figueiras, Maria Casais, Amadeo Diaz, Ivan Sanz-Falque, and Joan Vilanova. "Imaging in Drug Side Effects." Current Medical Imaging Reviews 11, no. 1 (April 23, 2015): 23–37. http://dx.doi.org/10.2174/157340561101150423104805.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Lang, Paul, Karen Yeow, Anthony Nichols, and Alexander Scheer. "Cellular imaging in drug discovery." Nature Reviews Drug Discovery 5, no. 4 (April 2006): 343–56. http://dx.doi.org/10.1038/nrd2008.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Willmann, Jürgen K., Nicholas van Bruggen, Ludger M. Dinkelborg, and Sanjiv S. Gambhir. "Molecular imaging in drug development." Nature Reviews Drug Discovery 7, no. 7 (July 2008): 591–607. http://dx.doi.org/10.1038/nrd2290.

Full text
APA, Harvard, Vancouver, ISO, and other styles
12

El-Deiry, Wafik S., Caroline C. Sigman, and Gary J. Kelloff. "Imaging and Oncologic Drug Development." Journal of Clinical Oncology 24, no. 20 (July 10, 2006): 3261–73. http://dx.doi.org/10.1200/jco.2006.06.5623.

Full text
Abstract:
For decades anatomic imaging with computed tomography or magnetic resonance imaging has facilitated drug development in medical oncology by providing quantifiable and objective evidence of response to cancer therapy. In recent years metabolic imaging with [18F]fluorodeoxyglucose–positron emission tomography has added an important component to the oncologist's armamentarium for earlier detection of response that is now widely used and appreciated. These modalities along with ultrasound and optical imaging (bioluminescence, fluorescence, near-infrared imaging, multispectral imaging) have become used increasingly in preclinical studies in animal models to document the effects of genetic alterations on cancer progression or metastases, the detection of minimal residual disease, and response to various therapeutics including radiation, chemotherapy, or biologic agents. The field of molecular imaging offers potential to deliver a variety of probes that can image noninvasively drug targets, drug distribution, cancer gene expression, cell surface receptor or oncoprotein levels, and biomarker predictors of prognosis, therapeutic response, or failure. Some applications are best suited to accelerate preclinical anticancer drug development, whereas other technologies may be directly transferable to the clinic. Efforts are underway to apply noninvasive in vivo imaging to specific preclinical or clinical problems to accelerate progress in the field. Because resources are limited, and patient suffering from failed or ineffective therapy continues, a concerted effort is being made to address these issues. Many simultaneous activities involving academia; the pharmaceutical, device, and biotechnology industries; US Food and Drug Administration; National Cancer Institute; Centers for Medicare and Medicaid Services; and specialized networks sponsored by the National Institutes of Health are beginning to address these issues to develop consensus recommendations and progress in this important area.
APA, Harvard, Vancouver, ISO, and other styles
13

Venkatanarasimha, N., B. Rock, R. D. Riordan, C. A. Roobottom, and W. M. Adams. "Imaging of illicit drug use." Clinical Radiology 65, no. 12 (December 2010): 1021–30. http://dx.doi.org/10.1016/j.crad.2010.06.013.

Full text
APA, Harvard, Vancouver, ISO, and other styles
14

Goins, Beth A., and William T. Phillips. "The Use of Scintigraphic Imaging During Liposome Drug Development." Journal of Pharmacy Practice 14, no. 5 (October 2001): 397–406. http://dx.doi.org/10.1106/da2m-fyju-1xxq-ppkk.

Full text
Abstract:
Liposomes, spherical lipid bilayers enclosing an aqueous space, have become an important class of drug carriers. This review describes the usefulness of scintigraphic imaging during the development of liposome-based drugs. This imaging modality is particularly helpful for tracking the distribution of liposomes in the body, monitoring the therapeutic responses following administration of liposome-based drugs, and investigating the physiological responses associated with liposome administration. Scintigraphy also can be used to monitor the therapeutic responses of patients given approved liposomal drugs. Several examples describing the potential of this imaging modality during both the preclinical formulation and clinical trial stages of liposomal drug development are included. Techniques for radiolabeling liposomes as well as methods for producing scintigraphic images are also described.
APA, Harvard, Vancouver, ISO, and other styles
15

Knudsen, G. "Pet imaging of receptor occupancy." European Psychiatry 64, S1 (April 2021): S6. http://dx.doi.org/10.1192/j.eurpsy.2021.39.

Full text
Abstract:
The discovery and development of drugs for treatment of brain disorders is an extremely challenging process requiring large resources, timelines, and associated costs. Positron Emission Tomography (PET) enables in vivo neuroimaging of various components of receptors, transporters, enzymatic activity and other types of proteins. PET also allows for studying the response to physiological or drug interventions in experimental medicine studies. Moreover, PET neuroimaging can assist to establish diagnoses in certain brain disorders and thereby improve patient selection and stratification for clinical trials. Over the past couple of decades, PET neuroimaging has thus become a central component of the evaluation of novel drugs for brain disorders, enabling decision-making in phase I studies, where early discharge of risk provides increased confidence to progress a candidate to a later phase testing at the right dose level or alternatively to kill a compound through failure to meet key criteria. The so called "3 pillars" of drug survival, namely; tissue exposure, target engagement, and pharmacologic activity, are particularly well suited for evaluation by PET imaging. Molecular neuroimaging has thus increasingly established itself as a unique tool that not only can demonstrate drug penetration and kinetics in the brain, but also identify pharmacodynamic effects, e.g., changes in glucose metabolism. It can also quantitate therapeutic action in vivo by determining, e.g., drug occupancy whereby the relevant dose ranges to be used in clinical efficacy trials can be determined.DisclosureNo significant relationships.
APA, Harvard, Vancouver, ISO, and other styles
16

de Vries, E. G. E., A. H. Brouwers, G. A. P. Hospers, C. P. Schröder, E. F. J. de Vries, S. F. Oosting, M. A. T. M. van Vugt, G. M. van Dam, and M. N. Lub-de Hooge. "15 Imaging Visualisation of Drug Target and Drug Effect." European Journal of Cancer 48 (November 2012): 8. http://dx.doi.org/10.1016/s0959-8049(12)71814-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Dolovich, M. "Imaging Drug Delivery and Drug Responses in the Lung." Proceedings of the American Thoracic Society 1, no. 4 (December 1, 2004): 329–37. http://dx.doi.org/10.1513/pats.200404-030ms.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

Berridge, Marc S., and Zhenghong Lee. "Scintigraphic Assessment of the Regional Distribution and Kinetics of Pharmaceuticals." Journal of Pharmacy Practice 14, no. 5 (October 2001): 416–26. http://dx.doi.org/10.1106/5k48-lmbc-g469-qgf3.

Full text
Abstract:
A lesser known use of imaging studies in drug development is to determine the patterns of deposition, biodistribution, and regional kinetics of drugs in the body. This kind of study is of most interest when the drug is intended for local action following topical administration by inhalation. Imaging provides a convenient noninvasive method for observing initial deposition patterns and their variations caused by variables of the drug’s formulation and delivery method. Though planar gamma imaging is the method that has most often been used, recent years have seen promising demonstrations of SPECT and PET imaging to provide three-dimensional and quantitative measurements of drug deposition. When the goal of a drug is direct local treatment of diseased tissue, delivery of that drug is an important therapeutic variable. Imaging studies allow the drug delivery to be measured and optimized before a drug formulation is committed to clinical trials.
APA, Harvard, Vancouver, ISO, and other styles
19

Sakai, Fumikazu, and Mizue Hasegawa. "Imaging Evaluation of Drug-induced Lung Injury by Anticancer Drugs." Haigan 48, no. 6 (2008): 721–26. http://dx.doi.org/10.2482/haigan.48.721.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Schmidt, Mark E. "The Future of Imaging in Drug Discovery." Journal of Pharmacy Practice 14, no. 5 (October 2001): 427–34. http://dx.doi.org/10.1177/089719001129040766.

Full text
Abstract:
The number of new chemical entities being registered by drug companies each year is declining, while at the same time, the number of new compounds, and thereby potential therapeutics, is increasing at an exponential rate. The need to demonstrate the safety, efficacy, and the “value” of these new compounds to a sophisticated pharmaceutical market, driven in turn by the forces of healthcare economics, make drug development difficult, resulting in a very lengthy and complex series of steps in the development of a drug. Many aspects of clinical pharmacology are more art than science, and detecting pharmacological effects at the level of living integrated systems is difficult. These challenges are most evident when developing new therapeutics for neuropsychiatric illnesses. We may at last be entering a postmonoamine era, exemplified by compounds such as NK-1 antagonists and metatropic glutamate receptor agonists. Such developments hold significant promise for the treatment of severe mental illness, while at the same time being confronted with completely unknown clinical pharmacologies. Functional imaging may not only be useful for the development of new CNS compounds, but it may in fact be essential for helping to define their clinical pharmacology. Several examples will be addressed that highlight the utility of functional imaging in the development of potentially new CNS drugs.
APA, Harvard, Vancouver, ISO, and other styles
21

Stone, James M., and Lyn S. Pilowsky. "Antipsychotic drug action: targets for drug discovery with neurochemical imaging." Expert Review of Neurotherapeutics 6, no. 1 (January 2006): 57–64. http://dx.doi.org/10.1586/14737175.6.1.57.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Waaijer, Stijn J. H., Iris C. Kok, Bertha Eisses, Carolina P. Schröder, Mathilde Jalving, Adrienne H. Brouwers, Marjolijn N. Lub-de Hooge, and Elisabeth G. E. de Vries. "Molecular Imaging in Cancer Drug Development." Journal of Nuclear Medicine 59, no. 5 (January 25, 2018): 726–32. http://dx.doi.org/10.2967/jnumed.116.188045.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Vaidyanathan, G., and M. Zalutsky. "Imaging Drug Resistance with Radiolabeled Molecules." Current Pharmaceutical Design 10, no. 24 (September 1, 2004): 2965–79. http://dx.doi.org/10.2174/1381612043383449.

Full text
APA, Harvard, Vancouver, ISO, and other styles
24

Pomper, M., and J. Lee. "Small Animal Imaging in Drug Development." Current Pharmaceutical Design 11, no. 25 (October 1, 2005): 3247–72. http://dx.doi.org/10.2174/138161205774424681.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

Dobrucki, Lawrence, Dipanjan Pan, and Andrew Smith. "Multiscale Imaging of Nanoparticle Drug Delivery." Current Drug Targets 16, no. 6 (July 16, 2015): 560–70. http://dx.doi.org/10.2174/1389450116666150202163022.

Full text
APA, Harvard, Vancouver, ISO, and other styles
26

Iseppon, Federico, John E. Linley, and John N. Wood. "Calcium imaging for analgesic drug discovery." Neurobiology of Pain 11 (January 2022): 100083. http://dx.doi.org/10.1016/j.ynpai.2021.100083.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Nobis, Max, David Herrmann, Paul Timpson, Zahra Erami, and Kurt Anderson. "Imaging Molecular Dynamics for Drug Discovery." Physiology News, Autumn 2016 (September 1, 2016): 27–29. http://dx.doi.org/10.36866/pn.104.27.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Fowler, Joanna S., and Nora D. Volkow. "PET Imaging Studies in Drug Abuse." Journal of Toxicology: Clinical Toxicology 36, no. 3 (January 1998): 163–74. http://dx.doi.org/10.3109/15563659809028936.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

van der Geest, Tessa, Peter Laverman, Josbert M. Metselaar, Gert Storm, and Otto C. Boerman. "Radionuclide imaging of liposomal drug delivery." Expert Opinion on Drug Delivery 13, no. 9 (July 7, 2016): 1231–42. http://dx.doi.org/10.1080/17425247.2016.1205584.

Full text
APA, Harvard, Vancouver, ISO, and other styles
30

Kazarian, S. G., K. W. T. Kong, M. Bajomo, J. Van Der Weerd, and K. L. A. Chan. "Spectroscopic Imaging Applied to Drug Release." Food and Bioproducts Processing 83, no. 2 (June 2005): 127–35. http://dx.doi.org/10.1205/fbp.04399.

Full text
APA, Harvard, Vancouver, ISO, and other styles
31

Nilsson, Anna, Richard J. A. Goodwin, Mohammadreza Shariatgorji, Theodosia Vallianatou, Peter J. H. Webborn, and Per E. Andrén. "Mass Spectrometry Imaging in Drug Development." Analytical Chemistry 87, no. 3 (January 14, 2015): 1437–55. http://dx.doi.org/10.1021/ac504734s.

Full text
APA, Harvard, Vancouver, ISO, and other styles
32

Szewczyk-Bieda, M. J., and T. B. Oliver. "RE: Imaging of illicit drug use." Clinical Radiology 66, no. 9 (September 2011): 897–98. http://dx.doi.org/10.1016/j.crad.2011.05.003.

Full text
APA, Harvard, Vancouver, ISO, and other styles
33

Ray, L. B. "Imaging to improve drug target mapping." Science 351, no. 6273 (February 4, 2016): 572–73. http://dx.doi.org/10.1126/science.351.6273.572-c.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

LEVENTIS, MARIA. "Genetics, Imaging Will Change Drug Treatments." Clinical Psychiatry News 33, no. 7 (July 2005): 77. http://dx.doi.org/10.1016/s0270-6644(05)70562-2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Bullen, Andrew. "Microscopic imaging techniques for drug discovery." Nature Reviews Drug Discovery 7, no. 1 (January 2008): 54–67. http://dx.doi.org/10.1038/nrd2446.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

PERKINS, A., and M. FRIER. "Nuclear medicine imaging and drug delivery." Nuclear Medicine Communications 21, no. 5 (May 2000): 415–16. http://dx.doi.org/10.1097/00006231-200005000-00001.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

Mountz, James M. "Imaging Drug Action in the Brain." Clinical Nuclear Medicine 19, no. 10 (October 1994): 926–27. http://dx.doi.org/10.1097/00003072-199410000-00027.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Stasiuk, Graeme J., Stephen Faulkner, and Nicholas J. Long. "Novel imaging chelates for drug discovery." Current Opinion in Pharmacology 12, no. 5 (October 2012): 576–82. http://dx.doi.org/10.1016/j.coph.2012.07.008.

Full text
APA, Harvard, Vancouver, ISO, and other styles
39

Smith, Geoffrey P. S., Cushla M. McGoverin, Sara J. Fraser, and Keith C. Gordon. "Raman imaging of drug delivery systems." Advanced Drug Delivery Reviews 89 (July 2015): 21–41. http://dx.doi.org/10.1016/j.addr.2015.01.005.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

Vinegoni, Claudio, Paolo Fumene Feruglio, Ignacy Gryczynski, Ralph Mazitschek, and Ralph Weissleder. "Fluorescence anisotropy imaging in drug discovery." Advanced Drug Delivery Reviews 151-152 (November 2019): 262–88. http://dx.doi.org/10.1016/j.addr.2018.01.019.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Caskey, Charles F. "Ultrasound Molecular Imaging and Drug Delivery." Molecular Imaging and Biology 19, no. 3 (March 2, 2017): 336–40. http://dx.doi.org/10.1007/s11307-017-1058-x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Matthews, Paul M., Robert Coatney, Hasan Alsaid, Beat Jucker, Sharon Ashworth, Christine Parker, and Kumar Changani. "Technologies: preclinical imaging for drug development." Drug Discovery Today: Technologies 10, no. 3 (September 2013): e343-e350. http://dx.doi.org/10.1016/j.ddtec.2012.04.004.

Full text
APA, Harvard, Vancouver, ISO, and other styles
43

Mudd, Sarah R., Robert A. Comley, Mats Bergstrom, Kyle D. Holen, Yanping Luo, Sabin Carme, Gerard B. Fox, Laurent Martarello, and John D. Beaver. "Molecular imaging in oncology drug development." Drug Discovery Today 22, no. 1 (January 2017): 140–47. http://dx.doi.org/10.1016/j.drudis.2016.09.020.

Full text
APA, Harvard, Vancouver, ISO, and other styles
44

Shaw, P., and L. S. Pilowsky. "Probing Cortical Sites of Antipsychotic Drug Action within vivoReceptor Imaging." Behavioural Neurology 12, no. 1-2 (2000): 3–9. http://dx.doi.org/10.1155/2000/184707.

Full text
Abstract:
Imaging receptors using radioactive ligands has allowed direct determination of the sites of action of antipsychotic drugs. Initial studies relating antipsychotic drug efficacy to action at striatal dopamine D2-like receptors have recently been undermined. Developments in imaging extrastriatal dopamine D2-like receptors suggest rather that antagonism of these receptors in the temporal cortex is the common site of action for antipsychotic drugs, with occupancy at striatal receptors relating more closely to extrapyramidal side effects. Further work into dopamine receptor subtypes and other receptor groups such as serotonin and GABA neurotransmitters awaits the development of suitable probes, but there are some initial finding. Again a main site of antipsychotic drug action is at cortical levels with high degrees of cortical D1 and 5HT2areceptor occupancy attained particularly by atypical antipsychotic drugs.
APA, Harvard, Vancouver, ISO, and other styles
45

Ng, Alvin Y. J., Jagath C. Rajapakse, Roy E. Welsch, Paul T. Matsudaira, Victor Horodincu, and James G. Evans. "A Cell Profiling Framework for Modeling Drug Responses from HCS Imaging." Journal of Biomolecular Screening 15, no. 7 (June 4, 2010): 858–68. http://dx.doi.org/10.1177/1087057110372256.

Full text
Abstract:
The authors present an unsupervised, scalable, and interpretable cell profiling framework that is compatible with data gathered from high-content screening. They demonstrate the effectiveness of their framework by modeling drug differential effects of IC-21 macrophages treated with microtubule and actin disrupting drugs. They identify significant features of cell phenotypes for unsupervised learning based on maximum relevancy and minimum redundancy criteria. A 2-stage clustering approach annotates, clusters cells, and then merges them together to form super-clusters. An interpretable cell profile consisting of super-cluster proportions profiled at each drug treatment, concentration, or duration is obtained. Differential changes in super-cluster profiles are the basis for understanding the drug’s differential effect and biology. The authors’ method is validated by significant chi-squared statistics obtained from similar drug-treated super-cluster profiles from a 5-fold cross-validation. In addition, drug profiles of 2 microtubule drugs with equivalent mechanisms of action are statistically similar. Several distinct trends are identified for the 5 cytoskeletal drugs profiled under different conditions.
APA, Harvard, Vancouver, ISO, and other styles
46

Dunphy, Mark P. S., and Nagavarakishore Pillarsetty. "The Unique Pharmacometrics of Small Molecule Therapeutic Drug Tracer Imaging for Clinical Oncology." Cancers 12, no. 9 (September 22, 2020): 2712. http://dx.doi.org/10.3390/cancers12092712.

Full text
Abstract:
Translational development of radiolabeled analogues or isotopologues of small molecule therapeutic drugs as clinical imaging biomarkers for optimizing patient outcomes in targeted cancer therapy aims to address an urgent and recurring clinical need in therapeutic cancer drug development: drug- and target-specific biomarker assays that can optimize patient selection, dosing strategy, and response assessment. Imaging the in vivo tumor pharmacokinetics and biomolecular pharmacodynamics of small molecule cancer drugs offers patient- and tumor-specific data which are not available from other pharmacometric modalities. This review article examines clinical research with a growing pharmacopoeia of investigational small molecule cancer drug tracers.
APA, Harvard, Vancouver, ISO, and other styles
47

Marik, Jan, Sandra M. Sanabria Bohorquez, Simon-Peter Williams, and Nicholas van Bruggen. "New imaging paradigms in drug development: the PET imaging approach." Drug Discovery Today: Technologies 8, no. 2-4 (June 2011): e63-e69. http://dx.doi.org/10.1016/j.ddtec.2011.11.004.

Full text
APA, Harvard, Vancouver, ISO, and other styles
48

Lamberts, Laetitia E., Simon P. Williams, Anton G. T. Terwisscha van Scheltinga, Marjolijn N. Lub-de Hooge, Carolien P. Schröder, Jourik A. Gietema, Adrienne H. Brouwers, and Elisabeth G. E. de Vries. "Antibody Positron Emission Tomography Imaging in Anticancer Drug Development." Journal of Clinical Oncology 33, no. 13 (May 1, 2015): 1491–504. http://dx.doi.org/10.1200/jco.2014.57.8278.

Full text
Abstract:
More than 50 monoclonal antibodies (mAbs), including several antibody–drug conjugates, are in advanced clinical development, forming an important part of the many molecularly targeted anticancer therapeutics currently in development. Drug development is a relatively slow and expensive process, limiting the number of drugs that can be brought into late-stage trials. Development decisions could benefit from quantitative biomarkers, enabling visualization of the tissue distribution of (potentially modified) therapeutic mAbs to confirm effective whole-body target expression, engagement, and modulation and to evaluate heterogeneity across lesions and patients. Such biomarkers may be realized with positron emission tomography imaging of radioactively labeled antibodies, a process called immunoPET. This approach could potentially increase the power and value of early trials by improving patient selection, optimizing dose and schedule, and rationalizing observed drug responses. In this review, we summarize the available literature and the status of clinical trials regarding the potential of immunoPET during early anticancer drug development.
APA, Harvard, Vancouver, ISO, and other styles
49

Samrot, Antony V., Saipriya C, Durga Sruthi P., A. Jenifer Selvarani, Raji P., Prakash P, Paulraj Ponnaiah, et al. "Production and Utilization of SPIONs for In-vitro Drug Release and X-ray Imaging." Journal of Pure and Applied Microbiology 14, no. 2 (June 25, 2020): 1317–22. http://dx.doi.org/10.22207/jpam.14.2.27.

Full text
APA, Harvard, Vancouver, ISO, and other styles
50

El-Boubbou, Kheireddine. "Multifunctional Magnetic Iron Oxide Nanoparticles for Intracellular Imaging and Drug Delivery to Cancer Cells." International Journal of Materials, Mechanics and Manufacturing 5, no. 4 (November 2017): 231–34. http://dx.doi.org/10.18178/ijmmm.2017.5.4.325.

Full text
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography